Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Sociol Health Illn ; 45(2): 298-316, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36239580

RESUMEN

During the early months of COVID-19, many people in the US turned to charitable crowdfunding to seek and provide assistance. Little is known about the needs, hopes or experiences that motivated US pandemic crowdfunding and how these were correlated with campaign success. This study uses a mixed-methods data analysis of a randomised cluster sample of 919 US GoFundMe campaigns during the first 7 months of the pandemic. Overall, most campaigns performed poorly, and 38% got no donations at all. The largest proportion of campaigns aimed to address individual, acute financial struggles, often arising from considerable challenges accessing or qualifying for government assistance. These campaigns, as well as those involving campaigners and beneficiaries of colour, tended to be least successful. Qualitative thematic analysis revealed three key crowdfunding motivations that reflect individualistic, agentive responses to the pandemic: struggling, helping and adapting. These motivations reveal a shift away from social suffering and collective mobilisation and towards largely individualised efforts of survival as digital crowdfunding becomes a key domain of crisis response. Crowdfunding platforms are playing an increasingly important role in mediating and influencing individual and collective responses to crisis, which has important political ramifications for how societies perceive and address health emergencies.


Asunto(s)
COVID-19 , Colaboración de las Masas , Humanos , Pandemias , Motivación , Colaboración de las Masas/métodos , Financiación de la Atención de la Salud , COVID-19/epidemiología
2.
Sensors (Basel) ; 22(4)2022 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-35214360

RESUMEN

This paper presents the performance improvement of a co-planar waveguide rectangularly notched UWB-MIMO antenna. The isolation and gain of the antenna are enhanced by using a parasitic isolator. The antenna consists of four microstrip patch antennas and an isolator. The UWB characteristic of the antenna is achieved by truncating the lower ends of the radiating patch by a semicircle. The rectangular notch characteristic is obtained by adding two electromagnetic bandgap structures on the backside of the antenna, which is attached to the radiator via shorting pin. The performance, especially the decoupling of the MIMO antenna is improved by using a novel parasitic decoupler, which is placed between the antennas to receive uncorrelated signals. The decoupling structure consists of a square shape metallic element with a circular slot inside and a half-semicircle slot edged at each corner. Four rectangular metallic stubs are extended from opposite parallel sides to improve further isolation. The simulated and measured results show that the antenna has a rectangular notch band (5.25-5.85 GHz) across the working band of 3-12.8 GHz. In addition, the antenna has a planar structure with an overall size of 60 × 60 × 1.52 mm3 and offers stable gain, reduced mutual coupling (<-21 dB), and lower envelop correlation (<0.001).

3.
J Neurooncol ; 130(3): 571-579, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27826680

RESUMEN

Antiangiogenic therapies for malignant gliomas often result in transient response, and recurrent disease is characterized by adoption of invasive and hypoxic phenotype. The notch signaling pathway is activated in gliomas, and augments cell migration and hypoxic response. Here we report a clinical study of the combination of bevacizumab and RO4929097, an inhibitor of the notch signaling cascade. A phase I clinical trial was conducted through the Adult Brain Tumor Consortium in subjects with recurrent malignant glioma. Primary objectives were to assess safety and to define the maximum tolerated dose of RO4929097 in combination with bevacizumab. Secondary objectives were to determine overall survival, progression free survival, radiographic response, pharmacokinetic evaluation, and tissue biomarker analysis. Thirteen subjects were enrolled. Of the three subjects treated with the highest dose of RO4929097, one grade 3 toxicity and one grade 2 toxicity were observed. Definitive maximum tolerated dose of RO4929097 in combination with bevacizumab was not identified due to manufacturer's decision to halt drug production. 2 of 12 evaluable subjects demonstrated radiographic response; one subject experienced CR and the second PR. The median overall survival was 10.9 months with a median progression-free survival of 3.7 months. Two subjects remained free of disease progression at 6 months from treatment initiation. PK evaluation did not identify clinically significant drug-drug interactions. All analyzed tissue specimens revealed activation of notch signaling. Combination of RO4929097 and bevacizumab was well-tolerated. Given the compelling scientific rationale, additional studies of antiangiogenic and notch signaling inhibitors should be considered.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzazepinas/uso terapéutico , Bevacizumab/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Adulto , Neoplasias Encefálicas/mortalidad , Femenino , Glioma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo
4.
Cancers (Basel) ; 16(11)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38893149

RESUMEN

Glioblastoma (GBM) cells are highly invasive, infiltrating the surrounding normal brain tissue, thereby limiting the efficacy of surgical resection and focal radiotherapy. Cysteamine, a small aminothiol molecule that is orally bioavailable and approved for cystinosis, has potential as a cancer treatment by inhibiting tumor cell invasion and metastasis. Here we demonstrate that these potential therapeutic effects of cysteamine are likely due to the inhibition of matrix metalloproteinases (MMPs) in GBM. In vitro assays confirmed that micromolar concentrations of cysteamine were not cytotoxic, enabling the interrogation of the cellular effects without confounding tumor cell loss. Cysteamine's inhibition of MMP activity, especially the targeting of MMP2, MMP9, and MMP14, was observed at micromolar concentrations, suggesting the mechanism of action in suppressing invasion and cell migration is by inhibition of these MMPs. These findings suggest that achievable micromolar concentrations of cysteamine effectively inhibit cancer cell invasion and migration in GBM, supporting the potential for use as an adjunct cancer treatment.

5.
Sci Rep ; 14(1): 14175, 2024 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898077

RESUMEN

Central nervous system tumors have resisted effective chemotherapy because most therapeutics do not penetrate the blood-tumor-brain-barrier. Nanomedicines between ~ 10 and 100 nm accumulate in many solid tumors by the enhanced permeability and retention effect, but it is controversial whether the effect can be exploited for treatment of brain tumors. PLX038A is a long-acting prodrug of the topoisomerase 1 inhibitor SN-38. It is composed of a 15 nm 4-arm 40 kDa PEG tethered to four SN-38 moieties by linkers that slowly cleave to release the SN-38. The prodrug was remarkably effective at suppressing growth of intracranial breast cancer and glioblastoma (GBM), significantly increasing the life span of mice harboring them. We addressed the important issue of whether the prodrug releases SN-38 systemically and then penetrates the brain to exert anti-tumor effects, or whether it directly penetrates the blood-tumor-brain-barrier and releases the SN-38 cargo within the tumor. We argue that the amount of SN-38 formed systemically is insufficient to inhibit the tumors, and show by PET imaging that a close surrogate of the 40 kDa PEG carrier in PLX038A accumulates and is retained in the GBM. We conclude that the prodrug penetrates the blood-tumor-brain-barrier, accumulates in the tumor microenvironment and releases its SN-38 cargo from within. Based on our results, we pose the provocative question as to whether the 40 kDa nanomolecule PEG carrier might serve as a "Trojan horse" to carry other drugs past the blood-tumor-brain-barrier and release them into brain tumors.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Irinotecán , Profármacos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Irinotecán/farmacocinética , Barrera Hematoencefálica/metabolismo , Ratones , Profármacos/farmacocinética , Profármacos/química , Profármacos/metabolismo , Humanos , Línea Celular Tumoral , Femenino , Ensayos Antitumor por Modelo de Xenoinjerto , Glioblastoma/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Camptotecina/uso terapéutico
6.
Mol Oncol ; 16(9): 1777-1794, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34856072

RESUMEN

Nutritional intervention is becoming more prevalent as adjuvant therapy for many cancers in view of the tumor dependence on external sources for some nutrients. However, little is known about the mechanisms that make cancer cells require certain nutrients from the microenvironment. Herein, we report the dependence of glioma cells on exogenous cysteine/cystine, despite this amino acid being nonessential. Using several 13 C-tracers and analysis of cystathionine synthase and cystathioninase levels, we revealed that glioma cells were not able to support glutathione synthesis through the transsulfuration pathway, which allows methionine to be converted to cysteine in cysteine/cystine-deprived conditions. Therefore, we explored the nutritional deprivation in a mouse model of glioma. Animals subjected to a cysteine/cystine-free diet survived longer, although this increase did not attain statistical significance, with concomitant reductions in plasma glutathione and cysteine levels. At the end point, however, tumors displayed the ability to synthesize glutathione, even though higher levels of oxidative stress were detected. We observed a compensation from the nutritional intervention revealed as the recovery of cysteine-related metabolite levels in plasma. Our study highlights a time window where cysteine deprivation can be exploited for additional therapeutic strategies.


Asunto(s)
Cisteína , Glioma , Animales , Proliferación Celular , Cisteína/metabolismo , Cistina/metabolismo , Glutatión/metabolismo , Humanos , Ratones , Microambiente Tumoral
7.
Soc Work Health Care ; 50(9): 682-93, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21985110

RESUMEN

Various factors have made rural access to maternity care a significant problem for rural women. The geographic distance between a mother's county of residence and the county in which she gave birth was examined in a rural state. Analyzing North Dakota county-level data using geographic information system (GIS) software, women from over half of the state's counties, making up nearly 18% of all births, were found to be over 40 miles to the hospital in which they gave birth. These findings suggest that rural women may experience significant geographic barriers as they receive health services in the prenatal, delivery, and postpartum periods of their pregnancy. We highlight the value of GIS, particularly geovisualization power, and note models of care that may be effective for rural women.


Asunto(s)
Sistemas de Información Geográfica , Accesibilidad a los Servicios de Salud/estadística & datos numéricos , Atención Prenatal/estadística & datos numéricos , Población Rural/estadística & datos numéricos , Femenino , Humanos , North Dakota , Embarazo , Salud Rural
8.
Soc Sci Med ; 282: 114105, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34139479

RESUMEN

During the first seven months of the COVID-19 pandemic, more than 175,000 crowdfunding campaigns were established in the US for coronavirus-related needs using the platform GoFundMe. Though charitable crowdfunding has been popular in recent years, the widespread creation of COVID-19 related campaigns points to potential shifts in how the platform is being used, and the volume of needs users have brought to the site during a profound economic, social, and epidemiological crisis. This study offers a systematic examination of the scope and impacts of COVID-19 related crowdfunding in the early months of the pandemic and assesses how existing social and health inequities shaped crowdfunding use and outcomes. Using data collected from all US-based GoFundMe campaigns mentioning COVID or coronavirus, we used descriptive analysis and a series of negative binomial and linear models to assess the contributions of demographic factors and COVID-19 impacts to campaign creation and outcome. We find significant evidence of growing inequalities in outcomes for campaigners. We find that crowdfunding provides substantially higher benefits in wealthier counties with higher levels of education. People from these areas are more likely to initiate campaigns in response to adverse health and economic impacts of COVID-19, and they also receive more funding compared to people living in areas with lower income and education. Modeling also indicates differential outcomes based on the racial and ethnic composition of county population, though without more detail about who is creating and funding campaigns we cannot explain causality. A targeted qualitative analysis of the top earning COVID-19 campaigns offers further evidence of how user privilege and corporate practices contribute to highly unequal outcomes. Taken together, these findings demonstrate how a market-oriented digital technology used to respond to large-scale crisis can exacerbate inequalities and further benefit already privileged groups.


Asunto(s)
COVID-19 , Colaboración de las Masas , Humanos , Pandemias , SARS-CoV-2
9.
Front Oncol ; 11: 719091, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336705

RESUMEN

Glioblastoma (GBM) is an aggressive brain malignancy with a dismal prognosis. With emerging evidence to disprove brain-immune privilege, there has been much interest in examining immunotherapy strategies to treat central nervous system (CNS) cancers. Unfortunately, the limited success of clinical studies investigating immunotherapy regimens, has led to questions about the suitability of immunotherapy for these cancers. Inadequate inherent populations of tumor infiltrating lymphocytes (TILs) and limited trafficking of systemic, circulating T cells into the CNS likely contribute to the poor response to immunotherapy. This paucity of TILs is in concert with the finding of epigenetic silencing of genes that promote immune cell movement (chemotaxis) to the tumor. In this study we evaluated the ability of GSK126, a blood-brain barrier (BBB) permeable small molecule inhibitor of EZH2, to reverse GBM immune evasion by epigenetic suppression of T cell chemotaxis. We also evaluated the in vivo efficacy of this drug in combination with anti-PD-1 treatment on tumor growth, survival and T cell infiltration in syngeneic mouse models. GSK126 reversed H3K27me3 in murine and human GBM cell lines. When combined with anti-PD-1 treatment, a significant increase in activated T cell infiltration into the tumor was observed. This resulted in decreased tumor growth and enhanced survival both in sub-cutaneous and intracranial tumors of immunocompetent, syngeneic murine models of GBM. Additionally, a significant increase in CXCR3+ T cells was also seen in the draining lymph nodes, suggesting their readiness to migrate to the tumor. Closer examination of the mechanism of action of GSK126 revealed its ability to promote the expression of IFN-γ driven chemokines CXCL9 and CXCL10 from the tumor cells, that work to traffic T cells without directly affecting T maturation and/or proliferation. The loss of survival benefit either with single agent or combination in immunocompromised SCID mice, suggest that the therapeutic efficacy of GSK126 in GBM is primarily driven by lymphocytes. Taken together, our data suggests that in glioblastoma, epigenetic modulation using GSK126 could improve current immunotherapy strategies by reversing the epigenetic changes that enable immune cell evasion leading to enhanced immune cell trafficking to the tumor.

10.
Cancers (Basel) ; 12(7)2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708903

RESUMEN

We examined the efficacy of selective inhibition of cyclin-dependent kinase 5 (CDK5) in glioblastoma by TP5. We analyzed its impact in vitro on CDK5 expression and activity, cell survival, apoptosis and cell cycle. DNA damage was analyzed using the expression of γH2A.X and phosphorylated ATM. Its tolerance and efficacy were assessed on in vivo xenograft mouse models. We showed that TP5 decreased the activity but not the expression of CDK5 and p35. TP5 alone impaired cell viability and colony formation of glioblastoma cell lines and induced apoptosis. TP5 increased DNA damage by inhibiting the phosphorylation of ATM, leading to G1 arrest. Whereas CDK5 activity is increased by DNA-damaging agents such as temozolomide and irradiation, TP5 was synergistic with either temozolomide or irradiation due to an accumulation of DNA damage. Concomitant use of TP5 and either temozolomide or irradiation reduced the phosphorylation of ATM, increased DNA damage, and inhibited the G2/M arrest induced by temozolomide or irradiation. TP5 alone suppressed the tumor growth of orthotopic glioblastoma mouse model. The treatment was well tolerated. Finally, alone or in association with irradiation or temozolomide, TP5 prolonged mouse survival. TP5 alone or in association with temozolomide and radiotherapy is a promising therapeutic option for glioblastoma.

11.
Cancer Res ; 79(20): 5218-5232, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31488423

RESUMEN

Cancer cells rely on mitochondrial functions to regulate key survival and death signals. How cancer cells regulate mitochondrial autophagy (mitophagy) in the tumor microenvironment as well as utilize mitophagy as a survival signal is still not well understood. Here, we elucidate a key survival mechanism of mitochondrial NIX-mediated mitophagy within the hypoxic region of glioblastoma, the most malignant brain tumor. NIX was overexpressed in the pseudopalisading cells that envelop the hypoxic-necrotic regions, and mitochondrial NIX expression was robust in patient-derived glioblastoma tumor tissues and glioblastoma stem cells. NIX was required for hypoxia and oxidative stress-induced mitophagy through NFE2L2/NRF2 transactivation. Silencing NIX impaired mitochondrial reactive oxygen species clearance, cancer stem cell maintenance, and HIF/mTOR/RHEB signaling pathways under hypoxia, resulting in suppression of glioblastoma survival in vitro and in vivo. Clinical significance of these findings was validated by the compelling association between NIX expression and poor outcome for patients with glioblastoma. Taken together, our findings indicate that the NIX-mediated mitophagic pathway may represent a key therapeutic target for solid tumors, including glioblastoma. SIGNIFICANCE: NIX-mediated mitophagy regulates tumor survival in the hypoxic niche of glioblastoma microenvironment, providing a potential therapeutic target for glioblastoma.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5218/F1.large.jpg.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Hipoxia de la Célula/fisiología , Glioblastoma/metabolismo , Proteínas de la Membrana/fisiología , Mitocondrias/metabolismo , Mitofagia/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Neoplasias Encefálicas/patología , Glioblastoma/patología , Glioma/metabolismo , Glioma/patología , Xenoinjertos , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteína Homóloga de Ras Enriquecida en el Cerebro/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología , Microambiente Tumoral , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
12.
J Mol Med (Berl) ; 97(8): 1183-1193, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31201471

RESUMEN

Chordoma, a malignant bone cancer, is highly resistant to conventional therapeutic approaches; this greatly limits radio- and chemotherapeutic options and disease management. In the present study, we investigated three patient-derived chordoma cell lines to elucidate the molecular mechanism of resistance to therapeutics. An in vitro high-throughput chemical screening assay and an in vivo xenograft model were used to identify novel chemosensitizers for chordoma. We found that patient-derived chordoma cell lines recapitulated disease phenotypes, which were highlighted by robust resistance to medical therapy manifested as lack of DNA damage accumulation. Mechanistically, the PARP DNA repair pathway was found to play a central role in this resistance. Chemical screening confirmed that PARP inhibitors could strikingly enhance temozolomide (TMZ) therapy in chordoma cells. Combining the FDA-approved PARP inhibitor, olaparib, with chemotherapeutics not only potentiated DNA damage accumulation, cell cycle arrest, and apoptosis in vitro but also suppressed chordoma xenograft expansion in vivo. We conclude that combining PARP inhibition with TMZ could be an effective therapeutic approach for the clinical management of chordoma. KEY MESSAGES: The PARP DNA repair pathway enhances chemoresistance in chordoma cells. Combining PARP inhibitors with genotoxic agents induces chordoma cell cytotoxicity. PARP inhibitor combining with temozolomide suppresses growth of chordoma in vivo.


Asunto(s)
Cordoma , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Temozolomida/farmacología , Animales , Línea Celular Tumoral , Cordoma/tratamiento farmacológico , Cordoma/metabolismo , Cordoma/patología , Daño del ADN , Reparación del ADN/efectos de los fármacos , Femenino , Humanos , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31536478

RESUMEN

Meningiomas are the most common adult primary tumor of the central nervous system, but there are no known effective medical therapies for recurrent meningioma, particularly for World Health Organization grade II and III tumors. Meningiomas arise from the meninges, located outside the blood-brain barrier, and therefore may be directly targeted by antibody-mediated immunotherapy. We found that programmed cell death ligand 1 (PD-L1) was highly expressed in multiple human malignant meningioma cell lines and patient tumor samples. PD-L1 was targeted with the anti-PD-L1 antibody avelumab and directed natural killer cells to mediate antibody-dependent cellular cytotoxicity (ADCC) of PD-L1-expressing meningioma tumors both in vitro and in vivo. ADCC of meningioma cells was significantly increased in target cells that upregulated PD-L1 expression and, conversely, abrogated in tumor cells that were depleted of PD-L1. Additionally, the high-affinity natural killer cell line, haNK, outperformed healthy donor NK cells in meningioma ADCC. Together, these data support a clinical trial designed to target PD-L1 with avelumab and haNK cells, potentially offering a novel immunotherapeutic approach for patients with malignant meningioma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Inmunoterapia/métodos , Células Asesinas Naturales/trasplante , Neoplasias Meníngeas/terapia , Meningioma/terapia , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias Meníngeas/inmunología , Meningioma/inmunología , Ratones , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Cancer Res ; 25(7): 2305-2313, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559168

RESUMEN

PURPOSE: Dopamine receptor D2 (DRD2) is a G protein-coupled receptor antagonized by ONC201, an anticancer small molecule in clinical trials for high-grade gliomas and other malignancies. DRD5 is a dopamine receptor family member that opposes DRD2 signaling. We investigated the expression of these dopamine receptors in cancer and their influence on tumor cell sensitivity to ONC201. EXPERIMENTAL DESIGN: The Cancer Genome Atlas was used to determine DRD2/DRD5 expression broadly across human cancers. Cell viability assays were performed with ONC201 in >1,000 Genomic of Drug Sensitivity in Cancer and NCI60 cell lines. IHC staining of DRD2/DRD5 was performed on tissue microarrays and archival tumor tissues of glioblastoma patients treated with ONC201. Whole exome sequencing was performed in RKO cells with and without acquired ONC201 resistance. Wild-type and mutant DRD5 constructs were generated for overexpression studies. RESULTS: DRD2 overexpression broadly occurs across tumor types and is associated with a poor prognosis. Whole exome sequencing of cancer cells with acquired resistance to ONC201 revealed a de novo Q366R mutation in the DRD5 gene. Expression of Q366R DRD5 was sufficient to induce tumor cell apoptosis, consistent with a gain-of-function. DRD5 overexpression in glioblastoma cells enhanced DRD2/DRD5 heterodimers and DRD5 expression was inversely correlated with innate tumor cell sensitivity to ONC201. Investigation of archival tumor samples from patients with recurrent glioblastoma treated with ONC201 revealed that low DRD5 expression was associated with relatively superior clinical outcomes. CONCLUSIONS: These results implicate DRD5 as a negative regulator of DRD2 signaling and tumor sensitivity to ONC201 DRD2 antagonism.


Asunto(s)
Antagonistas de los Receptores de Dopamina D2/farmacología , Neoplasias/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D5/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Medicamentos/genética , Expresión Génica , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Inmunohistoquímica , Imagen por Resonancia Magnética , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/mortalidad , Pronóstico , Unión Proteica , Piridinas/farmacología , Piridinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Receptores de Dopamina D2/genética , Receptores de Dopamina D5/química , Receptores de Dopamina D5/genética , Transducción de Señal
15.
Korean J Fam Med ; 39(6): 333-339, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30369217

RESUMEN

BACKGROUND: There is a distinction in alcohol consumption behavior between adults and college students. This study aims to verify the usability and the optimal cutoff point of Alcohol Use Disorders Identification Test-Korean revised version (AUDIT-KR) for screening alcohol use disorder in college students when the diagnostic and statistical manual of mental disorders (DSM), 5th edition diagnostic criteria is applied. METHODS: A total of 922 college students living in Daejeon were enrolled and divided into two groups based on how many items they corresponded to among DSM-5 alcohol use disorder diagnostic criteria: those who corresponded to ≥2 of the 11 items were classified into the patient group (107 males, 89 females) while the others into the control group (311 males, 415 females). The participants were evaluated using AUDIT-KR to find the optimal cutoff point for screening alcohol use disorder, sensitivity, and specificity. RESULTS: The mean±standard deviation scores in the AUDIT-KR were 12.76±7.27, 10.72±4.62 for males and females, respectively, in the patient group. In contrast, in the control group the scores were 6.26±5.23 and 3.95±3.59 in males and females, respectively. The area under the receiver operating characteristic curve (95% confidence interval) regarding alcohol use disorder screening by AUDIT-KR was 0.768 (0.715-0.821) and 0.883 (0.848-0.919) for males and females, respectively. The optimal cutoff point of alcohol use disorder for males was >9, sensitivity 64.49%, and specificity 76.85%. The optimal cutoff point for females was >6, sensitivity 82.02%, and specificity 80.48%. CONCLUSION: This study suggested that AUDIT-KR can be used as a screening tool for alcohol use disorder in groups of college students when DSM-5 diagnosis criteria are applied.

16.
J Immunother Cancer ; 6(1): 51, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29891009

RESUMEN

BACKGROUND: Corticosteroids are routinely utilized to alleviate edema in patients with intracranial lesions and are first-line agents to combat immune-related adverse events (irAEs) that arise with immune checkpoint blockade treatment. However, it is not known if or when corticosteroids can be administered without abrogating the efforts of immunotherapy. The purpose of this study was to evaluate the impact of dexamethasone on lymphocyte activation and proliferation during checkpoint blockade to provide guidance for corticosteroid use while immunotherapy is being implemented as a cancer treatment. METHODS: Lymphocyte proliferation, differentiation, and cytokine production were evaluated during dexamethasone exposure. Human T cells were stimulated through CD3 ligation and co-stimulated either directly by CD28 ligation or by providing CD80, a shared ligand for CD28 and CTLA-4. CTLA-4 signaling was inhibited by antibody blockade using ipilimumab which has been approved for the treatment of several solid tumors. The in vivo effects of dexamethasone during checkpoint blockade were evaluated using the GL261 syngeneic mouse intracranial model, and immune populations were profiled by flow cytometry. RESULTS: Dexamethasone upregulated CTLA-4 mRNA and protein in CD4 and CD8 T cells and blocked CD28-mediated cell cycle entry and differentiation. Naïve T cells were most sensitive, leading to a decrease of the development of more differentiated subsets. Resistance to dexamethasone was conferred by blocking CTLA-4 or providing strong CD28 co-stimulation prior to dexamethasone exposure. CTLA-4 blockade increased IFNγ expression, but not IL-2, in stimulated human peripheral blood T cells exposed to dexamethasone. Finally, we found that CTLA-4 blockade partially rescued T cell numbers in mice bearing intracranial gliomas. CTLA-4 blockade was associated with increased IFNγ-producing tumor-infiltrating T cells and extended survival of dexamethasone-treated mice. CONCLUSIONS: Dexamethasone-mediated T cell suppression diminishes naïve T cell proliferation and differentiation by attenuating the CD28 co-stimulatory pathway. However, CTLA-4, but not PD-1 blockade can partially prevent some of the inhibitory effects of dexamethasone on the immune response.


Asunto(s)
Dexametasona/uso terapéutico , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Animales , Dexametasona/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones
17.
Neuro Oncol ; 20(6): 799-809, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29294092

RESUMEN

Background: Standard therapy for chordoma consists of surgical resection followed by high-dose irradiation. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in signal transduction, cell cycle progression, cell differentiation, and DNA repair. LB100 is a small-molecule inhibitor of PP2A designed to sensitize cancer cells to DNA damage from irradiation and chemotherapy. A recently completed phase I trial of LB100 in solid tumors demonstrated its safety. Here, we show the therapeutic potential of LB100 in chordoma. Methods: Three patient-derived chordoma cell lines were used: U-CH1, JHC7, and UM-Chor1. Cell proliferation was determined with LB100 alone and in combination with irradiation. Cell cycle progression was assessed by flow cytometry. Quantitative γ-H2AX immunofluorescence and immunoblot evaluated the effect of LB100 on radiation-induced DNA damage. Ultrastructural evidence for nuclear damage was investigated using Raman imaging and transmission electron microscopy. A xenograft model was established to determine potential clinical utility of adding LB100 to irradiation. Results: PP2A inhibition in concert with irradiation demonstrated in vitro growth inhibition. The combination of LB100 and radiation also induced accumulation at the G2/M phase of the cell cycle, the stage most sensitive to radiation-induced damage. LB100 enhanced radiation-induced DNA double-strand breaks. Animals implanted with chordoma cells and treated with the combination of LB100 and radiation demonstrated tumor growth delay. Conclusions: Combining LB100 and radiation enhanced DNA damage-induced cell death and delayed tumor growth in an animal model of chordoma. PP2A inhibition by LB100 treatment may improve the effectiveness of radiation therapy for chordoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Cordoma/tratamiento farmacológico , Cordoma/patología , Piperazinas/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Animales , Apoptosis , Movimiento Celular , Proliferación Celular , Cordoma/enzimología , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncotarget ; 8(25): 40233-40245, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28410215

RESUMEN

Tissue hypoxia contributes to solid tumor pathogenesis by activating a series of adaptive programs. We previously showed that hypoxia promotes the preferential expansion and maintenance of CD133 positive human glioma stem cells (GSC) in a hypoxia inducible factor 1 alpha (HIF-1α)-dependent mechanism. Here, we examined the activity of digitoxin (DT), a cardiac glycoside and a putative inhibitor of HIF-1α, on human GSC in vitro and in vivo. During hypoxic conditions (1% O2), we observed the effect of DT on the intracellular level of HIF-1α and the extracellular level of vascular endothelial growth factor (VEGF) in human GSC. We found that DT at clinically achievable concentrations, suppressed HIF-1α accumulation during hypoxic conditions in human GSC and established glioma cell lines. DT treatment also significantly attenuated hypoxia-induced expression of VEGF, a downstream target of HIF-1α. Exposure to DT also reduced hypoxia-induced activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. Furthermore, DT potently inhibited neurosphere formation, and decreased CD133 expression even at concentrations that were not overtly cytotoxic. Lastly, treatment with DT reduced GSC engraftment in an in vivo xenograft model of glioblastoma. Intraperitoneal injections of DT significantly inhibited the growth of established glioblastoma xenografts, and suppressed expression of HIF-1α and carbonic anhydrase (CA9), a surrogate marker of hypoxia. Taken together, these results suggest that DT at clinically achievable concentration functions as an inhibitor of HIF-1α, worthy of further investigations in the therapy of glioblastoma.


Asunto(s)
Anhidrasas Carbónicas/biosíntesis , Digitoxina/farmacología , Glioblastoma/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Células Madre Neoplásicas/patología , Antígeno AC133/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Ratones SCID , Interferencia de ARN , ARN Interferente Pequeño/genética , Esferoides Celulares , Células Tumorales Cultivadas , Hipoxia Tumoral/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
JCI Insight ; 2(10)2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28515364

RESUMEN

Metabolic dysregulation promotes cancer growth through not only energy production, but also epigenetic reprogramming. Here, we report that a critical node in methyl donor metabolism, nicotinamide N-methyltransferase (NNMT), ranked among the most consistently overexpressed metabolism genes in glioblastoma relative to normal brain. NNMT was preferentially expressed by mesenchymal glioblastoma stem cells (GSCs). NNMT depletes S-adenosyl methionine (SAM), a methyl donor generated from methionine. GSCs contained lower levels of methionine, SAM, and nicotinamide, but they contained higher levels of oxidized nicotinamide adenine dinucleotide (NAD+) than differentiated tumor cells. In concordance with the poor prognosis associated with DNA hypomethylation in glioblastoma, depletion of methionine, a key upstream methyl group donor, shifted tumors toward a mesenchymal phenotype and accelerated tumor growth. Targeting NNMT expression reduced cellular proliferation, self-renewal, and in vivo tumor growth of mesenchymal GSCs. Supporting a mechanistic link between NNMT and DNA methylation, targeting NNMT reduced methyl donor availability, methionine levels, and unmethylated cytosine, with increased levels of DNA methyltransferases, DNMT1 and DNMT3A. Supporting the clinical significance of these findings, NNMT portended poor prognosis for glioblastoma patients. Collectively, our findings support NNMT as a GSC-specific therapeutic target in glioblastoma by disrupting oncogenic DNA hypomethylation.

20.
Methods Mol Biol ; 1516: 361-369, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27044042

RESUMEN

Gliomas are characterized by striking intratumoral cellular heterogeneity. A consistent method to isolate undifferentiated GSC fraction from clinical specimens provides an opportunity to establish in vitro models that more closely mirror the in vivo state compared to traditional serum containing culture methods. Here we describe techniques involved with isolation, identification, and expansion of human malignant glioma-derived stem cells using a serum-free growth condition.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Glioma/patología , Células Madre Neoplásicas/patología , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Glioma/cirugía , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA