Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Endocr Relat Cancer ; 28(10): T193-T213, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34259647

RESUMO

Cloning of the sodium iodide symporter (NIS) 25 years ago has opened an exciting chapter in molecular thyroidology with the characterization of NIS as one of the most powerful theranostic genes and the development of a promising gene therapy strategy based on image-guided selective NIS gene transfer in non-thyroidal tumors followed by application of 131I or alternative radionuclides, such as 188Re and 211At. Over the past two decades, significant progress has been made in the development of the NIS gene therapy concept, from local NIS gene delivery towards promising new applications in disseminated disease, in particular through the use of oncolytic viruses, non-viral polyplexes, and genetically engineered MSCs as highly effective, highly selective and flexible gene delivery vehicles. In addition to allowing the robust therapeutic application of radioiodine in non-thyroid cancer settings, these studies have also been able to take advantage of NIS as a sensitive reporter gene that allows temporal and spatial monitoring of vector biodistribution, replication, and elimination - critically important issues for preclinical development and clinical translation.


Assuntos
Radioisótopos do Iodo , Cintilografia , Simportadores , Astato , Linhagem Celular Tumoral , Humanos , Radioisótopos do Iodo/uso terapêutico , Radioisótopos , Rênio , Simportadores/genética , Simportadores/metabolismo , Distribuição Tecidual
2.
Acta Virol ; 63(1): 111-116, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30879320

RESUMO

Recently, mitochondria have been shown to have a vital role in the innate immune response to viral infection. In response, viruses such as adenovirus, have developed mechanisms to alter mitochondrial function through direct or indirect interaction with the mitochondria. The interaction of human recombinant adenoviral vectors directly with human mitochondria has not previously been shown. We demonstrate that human recombinant adenoviral vectors co-localize to mitochondria. We show that the adenoviral vectors are present within the membranes of the mitochondria and that they cause ultrastructural changes to the cristae. Further, we show that the adenoviral genome is also present in intact mitochondria. We have posited that the interaction between the adenovirus and the mitochondria may act to inhibit mitochondrial function. We have also posited that the transport of the adenoviral genome to the mitochondria may allow the future use of this vector as a tool for gene therapy of mitochondrial diseases. Keywords: mitochondria; human recombinant adenovirus; gene therapy; viral vectors; mitochondrial DNA; electron microscopy.


Assuntos
Adenoviridae , Vetores Genéticos , Mitocôndrias , Adenoviridae/genética , Linhagem Celular , Terapia Genética , Vetores Genéticos/genética , Humanos , Mitocôndrias/virologia
3.
Mol Imaging Biol ; 20(1): 4-20, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28971346

RESUMO

The 6th annual meeting to address key issues in positron emission tomography (PET)/magnetic resonance imaging (MRI) was held again in Tübingen, Germany, from March 27 to 29, 2017. Over three days of invited plenary lectures, round table discussions and dialogue board deliberations, participants critically assessed the current state of PET/MRI, both clinically and as a research tool, and attempted to chart future directions. The meeting addressed the use of PET/MRI and workflows in oncology, neurosciences, infection, inflammation and chronic pain syndromes, as well as deeper discussions about how best to characterise the tumour microenvironment, optimise the complementary information available from PET and MRI, and how advanced data mining and bioinformatics, as well as information from liquid biomarkers (circulating tumour cells and nucleic acids) and pathology, can be integrated to give a more complete characterisation of disease phenotype. Some issues that have dominated previous meetings, such as the accuracy of MR-based attenuation correction (AC) of the PET scan, were finally put to rest as having been adequately addressed for the majority of clinical situations. Likewise, the ability to standardise PET systems for use in multicentre trials was confirmed, thus removing a perceived barrier to larger clinical imaging trials. The meeting openly questioned whether PET/MRI should, in all cases, be used as a whole-body imaging modality or whether in many circumstances it would best be employed to give an in-depth study of previously identified disease in a single organ or region. The meeting concluded that there is still much work to be done in the integration of data from different fields and in developing a common language for all stakeholders involved. In addition, the participants advocated joint training and education for individuals who engage in routine PET/MRI. It was agreed that PET/MRI can enhance our understanding of normal and disrupted biology, and we are in a position to describe the in vivo nature of disease processes, metabolism, evolution of cancer and the monitoring of response to pharmacological interventions and therapies. As such, PET/MRI is a key to advancing medicine and patient care.


Assuntos
Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Humanos , Biópsia Líquida , Radioterapia Guiada por Imagem , Microambiente Tumoral
4.
Oncogene ; 36(26): 3760-3771, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28218904

RESUMO

FTY720 (also called fingolimod) is recognized as an immunosuppressant and has been approved by the Food and Drug Administration to treat refractory multiple sclerosis. However, long-term administration of FTY720 potentially increases the risk for cancer in recipients. The underlying mechanisms remain poorly understood. Herein, we provided evidence that FTY720 administration potentiated tumor growth. Mechanistically, FTY720 enhanced extramedullary hematopoiesis and massive accumulation of myeloid-derived suppressor cells (MDSCs), which actively suppressed antitumor immune responses. Granulocyte-macrophage colony-stimulating factor (GM-CSF), mainly produced by MDSCs, was identified as a key factor to mediate these effects of FTY720 in tumor microenvironment. Furthermore, we showed that FTY720 triggers MDSCs to release GM-CSF via S1P receptor 3 (S1pr3) through Rho kinase and extracellular signal-regulated kinase-dependent pathway. Thus, our findings provide mechanistic explanation for the protumorigenic potentials of FTY720 and suggest that targeting S1pr3 simultaneously may be beneficial for the patients receiving FTY720 treatment.


Assuntos
Cloridrato de Fingolimode/efeitos adversos , Hematopoese Extramedular/efeitos dos fármacos , Imunossupressores/efeitos adversos , Células Mieloides/efeitos dos fármacos , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Cloridrato de Fingolimode/farmacologia , Humanos , Imunossupressores/farmacologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Células Mieloides/citologia , Células Mieloides/imunologia , Microambiente Tumoral/efeitos dos fármacos
5.
Oncogene ; 36(23): 3213-3222, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27991930

RESUMO

Rho GTPases are critical signal transducers of multiple pathways. They have been proposed to be useful anti-neoplastic targets for over two decades, especially in Ras-driven cancers. Until recently, however, few in vivo studies had been carried out to test this premise. Several recent mouse model studies have verified that Rac1, RhoA, and some of their effector proteins such as PAK and ROCK, are likely anti-cancer targets for treating K-Ras-driven tumours. Other seemingly contradictory studies have suggested that at least in certain instances inhibition of individual Rho GTPases may paradoxically result in pro-neoplastic effects. Significantly, both RhoA GTPase gain- and loss-of-function mutations have been discovered in primary leukemia/lymphoma and gastric cancer by human cancer genome sequencing efforts, suggesting both pro- and anti-neoplastic roles. In this review we summarize and integrate these unexpected findings and discuss the mechanistic implications in the design and application of Rho GTPase targeting strategies in future cancer therapies.


Assuntos
Antineoplásicos/uso terapêutico , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Animais , Humanos , Neoplasias/enzimologia
6.
Gene Ther ; 21(4): 393-401, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24572789

RESUMO

A number of antitumor vaccines have recently shown promise in upregulating immune responses against tumor antigens and improving patient survival. In this study, we examine the effectiveness of vaccination using interleukin (IL)-15-expressing tumor cells and also examine their ability to upregulate immune responses to tumor antigens. We demonstrated that the coexpression of IL-15 with its receptor, IL-15Rα, increased the cell-surface expression and secretion of IL-15. We show that a gene transfer approach using recombinant adenovirus to express IL-15 and IL-15Rα in murine TRAMP-C2 prostate or TS/A breast tumors induced antitumor immune responses. From this, we developed a vaccine platform, consisting of TRAMP-C2 prostate cancer cells or TS/A breast cancer cells coexpressing IL-15 and IL-15Rα that inhibited tumor formation when mice were challenged with tumor. Inhibition of tumor growth led to improved survival when compared with animals receiving cells expressing IL-15 alone or unmodified tumor cells. Animals vaccinated with tumor cells coexpressing IL-15 and IL-15Rα showed greater tumor infiltration with CD8(+) T and natural killer (NK) cells, as well as increased antitumor CD8(+) T-cell responses. Vaccination with IL-15/IL-15Rα-modified TS/A breast cancer cells provided a survival advantage to mice challenged with unrelated murine TUBO breast cancer cells, indicating the potential for allogeneic IL-15/IL-15Rα-expressing vaccines.


Assuntos
Neoplasias da Mama/genética , Subunidade alfa de Receptor de Interleucina-15/biossíntese , Interleucina-15/biossíntese , Neoplasias da Próstata/genética , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/transplante , Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Vacinação
7.
Case Rep Oncol Med ; 2013: 625243, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24303221

RESUMO

Introduction. Mucoepidermoid carcinoma (MEC) of the lung is a rare form of lung cancer that is classified into low grade and high grade based on histological features. Surgical resection is the primary treatment for low-grade MEC with excellent outcomes, while high-grade MEC is a more aggressive form of malignancy. Clinical Case. We report a case of a 46-year-old woman who presented with dyspnea on exertion. Imaging studies revealed a mass involving the right upper lobe bronchus. Bronchoscopy, surgical resection, and pathological examination revealed a low-grade MEC with tumor-free margins. No adjuvant treatment was given. Discussion. Primary pulmonary MEC is a rare type of lung cancer with only few reported cases. This patient illustrates a typical presentation for low-grade MEC wherein surgical resection is considered curative. In contrast, high-grade MEC is a more aggressive malignancy with a poorer outcome. The role of targeted therapy directed against EGFR or a novel CRTC1-MAML2 fusion protein expressed in some high-grade tumors is yet to be determined.

8.
Cancer Gene Ther ; 20(3): 195-200, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23412431

RESUMO

Our laboratory has investigated replicating adenovirus-human sodium iodide symporter (Ad-hNIS) vectors in a combinatorial oncolytic approach known as radiovirotherapy. However, hNIS-mediated iodide sequestration requires an intact cell membrane, and the enhancement of infectivity may alter the radioiodide accumulation in vivo. To assess these effects, we constructed Ad-NIS vectors expressing NIS from the major late promoter. Viral tropism was altered using a hybrid Ad5/3 fiber, and rates of viral spread altered through expression of the Ad death protein (ADP). The hybrid 5/3 fiber enhanced Ad-mediated cytolysis and radioisotope uptake in vitro. Replicating ADP-lacking viral vectors showed levels of uptake similar to non-replicating vectors that declined as cells lysed. ADP expression enhanced the rate of cell lysis and viral release, but reduced the peak and duration of radioiodide uptake. SPECT-computed tomography imaging showed the Ad5/3-noADP-hNIS vector induced significantly more isotope uptake than other vector structures, indicating that viral spread may not always make up for the reduced NIS expression as in our work with prostate cancer. These results indicate that replicating, infectivity-enhanced Ad-NIS vectors provide superior overall efficacy, but also indicate that the effect of replication speed requires tumor and model-specific testing.


Assuntos
Adenovírus Humanos/genética , Vetores Genéticos/genética , Radioisótopos do Iodo/metabolismo , Simportadores/genética , Replicação Viral , Linhagem Celular Tumoral , Efeito Citopatogênico Viral , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Humanos , Vírus Oncolíticos/genética , Simportadores/metabolismo , Transplante Heterólogo
9.
Gene Ther ; 20(5): 567-74, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22972493

RESUMO

We have constructed a prostate tumor-specific conditionally replicating adenovirus (CRAd), named Ad5PB_RSV-NIS, which expresses the human sodium iodine symporter (NIS) gene. LNCaP tumors were established in nude mice and infected with this CRAd to study tumor viral spread, NIS expression, and efficacy. Using quantitative PCR, we found a linear correlation between the viral dose and viral genome copy numbers recovered after tumor infection. Confocal microscopy showed a linear correlation between adenovirus density and NIS expression. Radioiodide uptake vs virus dose-response curves revealed that the dose response curve was not linear and displayed a lower threshold of detection at 10(7) vp (virus particles) and an upper plateau of uptake at 10(11) vp. The outcome of radiovirotherapy was highly dependent upon viral dose. At 10(10) vp, no significant differences were observed between virotherapy alone or radiovirotherapy. However, when radioiodide therapy was combined with virotherapy at a dose of 10(11) vp, significant improvement in survival was observed, indicating a relationship between viral dose-response uptake and the efficacy of radiovirotherapy. The reasons behind the differences in radioiodide therapy efficacy can be ascribed to more efficient viral tumor spread and a decrease in the rate of radioisotope efflux. Our results have important implications regarding the desirable and undesirable characteristics of vectors for clinical translation of virus-mediated NIS transfer therapy.


Assuntos
Radioisótopos do Iodo , Terapia Viral Oncolítica , Neoplasias da Próstata/radioterapia , Neoplasias da Próstata/virologia , Simportadores/genética , Adenoviridae/genética , Proteína de Ligação a Androgênios/genética , Animais , Linhagem Celular Tumoral , Terapia Combinada , Vetores Genéticos , Humanos , Masculino , Camundongos , Neoplasias da Próstata/patologia , Transfecção , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Gene Ther ; 19(12): 839-44, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23037808

RESUMO

The sodium-iodide symporter (NIS) directs the uptake and concentration of iodide in thyroid cells. We have extended the use of NIS-mediated radioiodine therapy to prostate cancer. We have developed a prostate tumor specific conditionally replicating adenovirus that expresses hNIS (Ad5PB_RSV-NIS). For radiovirotherapy to be effective in humans, the radioiodine dose administered in the pre-clinical animal model should scale to the range of acceptable doses in humans. We performed (131)I dose-response experiments aiming to determine the dose required in mice to achieve efficient radiovirotherapy. Efficacy was determined by measuring tumor growth and survival times. We observed that individual tumors display disparate growth rates that preclude averaging within a treatment modality indicating heterogeneity of growth rate. We further show that a statistic and stochastic approach must be used when comparing the effect of an anti-cancer therapy on a cohort of tumors. Radiovirotherapy improves therapeutic value over virotherapy alone by slowing the rate of tumor growth in a more substantial manner leading to an increase in survival time. We also show that the radioiodine doses needed to achieve this increase scaled well within the current doses used for treatment of thyroid cancer in humans.


Assuntos
Adenoviridae/fisiologia , Radioisótopos do Iodo/administração & dosagem , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/terapia , Simportadores/genética , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Processos de Crescimento Celular/efeitos da radiação , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Neoplasias da Próstata/virologia , Taxa de Sobrevida , Simportadores/biossíntese , Simportadores/metabolismo , Transfecção , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Gene Ther ; 19(9): 659-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22790962

RESUMO

Anaplastic thyroid cancer is an extremely aggressive disease resistant to radioiodine treatment because of loss of sodium iodide symporter (NIS) expression. To enhance prognosis of this fatal cancer, we validated the preclinical efficacy of measles virus (MV)-NIS, the vaccine strain of the oncolytic MV (MV-Edm), modified to include the NIS gene. Western blotting analysis confirmed that a panel of eight anaplastic thyroid cancer (ATC)-derived cell lines do not express NIS protein, but do express CD46, the MV receptor. In vitro cell death assays and in vivo xenograft studies demonstrate the oncolytic efficacy of MV-NIS in BHT-101 and KTC-3, ATC-derived cell lines. Radioactive iodine uptake along with single-photon emission computed tomography (SPECT)-computed tomography imaging of KTC-3 xenografts after (99)Tc(m) administration confirmed NIS expression in vitro and in vivo, respectively, after virus treatment. Adjuvant administration of RAI, to MV-NIS-treated KTC-3 tumors showed a trend for increased tumor cell killing. As current treatment for ATC is only palliative, and MV-NIS is currently Food and Drug Administration approved for human clinical trials in myeloma, our data indicate that targeting ATC with MV-NIS could prove to be a novel therapeutic strategy for effective treatment of iodine-resistant ATC and will expedite its testing in clinical trials for this aggressive disease.


Assuntos
Iodo/metabolismo , Vírus do Sarampo/metabolismo , Vírus Oncolíticos/metabolismo , Simportadores/uso terapêutico , Neoplasias da Glândula Tireoide/terapia , Animais , Western Blotting , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Terapia Genética/métodos , Humanos , Radioisótopos do Iodo/metabolismo , Radioisótopos do Iodo/uso terapêutico , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Nus , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Receptores Virais/metabolismo , Simportadores/genética , Simportadores/metabolismo , Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Neurology ; 75(2): 111-5, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20534887

RESUMO

OBJECTIVES: Diagnostic challenges exist for differentiating HIV dementia from Alzheimer disease (AD) in older HIV-infected (HIV+) individuals. Similar abnormalities in brain amyloid-beta42 (Alphabeta42) metabolism may be involved in HIV-associated neuropathology and AD. We evaluated the amyloid-binding agent (11)C-Pittsburgh compound B ((11)C-PiB), a biomarker for Alphabeta42 deposition, in cognitively unimpaired HIV+ (n = 10) participants and matched community controls without dementia (n = 20). METHODS: In this case-control study, all participants had an (11)C-PiB scan within 2 years of concomitant CSF studies and neuropsychometric testing. Statistical differences between HIV+ and community controls for demographic and clinical values were assessed by chi(2) tests. Participants were further divided into either low (<500 pg/mL) or normal (>or=500 pg/mL) CSF Alphabeta42 groups with Student t tests performed to determine if regional differences in fibrillar amyloid plaque deposition varied with CSF Alphabeta42. RESULTS: Regardless of CSF Alphabeta42 level, none of the HIV+ participants had fibrillar amyloid plaques as assessed by increased (11)C-PiB mean cortical binding potential (MCBP) or binding potential within 4 cortical regions. In contrast, some community controls with low CSF Alphabeta42 (<500 pg/mL) had high (11)C-PiB MCBP with elevated binding potentials (>0.18 arbitrary units) within cortical regions. CONCLUSIONS: Cognitively unimpaired HIV+ participants, even with low CSF Alphabeta42 (<500 pg/mL), do not have (11)C-PiB parameters suggesting brain fibrillar amyloid deposition. The dissimilarity between unimpaired HIV+ and preclinical AD may reflect differences in Abeta42 production and/or formation of diffuse plaques. Future longitudinal studies of HIV+ participants with low CSF Abeta42 and normal (11)C-PiB are required.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Benzotiazóis/metabolismo , Encéfalo/metabolismo , Transtornos Cognitivos/metabolismo , Infecções por HIV/metabolismo , Adulto , Análise de Variância , Compostos de Anilina , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Radioisótopos de Carbono , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Transtornos Cognitivos/diagnóstico por imagem , Feminino , HIV , Infecções por HIV/diagnóstico por imagem , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Cintilografia , Tiazóis
13.
Gene Ther ; 17(11): 1325-32, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20428214

RESUMO

The sodium iodide symporter (NIS) directs the uptake and concentration of iodide in thyroid cells. We have extended the use of NIS-mediated radioiodine therapy to other types of cancer, we transferred and expressed the NIS gene into prostate, colon and breast cancer cells using adenoviral vectors. To improve vector efficiency we have developed a conditionally replicating adenovirus (CRAd) in which the E1a gene is driven by the prostate-specific promoter, Probasin and the cassette RSV promoter human NIScDNA-bGH polyA replaces the E3 region (CRAd Ad5PB_RSV-NIS). In vitro infection of the prostate cancer cell line LnCaP resulted in virus replication, cytolysis and release of infective viral particles. Conversely, the prostate cancer cell line PC-3 (androgen receptor negative) and the pancreatic cancer cell line Panc-1 were refractory to the viral cytopathic effect and did not support viral replication. Radioiodine uptake was readily measurable in LnCaP cells infected with Ad5PB_RSV-NIS 24 h post-infection, confirming NIS expression. In vivo, LnCaP tumor xenografts in nude-mice injected intratumorally with Ad5PB_RSV_NIS CRAd expressed NIS actively as evidenced by 99Tc uptake and imaging. Administration of therapeutic ¹³¹I after virus injection significantly increased survival probability in mice carrying xenografted LnCaP tumors compared with virotherapy alone. These data indicate that Ad5PB_RSV_NIS replication is stringently restricted to androgen-positive prostate cancer cells and results in effective NIS expression and uptake of radioiodine. This construct may allow multimodal therapy, combining cytolytic virotherapy with radioiodine treatment, to be developed as a novel treatment for prostate cancer.


Assuntos
Adenoviridae/genética , Proteína de Ligação a Androgênios/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/terapia , Simportadores/genética , Adenoviridae/metabolismo , Animais , Linhagem Celular Tumoral , Vetores Genéticos/genética , Humanos , Radioisótopos do Iodo/química , Masculino , Camundongos , Camundongos Nus , Simportadores/metabolismo , Transfecção , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Neurology ; 74(2): 106-12, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20032288

RESUMO

OBJECTIVE: To investigate whether cancer is associated with Alzheimer disease (AD) and vascular dementia (VaD). METHODS: Cox proportional hazards models were used to test associations between prevalent dementia and risk of future cancer hospitalization, and associations between prevalent cancer and risk of subsequent dementia. Participants in the Cardiovascular Health Study-Cognition Substudy, a prospective cohort study, aged 65 years or older (n = 3,020) were followed a mean of 5.4 years for dementia and 8.3 years for cancer. RESULTS: The presence of any AD (pure AD + mixed AD/VaD; hazard ratio [HR] = 0.41, 95% confidence interval [CI] = 0.20-0.84) and pure AD (HR = 0.31, 95% CI = 0.12-0.86) was associated with a reduced risk of future cancer hospitalization, adjusted for demographic factors, smoking, obesity, and physical activity. No significant associations were found between dementia at baseline and rate of cancer hospitalizations for participants with diagnoses of VaD. Prevalent cancer was associated with reduced risk of any AD (HR = 0.72; 95% CI = 0.52-0.997) and pure AD (HR = 0.57; 95% CI = 0.36-0.90) among white subjects after adjustment for demographics, number of APOE epsilon4 alleles, hypertension, diabetes, and coronary heart disease; the opposite association was found among minorities, but the sample size was too small to provide stable estimates. No significant association was found between cancer and subsequent development of VaD. CONCLUSIONS: In white older adults, prevalent Alzheimer disease (AD) was longitudinally associated with a reduced risk of cancer, and a history of cancer was associated with a reduced risk of AD. Together with other work showing associations between cancer and Parkinson disease, these findings suggest the possibility that cancer is linked to neurodegeneration.


Assuntos
Doença de Alzheimer/epidemiologia , Demência Vascular/epidemiologia , Neoplasias/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Estudos de Coortes , Demência Vascular/genética , Feminino , Predisposição Genética para Doença/genética , Hospitalização/estatística & dados numéricos , Hospitalização/tendências , Humanos , Masculino , Neoplasias/genética , Degeneração Neural/epidemiologia , Degeneração Neural/genética , Doença de Parkinson/epidemiologia , Doença de Parkinson/genética , Prevalência , Modelos de Riscos Proporcionais , Estudos Prospectivos , Medição de Risco/métodos , Fatores de Risco , População Branca
15.
Gene Ther ; 15(3): 214-23, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17989705

RESUMO

Due to limited treatment options the prognosis of patients with advanced hepatocellular cancer (HCC) has remained poor. To investigate an alternative therapeutic approach, we examined the feasibility of radioiodine therapy of HCC following human sodium iodide symporter (NIS) gene transfer using a mouse alpha-fetoprotein (AFP) promoter construct to target NIS expression to HCC cells. For this purpose, the murine Hepa 1-6 and the human HepG2 hepatoma cell lines were stably transfected with NIS cDNA under the control of the tumor-specific AFP promoter. The stably transfected Hepa 1-6 cell line showed a 10-fold increase in iodide accumulation, while HepG2 cells accumulated (125)I approximately 60-fold. Tumor-specific NIS expression was confirmed on mRNA level by northern blot analysis, and on protein level by immunostaining, that revealed primarily membrane-associated NIS-specific immunoreactivity. In an in vitro clonogenic assay up to 78% of NIS-transfected Hepa 1-6 and 93% of HepG2 cells were killed by (131)I exposure, while up to 96% of control cells survived. In vivo NIS-transfected HepG2 xenografts accumulated 15% of the total (123)I administered per gram tumor with a biological half-life of 8.38 h, resulting in a tumor absorbed dose of 171 mGy MBq(-1) (131)I. After administration of a therapeutic (131)I dose (55.5 MBq) tumor growth of NIS expressing HepG2 xenografts was significantly inhibited. In conclusion, tumor-specific iodide accumulation was induced in HCC cells by AFP promoter-directed NIS expression in vitro and in vivo, which was sufficiently high to allow a therapeutic effect of (131)I. This study demonstrates the potential of tumor-specific NIS gene therapy as an innovative treatment strategy for HCC.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Neoplasias Hepáticas/terapia , Regiões Promotoras Genéticas , Simportadores/genética , alfa-Fetoproteínas/genética , Animais , Terapia Combinada , Expressão Gênica , Humanos , Radioisótopos do Iodo/uso terapêutico , Camundongos , Compostos Radiofarmacêuticos/uso terapêutico , Transfecção/métodos
17.
Gene Ther ; 13(1): 60-6, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16121204

RESUMO

Ovarian cancer represents the fifth leading cause of cancer death among women in the United States, with >16 000 deaths expected this year. This study was carried out to investigate the potential of sodium iodide symporter (NIS)-mediated radioiodide therapy as a novel approach for ovarian cancer treatment. Radioiodide is routinely and effectively used for the treatment of benign and malignant thyroid disease as a result of native thyroidal expression of NIS, which mediates iodide uptake. In vitro gene transfer studies in ovarian cancer cells revealed a 12- and five-fold increase in iodide uptake when transduced with Ad/CMV/NIS or Ad/MUC1/NIS, respectively. Western blot/immunohistochemistry confirmed NIS protein expression. In vivo ovarian tumor xenografts were infected with the adenoviral constructs. (123)I imaging revealed a clear image of the CMV/NIS-transduced tumor, with a less intense image apparent following infection with MUC1/NIS. Therapeutic doses of (131)I following CMV/NIS infection caused a mean 53% reduction in tumor volume (P<0.0001). MUC1/NIS-transduced tumors did not regress, although at 8 weeks following therapy, tumor volume was significantly less that of control animals (166 versus 332%, respectively, P<0.05). This study represents a promising first step investigating the potential for NIS-mediated radioiodide imaging and therapy of ovarian tumors.


Assuntos
Terapia Genética/métodos , Radioisótopos do Iodo , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/terapia , Simportadores/genética , Adenoviridae/genética , Animais , Western Blotting/métodos , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Imuno-Histoquímica/métodos , Radioisótopos do Iodo/uso terapêutico , Camundongos , Mucina-1/genética , Transplante de Neoplasias , Regiões Promotoras Genéticas , Cintilografia , Simportadores/metabolismo , Transdução Genética/métodos , Transplante Heterólogo
18.
J Clin Endocrinol Metab ; 91(1): 69-78, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16234306

RESUMO

CONTEXT: The sodium iodide symporter (NIS) mediates the active iodide uptake in the thyroid gland as well as lactating breast tissue. Recently induction of functional NIS expression was reported in the estrogen receptor-positive human breast cancer cell line MCF-7 by all-trans retinoic acid (atRA) treatment in vitro and in vivo, which might offer the potential to treat breast cancer with radioiodine. OBJECTIVE: In the current study, we examined the effect of dexamethasone (Dex) on atRA-induced NIS expression and therapeutic efficacy of 131-I in MCF-7 cells. DESIGN: For this purpose, NIS mRNA and protein expression levels in MCF-7 cells were examined by Northern and Western blot analysis after incubation with Dex (10(-9) to 10(-7) m) in the presence of atRA (10(-6) m) as well as immunostaining using a mouse monoclonal human NIS-specific antibody. In addition, NIS functional activity was measured by iodide uptake and efflux assay, and in vitro cytotoxicity of 131-I was examined by in vitro clonogenic assay. RESULTS: After incubation with Dex in the presence of atRA, NIS mRNA levels in MCF-7 cells were stimulated up to 11-fold in a concentration-dependent manner, whereas NIS protein levels increased up to 16-fold and iodide accumulation was stimulated up to 3- to 4-fold. Furthermore, iodide efflux was modestly decreased after stimulation with Dex in the presence of atRA. Furthermore, in the in vitro clonogenic assay, selective cytotoxicity of 131-I was significantly increased from approximately 17% in MCF-7 cells treated with atRA alone to 80% in MCF-7 cells treated with Dex in the presence of atRA. CONCLUSION: Treatment with Dex in the presence of atRA significantly increases functional NIS expression levels in addition to inhibiting iodide efflux, resulting in an enhanced selective killing effect of 131-I in MCF-7 breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Dexametasona/farmacologia , Radioisótopos do Iodo/uso terapêutico , Simportadores/biossíntese , Tretinoína/farmacologia , Northern Blotting , Western Blotting , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Feminino , Humanos , Imuno-Histoquímica , Iodetos/metabolismo , RNA Mensageiro/biossíntese , Receptores de Estrogênio/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
19.
Cytotherapy ; 7(4): 345-52, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16162456

RESUMO

BACKGROUND: There is ongoing controversy about the transdifferentiation of hematopoietic stem cells (HSC) into different tissues such as mesenchymal cells. This transdifferentiation or 'plasticity' would be an appealing concept for many therapeutic strategies. While studies in the murine model show encouraging results, reports from clinical allogeneic stem cell transplantations do not support the concept of HSC plasticity. Our aim was to determine whether transplantation of transduced autologous marrow CD34+ cells leads to long-term engraftment of gene-marked cells with mesenchymal characteristics in the baboon. METHODS: We analyzed marrow of two baboons that had received green fluorescence protein (GFP)-marked CD34+ autologous marrow cells after myeloablative conditioning. Marrow was obtained 1 and 2.5 years after transplantation and adherent CD11a- (pan-leukocyte Ab) cells were cultured for 3 weeks. Cultures were then analyzed by flow cytometry and fluorescence microscopy for the presence of GFP+ cells. For further analysis fresh and cultured cells were also labeled with multiple Ab and functional analysis was performed. RESULTS: Both animals showed persistent and stable GFP marking by flow cytometry in peripheral blood leukocytes as well as in CD34+ marrow cells at 1 and 2.5 years after transplantation. There was no evidence of GFP+ mesenchymal cells by either flow cytometry or fluorescence microscopy, while functional and phenotypical analysis identified mesenchymal stem cells in these cultures. DISCUSSION: We conclude that genetically modified CD34+ cells do not contribute to the adherent marrow-derived mesenchymal cell population after autologous transplantation.


Assuntos
Antígenos CD34/biossíntese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Animais , Células Cultivadas , Gammaretrovirus , Genes Reporter , Vetores Genéticos , Células-Tronco Hematopoéticas/imunologia , Mesoderma/citologia , Papio , Transdução Genética , Transplante Autólogo
20.
Neurology ; 64(5): 895-8, 2005 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-15753432

RESUMO

Cross-sectional studies raise the possibility of protective relationships between, or a common mechanism underlying, the development of dementia of the Alzheimer type (DAT) and cancer. Using a prospective longitudinal design, the authors found that the risk of developing cancer is less among participants with DAT vs nondemented participants (p < 0.001) and that the risk of developing DAT may be less for participants with a history of cancer (p = 0.060).


Assuntos
Doença de Alzheimer/epidemiologia , Predisposição Genética para Doença/epidemiologia , Neoplasias/epidemiologia , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/fisiopatologia , Causalidade , Comorbidade , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Neoplasias/fisiopatologia , Estudos Prospectivos , Análise de Regressão , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA