Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Neuroimage ; 216: 116352, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31730921

RESUMEN

Individuals with attention-deficit/hyperactivity disorder (ADHD) have difficulties navigating dynamic everyday situations that contain multiple sensory inputs that need to either be attended to or ignored. As conventional experimental tasks lack this type of everyday complexity, we administered a film-based multi-talker condition with auditory distractors in the background. ADHD-related aberrant brain responses to this naturalistic stimulus were identified using intersubject correlations (ISCs) in functional magnetic resonance imaging (fMRI) data collected from 51 adults with ADHD and 29 healthy controls. A novel permutation-based approach introducing studentized statistics and subject-wise voxel-level null-distributions revealed that several areas in cerebral attention networks and sensory cortices were desynchronized in participants with ADHD (n = 20) relative to healthy controls (n = 20). Specifically, desynchronization of the posterior parietal cortex occurred when irrelevant speech or music was presented in the background, but not when irrelevant white noise was presented, or when there were no distractors. We also show regionally distinct ISC signatures for inattention and impulsivity. Finally, post-scan recall of the film contents was associated with stronger ISCs in the default-mode network for the ADHD and in the dorsal attention network for healthy controls. The present study shows that ISCs can further our understanding of how a complex environment influences brain states in ADHD.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/diagnóstico por imagen , Atención , Percepción Auditiva , Encéfalo/diagnóstico por imagen , Películas Cinematográficas , Percepción Visual , Estimulación Acústica/métodos , Adulto , Atención/fisiología , Trastorno por Déficit de Atención con Hiperactividad/psicología , Percepción Auditiva/fisiología , Encéfalo/fisiología , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Estimulación Luminosa/métodos , Percepción Visual/fisiología
2.
Hum Brain Mapp ; 41(17): 4876-4891, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32813290

RESUMEN

The development of treatments for attention impairments is hampered by limited knowledge about the malleability of underlying neural functions. We conducted the first randomized controlled trial to determine the modulations of brain activity associated with working memory (WM) training in adults with attention-deficit hyperactivity disorder (ADHD). At baseline, we assessed the aberrant functional brain activity in the n-back WM task by comparing 44 adults with ADHD with 18 healthy controls using fMRI. Participants with ADHD were then randomized to train on an adaptive dual n-back task or an active control task. We tested whether WM training elicits redistribution of brain activity as observed in healthy controls, and whether it might further restore aberrant activity related to ADHD. As expected, activity in areas of the default-mode (DMN), salience (SN), sensory-motor (SMN), frontoparietal (FPN), and subcortical (SCN) networks was decreased in participants with ADHD at pretest as compared with healthy controls, especially when the cognitive load was high. WM training modulated widespread FPN and SN areas, restoring some of the aberrant activity. Training effects were mainly observed as decreased brain activity during the trained task and increased activity during the untrained task, suggesting different neural mechanisms for trained and transfer tasks.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Trastorno por Déficit de Atención con Hiperactividad/rehabilitación , Corteza Cerebral/fisiopatología , Remediación Cognitiva , Red en Modo Predeterminado/fisiopatología , Función Ejecutiva/fisiología , Memoria a Corto Plazo/fisiología , Red Nerviosa/fisiopatología , Desempeño Psicomotor/fisiología , Adulto , Trastorno por Déficit de Atención con Hiperactividad/diagnóstico por imagen , Cerebelo/diagnóstico por imagen , Cerebelo/fisiopatología , Corteza Cerebral/diagnóstico por imagen , Red en Modo Predeterminado/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Red Nerviosa/diagnóstico por imagen , Evaluación de Resultado en la Atención de Salud , Adulto Joven
3.
Mol Ther ; 24(7): 1323-32, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27039846

RESUMEN

The development of oncolytic viruses has recently made great progress towards being available to cancer patients. With the breakthrough into clinics, it is crucial to analyze the existing clinical experience and use it as a basis for treatment improvements. Here, we report clinical data from 290 patients treated with oncolytic adenovirus. Using clinical variables and treatment characteristics, we constructed statistical models with regard to treatment response and overall survival (OS). Additionally, we investigated effects of neutralizing antibodies, tumor burden, and peripheral blood leucocyte counts on these outcomes. We found the absence of liver metastases to correlate with an improved rate of disease control (P = 0.021). In multivariate evaluation, patients treated with viruses coding for immunostimulatory granulocyte macrophage colony-stimulating factor were linked to better prognosis (hazard ratio (HR) 0.378, P < 0.001), as well as women with any cancer type (HR 0.694, P = 0.017). In multivariate analysis for imaging response, patients treated via intraperitoneal injection were more likely to achieve disease control (odds ratio (OR) 3.246, P = 0.027). Patients with low neutrophil-to-lymphocyte ratio before treatment had significantly longer OS (P < 0.001). These findings could explain some of the variation seen in treatment outcomes after virotherapy. Furthermore, the results offer hypotheses for treatment optimization and patient selection in oncolytic adenovirus immunotherapy.


Asunto(s)
Adenoviridae , Neoplasias/mortalidad , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Adenoviridae/genética , Adenoviridae/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Biomarcadores , Femenino , Expresión Génica , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Recuento de Leucocitos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/secundario , Masculino , Metástasis de la Neoplasia , Neoplasias/diagnóstico , Neoplasias/genética , Oportunidad Relativa , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Pronóstico , Modelos de Riesgos Proporcionales , Tomografía Computarizada por Rayos X , Transgenes , Resultado del Tratamiento , Carga Tumoral
4.
Mol Ther ; 24(1): 175-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26310629

RESUMEN

Despite many clinical trials conducted with oncolytic viruses, the exact tumor-level mechanisms affecting therapeutic efficacy have not been established. Currently there are no biomarkers available that would predict the clinical outcome to any oncolytic virus. To assess the baseline immunological phenotype and find potential prognostic biomarkers, we monitored mRNA expression levels in 31 tumor biopsy or fluid samples from 27 patients treated with oncolytic adenovirus. Additionally, protein expression was studied from 19 biopsies using immunohistochemical staining. We found highly significant changes in several signaling pathways and genes associated with immune responses, such as B-cell receptor signaling (P < 0.001), granulocyte macrophage colony-stimulating factor (GM-CSF) signaling (P < 0.001), and leukocyte extravasation signaling (P < 0.001), in patients surviving a shorter time than their controls. In immunohistochemical analysis, markers CD4 and CD163 were significantly elevated (P = 0.020 and P = 0.016 respectively), in patients with shorter than expected survival. Interestingly, T-cell exhaustion marker TIM-3 was also found to be significantly upregulated (P = 0.006) in patients with poor prognosis. Collectively, these data suggest that activation of several functions of the innate immunity before treatment is associated with inferior survival in patients treated with oncolytic adenovirus. Conversely, lack of chronic innate inflammation at baseline may predict improved treatment outcome, as suggested by good overall prognosis.


Asunto(s)
Adenoviridae/fisiología , Perfilación de la Expresión Génica/métodos , Inmunidad Innata , Neoplasias/genética , Neoplasias/terapia , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos CD4/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Pronóstico , Receptores de Superficie Celular/metabolismo , Resultado del Tratamiento
5.
Mol Ther ; 23(2): 321-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25381801

RESUMEN

Oncolytic immunotherapy with cytokine armed replication competent viruses is an emerging approach in cancer treatment. In a recent randomized trial, an increase in response rate was seen but the effect on overall survival is not known with any virus. To facilitate randomized trials, we performed a case-control study assessing the survival of 270 patients treated in an Advanced Therapy Access Program (ATAP), in comparison to matched concurrent controls from the same hospital. The overall survival of all virus treated patients was not increased over controls. However, when analysis was restricted to GMCSF-sensitive tumor types treated with GMSCF-coding viruses, a significant improvement in median survival was present (from 170 to 208 days, P = 0.0012, N = 148). An even larger difference was seen when analysis was restricted to good performance score patients (193 versus 292 days, P = 0.034, N = 90). The survival of ovarian cancer patients was especially promising as median survival nearly quadrupled (P = 0.0003, N = 37). These preliminary data lend support to initiation of randomized clinical trials with GMCSF-coding oncolytic adenoviruses.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Vectores Genéticos/administración & dosificación , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neoplasias/diagnóstico , Neoplasias/mortalidad , Modelos de Riesgos Proporcionales , Resultado del Tratamiento , Adulto Joven
6.
Mol Ther ; 23(10): 1641-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26156245

RESUMEN

In clinical trials with oncolytic adenoviruses, there has been no mortality associated with treatment vectors. Likewise, in the Advanced Therapy Access Program (ATAP), where 290 patients were treated with 10 different viruses, no vector-related mortality was observed. However, as the patient population who received adenovirus treatments in ATAP represented heavily pretreated patients, often with very advanced disease, some patients died relatively soon after receiving their virus treatment mandating autopsy to investigate cause of death. Eleven such autopsies were performed and confirmed disease progression as the cause of death in each case. The regulatory requirement for investigating the safety of advanced therapy medical products presented a unique opportunity to study tissue samples collected as a routine part of the autopsies. Oncolytic adenoviral DNA was recovered in a wide range of tissues, including injected and noninjected tumors and various normal tissues, demonstrating the ability of the vector to disseminate through the vascular route. Furthermore, we recovered and cultured viable virus from samples of noninjected brain metastases of an intravenously treated patient, confirming that oncolytic adenovirus can reach tumors through the intravascular route. Data presented here give mechanistic insight into mode of action and biodistribution of oncolytic adenoviruses in cancer patients.


Asunto(s)
Transducción Genética , Adenoviridae/genética , Adolescente , Adulto , Anciano , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Autopsia , Línea Celular Tumoral , Niño , Preescolar , ADN Viral , Femenino , Dosificación de Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/farmacocinética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Factores de Tiempo , Distribución Tisular , Proteínas Virales/genética , Proteínas Virales/metabolismo , Adulto Joven
7.
Mol Ther ; 23(5): 964-973, 2015 05.
Artículo en Inglés | MEDLINE | ID: mdl-25655312

RESUMEN

The quality of the antitumor immune response is decisive when developing new immunotherapies for cancer. Oncolytic adenoviruses cause a potent immunogenic stimulus and arming them with costimulatory molecules reshapes the immune response further. We evaluated peripheral blood T-cell subsets of 50 patients with refractory solid tumors undergoing treatment with oncolytic adenovirus. These data were compared to changes in antiviral and antitumor T cells, treatment efficacy, overall survival, and T-cell subsets in pre- and post-treatment tumor biopsies. Treatment caused a significant (P < 0.0001) shift in T-cell subsets in blood, characterized by a proportional increase of CD8(+) cells, and decrease of CD4(+) cells. Concomitant treatment with cyclophosphamide and temozolomide resulted in less CD4(+) decrease (P = 0.041) than cyclophosphamide only. Interestingly, we saw a correlation between T-cell changes in peripheral blood and the tumor site. This correlation was positive for CD8(+) and inverse for CD4(+) cells. These findings give insight to the interconnections between peripheral blood and tumor-infiltrating lymphocyte (TIL) populations regarding oncolytic virotherapy. In particular, our data suggest that induction of T-cell response is not sufficient for clinical response in the context of immunosuppressive tumors, and that peripheral blood T cells have a complicated and potentially misleading relationship with TILs.


Asunto(s)
Adenoviridae , Terapia Genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Subgrupos de Linfocitos T/inmunología , Adenoviridae/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Niño , Femenino , Humanos , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/diagnóstico , Neoplasias/genética , Virus Oncolíticos/genética , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Transgenes , Adulto Joven
8.
Int J Cancer ; 137(7): 1775-83, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25821063

RESUMEN

Metastatic melanoma is refractory to irradiation and chemotherapy, but amenable to immunological approaches such as immune-checkpoint-inhibiting antibodies or adoptive cell therapies. Oncolytic virus replication is an immunogenic phenomenon, and viruses can be armed with immunostimulatory molecules. Therefore, oncolytic immuno-virotherapy of malignant melanoma is an appealing approach, which was recently validated by a positive phase 3 trial. We investigated the potency of oncolytic adenovirus Ad5/3-D24-GMCSF on a panel of melanoma cell lines and animal models, and summarized the melanoma-specific human data from the Advanced Therapy Access Program (ATAP). The virus effectively eradicated human melanoma cells in vitro and subcutaneous SK-MEL-28 melanoma xenografts in nude mice when combined with low-dose cyclophosphamide. Furthermore, virally-expressed granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulated the differentiation of human monocytes into macrophages. In contrast to human cells, RPMI 1846 hamster melanoma cells exhibited no response to oncolytic viruses and the chimeric 5/3 fiber failed to increase the efficacy of transduction, suggesting limited utility of the hamster model in the context of viruses with this capsid. In ATAP, treatments appeared safe and well-tolerated. Four out of nine melanoma patients treated were evaluable for possible therapy benefit with modified RECIST criteria: one patient had minor response, two had stable disease, and one had progressive disease. Two patients were alive at 559 and 2,149 days after treatment. Ad5/3-D24-GMCSF showed promising efficacy in preclinical studies and possible antitumor activity in melanoma patients refractory to other forms of therapy. This data supports continuing the clinical development of oncolytic adenoviruses for treatment of malignant melanoma.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Cricetinae , Ciclofosfamida/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Macrófagos/patología , Macrófagos/virología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/virología , Ratones , Ratones Desnudos , Monocitos/patología , Monocitos/virología , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Int J Cancer ; 135(3): 720-30, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24374597

RESUMEN

Sarcomas are a relatively rare cancer, but often incurable at the late metastatic stage. Oncolytic immunotherapy has gained attention over the past years, and a wide range of oncolytic viruses have been delivered via intratumoral injection with positive safety and promising efficacy data. Here, we report preclinical and clinical results from treatment of sarcoma with oncolytic adenovirus Ad5/3-D24-GMCSF (CGTG-102). Ad5/3-D24-GMCSF is a serotype chimeric oncolytic adenovirus coding for human granulocyte-macrophage colony-stimulating factor (GM-CSF). The efficacy of Ad5/3-D24-GMCSF was evaluated on a panel of soft-tissue sarcoma (STS) cell lines and in two animal models. Sarcoma specific human data were also collected from the Advanced Therapy Access Program (ATAP), in preparation for further clinical development. Efficacy was seen in both in vitro and in vivo STS models. Fifteen patients with treatment-refractory STS (13/15) or primary bone sarcoma (2/15) were treated in ATAP, and treatments appeared safe and well-tolerated. A total of 12 radiological RECIST response evaluations were performed, and two cases of minor response, six cases of stable disease and four cases of progressive disease were detected in patients progressing prior to virus treatment. Overall, the median survival time post treatment was 170 days. One patient is still alive at 1,459 days post virus treatment. In summary, Ad5/3-D24-GMCSF appears promising for the treatment of advanced STS; a clinical trial for treatment of refractory injectable solid tumors including STS is ongoing.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Viroterapia Oncolítica , Sarcoma/terapia , Animales , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inyecciones Intralesiones , Mesocricetus , Ratones , Ratones Desnudos , Pronóstico , Sarcoma/sangre , Sarcoma/mortalidad , Tasa de Supervivencia , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Ther ; 21(6): 1212-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23546299

RESUMEN

Oncolytic adenoviruses and certain chemotherapeutics can induce autophagy and immunogenic cancer cell death. We hypothesized that the combination of oncolytic adenovirus with low-dose temozolomide (TMZ) is safe, effective, and capable of inducing antitumor immune responses. Metronomic low-dose cyclophosphamide (CP) was added to selectively reduce regulatory T-cells. Preclinically, combination therapy inhibited tumor growth, increased autophagy, and triggered immunogenic cell death as indicated by elevated calreticulin, adenosine triphosphate (ATP) release, and nuclear protein high-mobility group box-1 (HMGB1) secretion. A total of 41 combination treatments given to 17 chemotherapy-refractory cancer patients were well tolerated. We observed anti- and proinflammatory cytokine release, evidence of virus replication, and induction of neutralizing antibodies. Tumor cells showed increased autophagy post-treatment. Release of HMGB1 into serum--a possible indicator of immune response--increased in 60% of treatments, and seemed to correlate with tumor-specific T-cell responses, observed in 10/15 cases overall (P = 0.0833). Evidence of antitumor efficacy was seen in 67% of evaluable treatments with a trend for increased survival over matched controls treated with virus only. In summary, the combination of oncolytic adenovirus with low-dose TMZ and metronomic CP increased tumor cell autophagy, elicited antitumor immune responses, and showed promising safety and efficacy.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Dacarbazina/análogos & derivados , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenosina Trifosfato/metabolismo , Adenoviridae/fisiología , Adolescente , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/sangre , Calreticulina/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Niño , Terapia Combinada/métodos , Ciclofosfamida/farmacología , Citocinas/sangre , ADN Viral/sangre , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica , Persona de Mediana Edad , Virus Oncolíticos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Temozolomida , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
11.
Mol Ther ; 20(1): 221-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22044933

RESUMEN

Calcium channel blockers including verapamil have been proposed to enhance release and antitumor efficacy of oncolytic adenoviruses in preclinical studies but this has not been studied in humans before. Here, we studied if verapamil leads to increased replication of oncolytic adenovirus in cancer patients, as measured by release of virions from tumor cells into the systemic circulation. The study was conducted as a matched case-control study of advanced cancer patients treated with oncolytic adenoviruses with or without verapamil. We observed that verapamil increased mean virus titers present in blood after treatment (P < 0.05). The frequency or severity of adverse events was not increased, nor were cytokine responses or neutralizing antibody levels different between groups. Signs of possible treatment-related clinical benefits were observed in both groups, but there was no significant difference in responses or survival. Thus, our data suggests that the combination of verapamil with oncolytic adenoviruses is safe and well tolerated. Moreover, verapamil treatment seems to result in higher virus titers in blood, indicating enhanced overall replication in tumors. A randomized trial is needed to confirm these findings and to study if enhanced replication results in benefits to patients.


Asunto(s)
Adenoviridae/genética , Bloqueadores de los Canales de Calcio/uso terapéutico , Vectores Genéticos , Neoplasias/terapia , Virus Oncolíticos/genética , Verapamilo/uso terapéutico , Adenoviridae/inmunología , Adolescente , Adulto , Anciano , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Bloqueadores de los Canales de Calcio/efectos adversos , Terapia Combinada , ADN Viral/sangre , Sinergismo Farmacológico , Femenino , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/mortalidad , Neoplasias/patología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/inmunología , Análisis de Supervivencia , Resultado del Tratamiento , Verapamilo/efectos adversos , Carga Viral/efectos de los fármacos , Adulto Joven
12.
Netw Neurosci ; 7(4): 1302-1325, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38144696

RESUMEN

Current knowledge of white matter changes in large-scale brain networks in adult attention-deficit/hyperactivity disorder (ADHD) is scarce. We collected diffusion-weighted magnetic resonance imaging data in 40 adults with ADHD and 36 neurotypical controls and used constrained spherical deconvolution-based tractography to reconstruct whole-brain structural connectivity networks. We used network-based statistic (NBS) and graph theoretical analysis to investigate differences in these networks between the ADHD and control groups, as well as associations between structural connectivity and ADHD symptoms assessed with the Adult ADHD Self-Report Scale or performance in the Conners Continuous Performance Test 2 (CPT-2). NBS revealed decreased connectivity in the ADHD group compared to the neurotypical controls in widespread unilateral networks, which included subcortical and corticocortical structures and encompassed dorsal and ventral attention networks and visual and somatomotor systems. Furthermore, hypoconnectivity in a predominantly left-frontal network was associated with higher amount of commission errors in CPT-2. Graph theoretical analysis did not reveal topological differences between the groups or associations between topological properties and ADHD symptoms or task performance. Our results suggest that abnormal structural wiring of the brain in adult ADHD is manifested as widespread intrahemispheric hypoconnectivity in networks previously associated with ADHD in functional neuroimaging studies.

13.
Int J Cancer ; 131(1): 253-63, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21834073

RESUMEN

Conventional cancer treatments often have little impact on the course of advanced pancreatic cancer. Although cancer gene therapy with adenoviruses is a promising developmental approach, the primary receptor is poorly expressed in pancreatic cancers which might compromise efficacy and thus targeting to other receptors could be beneficial. Extended stealth delivery, combination with standard chemotherapy or circumvention of host antiadenoviral immune response might improve efficacy further. In this work, capsid-modified adenoviruses were studied for transduction of cell lines and clinical normal and tumor tissue samples. The respective oncolytic viruses were tested for oncolytic activity in vitro and in vivo. Survival was studied in a peritoneally disseminated pancreas cancer model, with or without concurrent gemcitabine while silica implants were utilized for extended intraperitoneal virus delivery. Immunocompetent mice and Syrian hamsters were used to study the effect of silica mediated delivery on antiviral immune responses and subsequent in vivo gene delivery. Capsid modifications selectively enhanced gene transfer to malignant pancreatic cancer cell lines and clinical samples. The respective oncolytic viruses resulted in increased cell killing in vitro, which translated into a survival benefit in mice. Early proinfammatory cytokine responses and formation of antiviral neutralizing antibodies was partially avoided with silica implants. The implant also shielded the virus from pre-existing neutralizing antibodies, while increasing the pancreas/liver gene delivery ratio six-fold. In conclusion, capsid modified adenoviruses would be useful for testing in pancreatic cancer trials. Silica implants might increase the safety and efficacy of the approach.


Asunto(s)
Adenovirus Humanos , Desoxicitidina/análogos & derivados , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Neoplasias Pancreáticas/terapia , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Anticuerpos Neutralizantes , Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Terapia Combinada , Cricetinae , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Femenino , Humanos , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Gel de Sílice , Transfección , Gemcitabina
14.
Mol Ther ; 19(9): 1737-46, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21673660

RESUMEN

Patients with advanced solid tumors refractory to and progressing after conventional therapies were treated with three different regimens of low-dose cyclophosphamide (CP) in combination with oncolytic adenovirus. CP was given with oral metronomic dosing (50 mg/day, N = 21), intravenously (single 1,000 mg dose, N = 7) or both (N = 7). Virus was injected intratumorally. Controls (N = 8) received virus without CP. Treatments were well tolerated and safe regardless of schedule. Antibody formation and virus replication were not affected by CP. Metronomic CP (oral and oral + intravenous schedules) decreased regulatory T cells (T(regs)) without compromising induction of antitumor or antiviral T-cell responses. Oncolytic adenovirus given together with metronomic CP increased cytotoxic T cells and induced Th1 type immunity on a systemic level in most patients. All CP regimens resulted in higher rates of disease control than virus only (all P < 0.0001) and the best progression-free (PFS) and overall survival (OS) was seen in the oral + intravenous group. One year PFS and OS were 53 and 42% (P = 0.0016 and P < 0.02 versus virus only), respectively, both which are unusually high for chemotherapy refractory patients. We conclude that low-dose CP results in immunological effects appealing for oncolytic virotherapy. While these first-in-human data suggest good safety, intriguing efficacy and extended survival, the results should be confirmed in a randomized trial.


Asunto(s)
Antineoplásicos/administración & dosificación , Ciclofosfamida/administración & dosificación , Neoplasias/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Linfocitos T Reguladores/inmunología , Adenoviridae/genética , Adolescente , Adulto , Anciano , Animales , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Niño , Terapia Combinada , Cricetinae , Ciclofosfamida/inmunología , Ciclofosfamida/uso terapéutico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Vectores Genéticos , Humanos , Masculino , Mesocricetus , Persona de Mediana Edad , Neoplasias/inmunología , Resultado del Tratamiento , Adulto Joven
15.
Mol Ther ; 18(10): 1874-84, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20664527

RESUMEN

Augmenting antitumor immunity is a promising way to enhance the potency of oncolytic adenoviral therapy. Granulocyte-macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific CD8(+) cytotoxic T-lymphocytes. Serotype 5 adenoviruses (Ad5) are commonly used in cancer gene therapy. However, expression of the coxsackie-adenovirus receptor is variable in many advanced tumors and preclinical data have demonstrated an advantage for replacing the Ad5 knob with the Ad3 knob. Here, a 5/3 capsid chimeric and p16-Rb pathway selective oncolytic adenovirus coding for GMCSF was engineered and tested preclinically. A total of 21 patients with advanced solid tumors refractory to standard therapies were then treated intratumorally and intravenously with Ad5/3-D24-GMCSF, which was combined with low-dose metronomic cyclophosphamide to reduce regulatory T cells. No severe adverse events occurred. Analysis of pretreatment samples of malignant pleural effusion and ascites confirmed the efficacy of Ad5/3-D24-GMCSF in transduction and cell killing. Evidence of biological activity of the virus was seen in 13/21 patients and 8/12 showed objective clinical benefit as evaluated by radiology with Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Antiadenoviral and antitumoral immune responses were elicited after treatment. Thus, Ad5/3-D24-GMCSF seems safe in treating cancer patients and promising signs of efficacy were seen.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adolescente , Adulto , Anciano , Animales , Línea Celular , Línea Celular Tumoral , Cricetinae , Ciclofosfamida/uso terapéutico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Inmunosupresores/uso terapéutico , Masculino , Mesocricetus , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
16.
J Gene Med ; 11(11): 966-77, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19670332

RESUMEN

BACKGROUND: Rapid clearance of adenoviruses from blood by macrophage lineage cells of the liver and spleen, and binding to platelets, hinder their successful systemic use for cancer gene therapy. Vitamin K dependent coagulation factors are important mediators for the adenovirus liver tropism. Here we aim to determine the effects of coagulation factor, thrombocyte and liver macrophage (Kupffer cell) ablation on biodistribution of serotype 5 adenoviruses in mice with orthotopic breast tumors. METHODS: Prior to intravenous injection of adenoviruses, mice bearing orthotopic breast tumors were pretreated with warfarin to inhibit vitamin K dependent coagulation factor synthesis, an anti-platelet antibody causing thrombocytopenia or an inhibitor of the Kupffer cell scavenger receptor or their combination. Virus availability in blood after injection was determined from blood samples and transgene expression in tissues analyzed 72 hours afterwards with In Vivo Imaging and luciferase assays. RESULTS: Warfarin pretreatment reduced gene delivery to liver, spleen and lung. Kupffer cell ablation increased persistence of adenoviruses in blood but didn't affect biodistribution significantly. Depletion of Kupffer cells combined with thrombocytopenia doubled the systemic gene delivery of 5/3 chimeric adenovirus to tumors (p < 0.05). Triple ablation of platelets, Kupffer cells and coagulation factors increased the tumor to liver ratio of systemic adenovirus gene delivery by 81% (p < 0.05). CONCLUSIONS: Depletion of coagulation factors can reduce transduction of liver, spleen and lung. Kupffer cell depletion is the most feasible method of increasing amount adenovirus in systemic blood flow and in combination with ablation of thrombocytes can increase the transduction of adenovirus to tumors.


Asunto(s)
Adenoviridae/genética , Neoplasias Mamarias Experimentales/genética , Virus Oncolíticos/genética , Transducción Genética , Adenoviridae/metabolismo , Animales , Factores de Coagulación Sanguínea/antagonistas & inhibidores , Plaquetas/efectos de los fármacos , Femenino , Macrófagos del Hígado/efectos de los fármacos , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/terapia , Ratones , Viroterapia Oncolítica/métodos , Warfarina/farmacología
17.
Hum Gene Ther ; 30(6): 740-752, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30672366

RESUMEN

After the discovery and characterization of the adenovirus in the 1950s, this prevalent cause of the common cold and other usually mild diseases has been modified and utilized in biomedicine in several ways. To date, adenoviruses are the most frequently used vectors and therapeutic (e.g., oncolytic) agents with a number of beneficial features. They infect both dividing and nondividing cells, enable high-level, transient protein expression, and are easy to amplify to high concentrations. As an important and versatile research tool, it is of essence to understand the limits and advantages that genetic modification of adenovirus vectors may entail. Therefore, a retrospective analysis was performed of adenoviral gene therapy constructs produced in the same laboratory with similar methods. The aim was to assess the impact of various modifications on the physical and functional titer of the virus. It was found that genome size (designed within "the 105% golden rule") did not significantly affect the physical titer of the adenovirus preparations, regardless of the type of transgene (e.g., immunostimulatory vs. other), number of engineered changes, and size of the mutated virus genome. One statistically significant exception was noted, however. Chimeric adenoviruses (5/3) had a slightly lower physical titer compared to Ad5-based viruses, although a trend for the opposite was true for functional titers. Thus, 5/3 chimeric viruses may in fact be appealing from a safety versus efficacy viewpoint. Armed viruses had lower functional and physical titers than unarmed viruses, while five genomic modifications started to decrease functional titer. Importantly, even highly modified armed viruses generally had good titers compatible with clinical testing. In summary, this paper shows the plasticity of adenovirus for various vector, oncolytic, and armed oncolytic uses. These results inform future generations of adenovirus-based drugs for human use. This information is directly transferable to academic laboratories and the biomedical industry involved in vector design and production optimization.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Vectores Genéticos/genética , Neoplasias/genética , Neoplasias/terapia , Adenoviridae/clasificación , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Ingeniería Genética , Terapia Genética/métodos , Tamaño del Genoma , Humanos , Ratones , Neoplasias/patología , Transducción Genética , Transgenes
18.
Mol Ther Oncolytics ; 9: 41-50, 2018 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-29989063

RESUMEN

The first US Food and Drug Administration (FDA)- and EMA-approved oncolytic virus has been available since 2015. However, there are no markers available that would predict benefit for the individual patient. During 2007-2012, we treated 290 patients with advanced chemotherapy-refractory cancers, using 10 different oncolytic adenoviruses. Treatments were given in a Finnish Medicines Agency (FIMEA)-regulated individualized patient treatment program (the Advanced Therapy Access Program [ATAP]), which required long-term follow-up of patients, which is presented here. Focusing on the longest surviving patients, some key clinical and biological features are presented as "oncograms." Some key attributes that could be captured in the oncogram are suggested to predict treatment response and survival after oncolytic adenovirus treatment. The oncogram includes immunological laboratory parameters assessed in peripheral blood (leukocytes, neutrophil-to-lymphocyte ratio, interleukin-8 [IL-8], HMGB1, anti-viral neutralizing antibody status), features of the patient (gender, performance status), tumor features (histological tumor type, tumor load, region of metastases), and oncolytic virus-specific features (arming of the virus). The retrospective approach used here facilitates verification in a prospective controlled trial setting. To our knowledge, the oncogram is the first holistic attempt to identify the patients most likely to benefit from adenoviral oncolytic virotherapy.

19.
Oncotarget ; 9(5): 6320-6335, 2018 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-29464075

RESUMEN

After the landmark approval of T-VEC, oncolytic viruses are finding their way to the clinics. However, response rates have still room for improvement, and unfortunately there are currently no available markers to predict responses for oncolytic immunotherapy. Interleukin 8 (IL-8) production is upregulated in many cancers and it also connects to several pathways that have been shown to impair the efficacy of adenoviral immunotherapy. We studied the role of IL-8 in 103 cancer patients treated with oncolytic adenoviruses. We found high baseline serum IL-8 concentration to be independently associated with poor prognosis (p<0.001). Further, normal baseline IL-8 was associated with improved prognostic potential of calculation of the neutrophil-to-lymphocyte ratio (p<0.001). Interestingly, a decrease in IL-8 concentration after treatment with oncolytic adenovirus predicted better overall survival (p<0.001) and higher response rate, although this difference was not significant (p=0.066). We studied the combination of adenovirus and IL-8 neutralizing antibody ex vivo in single cell suspensions and in co-cultures of tumor-associated CD15+ neutrophils and CD3+ tumor-infiltrating lymphocytes derived from fresh patient tumor samples. These results indicate a role for IL-8 as a biomarker in oncolytic virotherapy, but additionally provide a rationale for targeting IL-8 to improve treatment efficacy. In conclusion, curtailing the activity of IL-8 systemically or locally in the tumor microenvironment could improve anti-tumor immune responses resulting in enhanced efficacy of adenoviral immunotherapy of cancer.

20.
Oncoimmunology ; 5(2): e1078057, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27057453

RESUMEN

Breast cancer is a heterogeneous disease, characterized by several distinct biological subtypes, among which triple-negative breast cancer (TNBC) is one associated with a poor prognosis. Oncolytic virus replication is an immunogenic phenomenon, and viruses can be armed with immunostimulatory molecules to boost virus triggered antitumoral immune responses. Cyclophosphamide (CP) is a chemotherapy drug that is associated with cytotoxicity and immunosuppression at higher doses, whereas immunostimulatory and anti-angiogenic properties are observed at low continuous dosage. Therefore, the combination of oncolytic immuno-virotherapy with low-dose CP is an appealing approach. We investigated the potency of oncolytic adenovirus Ad5/3-D24-GMCSF on a TNBC cell line and in vivo in an orthotopic xenograft mouse model, in combination with low-dose CP or its main active metabolite 4-hydroperoxycyclophosphamide (4-HP-CP). Furthermore, we summarized the breast cancer-specific human data on this virus from the Advanced Therapy Access Program (ATAP). Low-dose CP increased the efficacy of Ad5/3-D24-GMCSF in vitro and in a TNBC mouse model. In ATAP, treatments appeared safe and well-tolerated. Thirteen out of 16 breast cancer patients treated were evaluable for possible benefits with modified RECIST 1.1 criteria: 1 patient had a minor response, 2 had stable disease (SD), and 10 had progressive disease (PD). One patient is alive at 1,771 d after treatment. Ad5/3-D24-GMCSF in combination with low-dose CP showed promising efficacy in preclinical studies and possible antitumor activity in breast cancer patients refractory to other forms of therapy. This preliminary data supports continuing the clinical development of oncolytic adenoviruses for treatment of breast cancer, including TNBC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA