Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Eur J Nucl Med Mol Imaging ; 50(3): 692-700, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36350400

RESUMEN

PURPOSE: Bladder cancer represents 3% of all new cancer diagnoses per year. We propose intravesical radionuclide therapy using the ß-emitter 90Y linked to DOTA-biotin-avidin ([90Y]DBA) to deliver short-range radiation against non-muscle invasive bladder cancer (NMIBC). MATERIAL AND METHODS: Image-guided biodistribution of intravesical DBA was investigated in an animal model by radiolabeling DBA with the 68Ga and dynamic microPET imaging following intravesical infusion of [68Ga]DBA for up to 4 h and post-necropsy γ-counting of organs. The antitumor activity of [90Y]DBA was investigated using an orthotopic MB49 murine bladder cancer model. Mice were injected with luciferase-expressing MB49 cells and treated via intravesical administration with 9.2 MBq of [90Y]DBA or unlabeled DBA 3 days after the tumor implantation. Bioluminescence imaging was conducted after tumor implantation to monitor the bladder tumor growth. In addition, we investigated the effects of [90Y]DBA radiation on urothelial histology with immunohistochemistry analysis of bladder morphology. RESULTS: Our results demonstrated that DBA is contained in the bladder for up to 4 h after intravesical infusion. A single dose of [90Y]DBA radiation treatment significantly reduced growth of MB49 bladder carcinoma. Attaching 90Y-DOTA-biotin to avidin prevents its re-absorption into the blood and distribution throughout the rest of the body. Furthermore, immunohistochemistry demonstrated that [90Y]DBA radiation treatment did not cause short-term damage to urothelium at day 10, which appeared similar to the normal urothelium of healthy mice. CONCLUSION: Our data demonstrates the potential of intravesical [90Y]DBA as a treatment for non-muscle invasive bladder cancer.


Asunto(s)
Neoplasias Vesicales sin Invasión Muscular , Neoplasias de la Vejiga Urinaria , Animales , Ratones , Avidina/uso terapéutico , Distribución Tisular , Radioisótopos de Galio , Ratones Endogámicos DBA , Neoplasias de la Vejiga Urinaria/diagnóstico por imagen , Neoplasias de la Vejiga Urinaria/radioterapia , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
2.
Nature ; 535(7611): 246-51, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27383785

RESUMEN

Poor prognosis in neuroblastoma is associated with genetic amplification of MYCN. MYCN is itself a target of let-7, a tumour suppressor family of microRNAs implicated in numerous cancers. LIN28B, an inhibitor of let-7 biogenesis, is overexpressed in neuroblastoma and has been reported to regulate MYCN. Here we show, however, that LIN28B is dispensable in MYCN-amplified neuroblastoma cell lines, despite de-repression of let-7. We further demonstrate that MYCN messenger RNA levels in amplified disease are exceptionally high and sufficient to sponge let-7, which reconciles the dispensability of LIN28B. We found that genetic loss of let-7 is common in neuroblastoma, inversely associated with MYCN amplification, and independently associated with poor outcomes, providing a rationale for chromosomal loss patterns in neuroblastoma. We propose that let-7 disruption by LIN28B, MYCN sponging, or genetic loss is a unifying mechanism of neuroblastoma development with broad implications for cancer pathogenesis.


Asunto(s)
Amplificación de Genes/genética , MicroARNs/genética , Neuroblastoma/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3'/genética , Animales , Deleción Cromosómica , Femenino , Eliminación de Gen , Genes Relacionados con las Neoplasias/genética , Humanos , Ratones , MicroARNs/metabolismo , Modelos Genéticos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Genes Dev ; 28(9): 971-82, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24732380

RESUMEN

Wilms Tumor, the most common pediatric kidney cancer, evolves from the failure of terminal differentiation of the embryonic kidney. Here we show that overexpression of the heterochronic regulator Lin28 during kidney development in mice markedly expands nephrogenic progenitors by blocking their final wave of differentiation, ultimately resulting in a pathology highly reminiscent of Wilms tumor. Using lineage-specific promoters to target Lin28 to specific cell types, we observed Wilms tumor only when Lin28 is aberrantly expressed in multiple derivatives of the intermediate mesoderm, implicating the cell of origin as a multipotential renal progenitor. We show that withdrawal of Lin28 expression reverts tumorigenesis and markedly expands the numbers of glomerulus-like structures and that tumor formation is suppressed by enforced expression of Let-7 microRNA. Finally, we demonstrate overexpression of the LIN28B paralog in a significant percentage of human Wilms tumor. Our data thus implicate the Lin28/Let-7 pathway in kidney development and tumorigenesis.


Asunto(s)
Diferenciación Celular , Neoplasias Renales/genética , Neoplasias Renales/fisiopatología , Proteínas de Unión al ARN/genética , Células Madre/citología , Tumor de Wilms/genética , Tumor de Wilms/fisiopatología , Animales , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/embriología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
Nature ; 499(7457): 219-22, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23748443

RESUMEN

The mammalian gut ecosystem has considerable influence on host physiology, but the mechanisms that sustain this complex environment in the face of different stresses remain obscure. Perturbations to the gut ecosystem, such as through antibiotic treatment or diet, are at present interpreted at the level of bacterial phylogeny. Less is known about the contributions of the abundant population of phages to this ecological network. Here we explore the phageome as a potential genetic reservoir for bacterial adaptation by sequencing murine faecal phage populations following antibiotic perturbation. We show that antibiotic treatment leads to the enrichment of phage-encoded genes that confer resistance via disparate mechanisms to the administered drug, as well as genes that confer resistance to antibiotics unrelated to the administered drug, and we demonstrate experimentally that phages from treated mice provide aerobically cultured naive microbiota with increased resistance. Systems-wide analyses uncovered post-treatment phage-encoded processes related to host colonization and growth adaptation, indicating that the phageome becomes broadly enriched for functionally beneficial genes under stress-related conditions. We also show that antibiotic treatment expands the interactions between phage and bacterial species, leading to a more highly connected phage-bacterial network for gene exchange. Our work implicates the phageome in the emergence of multidrug resistance, and indicates that the adaptive capacity of the phageome may represent a community-based mechanism for protecting the gut microflora, preserving its functional robustness during antibiotic stress.


Asunto(s)
Antibacterianos/farmacología , Bacteriófagos/efectos de los fármacos , Farmacorresistencia Microbiana/efectos de los fármacos , Heces/microbiología , Heces/virología , Genoma Viral/genética , Metagenoma/genética , Aerobiosis , Ampicilina/farmacología , Animales , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Ciprofloxacina/farmacología , Farmacorresistencia Microbiana/genética , Femenino , Transferencia de Gen Horizontal/efectos de los fármacos , Transferencia de Gen Horizontal/genética , Genes Virales/efectos de los fármacos , Genes Virales/genética , Especificidad del Huésped/efectos de los fármacos , Metagenoma/efectos de los fármacos , Ratones , Simbiosis/efectos de los fármacos , Simbiosis/genética
5.
Nat Methods ; 11(6): 663-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24793454

RESUMEN

Current in vitro hematopoiesis models fail to demonstrate the cellular diversity and complex functions of living bone marrow; hence, most translational studies relevant to the hematologic system are conducted in live animals. Here we describe a method for fabricating 'bone marrow-on-a-chip' that permits culture of living marrow with a functional hematopoietic niche in vitro by first engineering new bone in vivo, removing it whole and perfusing it with culture medium in a microfluidic device. The engineered bone marrow (eBM) retains hematopoietic stem and progenitor cells in normal in vivo-like proportions for at least 1 week in culture. eBM models organ-level marrow toxicity responses and protective effects of radiation countermeasure drugs, whereas conventional bone marrow culture methods do not. This biomimetic microdevice offers a new approach for analysis of drug responses and toxicities in bone marrow as well as for study of hematopoiesis and hematologic diseases in vitro.


Asunto(s)
Médula Ósea/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Técnicas Analíticas Microfluídicas , Animales , Médula Ósea/química , Técnicas de Cultivo de Célula , Ratones
6.
Adv Radiat Oncol ; 9(3): 101399, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38292890

RESUMEN

Purpose: The emerging online adaptive radiation therapy (OART) treatment strategy based on cone beam computed tomography allows for real-time replanning according to a patient's current anatomy. However, implementing this procedure requires a new approach across the patient's care path and monitoring of the "black box" adaptation process. This study identifies high-risk failure modes (FMs) associated with AI-driven OART and proposes an interdisciplinary workflow to mitigate potential medical errors from highly automated processes, enhance treatment efficiency, and reduce the burden on clinicians. Methods and Materials: An interdisciplinary working group was formed to identify safety concerns in each process step using failure mode and effects analysis (FMEA). Based on the FMEA results, the team designed standardized procedures and safety checklists to prevent errors and ensure successful task completion. The Risk Priority Numbers (RPNs) for the top twenty FMs were calculated before and after implementing the proposed workflow to evaluate its effectiveness. Three hundred seventy-four adaptive sessions across 5 treatment sites were performed, and each session was evaluated for treatment safety and FMEA assessment. Results: The OART workflow has 4 components, each with 4, 8, 13, and 4 sequentially executed tasks and safety checklists. Site-specific template preparation, which includes disease-specific physician directives and Intelligent Optimization Engine template testing, is one of the new procedures introduced. The interdisciplinary workflow significantly reduced the RPNs of the high-risk FMs, with an average decrease of 110 (maximum reduction of 305.5 and minimum reduction of 27.4). Conclusions: This study underscores the importance of addressing high-risk FMs associated with AI-driven OART and emphasizes the significance of safety measures in its implementation. By proposing a structured interdisciplinary workflow and integrated checklists, the study provides valuable insights into ensuring the safe and efficient delivery of OART while facilitating its effective integration into clinical practice.

7.
J Clin Invest ; 133(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37279067

RESUMEN

Patients with cancer who have high serum levels of squamous cell carcinoma antigen 1 (SCCA1, now referred to as SERPINB3) commonly experience treatment resistance and have a poor prognosis. Despite being a clinical biomarker, the modulation of SERPINB3 in tumor immunity is poorly understood. We found positive correlations of SERPINB3 with CXCL1, CXCL8 (CXCL8/9), S100A8, and S100A9 (S100A8/A9) myeloid cell infiltration through RNA-Seq analysis of human primary cervical tumors. Induction of SERPINB3 resulted in increased CXCL1/8 and S100A8/A9 expression, which promoted monocyte and myeloid-derived suppressor cell (MDSC) migration in vitro. In mouse models, Serpinb3a tumors showed increased MDSC and tumor-associated macrophage (TAM) infiltration, contributing to T cell inhibition, and this was further augmented upon radiation. Intratumoral knockdown (KD) of Serpinb3a resulted in tumor growth inhibition and reduced CXCL1 and S100A8/A expression and MDSC and M2 macrophage infiltration. These changes led to enhanced cytotoxic T cell function and sensitized tumors to radiotherapy (RT). We further revealed that SERPINB3 promoted STAT-dependent expression of chemokines, whereby inhibition of STAT activation by ruxolitinib or siRNA abrogated CXCL1/8 and S100A8/ A9 expression in SERPINB3 cells. Patients with elevated pretreatment SCCA levels and high phosphorylated STAT3 (p-STAT3) had increased intratumoral CD11b+ myeloid cells compared with patients with low SCCA levels and p-STAT3, who had improved overall survival after RT. These findings provide a preclinical rationale for targeting SERPINB3 in tumors to counteract immunosuppression and improve the response to RT.


Asunto(s)
Calgranulina A , Serpinas , Ratones , Animales , Humanos , Calgranulina A/genética , Calgranulina B/genética , Serpinas/genética , Quimiocinas/metabolismo
8.
Semin Radiat Oncol ; 31(3): 205-216, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34090647

RESUMEN

Radiation is a known immune modulator that drives both local and systemic immunologic effects. There is increasing interest and investigation into harnessing the pro-immunogenic effects of radiation for patients with metastatic cancer to improve systemic disease control and clinical outcomes. Here, we review fundamental immunology concepts in the context of our current understanding of both the pro-immunogenic and the less well-appreciated immunosuppressive effects of radiation therapy. Our aim is to offer the radiation oncology community a lens into the progress the field has made understanding the complex interaction between tumor-directed irradiation and immune-mediated tumor control, thus promoting further discovery and translation of radio-immuno-oncology innovation.


Asunto(s)
Inmunoterapia , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico
9.
Br J Radiol ; 94(1119): 20200433, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33586999

RESUMEN

OBJECTIVE: We aim to test the hypothesis that neurovascular bundle (NVB) displacement by rectal hydrogel spacer combined with NVB delineation as an organ at risk (OAR) is a feasible method for NVB-sparing stereotactic body radiotherapy. METHODS: Thirty-five men with low- and intermediate-risk prostate cancer who underwent rectal hydrogel spacer placement and pre-, post-spacer prostate MRI studies were treated with prostate SBRT (36.25 Gy in five fractions). A prostate radiologist contoured the NVB on both the pre- and post-spacer T2W MRI sequences that were then registered to the CT simulation scan for NVB-sparing radiation treatment planning. Three SBRT treatment plans were developed for each patient: (1) no NVB sparing, (2) NVB-sparing using pre-spacer MRI, and (3) NVB-sparing using post-spacer MRI. NVB dose constraints include maximum dose 36.25 Gy (100%), V34.4 Gy (95% of dose) <60%, V32Gy <70%, V28Gy <90%. RESULTS: Rectal hydrogel spacer placement shifted NVB contours an average of 3.1 ± 3.4 mm away from the prostate, resulting in a 10% decrease in NVB V34.4 Gy in non-NVB-sparing plans (p < 0.01). NVB-sparing treatment planning reduced the NVB V34.4 by 16% without the spacer (p < 0.01) and 25% with spacer (p < 0.001). NVB-sparing did not compromise PTV coverage and OAR endpoints. CONCLUSIONS: NVB-sparing SBRT with rectal hydrogel spacer significantly reduces the volume of NVB treated with high-dose radiation. Rectal spacer contributes to this effect through a dosimetrically meaningful displacement of the NVB that may significantly reduce RiED. These results suggest that NVB-sparing SBRT warrants further clinical evaluation. ADVANCES IN KNOWLEDGE: This is a feasibility study showing that the periprostatic NVBs can be spared high doses of radiation during prostate SBRT using a hydrogel spacer and nerve-sparing treatment planning.


Asunto(s)
Disfunción Eréctil/prevención & control , Hidrogeles/uso terapéutico , Neoplasias de la Próstata/radioterapia , Radiocirugia/métodos , Planificación de la Radioterapia Asistida por Computador/métodos , Anciano , Estudios de Factibilidad , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Órganos en Riesgo/diagnóstico por imagen , Próstata/diagnóstico por imagen , Neoplasias de la Próstata/diagnóstico por imagen , Dosificación Radioterapéutica , Recto/diagnóstico por imagen , Estudios Retrospectivos
10.
Int J Radiat Oncol Biol Phys ; 109(3): 813-818, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33190969

RESUMEN

PURPOSE: Radiation therapy (RT) modulates the immune characteristics of the tumor microenvironment (TME). It is not known whether these effects are dependent on the type of RT used. METHODS AND MATERIALS: We evaluated the immunomodulatory effects of carbon-ion therapy (CiRT) compared with biologically equivalent doses of photon therapy (PhRT) on solid tumors. Orthotopic 4T1 mammary tumors in immunocompetent hosts were treated with CiRT or biologically equivalent doses of PhRT. Seventy-two hours after RT, tumors were harvested and the immune characteristics of the TME were quantified by flow cytometry and multiplex cytokine analyses. RESULTS: PhRT decreased the abundance of CD4+ and CD8+ T cells in the TME at all doses tested, with compensatory increases in proliferation. By contrast, CiRT did not significantly alter CD8+ T-cell infiltration. High-dose CiRT increased secretion of proinflammatory cytokines by tumor-infiltrating CD8+ T cells, including granzyme B, IL-2, and TNF-α, with no change in IFN-γ. Conversely, high-dose PhRT increased CD8+ T-cell secretion of IFN-γ only. At most of the doses studied, PhRT increased proliferation of immunosuppressive regulatory T cells; this was only seen with high-dose CiRT. Cytokine analyses of bulk dissociated tumors showed that CiRT significantly increased levels of IFN-γ, IL-2, and IL-1ß, whereas PhRT increased IL-6 levels alone. CONCLUSIONS: At low doses, lymphocytes differ in their sensitivity to CiRT compared with PhRT. Unlike PhRT, low-dose CiRT is generally lymphocyte-sparing. At higher doses, CiRT is a more potent inducer of proinflammatory cytokines and merits further study as a modulator of the immunologic characteristics of the TME.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Radioterapia de Iones Pesados , Neoplasias Mamarias Animales/radioterapia , Fotones/uso terapéutico , Microambiente Tumoral/efectos de la radiación , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Citometría de Flujo , Granzimas/metabolismo , Granzimas/efectos de la radiación , Inmunocompetencia , Interferón gamma/metabolismo , Interferón gamma/efectos de la radiación , Interleucina-1beta/metabolismo , Interleucina-1beta/efectos de la radiación , Interleucina-2/metabolismo , Interleucina-2/efectos de la radiación , Interleucina-6/metabolismo , Interleucina-6/efectos de la radiación , Neoplasias Mamarias Animales/inmunología , Ratones , Efectividad Biológica Relativa , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de la radiación , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/efectos de la radiación
11.
Clin Cancer Res ; 27(2): 608-621, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33148676

RESUMEN

PURPOSE: Intratumoral immunosuppression mediated by myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) represents a potential mechanism of immune checkpoint inhibitor (ICI) resistance in solid tumors. By promoting TAM and MDSC infiltration, IL1ß may drive adaptive and innate immune resistance in renal cell carcinoma (RCC) and in other tumor types. EXPERIMENTAL DESIGN: Using the RENCA model of RCC, we evaluated clinically relevant combinations of anti-IL1ß plus either anti-PD-1 or the multitargeted tyrosine kinase inhibitor (TKI), cabozantinib. We performed comprehensive immune profiling of established RENCA tumors via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA sequencing (RNA-seq). Similar analyses were extended to the MC38 tumor model. RESULTS: Analyses via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA-seq showed that anti-IL1ß reduces infiltration of polymorphonuclear MDSCs and TAMs. Combination treatment with anti-IL1ß plus anti-PD-1 or cabozantinib showed increased antitumor activity that was associated with decreases in immunosuppressive MDSCs and increases in M1-like TAMs. CONCLUSIONS: Single-cell RNA-seq analyses show that IL1ß blockade and ICI or TKI remodel the myeloid compartment through nonredundant, relatively T-cell-independent mechanisms. IL1ß is an upstream mediator of adaptive myeloid resistance and represents a potential target for kidney cancer immunotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Modelos Animales de Enfermedad , Interleucina-1beta/antagonistas & inhibidores , Neoplasias Renales/tratamiento farmacológico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Anilidas/administración & dosificación , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Ratones , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide/metabolismo , Piridinas/administración & dosificación , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Macrófagos Asociados a Tumores/clasificación , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/metabolismo
12.
Radiat Oncol ; 14(1): 136, 2019 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-31375119

RESUMEN

BACKGROUND: Multiple phase I-II clinical trials have reported on the efficacy and safety of prostate stereotactic body radiotherapy (SBRT) for the treatment of prostate cancer. However, few have reported outcomes for prostate SBRT using periprostatic hydrogel spacer (SpaceOAR; Augmenix). Herein, we report safety and efficacy outcomes from our institutional prostate SBRT experience with SpaceOAR placement. METHODS: Fifty men with low- or intermediate-risk prostate cancer treated at a single institution with linear accelerator-based SBRT to 3625 cGy in 5 fractions, with or without androgen deprivation therapy (ADT) were included. All patients underwent SpaceOAR and fiducial marker placement followed by pre-treatment MRI. Toxicity assessments were conducted at least weekly while on treatment, 1 month after treatment, and every follow-up visit thereafter. Post-treatment PSA measurements were obtained 4 months after SBRT, followed by every 3-6 months thereafter. Acute toxicity was documented per RTOG criteria. RESULTS: Median follow up time was 20 (range 4-44) months. Median PSA at time of diagnosis was 7.4 (2.7-19.5) ng/ml. Eighteen men received 6 months of ADT for unfavorable intermediate risk disease. No PSA failures were recorded. Median PSA was 0.9 ng/mL at 20 months; 0.08 and 1.32 ng/mL in men who did and did not receive ADT, respectively. Mean prostate-rectum separation achieved with SpaceOAR was 9.6 ± 4 mm at the prostate midgland. No grade ≥ 3 GU or GI toxicity was recorded. During treatment, 30% of men developed new grade 2 GU toxicity (urgency or dysuria). These symptoms were present in 30% of men at 1 month and in 12% of men at 1 year post-treatment. During treatment, GI toxicity was limited to grade 1 symptoms (16%), although 4% of men developed grade 2 symptoms during the first 4 weeks after SBRT. All GI symptoms were resolving by the 1 month post-treatment assessment and no acute or late rectal toxicity was reported > 1 month after treatment. CONCLUSIONS: Periprostatic hydrogel placement followed by prostate SBRT resulted in minimal GI toxicity, and favorable early oncologic outcomes. These results indicate that SBRT with periprostatic spacer is a well-tolerated, safe, and convenient treatment option for localized prostate cancer.


Asunto(s)
Hidrogeles/efectos adversos , Enfermedades Urogenitales Masculinas/diagnóstico , Complicaciones Posoperatorias , Neoplasias de la Próstata/cirugía , Radiocirugia/efectos adversos , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Humanos , Hidrogeles/química , Masculino , Enfermedades Urogenitales Masculinas/sangre , Enfermedades Urogenitales Masculinas/etiología , Persona de Mediana Edad , Pronóstico , Antígeno Prostático Específico/sangre , Estudios Retrospectivos
14.
Int J Radiat Oncol Biol Phys ; 102(4): 841-847, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29891199

RESUMEN

PURPOSE: To implement Velocity-based image fusion and adaptive deformable registration to enable treatment planning for preclinical murine models of fractionated stereotactic radiosurgery (fSRS) using the small animal radiation research platform (SARRP). METHODS AND MATERIALS: C57BL6 mice underwent 3 unique cone beam computed tomography (CBCT) scans: 2 in the prone position and a third supine. A single T1-weighted post-contrast magnetic resonance imaging (MRI) series of a murine metastatic brain tumor model was selected for MRI-to-CBCT registration and gross tumor volume (GTV) identification. Two arms were compared: Arm 1, where we performed 3 individual MRI-to-CBCT fusions using rigid registration, contouring GTVs on each, and Arm 2, where the authors performed MRI-to-CBCT fusion and contoured GTV on the first CBCT followed by Velocity-based adaptive registration. The first CBCT and associated GTV were exported from MuriPlan (Xstrahl Life Sciences) into Velocity (Varian Medical Systems, Inc, Palo Alto, CA). In Arm 1, the second and third CBCTs were exported similarly along with associated GTVs (Arm 1), while in Arm 2, the first (prone) CBCT was fused separately to the second (prone) and third (supine) CBCTs, performing deformable registrations on initial CBCTs and applying resulting matrices to the contoured GTV. Resulting GTVs were compared between Arms 1 and 2. RESULTS: Comparing GTV overlays using repeated MRI fusion and GTV delineation (Arm 1) versus those of Velocity-based CBCT and GTV adaptive fusion (Arm 2), mean deviations ± standard deviation in the axial, sagittal, and coronal planes were 0.46 ± 0.16, 0.46 ± 0.22, and 0.37 ± 0.22 mm for prone-to-prone and 0.52 ± 0.27, 0.52 ± 0.36, and 0.68 ± 0.31 mm for prone-to-supine adaptive fusions, respectively. CONCLUSIONS: Velocity-based adaptive fusion of CBCTs and contoured volumes allows for efficient fSRS planning using a single MRI-to-CBCT fusion. This technique is immediately implementable on current SARRP systems, facilitating advanced preclinical treatment paradigms using existing clinical treatment planning software.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Tomografía Computarizada de Haz Cónico/métodos , Imagen por Resonancia Magnética/métodos , Radiocirugia/métodos , Planificación de la Radioterapia Asistida por Computador/métodos , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL , Carga Tumoral
15.
Aging Cell ; 13(2): 303-10, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24305501

RESUMEN

Because of its anabolic effects on muscle, testosterone is being explored as a function-promoting anabolic therapy for functional limitations associated with aging; however, concerns about testosterone's adverse effects on prostate have inspired efforts to develop strategies that selectively increase muscle mass while sparing the prostate. Testosterone's promyogenic effects are mediated through upregulation of follistatin. We show here that the administration of recombinant follistatin (rFst) increased muscle mass in mice, but had no effect on prostate mass. Consistent with the results of rFst administration, follistatin transgenic mice with constitutively elevated follistatin levels displayed greater muscle mass than controls, but had similar prostate weights. To elucidate signaling pathways regulated differentially by testosterone and rFst in prostate and muscle, we performed microarray analysis of mRNAs from prostate and levator ani of castrated male mice treated with vehicle, testosterone, or rFst. Testosterone and rFst shared the regulation of many transcripts in levator ani; however, in prostate, 593 transcripts in several growth-promoting pathways were differentially expressed after testosterone treatment, while rFst showed a negligible effect with only 9 transcripts differentially expressed. Among pathways that were differentially responsive to testosterone in prostate, we identified ornithine decarboxylase (Odc1), an enzyme in polyamine biosynthesis, as a testosterone-responsive gene that is unresponsive to rFst. Accordingly, we administered testosterone with and without α-difluoromethylornithine (DFMO), an Odc1 inhibitor, to castrated mice. DFMO selectively blocked testosterone's effects on prostate, but did not affect testosterone's anabolic effects on muscle. Co-administration of testosterone and Odc1 inhibitor presents a novel therapeutic strategy for prostate-sparing anabolic therapy.


Asunto(s)
Anabolizantes/farmacología , Inhibidores Enzimáticos/farmacología , Tratamientos Conservadores del Órgano , Inhibidores de la Ornitina Descarboxilasa , Próstata/patología , Testosterona/administración & dosificación , Anabolizantes/administración & dosificación , Animales , Peso Corporal/efectos de los fármacos , Eflornitina/administración & dosificación , Eflornitina/farmacología , Inhibidores Enzimáticos/administración & dosificación , Folistatina/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de los Órganos/efectos de los fármacos , Ornitina Descarboxilasa/metabolismo , Próstata/efectos de los fármacos , Próstata/crecimiento & desarrollo , Próstata/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Testosterona/farmacología
16.
17.
Nat Biotechnol ; 31(2): 160-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23292609

RESUMEN

The ever-increasing incidence of antibiotic-resistant infections combined with a weak pipeline of new antibiotics has created a global public health crisis. Accordingly, novel strategies for enhancing our antibiotic arsenal are needed. As antibiotics kill bacteria in part by inducing reactive oxygen species (ROS), we reasoned that targeting microbial ROS production might potentiate antibiotic activity. Here we show that ROS production can be predictably enhanced in Escherichia coli, increasing the bacteria's susceptibility to oxidative attack. We developed an ensemble approach of genome-scale, metabolic models capable of predicting ROS production in E. coli. The metabolic network was systematically perturbed and its flux distribution analyzed to identify targets predicted to increase ROS production. Targets that were predicted in silico were experimentally validated and further shown to confer increased susceptibility to oxidants. Validated targets also increased susceptibility to killing by antibiotics. This work establishes a systems-based method to tune ROS production in bacteria and demonstrates that increased microbial ROS production can potentiate killing by oxidants and antibiotics.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Pruebas de Sensibilidad Microbiana , Oxidantes/farmacología
18.
Sci Transl Med ; 5(190): 190ra81, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23785037

RESUMEN

A declining pipeline of clinically useful antibiotics has made it imperative to develop more effective antimicrobial therapies, particularly against difficult-to-treat Gram-negative pathogens. Silver has been used as an antimicrobial since antiquity, yet its mechanism of action remains unclear. We show that silver disrupts multiple bacterial cellular processes, including disulfide bond formation, metabolism, and iron homeostasis. These changes lead to increased production of reactive oxygen species and increased membrane permeability of Gram-negative bacteria that can potentiate the activity of a broad range of antibiotics against Gram-negative bacteria in different metabolic states, as well as restore antibiotic susceptibility to a resistant bacterial strain. We show both in vitro and in a mouse model of urinary tract infection that the ability of silver to induce oxidative stress can be harnessed to potentiate antibiotic activity. Additionally, we demonstrate in vitro and in two different mouse models of peritonitis that silver sensitizes Gram-negative bacteria to the Gram-positive-specific antibiotic vancomycin, thereby expanding the antibacterial spectrum of this drug. Finally, we used silver and antibiotic combinations in vitro to eradicate bacterial persister cells, and show both in vitro and in a mouse biofilm infection model that silver can enhance antibacterial action against bacteria that produce biofilms. This work shows that silver can be used to enhance the action of existing antibiotics against Gram-negative bacteria, thus strengthening the antibiotic arsenal for fighting bacterial infections.


Asunto(s)
Antibacterianos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Plata/farmacología , Animales , Antibacterianos/uso terapéutico , Biopelículas/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Disulfuros/metabolismo , Sinergismo Farmacológico , Bacterias Gramnegativas/fisiología , Bacterias Gramnegativas/ultraestructura , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/patología , Humanos , Radical Hidroxilo/metabolismo , Hierro/metabolismo , Ratones , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Plata/uso terapéutico , Plata/toxicidad
19.
Sci Transl Med ; 5(192): 192ra85, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23825301

RESUMEN

Prolonged antibiotic treatment can lead to detrimental side effects in patients, including ototoxicity, nephrotoxicity, and tendinopathy, yet the mechanisms underlying the effects of antibiotics in mammalian systems remain unclear. It has been suggested that bactericidal antibiotics induce the formation of toxic reactive oxygen species (ROS) in bacteria. We show that clinically relevant doses of bactericidal antibiotics-quinolones, aminoglycosides, and ß-lactams-cause mitochondrial dysfunction and ROS overproduction in mammalian cells. We demonstrate that these bactericidal antibiotic-induced effects lead to oxidative damage to DNA, proteins, and membrane lipids. Mice treated with bactericidal antibiotics exhibited elevated oxidative stress markers in the blood, oxidative tissue damage, and up-regulated expression of key genes involved in antioxidant defense mechanisms, which points to the potential physiological relevance of these antibiotic effects. The deleterious effects of bactericidal antibiotics were alleviated in cell culture and in mice by the administration of the antioxidant N-acetyl-l-cysteine or prevented by preferential use of bacteriostatic antibiotics. This work highlights the role of antibiotics in the production of oxidative tissue damage in mammalian cells and presents strategies to mitigate or prevent the resulting damage, with the goal of improving the safety of antibiotic treatment in people.


Asunto(s)
Antibacterianos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo , Adenosina Trifosfato/metabolismo , Aminoglicósidos/efectos adversos , Aminoglicósidos/farmacología , Animales , Antibacterianos/efectos adversos , Línea Celular , Células Cultivadas , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Quinolonas/efectos adversos , Quinolonas/farmacología , beta-Lactamas/efectos adversos , beta-Lactamas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA