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











Base de dados
Intervalo de ano de publicação
2.
Mol Ther ; 32(9): 3114-3127, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-38910324

RESUMO

T cell-focused cancer immunotherapy including checkpoint inhibitors and cell therapies has been rapidly evolving over the past decade. Nevertheless, there remains a major unmet medical need in oncology generally and immuno-oncology specifically. We have constructed an oncolytic adenovirus, Ad5/3-E2F-d24-aMUC1aCD3-IL-2 (TILT-322), which is armed with a human aMUC1aCD3 T cell engager and IL-2. TILT-322 treatment stimulated T cell cytotoxicity through the increased presence of granzyme B, perforin, and interferon-gamma. Additional immune profiling indicated TILT-322 increased gamma delta T cell activation and impacted other cell types such as natural killer cells and natural killer-like T cells that are traditionally involved in cancer immunotherapy. TILT-322 treatment also decreased the proportion of exhausted CD8+ T cells as demarked by immune checkpoint expression in ovarian ascites samples. Overall, our data showed that TILT-322 treatment led to an enhanced T cell activation and reversed T cell exhaustion translating into high antitumor efficacy when given locally or intravenously. The analysis of blood and tumors isolated from an in vivo patient-derived ovarian cancer xenograft model suggested TILT-322 mediated tumor control through improved T cell functions. Therefore, TILT-322 is a promising novel anti-tumor agent for clinical translation.


Assuntos
Adenoviridae , Anticorpos Biespecíficos , Ascite , Interleucina-2 , Mucina-1 , Neoplasias Ovarianas , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Neoplasias Ovarianas/terapia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/genética , Feminino , Animais , Adenoviridae/genética , Camundongos , Linhagem Celular Tumoral , Ascite/terapia , Ascite/imunologia , Interleucina-2/metabolismo , Mucina-1/genética , Mucina-1/imunologia , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Modelos Animais de Doenças , Imunoterapia/métodos , Exaustão das Células T
3.
Clin Cancer Res ; 30(17): 3715-3725, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-38546220

RESUMO

PURPOSE: TILT-123 (igrelimogene litadenorepvec) is an oncolytic adenovirus armed with TNFa and IL2, designed to induce T-cell infiltration and cytotoxicity in solid tumors. PATIENTS AND METHODS: TUNIMO (NCT04695327) was a single-arm, multicenter phase I dose-escalation trial designed to assess the safety of TILT-123 in advanced solid cancers refractory to standard therapy. Patients received intravenous and intratumoral TILT-123. The primary endpoint was safety by adverse events (AE), laboratory values, vital signs, and electrocardiograms. Secondary endpoints included tumor response, pharmacokinetics, and predictive biomarkers. RESULTS: Twenty patients were enrolled, with a median age of 58 years. Most prevalent cancer types included sarcomas (35%), melanomas (15%) and ovarian cancers (15%). No dose-limiting toxicities were observed. The most frequent treatment-related AEs included fever (16.7%), chills (13.0%), and fatigue (9.3%). Ten patients were evaluable for response on day 78 with RECIST 1.1, iRECIST or PET-based evaluation. The disease control rate by PET was 6/10 (60% of evaluable patients) and 2/10 by RECIST 1.1 and iRECIST(20%of evaluable patients). Tumor size reductions occurred in both injected and non-injected lesions. TILT-123 was detected in injected and non-injected tumors, and virus was observed in blood after intravenous and intratumoral injections. Treatment resulted in reduction of lymphocytes in blood, with concurrent lymphocyte increases in tumors, findings compatible with trafficking. CONCLUSIONS: TILT-123 was safe and able to produce antitumor effects in local and distant lesions in heavily pre-treated patients. Good tolerability of TILT-123 facilitates combination studies, several of which are ongoing (NCT04217473, NCT05271318, NCT05222932, and NCT06125197). See related commentary by Silva-Pilipich and Smerdou, p. 3649.


Assuntos
Adenoviridae , Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Linfócitos T , Humanos , Feminino , Pessoa de Meia-Idade , Masculino , Neoplasias/terapia , Neoplasias/patologia , Vírus Oncolíticos/genética , Terapia Viral Oncolítica/métodos , Terapia Viral Oncolítica/efeitos adversos , Idoso , Linfócitos T/imunologia , Adenoviridae/genética , Adulto , Resultado do Tratamento , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos
4.
Oncoimmunology ; 12(1): 2241710, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37546696

RESUMO

Lung cancer remains among the most difficult-to-treat malignancies and is the leading cause of cancer-related deaths worldwide. The introduction of targeted therapies and checkpoint inhibitors has improved treatment outcomes; however, most patients with advanced-stage non-small cell lung cancer (NSCLC) eventually fail these therapies. Therefore, there is a major unmet clinical need for checkpoint refractory/resistant NSCLC. Here, we tested the combination of aPD-1 and adenovirus armed with TNFα and IL-2 (Ad5-CMV-mTNFα/mIL-2) in an immunocompetent murine NSCLC model. Moreover, although local delivery has been standard for virotherapy, treatment was administered intravenously to facilitate clinical translation and putative routine use. We showed that treatment of tumor-bearing animals with aPD-1 in combination with intravenously injected armed adenovirus significantly decreased cancer growth, even in the presence of neutralizing antibodies. We observed an increased frequency of cytotoxic tumor-infiltrating lymphocytes, including tumor-specific cells. Combination treatment led to a decreased percentage of immunosuppressive tumor-associated macrophages and an improvement in dendritic cell maturation. Moreover, we observed expansion of the tumor-specific memory T cell compartment in secondary lymphoid organs in the group that received aPD-1 with the virus. However, although the non-replicative Ad5-CMV-mTNFα/mIL-2 virus allows high transgene expression in the murine model, it does not fully reflect the clinical outcome in humans. Thus, we complemented our findings using NSCLC ex vivo models fully permissive for the TNFα and IL-2- armed oncolytic adenovirus TILT-123. Overall, our data demonstrate the ability of systemically administered adenovirus armed with TNFα and IL-2 to potentiate the anti-tumor efficacy of aPD-1 and warrant further investigation in clinical trials.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Interleucina-2 , Neoplasias Pulmonares , Fator de Necrose Tumoral alfa , Animais , Humanos , Camundongos , Adenoviridae/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Interleucina-2/genética , Interleucina-2/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/uso terapêutico , Inibidores de Checkpoint Imunológico
5.
Front Immunol ; 14: 1171083, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37475863

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a highly treatment-resistant cancer. Currently, the only curative treatment for PDAC is surgery, but most patients are diagnosed with metastatic disease and thus outside the scope of surgery. The majority of metastatic patients receive chemotherapy, but responses are limited. New therapeutics are thus urgently needed for PDAC. One major limitation in treating PDAC has been the highly immunosuppressive tumor microenvironment (TME) which inhibits anti-cancer immune responses. We have constructed an oncolytic adenovirus coding for a variant the interleukin 2 molecule, Ad5/3-E2F-d24-vIL2 (also known as TILT-452, and "vIL-2 virus"), with preferential binding to IL-2 receptors on the surface of effector lymphocytes over T regulatory cells (T regs). In the present study this virus was evaluated in combination with nab-paclitaxel and gemcitabine chemotherapy in Panc02 mouse model. Ad5/3-E2F-d24-vIL2 showed marked PDAC cell killing in vitro, alongside induction of mitotic slippage and immunogenic cell death in PDAC cell lines, when combined with chemotherapy. Increased survival was seen in vivo with 80% of animals surviving long term, when compared to chemotherapy alone. Moreover, combination therapy mediated enhanced tumor growth control, without observable toxicities in internal organs or external features. Survival and tumor control benefits were associated with activation of tumor infiltrating immune cells, downregulation of inhibitory signals, change in fibroblast populations in the tumors and changes in intratumoral cytokines, with increased chemokine amounts (CCL2, CCL3, CCL4) and anti-tumor cytokines (IFN-γ and TNFα). Furthermore, vIL-2 virus in combination with chemotherapy efficiently induced tumor protection upon rechallenge, that was extended to a previously non-encountered cancer cell line. In conclusion, Ad5/3-E2F-d24-vIL2 is a promising immunotherapy candidate when combined with nab-paclitaxel and gemcitabine.


Assuntos
Infecções por Adenoviridae , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Camundongos , Adenoviridae , Citocinas/uso terapêutico , Interleucina-2/genética , Interleucina-2/farmacologia , Interleucina-2/uso terapêutico , Linhagem Celular Tumoral , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/tratamento farmacológico , Gencitabina , Linfócitos/patologia , Fibroblastos/patologia , Microambiente Tumoral , Neoplasias Pancreáticas
6.
Front Immunol ; 14: 1060540, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36817448

RESUMO

Introduction: Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer, but preclinical testing of hypotheses such as combination therapies has been complicated, in part due to species incompatibility issues. For example, one of few known permissive animal models for oncolytic adenoviruses is the Syrian hamster, for which an ICI, mainly an anti-PD-L1 monoclonal antibody (mAb) was not previously available. In this study, we developed an anti-Syrian hamster PD-L1 mAb to enable the evaluation of safety and efficacy, when combining anti-PD-L1 with an oncolytic adenovirus encoding tumour necrosis factor alpha (TNFα) and interleukin-2 (IL-2) (Ad5/3-E2F-D24-hTNFα-IRES-hIL-2 or TILT-123). Methods: Recombinant Syrian hamster PD-L1 was expressed and mice immunized for mAb formation using hybridoma technology. Clonal selection through binding and functional studies in vitro, in silico and in vivo identified anti-PD-L1 clone 11B12-1 as the primary mAb candidate for immunotherapy modelling. The oncolytic virus (OV) and ICI combination approach was then evaluated using 11B12-1 and TILT-123 in a Syrian hamster model of pancreatic ductal adenocarcinoma (PDAC). Results: Supernatants from hybridoma parent subclone 11B12B4 provided the highest positive PD-L1 signal, on Syrian hamster PBMCs and three cancer cell lines (HT100, HapT1 and HCPC1). In vitro co-cultures revealed superior immune modulated profiles of cell line matched HT100 tumour infiltrating lymphocytes when using subclones of 7G2, 11B12 and 12F1. Epitope binning and epitope prediction using AlphaFold2 and ColabFold revealed two distinct functional epitopes for clone 11B12-1 and 12F1-1. Treatment of Syrian hamsters bearing HapT1 tumours, with 11B12-1 induced significantly better (p<0.05) tumour growth control than isotype control by day 12. 12F1-1 did not induce significant tumour growth control. The combination of 11B12-1 with oncolytic adenovirus TILT-123 improved tumour growth control further, when compared to monotherapy (p<0.05) by day 26. Conclusions: Novel Syrian hamster anti-PD-L1 clone 11B12-1 induces tumour growth control in a hamster model of PDAC. Combining 11B12-1 with oncolytic adenovirus TILT-123 improves tumour growth control further and demonstrates good safety and toxicity profiles.


Assuntos
Carcinoma Ductal Pancreático , Vírus Oncolíticos , Neoplasias Pancreáticas , Cricetinae , Animais , Camundongos , Mesocricetus , Inibidores de Checkpoint Imunológico , Adenoviridae , Neoplasias Pancreáticas/terapia , Imunoterapia , Anticorpos Monoclonais , Replicação Viral , Neoplasias Pancreáticas
7.
Mol Ther Oncolytics ; 28: 59-73, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36699617

RESUMO

Immunotherapy with bispecific T cell engagers has shown efficacy in patients with hematologic malignancies and uveal melanoma. Antitumor effects of bispecific T cell engagers in most solid tumors are limited due to their short serum half-life and insufficient tumor concentration. We designed a novel serotype 5/3 oncolytic adenovirus encoding a human mucin1 antibody and the human CD3 receptor, Ad5/3-E2F-d24-aMUC1aCD3 (TILT-321). TILT-321 is engineered to replicate only in cancer cells, leading to a high concentration of the aMUC1aCD3 molecule in the tumor microenvironment. Infection and cell viability assays were performed to determine the oncolytic potential of the novel construct. The functionality of the virus-derived aMUC1aCD3 was evaluated in vitro. When TILT-321 was combined with allogeneic T cells, rapid tumor cell lysis was observed. TILT-321-infected cells secreted functional aMUC1aCD3, as shown by increased T cell activity and its binding to MUC1 and CD3. In vivo, TILT-321 treatment led to effective antitumor efficacy mediated by increased intratumoral T cell activity in an A549 and patient-derived ovarian cancer xenograft mouse model humanized with peripheral blood mononuclear cells (PBMC). This study provides a proof of concept for an effective strategy to overcome the key limitations of recombinant bispecific T cell engager delivery for solid tumor treatment.

8.
Oncoimmunology ; 11(1): 2096572, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35845722

RESUMO

Cytokines have proven to be effective for cancer therapy, however whilst low-dose monotherapy with cytokines provides limited therapeutic benefit, high-dose treatment can lead to a number of adverse events. Interleukin 7 has shown promising results in clinical trials, but anti-cancer effect was limited, in part due to a low concentration of the cytokine within the tumor. We hypothesized that arming an oncolytic adenovirus with Interleukin 7, enabling high expression localized to the tumor microenvironment, would overcome systemic delivery issues and improve therapeutic efficacy. We evaluated the effects of Ad5/3-E2F-d24-hIL7 (TILT-517) on tumor growth, immune cell activation and cytokine profiles in the tumor microenvironment using three clinically relevant animal models and ex vivo tumor cultures. Our data showed that local treatment of tumor bearing animals with Ad5/3- E2F-d24-hIL7 significantly decreased cancer growth and increased frequency of tumor-infiltrating cells. Ad5/3-E2F-d24-hIL7 promoted notable upregulation of pro-inflammatory cytokines, and concomitant activation and migration of CD4+ and CD8 + T cells. Interleukin 7 expression within the tumor was positively correlated with increased number of cytotoxic CD4+ cells and IFNg-producing CD4+ and CD8+ cells. These findings offer an approach to overcome the current limitations of conventional IL7 therapy and could therefore be translated to the clinic.


Assuntos
Infecções por Adenoviridae , Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Citocinas , Interleucina-7 , Linfócitos do Interstício Tumoral , Terapia Viral Oncolítica/métodos
9.
Front Immunol ; 13: 794251, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35355980

RESUMO

Immune checkpoint inhibitors (ICI) have provided significant improvement in clinical outcomes for some patients with solid tumors. However, for patients with head and neck cancer, the response rate to ICI monotherapy remains low, leading to the exploration of combinatorial treatment strategies. In this preclinical study, we use an oncolytic adenovirus (Ad5/3) encoding hTNFα and hIL-2 and non-replicate adenoviruses (Ad5) encoding mTNFα and mIL-2 with ICI to achieve superior tumor growth control and improved survival outcomes. The in vitro effect of Ad5/3-E2F-D24-hTNFa-IRES-hIL-2 was characterized through analyses of virus replication, transgene expression and lytic activity using head and neck cancer patient derived cell lines. Mouse models of ICI naïve and refractory oral cavity squamous cell carcinoma were established to evaluate the local and systemic anti-tumor immune response upon ICI treatment with or without the non-replicative adenovirus encoding mTNFα and mIL-2. We delineated the mechanism of action by measuring the metabolic activity and effector function of CD3+ tumor infiltrating lymphocytes (TIL) and transcriptomic profile of the CD45+ tumor immune compartment. Ad5/3-E2F-D24-hTNFa-IRES-hIL-2 demonstrated robust replicative capability in vitro across all head and neck cell lines screened through potent lytic activity, E1a and transgene expression. In vivo, in both ICI naïve and refractory models, we observed improvement to tumor growth control and long-term survival when combining anti-PD-1 or anti-PD-L1 with the non-replicative adenovirus encoding mTNFα and mIL-2 compared to monotherapies. This observation was verified by striking CD3+ TIL derived mGranzyme b and interferon gamma production complemented by increased T cell bioenergetics. Notably, interrogation of the tumor immune transcriptome revealed the upregulation of a gene signature distinctive of tertiary lymphoid structure formation upon treatment of murine anti-PD-L1 refractory tumors with non-replicative adenovirus encoding mTNFα and mIL-2. In addition, we detected an increase in anti-tumor antibody production and expansion of the memory T cell compartment in the secondary lymphoid organs. In summary, a non-replicative adenovirus encoding mTNFα and mIL-2 potentiates ICI therapy, demonstrated by improved tumor growth control and survival in head and neck tumor-bearing mice. Moreover, the data reveals a potential approach for inducing tertiary lymphoid structure formation. Altogether our results support the clinical potential of combining this adenovirotherapy with anti-PD-1 or anti-PD-L1.


Assuntos
Neoplasias de Cabeça e Pescoço , Terapia Viral Oncolítica , Estruturas Linfoides Terciárias , Adenoviridae/genética , Animais , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Interleucina-2/genética , Camundongos , Terapia Viral Oncolítica/métodos , Fator de Necrose Tumoral alfa/genética
10.
Oncoimmunology ; 11(1): 2028960, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35083096

RESUMO

Intratumoral immunotherapies are entering clinical use but concerns remain regarding their effects on non-injected tumors. Here, we studied the impact of local treatment with an adenovirus coding for TNFa and IL-2 on systemic antitumor response in animals receiving aPD-1 (anti-programmed cell death protein 1) therapy. Using bilateral murine melanoma models, we tested systemic tumor response to combined therapy with anti-PD-1 and an adenovirus coding for TNFa and IL-2 ("virus"). Virus was given intratumorally (to one of the two tumors only) and aPD-1 monoclonal antibody systemically. We evaluated both tumors' response to treatment, overall survival, metastasis development, and immunological mechanisms involved with response. Consistent tumor control was observed in both injected and non-injected tumors, including complete response in all treated animals receiving aPD-1+ virus therapy. Mechanistically, virus injections enabled potent effector lymphocyte response locally, with systemic effects in non-injected tumors facilitated by aPD-1 treatment. Moreover, adenovirus therapy demonstrated immunological memory formation. Virus therapy was effective in preventing metastasis development. Local treatment with TNFa and IL-2 coding adenovirus enhanced systemic response to aPD-1 therapy, by re-shaping the microenvironment of both injected and non-injected tumors. Therefore, our pre-clinical data support the rationale for a trial utilizing a combination of aPD-1 plus virus for the treatment of human cancer.


Assuntos
Infecções por Adenoviridae , Melanoma , Terapia Viral Oncolítica , Adenoviridae/genética , Animais , Imunoterapia , Interleucina-2 , Melanoma/terapia , Camundongos , Microambiente Tumoral
11.
Mol Ther Oncolytics ; 17: 47-60, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32322662

RESUMO

Despite some promising results, the majority of patients do not benefit from T cell therapies, as tumors prevent T cells from entering the tumor, shut down their activity, or downregulate key antigens. Due to their nature and mechanism of action, oncolytic viruses have features that can help overcome many of the barriers currently facing T cell therapies of solid tumors. This study aims to understand how four different oncolytic viruses (adenovirus, vaccinia virus, herpes simplex virus, and reovirus) perform in that task. For that purpose, an immunocompetent in vivo tumor model featuring adoptive tumor-infiltrating lymphocyte (TIL) therapy was used. Tumor growth control (p < 0.001) and survival analyses suggest that adenovirus was most effective in enabling T cell therapy. The complete response rate was 62% for TILs + adenovirus versus 17.5% for TILs + PBS. Of note, TIL biodistribution did not explain efficacy differences between viruses. Instead, immunostimulatory shifts in the tumor microenvironment mirrored efficacy results. Overall, the use of oncolytic viruses can improve the utility of T cell therapies, and additional virus engineering by arming with transgenes can provide further antitumor effects. This phenomenon was seen when an unarmed oncolytic adenovirus was compared to Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123). A clinical trial is ongoing, where patients receiving TIL treatment also receive TILT-123 (ClinicalTrials.gov: NCT04217473).

12.
Water Res ; 35(5): 1240-4, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11268844

RESUMO

Sewage effluent discharged to surface water has been shown to contain human hormones, particularly oestrogens, and synthetic chemicals which may be able to disrupt the endocrine system. Since many surface waters which receive sewage effluent are subsequently used as drinking water sources, it is important to demonstrate that treated drinking water is not contaminated. Oestrogenic activity in rivers and drinking water in the region of Severn Trent Water was studied using a combination of bioassay, to integrate exposure over time, and advanced chemical analysis. There was little or no evidence of substances that were oestrogenic, even in waters receiving significant amounts of sewage effluent. Oestrogenic activity, as measured in the rainbow trout vitellogenin assay, was seen at the Tame/Trent confluence but this activity was relatively weak. There was no activity detected at raw water intakes and no hormones or substances that are oestrogenic were detected in the final drinking water.


Assuntos
Estrogênios/análise , Água Doce/análise , Esgotos , Purificação da Água , Abastecimento de Água , Animais , Bioensaio , Inglaterra , Ensaio de Imunoadsorção Enzimática/métodos , Estradiol/análise , Estrona/análise , Etinilestradiol/análise , Cromatografia Gasosa-Espectrometria de Massas/métodos , Humanos , Oncorhynchus mykiss , Radioimunoensaio/métodos , Sensibilidade e Especificidade , Esterilização/métodos , Ultrafiltração/métodos , Poluentes Químicos da Água/análise
13.
Expert Opin Investig Drugs ; 9(5): 1009-20, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-11060723

RESUMO

Ribozymes are RNA molecules that possess the ability to cleave and thus destroy other RNA molecules. As a result of this ability, they are ideal specific agents to use against the messenger RNAs of important genes found to be linked with disease (of cellular and viral origin). This review will briefly describe the different types of ribozyme and the potential they have as therapeutic compounds against viruses, oncogenes and drug resistance in haematological settings. The latest news from the various Phase I and II ribozyme clinical trials is discussed, as is the potential for the ribozymes' future as therapeutic agents.


Assuntos
Doenças Hematológicas/tratamento farmacológico , RNA Catalítico/uso terapêutico , Animais , Fármacos Anti-HIV/uso terapêutico , Humanos , RNA Catalítico/química
14.
J Leukoc Biol ; 66(3): 361-8, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10496304

RESUMO

With the identification and increasing understanding of the genes involved in neoplastic transformation has come the realization that abrogation of these genes' products may lead to cell death or a return to normalcy. The use of ribozymes and their nucleic acid cousins, antisense oligodeoxynucleotides (ODNs), are two such ways of perturbing the disease-related gene expression. This review will look at the development and application of ribozymes to abrogate gene expression, with particular relevance to hematological settings. Some examples of antisense ODNs will also be mentioned where appropriate.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Doenças Hematológicas/terapia , RNA Catalítico/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Doenças Hematológicas/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Humanos , Doenças do Sistema Imunitário/terapia , Conformação de Ácido Nucleico , Oligonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas de Fusão Oncogênica/genética , Oncogenes/genética , RNA Catalítico/química , RNA Catalítico/farmacocinética , RNA Catalítico/farmacologia , Viroses/terapia , Replicação Viral/efeitos dos fármacos
15.
Blood ; 91(2): 371-82, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9427689

RESUMO

Ribozymes are catalytic RNA molecules that recognize their target RNA in a highly sequence-specific manner. They can therefore be used to inhibit deleterious gene expression (by cleavage of the target mRNA) or even repair mutant cellular RNAs. Targets such as the mRNAs of oncogenes (resulting from base mutations or chromosome translocations, eg, ras or bcr-abl) and viral genomes and transcripts (human immunodeficiency virus-type 1 [HIV-1]) are ideal targets for such sequence-specific agents. The aim of this review is therefore to introduce the different classes of ribozymes, highlighting some of the chemistry of the reactions they catalyze, to address the specific inhibition of genes by ribozymes, the problems yet to be resolved, and how new developments in the field give hope to the future for ribozymes in the therapeutic field.


Assuntos
Terapia Genética , RNA Catalítico/uso terapêutico , Regulação da Expressão Gênica , Humanos , RNA Catalítico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA