Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Natl Compr Canc Netw ; 22(2)2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38364363

RESUMO

More than 90 distinct fusion partners of ALK rearrangement have been identified. Different ALK fusions may exhibit different sensitivities to ALK tyrosine kinase inhibitors. The emergence of rare fusions poses significant challenges to targeted therapies. This study aimed to investigate the response of KANK1::ALK fusion to alectinib in an advanced lung adenocarcinoma. A novel KANK1::ALK fusion was identified by next-generation sequencing (NGS) and Ventana immunohistochemistry assessments. A 73-year-old woman who had never smoked was admitted with hemoptysis in May 2020. PET/CT revealed a nodule in the left upper lobe, with bilateral pulmonary and multiple lymph node metastases. The upper lobe nodule of the left lung was diagnosed as adenocarcinoma through bronchofiberscopy biopsy, resulting in a clinical diagnosis of stage IVA (cT1c,N3,M1a). Because the biopsy tissue was insufficient for NGS analysis, a blood-based genetic analysis was performed, revealing the presence of KRAS p.Q61R mutations. The patient received carboplatin and pemetrexed with pembrolizumab as first-line therapy, followed by maintenance therapy of pembrolizumab monotherapy. Although the tumor initially showed significant shrinkage, it unfortunately progressed further after 11 months. Subsequently, the patient was given carboplatin and pemetrexed with pembrolizumab again, but the tumor progression continued. An NGS using a rebiopsy of the left upper lobe tumor suggested a KANK1::ALK fusion. Alectinib was prescribed in January 2022, and a durable partial response was observed after 18 months. ALK rearrangements were observed in the broader spectrum of lung cancers. This study provided a potential treatment option for patients with KANK1::ALK fusions. Further studies are needed to understand the function of these fusions.


Assuntos
Adenocarcinoma de Pulmão , Carbazóis , Neoplasias Pulmonares , Piperidinas , Feminino , Humanos , Idoso , Pemetrexede , Carboplatina/uso terapêutico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Quinase do Linfoma Anaplásico/genética , Inibidores de Proteínas Quinases/uso terapêutico , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas do Citoesqueleto/uso terapêutico , Proteínas Adaptadoras de Transdução de Sinal/uso terapêutico
2.
Phytomedicine ; 116: 154895, 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37229890

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is a major cause of cancer-associated mortality worldwide. Myosin-9's role in HCC and the anti-HCC effect of the drugs targeting Myosin-9 remain poorly understood so far. Candidate antitumor agents obtained from natural products have attracted worldwide attention. Usenamine A is a novel product, which was first extracted in our laboratory from the lichen Usnea longissima. According to published reports, usenamine A exhibits good antitumor activity, while the mechanisms underlying its antitumor effects remain to be elucidated. PURPOSE: The present study investigated the anti-hepatoma effect of usenamine A and the underlying molecular mechanisms, along with evaluating the therapeutic potential of targeting Myosin-9 in HCC. METHODS: The CCK-8, Hoechst staining, and FACS assays were conducted in the present study to investigate how usenamine A affected the growth and apoptosis of human hepatoma cells. Moreover, TEM, acridine orange staining, and immunofluorescence assay were performed to explore the induction of autophagy by usenamine A in human hepatoma cells. The usenamine A-mediated regulation of protein expression in human hepatoma cells was analyzed using immunoblotting. MS analysis, SPR assay, CETSA, and molecular modeling were performed to identify the direct target of usenamine A. Immunofluorescence assay and co-immunoprecipitation assay were conducted to determine whether usenamine A affected the interaction between Myosin-9 and the actin present in human hepatoma cells. In addition, the anti-hepatoma effect of usenamine A was investigated in vivo using a xenograft tumor model and the IHC analysis. RESULTS: The present study initially revealed that usenamine A could suppress the proliferation of HepG2 and SK-HEP-1 cells (hepatoma cell lines). Furthermore, usenamine A induced cell apoptosis via the activation of caspase-3. In addition, usenamine A enhanced autophagy. Moreover, usenamine A administration could dramatically suppress the carcinogenic ability of HepG2 cells, as evidenced by the nude mouse xenograft tumor model. Importantly, it was initially revealed that Myosin-9 was a direct target of usenamine A. Usenamine A could block cytoskeleton remodeling through the disruption of the interaction between Myosin-9 and actin. Myosin-9 participated in suppressing proliferation while inducing apoptosis and autophagy in response to treatment with usenamine A. In addition, Myosin-9 was revealed as a potential oncogene in HCC. CONCLUSIONS: Usenamine A was initially revealed to suppress human hepatoma cells growth by interfering with the Myosin-9/actin-dependent cytoskeleton remodeling through the direct targeting of Myosin-9. Myosin-9 is, therefore, a promising candidate target for HCC treatment, while usenamine A may be utilized as a possible anti-HCC therapeutic, particularly in the treatment of HCC with aberrant Myosin-9.


Assuntos
Morte Celular Autofágica , Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Humanos , Carcinoma Hepatocelular/patologia , Actinas , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Hepáticas/patologia , Apoptose , Células Hep G2 , Proteínas do Citoesqueleto/farmacologia , Proteínas do Citoesqueleto/uso terapêutico , Citoesqueleto/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Nat Prod ; 85(8): 2006-2017, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-35976233

RESUMO

Bladder cancer, specifically, muscle-invasive bladder cancer (MIBC), is among the most common malignant tumors. Patients with MIBC who cannot tolerate standard drugs require novel treatments. Targeting apoptosis may help treat cancer, which may be achieved with the use of some natural products. Nodosin, found in Isodon serra (Maxim.) Kudo (known as Xihuangcao), may inhibit bladder cancer cells. Transcriptomics and proteomics dual-omic analyses revealed the network pharmacological mechanism: (1) blocking the S phase by up-regulating RPA2, CLSPN, MDC1, PDCD2L, and E2F6 gene expressions, suppressing cancer cell proliferation; (2) inducing apoptosis and autophagy and restraining ferroptosis by up-regulating HMOX1, G0S2, SQSTM1, FTL, SLC7A11, and AIFM2 gene expressions; (3) preventing cancer cell migration by down-regulating NEXN, LIMA1, CFL2, PALLD, and ITGA3 gene expressions. In vivo, nodosin inhibited bladder cancer cell growth in a model of xenograft tumor in nude mice. This study is the first to report basic research findings on the network pharmacological mechanism of cytotoxicity of bladder cancer cells by nodosin, providing novel evidence for the application of nodosin in the field of oncology; however, other mechanisms may be involved in the effects of nodosin for further research. These findings provide a foundation for the development of novel MIBC drugs.


Assuntos
Produtos Biológicos , Neoplasias da Bexiga Urinária , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/uso terapêutico , Animais , Produtos Biológicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/farmacologia , Proteínas do Citoesqueleto/uso terapêutico , Diterpenos , Humanos , Camundongos , Camundongos Nus , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/farmacologia , Proteínas dos Microfilamentos/uso terapêutico , Músculos/metabolismo , Músculos/patologia , Farmacologia em Rede , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia
4.
Phytomedicine ; 104: 154320, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35830758

RESUMO

BACKGROUND: After thrombosis, t-PA thrombolysis is the first choice, but the use of t-PA can easily lead to hemorrhagic injury and neurotoxicity. The combination of Danhong injection (DHI) and tissue plasminogen activator (t-PA) therapy may be a new strategy to find high-efficiency anti-thrombosis and low bleeding risk. However, nothing is about the effect of DHI plus t-PA on platelet activation. PURPOSE: The present research was to explore the optimal dose of DHI and t-PA in vivo and mechanisms involved with the treatment of combining DHI and t-PA for thrombotic disease and determined whether DHI plus t-PA affects thrombotic processes related to platelet activation. METHODS: Mice were induced by administering κ-carrageenan intraperitoneally, the ratio of different doses of DHI and t-PA in vivo, and the optimal dose effects on platelet aggregation, platelet adhesion, thrombosis formation, and platelet activation were determined. The effects of the αIIbß3 signaling pathway were analyzed in mice. RESULTS: In vitro, DHI (62% v/v), t-PA (1 mg/ml), and DHI + t-PA (62% v/v + 1 mg/ml) decreased rat platelet aggregation and adhesion, with a stronger effect from the combination as compared to t-PA monotherapy. In vivo, injections of κ-carrageenan were used to induce BALB/c mice. The optimal dose of DHI, t-PA, and DHI + t-PA is 12 ml/kg, 10 mg/kg, and 12 ml/kg + 7.5 mg/kg. The administration of DHI (12 ml/kg), t-PA (10 mg/kg), and DHI + t-PA (12 ml/kg + 7.5 mg/kg) decreased thrombi in mouse tissue vessels. Furthermore, the reduction of thrombosis formation by DHI, t-PA, and DHI + t-PA was related to lower collagen deposition, and lowered expressions of collagen I, matrix metalloproteinase 2 (MMP-2), and metalloproteinase 9 (MMP-9) in mouse tails, with increased efficacy in combination as compared to t-PA alone. The anti-thrombosis actions of DHI, t-PA, and their combination regulated the expression of CD41, purinergic receptor (P2Y12), guanine nucleotide-binding protein G (q) subunit alpha (GNAQ), phosphatidylinositol phospholipase c beta (PLCß), Ras-related protein 1 (Rap1), RIAM, talin1, fibrinogen alpha chain (FG), kindlin-3, and RAS guany1-releasing protein 1 (RasGRP1). CONCLUSIONS: Based on expression, the mechanism responsible for thrombosis may be attributed to platelet activation via the αIIbß3 signaling pathway. Combination therapy with DHI and t-PA exerted potent thrombolytic effects. Thus, our data can be used as a foundation for further clinical studies examining the efficacy of traditional Chinese medicines for the treatment of thrombosis.


Assuntos
Trombose , Ativador de Plasminogênio Tecidual , Animais , Carragenina , Proteínas do Citoesqueleto/uso terapêutico , Medicamentos de Ervas Chinesas , Fatores de Troca do Nucleotídeo Guanina/uso terapêutico , Metaloproteinase 2 da Matriz , Camundongos , Ratos , Cauda/metabolismo , Trombose/tratamento farmacológico , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tecidual/uso terapêutico
5.
Cancer Genet ; 264-265: 50-59, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35366592

RESUMO

Gene fusions involving the neurotrophic receptor tyrosine kinase genes NTRK1, NTRK2, and NTRK3, are well established oncogenic drivers in a broad range of pediatric and adult tumors. These fusions are also important actionable markers, predicting often dramatic response to FDA approved kinase inhibitors. Accurate interpretation of the clinical significance of NTRK fusions is a high priority for diagnostic laboratories, but remains challenging and time consuming given the rapid pace of new data accumulation, the diversity of fusion partners and tumor types, and heterogeneous and incomplete information in variant databases and knowledgebases. The ClinGen NTRK Fusions Somatic Cancer Variant Curation Expert Panel (SC-VCEP) was formed to systematically address these challenges and create an expert-curated resource to support clinicians, researchers, patients and their families in making accurate interpretations and informed treatment decisions for NTRK fusion-driven tumors. We describe a system for NTRK fusion interpretation (including compilation of key elements and annotations) developed by the NTRK fusions SC-VCEP. We illustrate this stepwise process on examples of LMNA::NTRK1 and KANK1::NTRK2 fusions. Finally, we provide detailed analysis of current representation of NTRK fusions in public fusion databases and the CIViC knowledgebase, performed by the NTRK fusions SC-VCEP to determine existing gaps and prioritize future curation activities.


Assuntos
Neoplasias , Receptor trkA , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/uso terapêutico , Adulto , Biomarcadores Tumorais/genética , Carcinogênese , Criança , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/uso terapêutico , Fusão Gênica , Humanos , Neoplasias/diagnóstico , Proteínas de Fusão Oncogênica/genética , Receptor trkA/genética , Receptor trkA/uso terapêutico
6.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-34769119

RESUMO

Human Ezrin Peptides (HEPs) are inhibitors of expression of IL-6 and other inflammatory cytokines, amplifiers of adaptive B cell and T cell immunity and enhancers of tissue repair. The mutation stable C-terminus of HIV gp120, mimics 69% of the "Hep-receptor", a zipped α-helical structure in the middle of the α domain of human ezrin protein. Synthetic peptides homologous to the Hep-receptor of ezrin of five to fourteen amino acids, activate anti-viral immunity against a wide range of viruses (HIV, HCV, herpes, HPV, influenza and other human respiratory viruses). Human Ezrin Peptide One (HEP1) TEKKRRETVEREKE (brand name Gepon, registered for human use in Russia from 2001) is a successful treatment for opportunistic infections in HIV-infected patients. That treats HEP1and prevents mucosal candidiasis, herpes zoster outbreaks and infection-induced chronic diarrhea. There are clinical publications in Russian on the successful treatments of chronic recurrent vaginal candidiasis, acute and chronic enterocolitis and dysbacteriosis, which are accompanied by normalization of the mucosal microbiome, and the decline or disappearance of inflammation. HEP1 is also an effective treatment and prevention for recurrent inflammation and ulceration in the stomach, duodenum and colon. HEP1 and RepG3 GEKKRRETVEREGG (a derivative of HEP1) have been used successfully as an inhaled spray peptide solution to treat a small number of human volunteers with mild-to-moderate COVID, resulting from SARS-CoV-2 infection, based on earlier successes in treating acute viral respiratory disease with inflammatory complications. Ezrin peptides seem to correct a dysregulation of innate immune responses to SARS-CoV-2. They are also adjuvants of B cell adaptive immunity and increase antibody titres, resulting in protection from lethal virus infection of mice. In a clinical study in Moscow, orally administered HEP1 was shown to enhance antibody-titres produced in response to hepatitis-B vaccination. These very preliminary but promising results with ezrin peptide treatment of COVID must be replicated in large-scale randomised placebo controlled clinical studies, to be verified.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Proteínas do Citoesqueleto/farmacologia , Proteínas do Citoesqueleto/uso terapêutico , Imunidade Adaptativa/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Animais , Antivirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Humanos , Camundongos , Infecções Respiratórias/tratamento farmacológico , Viroses/tratamento farmacológico
7.
Oncologist ; 25(12): e1968-e1979, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32926498

RESUMO

BACKGROUND: For stage II colorectal cancer (CRC), the efficacy of adjuvant chemotherapy remains controversial. Consensus molecular subtype (CMS) has been validated to be a prognostic tool for CRCs. In this study, CMS status was investigated as a prognostic biomarker for the efficacy of adjuvant chemotherapy for stage II colorectal cancer. MATERIALS AND METHODS: The tissue microarray was retrospectively constructed of 165 nonconsecutive, primary, and sporadic stage II CRCs. CMS status was determined by immunohistochemistry staining of CDX2, HTR2B, FRMD6, and ZEB1, combining with microsatellite instability testing. The prognostic for adjuvant chemotherapy efficacy of CMS status was calculated by Kaplan-Meier curves and Cox regression analysis. Subgroup analyses were conducted according to tumor location. RESULTS: Kaplan-Meier curves indicated that CMS was associated with overall survival (OS) and disease-free survival for stage II CRCs. Cox regression analysis showed that CMS was an independent risk factor for OS. Among high-risk clinicopathological factors, patients with CMS2/3 (hazard ratio [HR]: 0.445, 95% confidence interval [CI]: 0.227-0.875), left-sided tumors (HR: 0.488, 95% CI: 0.247-0.968), or fewer than 12 lymph nodes examined (HR: 0.307, 95% CI: 0.097-0.974) had survival benefit from adjuvant chemotherapy. Subgroup analysis showed that adjuvant chemotherapy only improved OS for patients with left-sided tumors of CMS2/3 subtype. Regardless of CMS, right-sided tumors had no benefit from adjuvant chemotherapy. CONCLUSION: CMS is a better prognostic factor for adjuvant chemotherapy for stage II CRCs. Together with tumor location, CMS classification will aid in personalized treatment for stage II CRCs. IMPLICATIONS FOR PRACTICE: For stage II colorectal cancer (CRC), the efficacy of adjuvant chemotherapy remains controversial, in that its minimal benefit (no more than 5% on average) is considered not worth the toxic effects of the drugs. There are still no effective prognostic and predictive biomarkers. This study showed that consensus molecular subtype (CMS) status is a predictive marker for adjuvant chemotherapy efficacy. Patients with left-sided tumors of CMS2/3 subtype have survival benefit by receiving adjuvant chemotherapy, which will aid in personalized treatment for stage II CRCs. Moreover, this test of CMS based on immunohistochemistry is cheap, not time consuming, and easily conducted in the laboratories of most hospitals.


Assuntos
Biomarcadores Tumorais , Neoplasias Colorretais , Biomarcadores Tumorais/genética , Quimioterapia Adjuvante , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Consenso , Proteínas do Citoesqueleto/uso terapêutico , Humanos , Imuno-Histoquímica , Proteínas de Membrana , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos
8.
Bioorg Med Chem ; 27(14): 3179-3193, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31200986

RESUMO

The spread of infections caused by multidrug-resistant (MDR) pathogens, such as methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant S. aureus (VRSA), has created a need for new antibiotics with novel mechanisms of action. The bacterial division protein FtsZ has been identified as a novel drug target that can be exploited clinically. As part of an ongoing effort to develop FtsZ-targeting antibacterial agents, we describe herein the design, synthesis and bioactivity of six series of novel 1,3,4-oxadiazol-2-one-containing, 1,2,4-triazol-3-one-containing and pyrazolin-5-one-containing benzamide derivatives. Among them, compound A14 was found to be the most potent antibacterial agent, much better than clinical drugs such as ciprofloxacin, linezolid and erythromycin against all the tested gram-positive strains, particularly methicillin-resistant, penicillin-resistant and clinical isolated S. aureus. Subsequent studies on biological activities and docking analyses proved that A14 functioned as an effective compound targeting FtsZ. Preliminary SAR indicated a general direction for further optimization of these novel analogues. Taken together, this research provides a promising chemotype for developing newer FtsZ-targeting bactericidal agents.


Assuntos
Proteínas de Bactérias/uso terapêutico , Benzamidas/uso terapêutico , Proteínas do Citoesqueleto/uso terapêutico , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Proteínas de Bactérias/farmacologia , Benzamidas/farmacologia , Proteínas do Citoesqueleto/farmacologia
10.
FEBS Lett ; 584(14): 3085-8, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20553717

RESUMO

Abnormal tau cleavage is prominent in the neurofibrillary degeneration characteristic of Alzheimer's disease (AD) and related tauopathies. We recently showed that cleaved human tau is secreted by specific mechanisms when overexpressed. Here we examined the effect of expressing N-terminal and full length tau constructs in transiently and stably transfected neuronal lines. We show that secreted tau exhibits a cleavage pattern similar to CSF-tau from human AD patients and that tau secretion is specifically inhibited by the presence of the exon 2 insert. These results suggest that tau secretion may play a hitherto unsuspected role in AD and related tauopathies.


Assuntos
Éxons , Doença de Alzheimer/genética , Animais , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/uso terapêutico , Formas de Dosagem , Vias de Administração de Medicamentos , Humanos , Camundongos , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Tauopatias/tratamento farmacológico , Tauopatias/genética
11.
Transplantation ; 87(10): 1488-96, 2009 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-19461485

RESUMO

BACKGROUND: Renal damage caused by cold preservation and warm reperfusion has been well documented and involves tissue edema, cell swelling, ATP depletion, calcium toxicity, and oxidative stress. However, more common proximal mechanisms have not been identified, which may limit the development of effective clinical treatment strategies. Previous work indicates that many cytoskeletal structures are affected by cold preservation and reperfusion, including membrane-rich ezrin-associated complexes. The aim of this study was to investigate whether the sublamellar cytoskeletal protein ezrin is causally involved in cold preservation injury in renal tubule epithelial cells. METHODS: We created a stably transfected cell line (LLC-EZ) using the pig kidney proximal tubular epithelial cell line (LLC-PK1), which constitutively overexpresses wild-type ezrin. These cells were cold stored in University of Wisconsin Solution and reperfused in vitro to model renal tubule preservation injury, which was assessed by biochemical, metabolic, functional, and structural endpoints. RESULTS: Overexpression of ezrin increased cell viability (lactate dehydrogenase release), mitochondrial activity (ATP synthesis, dehydrogenase activity, and inner mitochondrial membrane potential), and protected the structure of cell membrane microvilli and mitochondria after cold storage preservation injury. Reperfusion-induced apoptosis was also significantly reduced in LLC-EZ cells overexpressing ezrin. CONCLUSIONS: Enhanced ezrin expression protects tubule epithelial cells from cold storage preservation injury, possibly by membrane or mitochondrial mechanisms.


Assuntos
Proteínas do Citoesqueleto/uso terapêutico , Células Epiteliais/citologia , Túbulos Renais Proximais/citologia , Traumatismo por Reperfusão/prevenção & controle , Preservação de Tecido/métodos , Animais , Técnicas de Cultura de Células/métodos , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Temperatura Baixa , Proteínas do Citoesqueleto/genética , DNA Complementar/genética , Rim , Camundongos , Mitocôndrias/fisiologia , Mitocôndrias/ultraestrutura , Suínos , Transfecção
12.
Arzneimittelforschung ; 57(7): 497-504, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17803064

RESUMO

This report shows the therapeutic benefit of HEP1 (human ezrin peptide 324-337; TEKKRRETVEREKE) monotherapy of hepatitis C virus (HCV) infection in HIV infected patients in two clinical studies. In the Pilot Study I, 16 of 18 patients responded well to the treatment with significant reductions of HCV viral load and a normalization of serum liver enzymes. In 8 of 18 patients, HCV RNA became undetectable, and 3 of 8 interferon/ribavirin treatment failure patients showed undetectable HCV load following HEP1 treatment. In the second study, 8 of 10 patients responded well to the treatment with a pronounced reduction of the HCV viral load and a normalization of serum liver enzymes. Three of 15 patients (20%) showed an undetectable viral load 30 days after the end of a 30-day course of HEP1 treatment. In both studies, all genotypes of HCV were sensitive to HEP1 treatment. Analysis of the combined data from both studies showed the overall efficacy of HEP1 therapy: in 37 HCV+HIV patients, HEP1 therapy gave the following results: 10 of 37 (27%) HCV+HIV patients showed a reduction of viral load between -7 log (-10,000,000x) and -3 log (-1000x); 4 of 37 (11%) a reduction of -3 log (-1000x); 6 of 37 (16%) a reduction of -2 log (-100x); 11 of 37 (30%) a reduction of -1 log (-10x); 6 of 37 (16%) a reduction of less than -1 log (-10x); 0 of 37 (0%) had an increase in viral load, and the average reduction in viral load for all 37 patients was -2 log (-100x). No adverse reactions or side effects were detected and the improving CD4/CD8 ratio showed that the therapy had no negative impact on the immunological status. Thus, oral HEP1 therapy matches the efficacy results for injectable peginterferon/oral ribavirin therapy with the advantages of more rapid action and less side effects. HEP1 therapy should be used in patients where either peginterferon/ribavirin therapy fails or is contraindicated.


Assuntos
Antivirais/uso terapêutico , Proteínas do Citoesqueleto/uso terapêutico , Infecções por HIV/complicações , Hepatite C/complicações , Hepatite C/tratamento farmacológico , Adulto , Antivirais/efeitos adversos , Relação CD4-CD8 , Proteínas do Citoesqueleto/efeitos adversos , Método Duplo-Cego , Feminino , HIV-1/metabolismo , Hepacivirus/genética , Hepatite C/diagnóstico , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Projetos Piloto , RNA Viral/genética , RNA Viral/isolamento & purificação , Carga Viral
14.
Hum Gene Ther ; 13(13): 1631-46, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12228018

RESUMO

The sarcoglycanopathies are a group of four autosomal recessive limb girdle muscular dystrophies (LGMD 2D, 2E, 2C, and 2F), caused by mutations of the alpha-, beta-, gamma-, or delta-sarcoglycan genes, respectively. The delta-sarcoglycan-deficient hamster has been the most utilized model for gene delivery to muscle by recombinant adeno-associated virus (AAV) vectors; however, human patients with delta-sarcoglycan deficiency are exceedingly rare, with only two patients described in the United States. Here, we report construction and use of AAV vectors expressing either alpha- or beta-sarcoglycan, the genes responsible for the most common forms of the human sarcoglycanopathies. Both vectors showed successful short-term genetic, biochemical, and histological rescue of both alpha- and beta-sarcoglycan-deficient mouse muscle. However, comparison of persistence of expression in 51 injected mice showed substantial differences between AAV alpha-sarcoglycan (alpha-SG) and beta-sarcoglycan (beta-SG) vectors. AAV-beta-SG showed long-term expression with no decrease in expression for more than 21 months after injection, whereas AAV-alpha-SG showed a dramatic loss of positive fibers between 28 and 41 days post-injection (p = 0.006). Loss of immunopositive myofibers was correlated with significant inflammatory cell infiltrate, primarily macrophages. To determine whether the loss of alpha-sarcoglycan-positive fibers was due to an immune response or cytotoxic effect of alpha-sarcoglycan overexpression, severe combined immunodeficient (SCID) mouse muscle was assayed for cytotoxicity after injection with AAV-alpha-SG, AAV-beta-SG, or phosphate-buffered saline. The results were consistent with overexpression of alpha-sarcoglycan causing significant cytotoxicity. The cytotoxicity of alpha-sarcoglycan, and not beta- or delta-sarcoglycan overexpression, was consistent with biochemical studies of the hierarchical order of assembly of the sarcoglycan complex. Our data suggest that even closely related proteins might require different levels of expression to avoid toxicity and achieve long-term tissue rescue.


Assuntos
Proteínas do Citoesqueleto/genética , Dependovirus , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Glicoproteínas de Membrana/genética , Distrofias Musculares/terapia , Animais , Proteínas do Citoesqueleto/uso terapêutico , Distroglicanas , Glicoproteínas de Membrana/uso terapêutico , Camundongos , Camundongos Knockout , Camundongos SCID , Fibras Musculares Esqueléticas/citologia , Distrofias Musculares/genética , Sarcoglicanas
15.
Neuromuscul Disord ; 12 Suppl 1: S78-89, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12206801

RESUMO

Duchenne muscular dystrophy is an X-linked recessive muscle wasting disease caused by the absence of the muscle cytoskeletal protein, dystrophin. Dystrophin is a member of the spectrin superfamily of proteins and is closely related in sequence similarity and functional motifs to three proteins that constitute the dystrophin related protein family, including the autosomal homologue, utrophin. An alternative strategy circumventing many problems associated with somatic gene therapies for Duchenne muscular dystrophy has arisen from the demonstration that utrophin can functionally substitute for dystrophin and its over-expression in muscles of dystrophin-null transgenic mice completely prevents the phenotype arising from dystrophin deficiency. One potential approach to increase utrophin levels in muscle for possible therapeutic purpose in humans is to increase expression of the utrophin gene at a transcriptional level via promoter activation. This has lead to an interest in the identification and manipulation of important regulatory regions and/or molecules that increase the expression of utrophin and their delivery to dystrophin-deficient tissue. As pre-existing cellular mechanisms are utilized, this approach would avoid many problems associated with conventional gene therapies.


Assuntos
Proteínas do Citoesqueleto/genética , Terapia Genética/métodos , Proteínas de Membrana/genética , Distrofia Muscular de Duchenne/terapia , Animais , Proteínas do Citoesqueleto/uso terapêutico , Distrofina/genética , Humanos , Proteínas de Membrana/uso terapêutico , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Regiões Promotoras Genéticas , Regulação para Cima , Utrofina
17.
Am J Clin Oncol ; 24(1): 47-51, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11232949

RESUMO

The investigative therapy for a senior patient after radical subtotal gastroesophagectomy for regional lymph node and proximal esophagus metastasized adenocarcinoma (stage IIIA, T3, N 1 M0) of the cardioesophageal junction is reported. The case has several unusual features: (1) the patient is the author and is not a physician; (2) in the absence of codified postsurgical treatment, he used his academic biomedical background, commercial associations, and international contacts to find and prioritize six clinically tested options for investigative postsurgical therapy; (3) after unsuccessful efforts to append ongoing clinical trials of new immunotherapies for breast adenocarcinoma (the first two therapy options), an innovative protocol was designed and gained allowance by the U.S. Food and Drug Administration for his use of combined nonspecific immunotherapy and chemotherapy based on extensive trials in South Korea that showed the synergistic effect of the two postsurgical therapies used together. A potent, new, nonspecific immunostimulant (DetoxPC) was injected subcutaneously in 10 diminishing doses during 105 weeks. Two standard chemotherapeutic drugs (5-fluorouracil and mitomycin-C) were injected intravenously in six equal doses during three weeks. Five years after the surgery, the patient enjoys good health without signs or symptoms of recurrence or metastasis. He discusses his perspectives on future clinical trials and on a patient actively pursuing investigative postsurgical therapy for a malignancy when otherwise poor survival is indicated.


Assuntos
Adenocarcinoma/terapia , Adjuvantes Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteínas do Citoesqueleto/uso terapêutico , Lipídeo A/análogos & derivados , Lipídeo A/uso terapêutico , Neoplasias Gástricas/terapia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/cirurgia , Idoso , Terapia Combinada , Combinação de Medicamentos , Fluoruracila/administração & dosagem , Humanos , Masculino , Mitomicina/administração & dosagem , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/cirurgia
18.
Curr Neurol Neurosci Rep ; 1(1): 89-96, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11898504

RESUMO

Gene transfer research for Duchenne muscular dystrophy (DMD) has brought the goal of successful treatment of this devastating, inherited disease closer to being a reality. Although gene therapeutic approaches for DMD patients are not yet in clinical use, recent advances using DMD animal models are encouraging. Progress in vector design, such as high-capacity adenoviral vectors, targeted adenoviral vectors, and heterodimerization of DNA delivered by adeno-associated virus (AAV) vectors have advanced the field considerably. The recent studies into the pharmacologic-induced read-through of stop codons, the increased study of utrophin and its upregulation, and the introduction of point mutation correction using chimeric oligonucleotides have expanded the field, providing new avenues of inquiry.


Assuntos
Terapia Genética , Distrofia Muscular de Duchenne/terapia , Antibacterianos/uso terapêutico , Proteínas do Citoesqueleto/uso terapêutico , Vetores Genéticos , Gentamicinas/uso terapêutico , Humanos , Proteínas de Membrana/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Utrofina
19.
Hum Gene Ther ; 10(8): 1299-310, 1999 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-10365661

RESUMO

Utrophin is a close homolog of dystrophin, the protein whose mutations cause Duchenne muscular dystrophy (DMD). Utrophin is present at low levels in normal and dystrophic muscle, whereas dystrophin is largely absent in DMD. In such cases, the replacement of dystrophin using a utrophin gene transfer strategy could be more advantageous because utrophin would not be a neoantigen. To establish if adenovirus (AV)-mediated utrophin gene transfer is a possible option for the treatment of DMD, an AV vector expressing a shortened version of utrophin (AdCMV-Utr) was constructed. The effect of utrophin overexpression was investigated following intramuscular injection of this AV into mdx mice, the mouse model of DMD. When the tibialis anterior (TA) muscles of 3- to 5-day-old animals were injected with 5 microl of AdCMV-Utr (7.0 x 10(11) virus/ml), an average of 32% of fibers were transduced and the transduction level remained stable for at least 60 days. The presence of utrophin restored the normal histochemical pattern of the dystrophin-associated protein complex at the cell surface and resulted in a reduction in the number of centrally nucleated fibers. The transduced fibers were largely impermeable to the tracer dye Evans blue, suggesting that utrophin protects the surface membrane from breakage. In vitro measurements of the force decline in response to high-stress eccentric contractions demonstrated that the muscles overexpressing utrophin were more resistant to mechanical stress-induced injury. Taken together, these data indicate that AV-mediated utrophin gene transfer can correct various aspects of the dystrophic phenotype. However, a progressive reduction in the number of transduced fibers was observed when the TA muscles of 30- to 45-day-old mice were injected with 25 microl of AdCMV-Utr. This reduction coincides with a humoral response to the AV and transgene, which consists of a hybrid mouse-human cDNA.


Assuntos
Adenoviridae , Proteínas do Citoesqueleto/uso terapêutico , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas de Membrana/uso terapêutico , Distrofias Musculares/terapia , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/genética , Expressão Gênica , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos mdx , Músculos/patologia , Distrofias Musculares/patologia , Fenótipo , Utrofina
20.
Nat Med ; 5(4): 439-43, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10202936

RESUMO

Limb-girdle muscular dystrophies 2C-F represent a family of autosomal recessive diseases caused by defects in sarcoglycan genes. The cardiomyopathic hamster is a naturally occurring model for limb-girdle muscular dystrophy caused by a primary deficiency in delta-sarcoglycan. We show here that acute sarcolemmal disruption occurs in this animal model during forceful muscle contraction. A recombinant adeno-associated virus vector encoding human delta-sarcoglycan conferred efficient and stable genetic reconstitution in the adult cardiomyopathic hamster when injected directly into muscle. A quantitative assay demonstrated that vector-transduced muscle fibers are stably protected from sarcolemmal disruption; there was no associated inflammation or immunologic response to the vector-encoded protein. Efficient gene transduction with rescue of the sarcoglycan complex in muscle fibers of the distal hindlimb was also obtained after infusion of recombinant adeno-associated virus into the femoral artery in conjunction with histamine-induced endothelial permeabilization. This study provides a strong rationale for the development of gene therapy for limb-girdle muscular dystrophy.


Assuntos
Proteínas do Citoesqueleto/uso terapêutico , Terapia Genética/métodos , Histamina/uso terapêutico , Glicoproteínas de Membrana/uso terapêutico , Distrofia Muscular Animal/terapia , Animais , Permeabilidade da Membrana Celular , Cricetinae , Proteínas do Citoesqueleto/genética , Dependovirus/genética , Vetores Genéticos , Humanos , Glicoproteínas de Membrana/genética , Perfusão , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/uso terapêutico , Sarcoglicanas , Sarcolema/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA