RESUMO
Purpose: Spontaneously occurring glioma in pet dogs is increasingly recognized as a valuable translational model for human glioblastoma. Canine high grade glioma and human glioblastomas share many molecular similarities, including accumulation of immunosuppressive regulatory T cells (Tregs) that inhibit anti-tumor immune responses. Identifying in dog mechanisms responsible for Treg recruitment may afford targeting the cellular population driving immunosuppression, the results providing a rationale for translational clinical studies in human patients. Our group has previously identified C-C motif chemokine 2 (CCL2) as a glioma-derived T-reg chemoattractant acting on chemokine receptor 4 (CCR4) in a murine orthotopic model of glioma. Recently, we demonstrated a robust increase of CCL2 in the brain tissue of canine patients bearing high-grade glioma. Methods: We performed a series of in vitro experiments using canine Tregs and patient-derived canine glioma cell lines (GSC 1110, GSC 0514, J3T-Bg, G06A) to interrogate the CCL2-CCR4 signaling axis in the canine. Results: We established a flow cytometry gating strategy for identification and isolation of FOXP3+ Tregs in dogs. The canine CD4 + CD25high T-cell population was highly enriched in FOXP3 and CCR4 expression, indicating they are bona fide Tregs. Canine Treg migration was enhanced by CCL2 or by glioma cell line-derived supernatant. Blockade of the CCL2-CCR4 axis significantly reduced migration of canine Tregs. CCL2 mRNA was expressed in all glioma cell lines and expression increased when exposed to Tregs but not to CD4 + helper T-cells. Conclusion: Our study validates CCL2-CCR4 as a bi-directional Treg-glioma immunosuppressive and tumor-promoting axis in canine high-grade glioma.
RESUMO
PURPOSE: Spontaneously occurring glioma in pet dogs is increasingly recognized as a valuable translational model for human glioblastoma. Canine high-grade glioma and human glioblastomas share many molecular similarities, including the accumulation of immunosuppressive regulatory T cells (Tregs) that inhibit anti-tumor immune responses. Identifying in dog mechanisms responsible for Treg recruitment may afford to target the cellular population driving immunosuppression, the results providing a rationale for translational clinical studies in human patients. Our group has previously identified C-C motif chemokine 2 (CCL2) as a glioma-derived T-reg chemoattractant acting on chemokine receptor 4 (CCR4) in a murine orthotopic glioma model. Recently, we demonstrated a robust increase of CCL2 in the brain tissue of canine patients bearing high-grade glioma. METHODS: We performed a series of in vitro experiments using canine Tregs and patient-derived canine glioma cell lines (GSC 1110, GSC 0514, J3T-Bg, G06A) to interrogate the CCL2-CCR4 signaling axis in the canine. RESULTS: We established a flow cytometry gating strategy for identifying and isolating FOXP3+ Tregs in dogs. The canine CD4 + CD25high T-cell population was highly enriched in FOXP3 and CCR4 expression, indicating they are bona fide Tregs. Canine Treg migration was enhanced by CCL2 or by glioma cell line-derived supernatant. Blockade of the CCL2-CCR4 axis significantly reduced migration of canine Tregs. CCL2 mRNA was expressed in all glioma cell lines, and expression increased when exposed to Tregs but not CD4 + helper T-cells. CONCLUSION: Our study validates CCL2-CCR4 as a bi-directional Treg-glioma immunosuppressive and tumor-promoting axis in canine high-grade glioma.
Assuntos
Neoplasias Encefálicas , Quimiocina CCL2 , Glioma , Receptores CCR4 , Linfócitos T Reguladores , Animais , Cães , Humanos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Glioma/metabolismo , Glioma/imunologia , Glioma/patologia , Receptores CCR4/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismoRESUMO
Over the last decade, autophagy has emerged as one of the critical cellular systems that control homeostasis. Besides management of normal homeostatic processes, autophagy can also be induced by tissue damage stress or by rapidly progressing tumors. During tumor progression, autophagy mediates a cellular reaction to the changes inside and outside of cells, which leads to tumor adaptation. Even though the regulation of autophagy seems universal and is a well-described process, its dysregulation and role in glioma progression remain an important topic of investigation. In this review, we summarize recent evidence of autophagy regulation in brain tumor tissues and possible interconnection between signaling pathways that govern cellular responses. This perspective may help to assess the qualitative differences and various outcomes in response to autophagy stimulation.
Assuntos
Proteínas Relacionadas à Autofagia/genética , Autofagia/genética , Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Glioma/mortalidade , Glioma/patologia , Glioma/terapia , Humanos , MicroRNAs/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Terapia Viral Oncolítica , RNA Longo não Codificante/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genéticaRESUMO
Magnetic micro and nanoparticles are increasingly used in biotechnological applications due to the ability to control their behavior through an externally applied field. We demonstrate the fabrication of particles made from ultrathin perpendicularly magnetized CoFeB/Pt layers with antiferromagnetic interlayer coupling. The particles are characterized by zero moment at remanence, low susceptibility at low fields, and a large saturated moment created by the stacking of the basic coupled bilayer motif. We demonstrate the transfer of magnetic properties from thin films to lithographically defined 2 µm particles which have been lifted off into solution. We simulate the minimum energy state of a synthetic antiferromagnetic bilayer system that is free to rotate in an applied field and show that the low field susceptibility of the system is equal to the magnetic hard axis followed by a sharp switch to full magnetization as the field is increased. This agrees with the experimental results and explains the behaviour of the particles in solution.
RESUMO
In preclinical studies, neural stem cell (NSC)-based delivery of oncolytic virus has shown great promise in the treatment of malignant glioma. Ensuring the success of this therapy will require critical evaluation of the spatial distribution of virus after NSC transplantation. In this study, the patient-derived GBM43 human glioma line was established in the brain of athymic nude mice, followed by the administration of NSCs loaded with conditionally replicating oncolytic adenovirus (NSC-CRAd-S-pk7). We determined the tumor coverage potential of oncolytic adenovirus by examining NSC distribution using magnetic resonance (MR) imaging and by three-dimensional reconstruction from ex vivo tissue specimens. We demonstrate that unmodified NSCs and NSC-CRAd-S-pk7 exhibit a similar distribution pattern with most prominent localization occurring at the tumor margins. We were further able to visualize the accumulation of these cells at tumor sites via T2-weighted MR imaging as well as the spread of viral particles using immunofluorescence. Our analyses reveal that a single administration of oncolytic virus-loaded NSCs allows for up to 31% coverage of intracranial tumors. Such results provide valuable insights into the therapeutic potential of this novel viral delivery platform.
Assuntos
Rastreamento de Células , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/patologia , Imageamento por Ressonância Magnética , Células-Tronco Neurais/metabolismo , Vírus Oncolíticos/genética , Adenoviridae/genética , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Rastreamento de Células/métodos , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Glioblastoma/diagnóstico , Humanos , Camundongos , Transdução Genética , Carga Tumoral , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
Glioblastoma multiforme patients have a poor prognosis due to therapeutic resistance and tumor relapse. It has been suggested that gliomas are driven by a rare subset of tumor cells known as glioma stem cells (GSCs). This hypothesis states that only a few GSCs are able to divide, differentiate, and initiate a new tumor. It has also been shown that this subpopulation is more resistant to conventional therapies than its differentiated counterpart. In order to understand glioma recurrence post therapy, we investigated the behavior of GSCs after primary chemotherapy. We first show that exposure of patient-derived as well as established glioma cell lines to therapeutic doses of temozolomide (TMZ), the most commonly used antiglioma chemotherapy, consistently increases the GSC pool over time both in vitro and in vivo. Secondly, lineage-tracing analysis of the expanded GSC pool suggests that such amplification is a result of a phenotypic shift in the non-GSC population to a GSC-like state in the presence of TMZ. The newly converted GSC population expresses markers associated with pluripotency and stemness, such as CD133, SOX2, Oct4, and Nestin. Furthermore, we show that intracranial implantation of the newly converted GSCs in nude mice results in a more efficient grafting and invasive phenotype. Taken together, these findings provide the first evidence that glioma cells exposed to chemotherapeutic agents are able to interconvert between non-GSCs and GSCs, thereby replenishing the original tumor population, leading to a more infiltrative phenotype and enhanced chemoresistance. This may represent a potential mechanism for therapeutic relapse.
Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/efeitos dos fármacos , Dacarbazina/análogos & derivados , Glioblastoma/patologia , Células-Tronco Neoplásicas/fisiologia , Animais , Antineoplásicos Alquilantes/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Glioblastoma/tratamento farmacológico , Humanos , Masculino , Camundongos Nus , Fenótipo , Temozolomida , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
Myeloid-derived suppressor cells (MDSCs) accumulate in the glioma microenvironment during tumor progression and promote immunosuppression. Interleukin-12 (IL-12) immunogene therapy can alter MDSCs toward an antigen-presenting cell phenotype and these mature cells can have a central role in antigen presentation. It remains unclear, however, how MDSC depletion can affect glioma immunotherapy. In this study, we generated a replication-deficient adenoviral vector, Ad.5/3.cRGD-mIL12p70, that transduces the GL261-based murine glioma cell line, resulting in the induction of biologically active, murine IL12p70 expression. Ex vivo, IL-12 expressed by GL261 cells induced interferon-γ synthesis in CD8(+) T cells (P<0.001), CD4(+) T cells (P=0.009) and natural killer cells (P=0.036). When injected 1 week after tumor implantation, Ad.5/3.cRGD-mIL12p70 successfully prolonged the survival of glioma-bearing mice. Sixty percent of animals treated with IL-12 immunotherapy were long-term survivors over 175 days, whereas all the control group animals expired by 40 days after tumor implantation (P=0.026). Mice receiving Ad.5/3.cRGD-mIL12p70 also accumulated 50% less MDSCs in the brain than the control group (P=0.007). Moreover, in the IL-12 group, MDSCs significantly overexpressed CD80 and major histocompatibility complex class II molecules (P=0.041). Depletion of MDSCs with Gr1(+) antibody had no survival benefit induced by IL-12-mediated immunotherapy. Of note, IL-12 therapy increased the presence of myeloid dendritic cells (mDCs) in the glioma microenvironment (P=0.0069). Ultimately, the data show that in the context of IL-12 immunogene therapy, MDSCs are dispensable and mDCs may provide the majority of antigen presentation in the brain.
Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Glioma/genética , Glioma/imunologia , Interleucina-12/genética , Células Mieloides/imunologia , Adenoviridae/genética , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/mortalidade , Glioma/terapia , Imunoterapia , Interleucina-12/biossíntese , Masculino , CamundongosRESUMO
Glioblastoma multiforme (GBM), the most common and aggressive form of primary brain tumor, presents a dismal prognosis. Current standard therapies are only able to improve patient survival by a few months. The search for alternative approaches in glioblastoma treatment, together with the recent discovery of a new class of small RNA molecules that are capable of regulating gene expression, prompted a race for a deeper and thorough understanding of how these molecules work. Today, it is known that microRNAs are involved in many cellular processes that are altered in GBM tumors, such as angiogenesis, invasion, cell proliferation and apoptosis. Research in this area is now gathering efforts to translate these findings into clinically relevant therapies that could improve the diagnosis and outcome of GBM patients. In this review, we discuss the use of microRNAs as potential diagnostic, prognostic and therapeutic tools against glioblastoma. We will also assess the current challenges and future perspectives of microRNA-based therapies, with a special focus on why this promising therapeutic approach is not yet in the clinic and how to overcome this limitation.
Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , MicroRNAs/genética , Genes Supressores de Tumor , Humanos , MicroRNAs/metabolismo , Prognóstico , Transdução de SinaisRESUMO
Conditionally replicating adenoviruses (CRAd) are a promising class of gene therapy agents that can overcome already known glioblastoma (GBM) resistance mechanisms but have limited distribution upon direct intratumoral (i.t.) injection. Collagen bundles in the extracellular matrix (ECM) have an important role in inhibiting virus distribution. In fact, ECM pre-treatment with collagenases improves virus distributions to tumor cells. Matrix metalloproteinases (MMPs) are an endogenous class of collagenases secreted by tumor cells whose function can be altered by different drugs including anti-angiogenic agents, such as bevacizumab. In this study we hypothesized that upregulation of MMP activity during anti-angiogenic therapy can improve CRAd-S-pk7 distribution in GBM. We find that MMP-2 activity in human U251 GBM xenografts increases (*P=0.03) and collagen IV content decreases (*P=0.01) during vascular endothelial growth factor (VEGF-A) antibody neutralization. After proving that collagen IV inhibits CRAd-S-pk7 distribution in U251 xenografts (Spearman rho=-0.38; **P=0.003), we show that VEGF-blocking antibody treatment followed by CRAd-S-pk7 i.t. injection reduces U251 tumor growth more than each individual agent alone (***P<0.0001). Our data propose a novel approach to improve virus distribution in tumors by relying on the early effects of anti-angiogenic therapy.
Assuntos
Adenoviridae/fisiologia , Inibidores da Angiogênese/farmacologia , Colágeno/metabolismo , Glioma/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Bloqueadores/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/genética , Glioma/patologia , Humanos , Proteínas Inibidoras de Apoptose/genética , Injeções Intralesionais , Estimativa de Kaplan-Meier , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Nus , Terapia Viral Oncolítica/métodos , Polilisina/genética , Polilisina/metabolismo , Regiões Promotoras Genéticas/genética , Proteólise , Survivina , Fator A de Crescimento do Endotélio Vascular/imunologia , Replicação Viral/efeitos dos fármacosRESUMO
Oncolytic virotherapy is a promising novel therapy for glioblastoma that needs to be optimized before introduced to clinic. The targeting of conditionally replicating adenoviruses (CRAds) can be improved by relying on the tumor-tropic properties of neural stem cells (NSCs). Here, we report the characterization of an FDA approved NSC, HB1.F3-CD, as a cell carrier for CRAd-S-pk7, a glioma-tropic oncolytic adenovirus. We show that NSCs replicate and release infectious CRAd-S-pk7 progeny capable of lysing glioma cell lines. Moreover, ex-vivo-loaded NSCs, injected intracranially in nude mice bearing human glioma xenografts (i) retained their tumor tropism, (ii) continued to replicate CRAd-S-pk7 for more than a week after reaching the tumor site and (iii) successfully handed off CRAd-S-pk7 to glioma cells in vivo. Delivery via carrier cells reduced non-specific adenovirus distribution in the mouse brain. Moreover, we assessed biodistribution of loaded NSCs after intracranial injection in animal models semi-permissive to adenovirus replication, the Syrian hamster and cotton rat. NSCs did not migrate to distant organs and high levels of CRAd-S-pk7 DNA were observed only in the injected hemisphere. In conclusion, this optimized carrier system, with high efficiency of adenovirus delivery and minimal systemic toxicity, poses considerable advantages for anti-glioma oncolytic virotherapy.
Assuntos
Adenoviridae/fisiologia , Neoplasias Encefálicas/terapia , Glioma/terapia , Células-Tronco Neurais/transplante , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Adenoviridae/genética , Proteínas E1A de Adenovirus/biossíntese , Proteínas E1A de Adenovirus/genética , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Sobrevivência Celular , Cricetinae , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/biossíntese , Humanos , Luciferases de Vaga-Lume/biossíntese , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Vírus Oncolíticos/genética , Organismos Geneticamente Modificados , Proteínas Recombinantes/biossíntese , Sigmodontinae , Carga Viral , Replicação ViralRESUMO
Functional nanoscale materials that possess specific physical or chemical properties can leverage energy transduction in vivo. Once these materials integrate with biomolecules they combine physical properties of inorganic material and the biorecognition capabilities of bio-organic moieties. Such nano-bio hybrids can be interfaced with living cells, the elementary functional units of life. These nano-bio systems are capable of bio-manipulation or actuation via altering intracellular biochemical pathways. Thus, nano-bio conjugates are appealing for a wide range of applications from the life sciences and nanomedicine to catalysis and clean energy production. Here we highlight recent progress in our efforts to develop smart nano-bio hybrid materials, and to study their performance within cellular machinery under application of external stimuli, such as light or magnetic fields.
RESUMO
Conditionally replicative adenoviruses (CRAds) represent a novel treatment strategy for malignant glioma. Recent studies suggest that the cytopathic effect elicited by these vectors is mediated through autophagy, a form of programmed cell death. Likewise, temozolomide (TMZ), a chemotherapeutic agent used for the treatment of malignant gliomas, also triggers autophagic cell death. In this study, we examined the potential to combine the two treatments in the setting of experimental glioma. In vitro, pretreatment with TMZ followed by CRAd-Surivin-pk7 enhanced cytotoxicity against a panel of glioma cell lines. Western blot analysis showed increased expression of BAX and p53, decreased expression of BCL2 and elevated level of APG5. Treatment with TMZ followed by CRAd-Survivin-pk7 (CRAd-S-pk7) led to a significant over-expression of autophagy markers, acidic vesicular organelles and light-chain 3 (LC3). These results were further evaluated in vivo, in which 90% of the mice with intracranial tumours were long-term survivors (>100 days) after treatment with TMZ and CRAd-S-pk7 (P<0.01). Analysis of tumours ex vivo showed expression of both LC3 and cleaved Caspase-3, proving that both autophagy and apoptosis are responsible for cell death in vivo. These results suggest that combination of chemovirotherapy offers a powerful tool against malignant glioma and should be further explored in the clinical setting.
Assuntos
Adenoviridae/fisiologia , Antineoplásicos Alquilantes/uso terapêutico , Apoptose , Autofagia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioma/terapia , Terapia Viral Oncolítica , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Dacarbazina/uso terapêutico , Feminino , Glioma/mortalidade , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Associadas aos Microtúbulos/análise , Mitose , Temozolomida , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
Conditionally replicative adenoviruses (CRAds) are often evaluated in mice; however, normal and cancerous mouse tissues are poorly permissive for human CRAds. As the cotton rat (CR) is a semipermissive animal and the Syrian hamster (SH) is a fully permissive model for adenoviral replication, we compared them in a single study following intracranial (i.c.) injection of a novel glioma-targeting CRAd. Viral genomic copies were quantified by real-time PCR in brain, blood, liver and lung. The studies were corroborated by immunohistochemical, serological and immunological assays. CR had a multiple log higher susceptibility for adenoviral infection than SH. A similar amount of genomic copies of CRAd-Survivin-pk7 and human adenovirus serotype 5 (AdWT) was found in the brain of CR and in all organs from SH. In blood and lung of CR, AdWT had more genomic copies than CRAd-Survivin-pk7 in some of the time points studied. Viral antigens were confirmed in brain slices, an elevation of serum transaminases was observed in both models, and an increase in anti-adenoviral antibodies was detected in SH sera. In conclusion, CR represents a sensitive model for studying biodistribution of CRAds after i.c. delivery, allowing for the detection of differences in the replication of CRAd-Survivin-pk7 and AdWT that were not evident in SH.
Assuntos
Adenoviridae/fisiologia , Vetores Genéticos , Vírus Oncolíticos/fisiologia , Replicação Viral , Adenoviridae/genética , Animais , Formação de Anticorpos/efeitos dos fármacos , Encéfalo/virologia , Neoplasias do Sistema Nervoso Central/terapia , Cricetinae , DNA Viral/sangue , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Glioma/terapia , Humanos , Fígado/virologia , Pulmão/virologia , Masculino , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Ratos , Especificidade da Espécie , Transaminases/sangueRESUMO
Adenoviral oncolytic virotherapy represents an attractive treatment modality for central nervous system (CNS) neoplasms. However, successful application of virotherapy in clinical trials has been hampered by inadequate distribution of oncolytic vectors. Neural stem cells (NSCs) have been shown as suitable vehicles for gene delivery because they track tumor foci. In this study, we evaluated the capability of NSCs to deliver a conditionally replicating adenovirus (CRAd) to glioma. We examined NSC specificity with respect to viral transduction, migration and capacity to deliver a CRAd to tumor cells. Fluorescence-activated cell sorter (FACS) analysis of NSC shows that these cells express a variety of surface receptors that make them amenable to entry by recombinant adenoviruses. Luciferase assays with replication-deficient vectors possessing a variety of transductional modifications targeted to these receptors confirm these results. Real-time PCR analysis of the replication profiles of different CRAds in NSCs and a representative glioma cell line, U87MG, identified the CRAd-Survivin (S)-pk7 virus as optimal vector for further delivery studies. Using in vitro and in vivo migration studies, we show that NSCs infected with CRAd-S-pk7 virus migrate and preferentially deliver CRAd to U87MG glioma. These results suggest that NSCs mediate an enhanced intratumoral distribution of an oncolytic vector in malignant glioma when compared with virus injection alone.
Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Neurônios/virologia , Terapia Viral Oncolítica/métodos , Transplante de Células-Tronco/métodos , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Neoplasias Encefálicas/virologia , Movimento Celular/fisiologia , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/farmacocinética , Glioma/virologia , Humanos , Proteínas Inibidoras de Apoptose , Masculino , Camundongos , Camundongos Nus , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Transplante de Neoplasias , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , RNA Mensageiro/genética , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Células-Tronco/virologia , Survivina , Transcrição Gênica , Células Tumorais Cultivadas , Replicação ViralRESUMO
Malignant glioma continues to be a major target for gene therapy and virotherapy due to its aggressive growth and the current lack of effective treatment. However, these approaches have been hampered by inefficient infection of glioma cells by viral vectors,particularly vectors derived from serotype 5 adenoviruses (Ad5). This results from limited cell surface expression of the primary adenovirus receptor, coxsackie-adenovirus-receptor (CAR), on tumor cells. To circumvent this problem, Ad fiber pseudotyping,the genetic replacement of either the entire fiber or fiber knob domain with its structural counterpart from another human Ad serotype that recognizes a cellular receptor other than CAR, has been shown to enhance Ad infectivity in a variety of tumor types,including human glioma. Here, we have extended the paradigm of genetic pseudotyping to include fiber domains from non-human or"xenotype" Ads for infectivity enhancement of human glioma cell populations. In this study, we evaluated the gene transfer efficiency of a panel of Ad vectors which express one of five different "xenotype"fiber knob domains, including those derived from murine,ovine, porcine and canine species, in both human glioma cell lines as well as primary glioma tumor cells from patients. Adenovirus vectors displaying either canine Ad or porcine Ad fiber elements had the highest gene transfer to both glioma cell lines and primary tumor cells. The correlation between the viral infectivity of modified adenovirus vectors and expression of human CAR and CD46(an adenovirus type B receptor) on the surfaces of tumor cells was also analyzed. Taken together, human adenovirus vectors modified with "xenotype" fiber elements could be excellent candidates to target human glioma.
Assuntos
Adenoviridae/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Animais , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Citomegalovirus/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/metabolismo , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Vírus/metabolismoRESUMO
Low pressure hydrocephalus (LPH) is a rare clinical condition. We report our experience with 10 patients treated at the Johns Hopkins Hospital. We reviewed the records of 10 patients (five men, five women; mean age 43 years) treated between 1996 and 2000. All underwent intracranial pressure (ICP) monitoring and subatmospheric cerebrospinal fluid (CSF) drainage with an intraventricular or lumbar catheter. All patients developed ventriculomegaly: five following aneurysmal subarachnoid haemorrhage; one after meningitis; one after intraventricular haemorrhage. Three patients presented with chronic aqueductal stenosis. Ventriculomegaly was clinically detected on average 12 days after presentation. Mean ICP was 4.8 mmHg (range 0-10). All patients improved only in the setting of negative pressure CSF drainage, and were subsequently treated with low pressure ventriculo- or lumboperitoneal shunts. At 1 year, eight patients (80%) showed good recovery to minimal disability; seven patients (70%) had resolving ventriculomegaly. The mechanism of low pressure hydrocephalus remains unclear. In our cohort, different aetiologies were responsible for the change in compliance/elastance of the brain parenchyma and subsequent development of ventriculomegaly. We propose that while ventriculomegaly (and therefore neuronal dysfunction) can be initiated in the setting of high ICP, the maintenance of ventriculomegaly at normal or low ICP is a physiological example of hysteresis. This behaviour, which has been characterized by the chaos theory of non-linear dynamics as a Hopf bifurcation, explains how a system can exhibit two different states (ventricular size) at a single parameter value (ICP). Most importantly, it helps to explain how lowering ICP in the setting of LPH can resolve ventriculomegaly and its neurologic sequelae.
Assuntos
Ventrículos Cerebrais/patologia , Derivações do Líquido Cefalorraquidiano/métodos , Hidrocefalia/cirurgia , Complicações Pós-Operatórias/etiologia , Adulto , Idoso , Estudos de Coortes , Feminino , Humanos , Hidrocefalia/fisiopatologia , Pressão Intraocular/fisiologia , Masculino , Meningite/etiologia , Pessoa de Meia-Idade , Hemorragia Pós-Operatória/etiologia , Estudos Retrospectivos , Hemorragia Subaracnóidea/etiologia , Fatores de Tempo , Tomografia Computadorizada por Raios X , Resultado do TratamentoRESUMO
Contemporary treatment of malignant brain tumors has been hampered by problems with drug delivery to the tumor bed. Inherent boundaries of the central nervous system, such as the blood-brain barrier or the blood-cerebrospinal fluid barrier, and a general lack of response to many chemotherapeutic agents have led to alternative treatment modalities. In general, all these modalities have sought to either disrupt or bypass the physiologic brain barriers and deliver the drug directly to the tumor. This article reviews past, as well as current, methods of drug delivery to tumors of the central nervous system. Special emphasis is placed on biodegradable polymers that can release chemotherapeutic agents against malignant gliomas. A variety of other nonchemotherapeutic drugs, including antiangiogenesis and immunotherapeutic agents, are presented in the context of new polymer technology. Finally, future directions in drug delivery are discussed with an overview on new advances in emerging biotechnology.
Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Biodegradação Ambiental , Barreira Hematoencefálica , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Carmustina/administração & dosagem , Carmustina/farmacocinética , Carmustina/uso terapêutico , Ensaios Clínicos como Assunto , Terapia Combinada , Método Duplo-Cego , Portadores de Fármacos , Implantes de Medicamento , Previsões , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Glioma/tratamento farmacológico , Glioma/mortalidade , Glioma/terapia , Humanos , Injeções Intralesionais , Injeções Espinhais , Tábuas de Vida , Compostos de Nitrosoureia/administração & dosagem , Compostos de Nitrosoureia/farmacocinética , Compostos de Nitrosoureia/uso terapêutico , Polímeros , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Pró-Fármacos/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Análise de Sobrevida , Resultado do TratamentoRESUMO
OBJECT: Local delivery of cytokines has been shown to have a potent antitumor activity against a wide range of malignant brain tumors. In this study, the authors examined the efficacy of treating central nervous system (CNS) tumors by transfecting poorly immunogenic B16/F10 melanoma cells with interleukin (IL)-2, IL-4, or granulocytemacrophage-colony stimulating factor (GM-CSF) gene, and using these cells to deliver the cytokine locally at the site of the CNS tumor. The object was to determine which cytokine would possess the greatest antitumor activity and to further elucidate its mechanism of action. METHODS: The transfected B16/F10 cells were irradiated to prevent replication and injected intracranially into C57BL/6 mice (10 mice per group) along with nonirradiated, nontransfected B16/F10 (wild-type) melanoma cells. Sixty percent of mice treated with IL-2 (p < 0.001 compared with control) and 10% treated with IL-4 (median survival = 31 days, p < 0.001 compared with control) were long term survivors (> 120 days). The median survival for animals treated with GM-CSF was 22 days with no long term survivors (p = 0.01 compared with control). Control animals that received only wild-type cells had a median survival of 18 days (range 15-20 days). Histopathological examination of brains from animals killed at different times showed minimal infiltration of tumor cells in the IL-2 group, moderate infiltration of tumor cells in the IL-4 group, and gross tumor invasion and tissue necrosis in the GM-CSF group. Animals treated with IL-2 showed a strong CD8 T cell-mediated response, whereas IL-4 evoked a prominent eosinophilic infiltrate in the area of the tumor. CONCLUSIONS: High levels of locally expressed IL-2 rather than IL-4 or GM-CSF stimulate a strong immunological cytotoxic antitumor response that leads to significant prolongation of survival in mice challenged with B16/F10 intracranial melanoma tumor cells. Consequently, IL-2 may be a superior candidate for use in paracrine immunotherapy.