Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc (Bayl Univ Med Cent) ; 28(1): 50-3, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25552798

RESUMO

We present the case of a 74-year-old woman with metastatic lobular carcinoma with an occult breast primary presenting as a suspected ampullary tumor due to its ampullary metastasis. The patient's clinical presentation is of interest in two aspects. First, lobular carcinoma of the breast metastatic to the ampulla is extremely rare. Second, in the absence of a detectable primary lesion, prior history of malignancy, or distinguishing clinical, radiological, and endoscopic features, histopathological assessments are pivotal for arriving at the appropriate diagnosis and for optimizing treatment.

2.
BMJ Case Rep ; 20142014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25452296

RESUMO

Polycythaemia vera (PV) is a clonal disorder of bone marrow stem cells characterised by erythrocytosis. Diagnosis of PV requires exclusion of secondary causes of polycythaemia. It has been held that an elevated erythropoietin (Epo) level strongly indicates secondary erythrocytosis and excludes PV diagnosis, to the extent that the reduced serum Epo level is currently listed as a minor criterion in the WHO classification scheme for PV. However, patients with PV who co-present with Budd-Chiari syndrome have been documented with elevated serum Epo levels. For these patients, identification of the Janus kinase 2 (JAK2) V617F point mutation along with the transient nature of the Epo elevation provides certainty of PV diagnosis, as illustrated by the proband. In this case report, the patient's positive response to cytoreductive therapy (hydroxyurea 500 mg daily) and phlebotomy (750 mL over three phlebotomies) further supports validity of PV diagnosis with elevated Epo. The patient remains on rivaroxaban (Xarelto) for treatment of her portal vein thrombosis.


Assuntos
Síndrome de Budd-Chiari/diagnóstico , Eritropoetina/sangue , Policitemia Vera/diagnóstico , Ascite/etiologia , Biomarcadores/sangue , Síndrome de Budd-Chiari/sangue , Síndrome de Budd-Chiari/complicações , Diagnóstico Diferencial , Feminino , Humanos , Pessoa de Meia-Idade , Policitemia Vera/sangue , Policitemia Vera/complicações
3.
BMJ Case Rep ; 20142014 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-24729118

RESUMO

A 74-year-old man was previously diagnosed with BCR-ABL1-positive pre-B cell acute lymphoblastic leukaemia (pre-B ALL) based on bone marrow cytology, flow cytometry, cytogenetics and fluorescent in situ hybridisation findings. Following a highly complicated hospital course, the patient achieved cytogenetic remission by consolidated chemotherapy and the tyrosine kinase inhibitor dasatinib. He subsequently presented with relapsed pre-B ALL after 3 years in remission. In consideration of his challenging clinical history, he was started on concurrent ponatinib (45 mg daily) and bortezomib (1.3 mg/m(2) intravenous weekly). The major molecular response was achieved (<0.0893% BCR-ABL1 transcripts) after 3 months. Bone marrow now demonstrates a BCR-ABL1-negative, complete cytogenetic response. The patient continues to do well with mild thrombocytopenia and improved anaemia on bortezomib and 30 mg daily ponatinib. Our experience with a single patient suggests the feasibility of combined targeted therapy with ponatinib and bortezomib. This novel treatment approach achieved clinical remission with a manageable toxicity profile.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Doença Aguda , Idoso , Ácidos Borônicos/administração & dosagem , Bortezomib , Humanos , Imidazóis/administração & dosagem , Masculino , Cromossomo Filadélfia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Pirazinas/administração & dosagem , Piridazinas/administração & dosagem , Recidiva , Indução de Remissão
4.
Curr Pharm Biotechnol ; 13(9): 1750-60, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21740355

RESUMO

Oncolytic virotherapy is an evolving but, as yet, unrealized treatment option for cancer. This approach harnesses the cancer-restricted replicative activity of engineered viruses to achieve tumor cell kill. Tumors that are resistant to chemotherapy or radiotherapy can be susceptible to viral oncolysis because of distinct cell kill mechanisms. There is now compelling evidence that collateral induction of anti-tumor immune responses contributes substantially to viral antitumor activities. In addition to the expected anti-viral immune clearance, the "danger" signal created by virus-infected cells can generate immune co-stimulation known to override immune suppression and reverse tolerance within the tumor microenvironment. Our recent findings indicate that immune activation augments the clinical outcomes of oncolytic virotherapy. Strikingly similar and robust clinical response rates ( > 25%) were observed among advanced cancer patients following intratumoral treatments with adenoviral (AdΔ24) and herpes simplex (JS1/34.5-/47) constructs armed with an integrated granulocyte-macrophage colony-stimulating factor (GMCSF) payload. Both agents produced regressions in injected as well as distant, uninjected lesions, demonstrating systemic effectiveness. We discuss the innate and adaptive immune activating events that may contribute to these clinical outcomes, and examine systemic delivery strategies to tilt the immunological balance from viral clearance to tumor elimination.


Assuntos
Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Imunidade Adaptativa/imunologia , Animais , Humanos , Imunidade Inata/imunologia , Neoplasias/virologia
5.
DNA Cell Biol ; 30(9): 715-26, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21612405

RESUMO

Bifunctional small hairpin RNAs (bi-shRNAs) are functional miRNA/siRNA composites that are optimized for posttranscriptional gene silencing through concurrent mRNA cleavage-dependent and -independent mechanisms (Rao et al., 2010 ). We have generated a novel bi-shRNA using the miR30 scaffold that is highly effective for knockdown of human stathmin (STMN1) mRNA. STMN1 overexpression well documented in human solid cancers correlates with their poor prognosis. Transfection with the bi-shSTMN1-encoding expression plasmid (pbi-shSTMN1) markedly reduced CCL-247 human colorectal cancer and SK-Mel-28 melanoma cell growth in vitro (Rao et al., 2010 ). We now examine in vivo the antitumor efficacy of this RNA interference-based approach with human tumor xenografted athymic mice. A single intratumoral (IT) injection of pbi-shSTMN1 (8 µg) reduced CCL-247 tumor xenograft growth by 44% at 7 days when delivered as a 1,2-dioleoyl-3-trimethyl-ammoniopropane:cholesterol liposomal complex. Extended growth reductions (57% at day 15; p < 0.05) were achieved with three daily treatments of the same construct. STMN1 protein reduction was confirmed by immunoblot analysis. IT treatments with pbi-shSTMN1 similarly inhibited the growth of tumorgrafts derived from low-passage primary melanoma (≥70% reduction for 2 weeks) and abrogated osteosarcoma tumorgraft growth, with the mature bi-shRNA effector molecule detectable for up to 16 days after last injection. Antitumor efficacy was evident for up to 25 days posttreatment in the melanoma tumorgraft model. The maximum tolerated dose by IT injection of >92 µg (Human equivalent dose [HED] of >0.3 mg/kg) in CCL-247 tumor xenograft-bearing athymic mice was ∼10-fold higher than the extrapolated IC(50) of 9 µg (HED of 0.03 mg/kg). Healthy, immunocompetent rats were used as biorelevant models for systemic safety assessments. The observed maximum tolerated dose of <100 µg for intravenously injected pbi-shSTMN1 (mouse equivalent of <26.5 µg; HED of <0.09 mg/kg) confirmed systemic safety of the therapeutic dose, hence supporting early-phase assessments of clinical safety and preliminary efficacy.


Assuntos
Neoplasias Colorretais/terapia , Técnicas de Silenciamento de Genes/métodos , Terapia Genética/métodos , Melanoma/terapia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estatmina/metabolismo , Animais , Primers do DNA/genética , Feminino , Humanos , Immunoblotting , Masculino , Dose Máxima Tolerável , Camundongos , Camundongos Nus , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Hum Gene Ther ; 22(11): 1331-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21517694

RESUMO

Hereditary inclusion body myopathy (HIBM) is an autosomal recessive adult-onset myopathy due to mutations in the GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase) gene. Affected patients have no therapeutic options. We have previously demonstrated in preclinical testing the ability to safely correct GNE gene function through liposomal delivery of the wild-type GNE gene. Results were verified in a single patient treated by intravenous infusion of GNE gene lipoplex. A single patient (patient 001) with severe HIBM treated with a compassionate investigational new drug received seven doses of GNE gene lipoplex via intravenous infusion at the following doses: 0.4, 0.4, 1.0, 4.0, 5.0, 6.0, and 7.0 mg of DNA. GNE transgene expression, downstream induction of sialic acid, safety, and muscle function were evaluated. Transient low-grade fever, myalgia, tachycardia, transaminase elevation, hyponatremia, and hypotension were observed after infusion of each dose of GNE gene lipoplex. Quadriceps muscle expression of the delivered GNE, plasmid, and RNA was observed 24 hr after the 5.0-mg dose and at significantly greater levels 72 hr after the 7.0-mg infusion in comparison with expression in quadriceps muscle immediately before infusion. Sialic acid-related proteins were increased and stabilization in the decline of muscle strength was observed. We conclude that clinical safety and activity have been demonstrated with intravenous infusion of GNE gene lipoplex. Further assessment will involve a phase I trial of intravenous administration of GNE gene lipoplex in individuals with less advanced HIBM with more muscle function.


Assuntos
Complexos Multienzimáticos/genética , Miosite de Corpos de Inclusão/terapia , Adulto , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Infusões Intravenosas , Lipossomos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miosite de Corpos de Inclusão/genética , RNA/metabolismo
7.
J Gene Med ; 12(5): 403-12, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20440751

RESUMO

BACKGROUND: Hereditary inclusion body myopathy (HIBM) is an autosomal recessive adult onset myopathy. It is characterized by mutations of the GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase) gene. Afflicted patients have no therapeutic options. In preclinical testing, we have previously demonstrated the ability to correct GNE gene function and the safety of delivery of wild type GNE gene using a liposomal delivery vehicle. METHODS: A single patient (subject #001) with severe HIBM treated by compassionate investigational new drug received four doses of GNE gene Lipoplex via intramuscular injection. GNE transgene expression, downstream induction of sialic acid, safety and muscle function were evaluated. RESULTS: Significant durable improvement in locoregional skeletal muscle function was observed in the injected left extensor carpi radialis longus of #001 in correlation with GNE transgene upregulation and local induction of sialic acid. Other than transient low grade fever and pain at the injection site, no significant toxicity was observed. CONCLUSIONS: Proof of principle for manufacturing of 'clinical grade' GNE gene Lipoplex, clinical safety and activity are demonstrated with GNE gene Lipoplex. Further assessment will involve intravenous administration and subsequent phase I trial involving additional but less severely afflicted HIBM patients.


Assuntos
Terapia Genética , Lipossomos/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/uso terapêutico , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/terapia , Adolescente , Adulto , Biópsia , Feminino , Terapia Genética/efeitos adversos , Humanos , Injeções Intramusculares , Força Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miosite de Corpos de Inclusão/fisiopatologia , Ácido N-Acetilneuramínico/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Adulto Jovem
8.
Mol Ther ; 18(2): 429-34, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19935775

RESUMO

A phase I clinical trial was conducted to determine the clinical safety of Telomelysin, a human telomerase reverse transcriptase (hTERT) promoter driven modified oncolytic adenovirus, in patients with advanced solid tumors. A single intratumoral injection (IT) of Telomelysin was administered to three cohorts of patients (1 x 10(10), 1 x 10(11), 1 x 10(12) viral particles). Safety, response and pharmacodynamics were evaluated. Sixteen patients with a variety of solid tumors were enrolled. IT of Telomelysin was well tolerated at all dose levels. Common grade 1 and 2 toxicities included injection site reactions (pain, induration) and systemic reactions (fever, chills). hTERT expression was demonstrated at biopsy in 9 of 12 patients. Viral DNA was transiently detected in plasma in 13 of 16 patients. Viral DNA was detectable in four patients in plasma or sputum at day 7 and 14 post-treatment despite below detectable levels at 24 h, suggesting viral replication. One patient had a partial response of the injected malignant lesion. Seven patients fulfilled Response Evaluation Criteria in Solid Tumors (RECIST) definition for stable disease at day 56 after treatment. Telomelysin was well tolerated. Evidence of antitumor activity was suggested.


Assuntos
Adenoviridae/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/metabolismo , Telomerase/metabolismo , Adenoviridae/genética , Adulto , Idoso , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias/genética , Neoplasias/metabolismo , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/genética , Telomerase/genética
9.
Gene Regul Syst Bio ; 3: 89-101, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19838336

RESUMO

Hereditary inclusion body myopathy-2 (HIBM2) is an adult-onset, muscular disease caused by mutations in the GNE gene. HIBM2-associated GNE mutations causing hyposialyation have been proposed to contribute to reduced muscle function in patients with HIBM2, though the exact cause of this disease is unknown. In the current studies we examined pre-clinical in vivo toxicity, and expression of the plasmid-based, CMV driven wild-type GNE plasmid vector. The plasmid vector was injected intramuscularly (IM) or systemically (IV) into BALB/c mice, following encapsulation in a cationic liposome (DOTAP:Cholesterol). Single IM injections of the GNE-lipoplex at 40 microg did not produce overt toxicity or deaths, indicating that the no observable adverse effect level (NOAEL) dose for IM injection was >or=40 microg. Single intravenous (IV) infusion of GNE-lipoplex was lethal in 33% of animals at 100 microg dose, with a small proportion of animals in the 40 microg cohort demonstrating transient toxicity. Thus the NOAEL dose by the IV route was greater than 10 microg and less than or equal to 40 microg. Real-time RT-qPCR analysis demonstrated recombinant human GNE mRNA expression in 100% of muscle tissues that received IM injection of 40 microg GNE-lipoplex, at 2 weeks. These results indicate that GNE-lipoplex gene transfer is safe and can produce durable transgene expression in treated muscles. Our findings support future exploration of the clinical efficacy of GNE-lipoplex for experimental gene therapy of HIBM2.

10.
Clin Cancer Res ; 15(15): 4847-56, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19622582

RESUMO

PURPOSE: The growth-inhibitory activity of recombinant CD40 ligand (CD40L) is well documented in human multiple myeloma (MM). We examined MM-targeted delivery of CD40L by a conditional replicative oncolytic adenovirus, AdEHCD40L. EXPERIMENTAL DESIGN: The growth-regulatory activity of AdEHCD40L was determined in vitro and in vivo. Differential analysis with AdEHCD40L and parental virus (AdEHNull)-infected cultures allowed the identification of cellular and molecular pathways modulated by the CD40L transgene. RESULTS: Conditional expression of viral E1A and CD40L transgene was shown in human MM lines RPMI 8226 [interleukin (IL)-6 independent] and Kas-6/1 (IL-6 dependent) under hypoxic conditions commonly found in MM in situ. AdEHCD40L inhibited MM cell growth more effectively than AdEHNull. This enhanced growth-inhibitory activity was abrogated by cotreatment with a CD40L antibody. Chemoresistant MM lines (MR20 and LR5) were similarly susceptible to AdEHCD40L treatment. AdEHCD40L induced apoptosis and S-phase cell cycle blockade while uniquely up-regulating the previously described proapoptotic elements tumor necrosis factor-related apoptosis-inducing ligand, Fas, and IL-8. Intratumoral injections of AdEHCD40L reduced the growth of severe combined immunodeficient/hu RPMI 8226 xenografts by >50% compared with 28% reduction by AdEHNull. Adenoviral hexon and CD40L were detected in AdEHCD40L-treated tumors at day 35 after infection primarily in necrotic areas, suggesting viral replicative activity. CONCLUSIONS: These findings show that CD40L acts in concert with viral oncolysis to produce MM growth inhibition through activation of cellular apoptosis. The direct growth-inhibitory activity of AdEHCD40L, together with the well-known immune-potentiating features of CD40L, may be clinically applicable for the experimental treatment of MM or plasma cell leukemia.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/metabolismo , Ligante de CD40/genética , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Infecções por Adenoviridae/virologia , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica , Transdução Genética , Transgenes/genética , Transgenes/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Curr Gene Ther ; 9(1): 45-60, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19275571

RESUMO

Tremendous strides have been made in proteogenomics and RNA interference technologies. Hence "personalized" cancer gene therapy has become a foreseeable rather than a predictable reality. Currently, the lack of an optimized, systemic gene delivery vehicle remains a key limiting factor for developing effective treatment applications. Since their introduction by Felgner in 1987, cationic lipids have been an attractive consideration for gene delivery, in view of their biocompatibility, biodegradability, low toxicity, and low immunogenicity. Successful in vivo transgene expression by cationic lipid- or cationic polymer-based delivery depends critically on a long circulating half life (>48 h), a definable systemic biodistribution with target-specific cancer localization, and efficient cell entry and internalization. Ideally, the agent should have a hydrophobic, stabilized core that ensures integrity of the therapeutic entity in vivo, a biocompatible, neutrally charged shell (zeta potential of approximately +/-10 mv) for enhanced, "stealth" circulation, and a suitable size (approximately 50-200 nm in diameter) for access into the tumor neovasculature and reduced reticuloendothelial system (RES) uptake. "Smart" receptor-targeting moieties can redirect intracellular trafficking. Additional engineered features have also been incorporated to minimize lysosomal degradation (membrane fusogenic lipids or proton sponge), promote endosomal escape into cytoplasm (cell penetrating peptides, triblock copolymer construction), and enhance nuclear entry and activate the endogenous transcriptional machinery (inclusion of a nuclear localization signal). Improvements in each of these respective areas of study have converged to yield promising in vivo results.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Lipídeos/administração & dosagem , Neoplasias/terapia , Animais , Humanos , Neoplasias/genética
12.
Clin Cancer Res ; 15(4): 1317-25, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19228733

RESUMO

PURPOSE: CD40 ligand (CD40L, CD154) plays a central role in immunoregulation and also directly modulates epithelial cell growth and differentiation. We previously showed that the CD40 receptor is commonly expressed in primary breast cancer tissues. In this proof-of-principle study, we examined the breast cancer growth-regulatory activities of an oncolytic adenoviral construct carrying the CD40L transgene (AdEHCD40L). EXPERIMENTAL DESIGN: In vitro and in vivo evaluations were carried out on AdEHCD40L to validate selective viral replication and CD40L transgene activity in hypoxia inducing factor-1alpha and estrogen receptor-expressing human breast cancer cells. RESULTS: AdEHCD40L inhibited the in vitro growth of CD40+ human breast cancer lines (T-47D, MDA-MB-231, and BT-20) by up to 80% at a low multiplicity of infection of 1. Incorporation of the CD40L transgene reduced the effective dose needed to achieve 50% growth inhibition (ED50) by approximately 10-fold. In contrast, viral and transgene expression of AdEHCD40L, as well its cytotoxicity, was markedly attenuated in nonmalignant cells. Intratumoral injections with AdEHCD40L reduced preexisting MDA-MB-231 xenograft growth in severe combined immunodeficient mice by >99% and was significantly more effective (P<0.003) than parental virus AdEH (69%) or the recombinant CD40L protein (49%). This enhanced antitumor activity correlated with cell cycle blockade and increased apoptosis in AdEHCD40L-infected tumor cells. CONCLUSIONS: These novel findings, together with the previously known immune-activating features of CD40L, support the potential applicability of AdEHCD40L for experimental treatment of human breast cancer.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/terapia , Ligante de CD40/genética , Terapia Genética/métodos , Transgenes , Proteínas E1A de Adenovirus/análise , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos SCID , Fenótipo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
DNA Cell Biol ; 26(5): 293-300, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17504025

RESUMO

MicroRNAs (miRNAs) are a recently discovered class of small (approximately 18-24 nt) nucleic acids that negatively regulate gene expression. This novel class of molecules modulates a wide array of growth and differentiation processes in human cancers. High throughput analyses, utilizing the solid phase, array platform, or liquid phase, bead-based hybridization have variously demonstrated that miRNA expression was commonly dysregulated in human cancer. miRNA expression profiling has shown promise in defining malignant status in retrospective studies. Considerable disagreement remains with respect to the miRNA signature for a specific cancer cell type, which appears to depend largely on the analytical platform. Nonetheless, various internally controlled studies have successfully identified the histotype of tumors of unknown origin according to miRNA expression profile. The evaluation of miRNAs expression may also be of prognostic value, as best exemplified by the correlation of let-7 and mir-155 levels with disease survival in nonsmall cell lung cancer.


Assuntos
MicroRNAs/genética , Neoplasias/diagnóstico , Neoplasias/genética , Animais , Biomarcadores/análise , Perfilação da Expressão Gênica , Humanos , MicroRNAs/análise , Valor Preditivo dos Testes , Prognóstico
14.
Cancer Res ; 66(19): 9736-43, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17018633

RESUMO

Despite successes in animal models, cancer gene therapy with small interfering RNAs (siRNA) is hindered by the lack of an optimal delivery platform. We examined the applicability of the replication-competent, oncolytic adenovirus, ONYX-411, to deliver a mutant K-ras siRNA transgene to human cancer cells. Proof-of-principle studies showed an additive tumor growth-inhibitory response through siRNA-mediated K-ras knockdown and ONYX-411-mediated cancer cell lysis. A novel construct, termed Internavec (for interfering RNA vector), was generated by cloning a K-ras(v12)-specific siRNA(ras-4) hairpin construct under the control of the human H1 promoter into the deleted E3b region of ONYX-411. Internavec acquired an increase in potency of approximately 10-fold in human cancer cells expressing the relevant K-ras(v12) mutation (H79, H441, and SW480), as defined by a reduction in the effective dose needed to achieve 50% growth inhibition (ED(50)). Internavec remained attenuated in nonmalignant epithelial cells. Daily intratumoral injections of Internavec (five daily injections of 1 x 10(8) plaque-forming units) significantly reduced the growth of s.c. H79 pancreatic cancer xenografts in nu/nu mice by 85.5%, including complete growth suppression in three of five mice. Parental ONYX-411 or ONYX-411-siRNA(GFP) was markedly less effective (47.8% growth reduction, P = 0.03; and 44.1% growth reduction, P = 0.03, respectively). siRNA(ras) transgene activity contributed to cell cycle blockage, increased apoptosis, and marked down-regulation of Ras signaling-related gene expression (AKT2, GSK3 beta, E2F2, and MAP4K5). These findings indicate that Internavec can generate a two-pronged attack on tumor cells through oncogene knockdown and viral oncolysis, resulting in a significantly enhanced antitumor outcome.


Assuntos
Adenoviridae/genética , Genes ras , Terapia Genética , Vetores Genéticos/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Adenoviridae/fisiologia , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Divisão Celular , Linhagem Celular Tumoral/efeitos dos fármacos , Neoplasias do Colo/patologia , Células Epiteliais/efeitos dos fármacos , Perfilação da Expressão Gênica , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Proteína Oncogênica p21(ras)/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Proteínas Proto-Oncogênicas c-raf/metabolismo , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Curr Mol Med ; 6(3): 339-49, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16712479

RESUMO

Patients with non-small cell lung cancer (NSCLC) are commonly diagnosed with advanced disease and have limited therapeutic options. Experimental treatment approaches including small molecule targeted therapeutics, gene modified tumor vaccines, and viral-based gene therapy have induced tumor regression in a small proportion of patients, suggesting that advanced NSCLC is susceptible to molecular perturbations. RNA interference (RNAi) has generated considerable excitement as a potential cancer therapeutic application. RNAi is the process by which small, double stranded RNA molecules (small interfering RNA, or siRNA) can initiate sequence-specific, post-transcriptional gene silencing (PTGS). Cancer growth inhibition was attained through siRNA-knockdown of unique or overexpressed cancer oncogenetic messages that are relevant to NSCLC pathophysiology. As with other loss-of-function cancer gene therapy approaches, clinical efficacy of siRNA depends largely on the extent of cell target coverage at the locoregional and/or systemic level. Cationic liposomes as well as viral vectors have been used successfully for siRNA delivery. However, viral delivery may have more immediate relevance due to its wider clinical acceptance in the cancer gene therapy arena. We advocate the use of conditional replicative, oncolytic adenovirus for siRNA delivery, which offers potential benefits of restricted and renewable siRNA expression within the tumor microenvironment, and an additive anti-tumor outcome through viral oncolysis and siRNA-mediated oncogene-silencing, which we have demonstrated with the A549 NSCLC cell line. Several oncolytic adenoviral constructs are potentially applicable clinical platforms with proven infectivity and safety, which are feasible also for the delivery of microRNAs (miRNA), a recently discovered group of endogenous, small RNA with PTGS activity that is downregulated in lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/terapia , MicroRNAs/genética , RNA Interferente Pequeno/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Terapia Genética/métodos , Humanos , Neoplasias Pulmonares/genética , MicroRNAs/uso terapêutico , Modelos Biológicos , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico
16.
Curr Opin Mol Ther ; 7(2): 114-24, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15844618

RESUMO

RNA interference describes the recently discovered process of sequence-specific, post-transcriptional gene silencing that is initiated by double-stranded RNA molecules known as small interfering RNAs (siRNAs). siRNAs have an acceptable half-life in vitro, a predictable biodistribution profile similar to that of single-stranded antisense oligonucleotides (ASOs), and have repeatedly been more robust than ASO techniques in terms of consistency of transcript knockdown and threshold concentration. Following validation in mammalian cells by Tuschl and co-workers in 2001, synthetic siRNAs have gained wide acceptance as a laboratory tool for target validation. Currently, there is considerable interest in the therapeutic use of siRNA, particularly in areas of infectious disease and cancer. In vitro and in vivo findings demonstrate the efficacy of siRNA knockdown of gene messages that are pivotal for tumor cell growth, metastasis, angiogenesis and chemoresistance, leading to tumor growth suppression. However, siRNA-based cancer therapy faces similar pharmacokinetic limitations to ASO therapy with respect to the extent that siRNA accesses primary and metastatic target cells. The recently identified 'off-target activity' of siRNAs is also of concern. The concept of carrier-restricted delivery of siRNA by conditionally replicative, oncolytic adenoviruses is discussed. Oncolytic adenoviral delivery offers the potential benefits of restricted and renewable siRNA expression within the tumor microenvironment, an additive antitumor outcome through viral oncolysis and siRNA-mediated oncogene silencing, and a proven clinical platform with respect to infectivity and safety.


Assuntos
Neoplasias/tratamento farmacológico , RNA Interferente Pequeno/uso terapêutico , Adenoviridae , Animais , Vetores Genéticos , Humanos , Neoplasias/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
17.
Clin Lymphoma ; 5(4): 285-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15794867

RESUMO

Immunoglobulin (Ig) M myeloma is a distinct entity with features of multiple myeloma (MM) and Waldenstrom's macroglobulinemia (WM). The malignant cells in IgM myeloma have a distinctive chromosomal translocation that differentiates them from WM. These cells are postgerminal-center in origin with isotype-switch transcripts. They appear to be arrested at a point of maturation between that of WM and MM. Preliminary data indicate that a pattern of osteoclast-activating factor and osteoprotegerin expression similar to that observed in classic MM is present in IgM myeloma. Additional studies on patients with this rare tumor may provide further insight into the pathogenesis of bone disease in plasma cell dyscrasias.


Assuntos
Imunoglobulina M/genética , Imunoglobulina M/imunologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Macroglobulinemia de Waldenstrom/genética , Macroglobulinemia de Waldenstrom/imunologia , Diferenciação Celular , Citocinas/biossíntese , Glicoproteínas/biossíntese , Humanos , Linfocinas/biossíntese , Masculino , Pessoa de Meia-Idade , Osteoprotegerina , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores do Fator de Necrose Tumoral , Translocação Genética
18.
Mol Ther ; 11(1): 160-72, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15585417

RESUMO

The mda-7 gene (approved gene symbol IL24) is a novel tumor suppressor gene with tumor-apoptotic and immune-activating properties. We completed a Phase I dose-escalation clinical trial, in which a nonreplicating adenoviral construct expressing the mda-7 transgene (INGN 241; Ad-mda7) was administered intratumorally to 22 patients with advanced cancer. Excised tumors were evaluated for vector-specific DNA and RNA, transgenic MDA-7 expression, and biological effects. Successful gene transfer as assessed by DNA- and RT-PCR was demonstrated in 100% of patients evaluated. DNA analyses demonstrated a dose-dependent penetration of INGN 241 (up to 4 x 10(8) copies/mug DNA at the 2 x 10(12) vp dose). A parallel distribution of vector DNA, vector RNA, MDA-7 protein expression, and apoptosis induction was observed in all tumors, with signals decreasing with distance away from the injection site. Additional evidence for bioactivity of INGN 241 was illustrated via regulation of the MDA-7 target genes beta-catenin, iNOS, and CD31. Transient increases (up to 20-fold) of serum IL-6, IL-10, and TNF-alpha were observed. Significantly higher elevations of IL-6 and TNF-alpha were observed in patients who responded clinically to INGN 241. Patients also showed marked increases of CD3+CD8+ T cells posttreatment, suggesting that INGN 241 increased systemic TH1 cytokine production and mobilized CD8+ T cells. Intratumoral delivery of INGN 241 induced apoptosis in a large volume of tumor and elicited tumor-regulatory and immune-activating events that are consistent with the preclinical features of MDA-7/IL-24.


Assuntos
Adenoviridae/genética , Terapia Genética , Vetores Genéticos/genética , Interleucinas/genética , Interleucinas/uso terapêutico , Neoplasias/genética , Neoplasias/terapia , Adenoviridae/fisiologia , Apoptose , Biomarcadores/análise , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Genes Supressores de Tumor , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Humanos , Imunidade Ativa/imunologia , Injeções , Melanoma/genética , Melanoma/patologia , Melanoma/terapia , Estadiamento de Neoplasias , Neoplasias/imunologia , Neoplasias/patologia , Transativadores/metabolismo , Transgenes/genética , Resultado do Tratamento , Replicação Viral , beta Catenina
19.
Cancer Gene Ther ; 10(5): 341-52, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12719704

RESUMO

ONYX-015 is an adenovirus that selectively replicates in p53 dysfunctional or mutated malignant cells. We performed a pilot trial to determine the safety and feasibility of treatment with ONYX-015 delivered intravenously in patients with advanced malignancy. One cohort of five patients received ONYX-015 once a week for 6 weeks at a dose of 2 x 10(12) particles per infusion in combination with weekly infusions of irinotecan (CPT11, 125 mg per week) and 5-fluorouracil (5FU, 500 mg per week). A second cohort of five patients received the combination of ONYX-015 at a dose of 2 x 10(11) particles per week for 6 weeks in combination with interleukin 2 (IL 2, 1.1 x 10(6) units daily via subcutaneous injection for 5 days each week for 4 weeks). Toxicity attributable to ONYX-015 was limited to transient fever. All patients demonstrated elevations in neutralizing antibody titers within 4 weeks of the infusion of ONYX-015. Serum levels of IL-6, IL-10, tumor necrosis factor-alpha, and interferon-gamma increased within 6 hours of viral infusion, suggesting immune activation. This response was more pronounced in the cohort of patients who received 2 x 10(12) particles per infusion. Two patients demonstrated uptake of viral particles in malignant tissue by quantitative PCR. Electron microscopy confirmed selective cytoplasmic viral particles within malignant cells but not within adjacent normal tissue in a third patient. In conclusion ONYX-015 can be administered safely in combination with CPT11, 5FU or low-dose IL 2 and is able to access malignant tissue following intravenous infusion. Further investigation of ONYX-015, possibly with agents that may modulate replication activity, or duration of virus survival, is indicated.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Camptotecina/análogos & derivados , Interleucina-2/uso terapêutico , Neoplasias/terapia , Vacinas Virais/uso terapêutico , Adenoviridae/genética , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Camptotecina/administração & dosagem , Terapia Combinada , Citocinas/sangue , Feminino , Fluoruracila/administração & dosagem , Terapia Genética , Humanos , Imunoterapia , Infusões Intravenosas , Irinotecano , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Replicação Viral
20.
Semin Oncol ; 30(2): 318-24, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12720161

RESUMO

Assays for specific antigen-binding activity were performed on sera from 172 patients with monoclonal macroglobulinemia defined by immunofixation electrophoresis. The sera were collected between 1970 and 2002. Mean IgM level was 1,409 mg/dL with a range from 70 to 6,800. Cryoglobulins were identified in 15.3% (26/170 sera: 12 trace, five single component, and nine mixed IgM-IgG). Rheumatoid factor (RF) was detected in 19 of 151 (12.6%) samples with titers ranging from 1:80 to 1:327,680. Among the nine mixed IgM-IgG cryos, eight were RF-positive and six of six displayed positivity for hepatitis C virus. Cold agglutinins (CA) were present in 8.5% (10/117) of sera with anti-I titers between 1:512 and 1:65,536. IgM binding to a series of glycosaminoglycan oligosaccharides, glycolipids, and glycoprotein antigens was found in 75 samples (43%). IgM binding to antigens having known associations to polyneuropathies occurred in 20 patients (12%). Antinuclear antibody (ANA) was documented in 10.7% (18/169) of sera. Anti-DNA activity was absent in all samples tested. Sera from 71% of patients with monoclonal macroglobulinemia in this series exhibited binding to autoantigens. Some of these immune complexes resulted in clinically significant manifestations. Our results suggest that many monoclonal immunoglobulins may be functional antibodies rather than "paraproteins." Characterization of antigen-binding activities may provide insight into the pathogenesis of monoclonal gammopathies.


Assuntos
Anticorpos/sangue , Macroglobulinemia de Waldenstrom/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Aglutininas/sangue , Anticorpos Antinucleares/sangue , Crioglobulinas , Feminino , Anticorpos Anti-Hepatite C/sangue , Humanos , Imunoglobulina M/sangue , Masculino , Pessoa de Meia-Idade , Fator Reumatoide/sangue , Macroglobulinemia de Waldenstrom/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...