Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 230
Filtrar
1.
J Biol Chem ; 300(1): 105509, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38042493

RESUMEN

Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.


Asunto(s)
Acetilcisteína , AMP Cíclico , Factores de Intercambio de Guanina Nucleótido , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Tionucleótidos , Animales , Niño , Humanos , Ratones , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , AMP Cíclico/uso terapéutico , ADN/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/agonistas , Ratones Endogámicos NOD , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Masculino , Femenino , Preescolar , Tionucleótidos/farmacología , Tionucleótidos/uso terapéutico , Daño del ADN , Quimioterapia Combinada
2.
Blood Adv ; 5(13): 2775-2787, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34251414

RESUMEN

Overexpression of B-cell leukemia/lymphoma 2 (BCL2) renders acute myeloid leukemia (AML) cells resistant to chemotherapy and has been associated with unfavorable outcomes. Oblimersen (G3139) is a phosphorothioate 18-mer antisense oligonucleotide directed against the first 6 BCL2 codons. In a phase 1 study of AML patients treated with G3139, cytarabine, and daunorubicin induction with cytarabine consolidation, no antisense-related toxicity was reported, and BCL2 downregulation occurred in patients achieving complete remission. In this phase 3 trial, untreated older AML patients were randomized to cytarabine (100 mg/m2 per day on days 4-10) and daunorubicin (60 mg/m2 per day on days 4-6) followed by cytarabine consolidation (2000 mg/m2 per day on days 4-8) with (arm A) or without (arm B) G3139 (7 mg/m2 per day on days 1-10 [induction] or days 1-8 [consolidation]). A total of 506 patients were enrolled. No differences in toxicity were observed between arms. Estimated overall survival (OS) at 1 year was 43% for arm A and 40% for arm B (1-sided log rank P = .13), with no differences in disease-free (DFS; P = .26) or event-free survival (P = .80). Subgroup analyses showed patients age <70 years in arm A had improved OS by 1 month vs those in arm B (P = .04), and patients with secondary AML in arm A had better DFS vs those in arm B (P = .04). We conclude that addition of G3139 to chemotherapy failed to improve outcomes of older AML patients. However, more effective means of inhibiting BCL2 are showing promising results in combination with chemotherapy in AML. This trial was registered at www.clinicaltrials.gov as #NCT00085124.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Leucemia Mieloide Aguda , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Citarabina/uso terapéutico , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Tionucleótidos/uso terapéutico
3.
Biochim Biophys Acta Rev Cancer ; 1875(2): 188500, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33385484

RESUMEN

Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.


Asunto(s)
Clusterina/genética , Clusterina/metabolismo , Neoplasias/metabolismo , Clusterina/antagonistas & inhibidores , Sinergismo Farmacológico , Quimioterapia , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Tionucleótidos/farmacología , Tionucleótidos/uso terapéutico
4.
Aliment Pharmacol Ther ; 51(12): 1286-1304, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32363674

RESUMEN

BACKGROUND: Thiopurines in combination with glucocorticoids are used as first-line, second-line and maintenance therapies in autoimmune hepatitis and opportunities exist to improve and expand their use. AIMS: To describe the metabolic pathways and key factors implicated in the efficacy and toxicity of the thiopurine drugs and to indicate the opportunities to improve outcomes by monitoring and manipulating metabolic pathways, individualising dosage and strengthening the response. METHODS: English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. RESULTS: Thiopurine methyltransferase activity and 6-tioguanine (6-thioguanine) nucleotide levels influence drug efficacy and safety, and they can be manipulated to improve treatment response and prevent myelosuppression. Methylated thiopurine metabolites are associated with hepatotoxicity, drug intolerance and nonresponse and their production can be reduced or bypassed. Universal pre-treatment assessment of thiopurine methyltransferase activity and individualisation of dosage to manipulate metabolite thresholds could improve outcomes. Early detection of thiopurine resistance by metabolite testing, accurate estimations of drug onset and strength by surrogate markers and adjunctive use of allopurinol could improve the management of refractory disease. Dose-restricted tioguanine (thioguanine) could expand treatment options by reducing methylated metabolites, increasing the bioavailability of 6-tioguanine nucleotides and ameliorating thiopurine intolerance or resistance. CONCLUSIONS: The efficacy and safety of thiopurines in autoimmune hepatitis can be improved by investigational efforts that establish monitoring strategies that allow individualisation of dosage and prediction of outcome, increase bioavailability of the active metabolites and demonstrate superiority to alternative agents.


Asunto(s)
Hepatitis Autoinmune/tratamiento farmacológico , Purinas/uso terapéutico , Alopurinol/uso terapéutico , Azatioprina/uso terapéutico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Nucleótidos de Guanina/uso terapéutico , Hepatitis Autoinmune/epidemiología , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Mercaptopurina/uso terapéutico , Mejoramiento de la Calidad , Tioguanina/uso terapéutico , Tionucleótidos/uso terapéutico , Resultado del Tratamiento
5.
Medicine (Baltimore) ; 98(6): e14254, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30732140

RESUMEN

Custirsen is the second-generation antisense oligonucleotide (ASO), which can reduce cellular levels of clusterin to increase the cytotoxic effect of chemotherapeutic drugs. Our study assessed the efficacy and safety of custirsen in patients with metastatic castration-resistant prostate cancer (mCRPC).We conducted a comprehensive search to identify all the randomized controlled trials (RCTs) of custirsen for the treatment of mCRPC. The reference lists of the retrieved studies were investigated.Three publications involving a total of 1709 patients were used in the analysis. We found that overall survival (OS) (P = .25) was not statistically significant in the comparison. Safety assessments indicated custirsen were often associated with complications resulting from neutropenia (P < .001), anaemia (P < .001), thrombocytopenia (P < .001), and diarrhea (P = .002).Our meta-analysis shows that custirsen has no obvious effect on improving the OS of patients with mCRPC. Adverse reactions were more common among those patients treated with custirsen as compared to those treated with placebo.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/patología , Tionucleótidos/uso terapéutico , Humanos , Masculino
6.
Biomaterials ; 197: 182-193, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30660994

RESUMEN

Antisense oligonucleotides (ASOs) usually contain a fully phosphorothioate (PS) backbone, which possibly interact with many genes and proteins under intracellular conditions. G3139 is an ASO that targets Bcl-2 mRNA and induces cell apoptosis. Here, we report a kind of cytidinyl-lipid combined with a cationic lipid (DNCA/CLD, molar ration, 28:3, named mix), which may interact with oligonucleotides via H-bond formation, pi-stacking and electrostatic interaction, accompanied by low zeta potentials. The IC50 value of G3139 delivered by mix-lipid reduced from above 20 µM to 0.158 µM for MCF-7/ADR, and exhibited stronger antiproliferation upon other cancer cell lines. In addition, PS modification in the 3'-half of G3139 (especially at positions 13-16) enhanced serum stability, target specificity and anticancer activity. Also, a locked nucleic acid (LNA) gapmer G3139 (LNA-G3139) showed superior antiproliferation (78.5%) and Bcl-2 mRNA suppression effects (85.5%) at 200 nM, mainly due to its high complementary RNA affinity. More apoptosis-associated targets were identified, and a lower level of non-specific protein binding (HSA) revealed that both antisense and aptamer mechanisms might simultaneously exist. A combination of a new delivery system and chemical modifications, such as in LNA-G3139, may have potential clinical application prospects in the future.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Citidina/análogos & derivados , Portadores de Fármacos/química , Tionucleótidos/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Lípidos/química , Células MCF-7 , Ratones Endogámicos BALB C , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Tionucleótidos/farmacocinética , Tionucleótidos/farmacología , Tionucleótidos/uso terapéutico
7.
Lancet Oncol ; 18(11): 1532-1542, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29033099

RESUMEN

BACKGROUND: Docetaxel and cabazitaxel improve overall survival compared with mitoxantrone in patients with metastatic castration-resistant prostate cancer. Custirsen (OGX011) is a second generation highly specific antisense oligonucleotide that inhibits the production of clusterin, an antiapoptotic protein that is upregulated in response to chemotherapy and that confers treatment resistance. We aimed to assess whether custirsen in combination with cabazitaxel and prednisone increases overall survival in patients with metastatic castration-resistant prostate cancer previously treated with docetaxel. METHODS: In this randomised, open-label, international, phase 3 trial, men with radiographically documented metastatic castration-resistant prostate cancer that had progressed after docetaxel treatment with a Karnofsky performance status of more than 70% and who were fit for chemotherapy, were recruited from 95 cancer treatment centres in eight countries. Patients were randomly assigned (1:1) centrally using permuted blocks (block size 8) to receive cabazitaxel plus prednisone (cabazitaxel 25 mg/m2 intravenously every 21 days plus oral prednisone 10 mg daily) with or without custirsen (640 mg intravenously on days 1, 8, and 15, plus three previous loading doses) until disease progression, unacceptable toxicity, or the completion of ten treatment cycles. Randomisation was stratified by use of opioids for prostate cancer-related pain at screening, disease progression following first-line docetaxel treatment established by radiographic evidence, and previous treatment with abiraterone or enzalutamide. The co-primary endpoints were overall survival in all randomly assigned patients and in a poor-prognosis subgroup. All analyses were intention to treat with the exception of safety, which was reported for patients who received any assigned treatment. The trial has been completed and the results presented here are the final analysis. This trial is registered with Clinicaltrials.gov, number NCT01578655. FINDINGS: Between Sept 9, 2012, and Sept 29, 2014, 795 patients were screened for enrolment. 635 men were eligible for inclusion and were randomly assigned (n=317 in the cabazitaxel and prednisone plus custirsen group and n=318 in the cabazitaxel and prednisone group). Median follow up was 28·3 months (IQR 24·4-34·5) for the custirsen group and 29·8 months (IQR 25·3-35·2) for the control group. Median overall survival in all randomly assigned patients did not differ between the two groups (14·1 months [95% CI 12·7-15·9] in the curtisen group vs 13·4 months [12·1-14·9] in the control group; hazard ratio [HR] 0·95 [95% CI 0·80-1·12]; log-rank p=0·53). In the poor prognosis subgroup, median overall survival also did not differ between the two treatment groups (11·0 months [95% CI 9·3-13·3] in the custursin group vs 10·9 months [8·2-12·4] in the control group; HR 0·97 [95% CI 0·80-1·21]; two-sided p=0·80). The most frequently reported grade 3 or worse adverse events in the custirsen versus control groups were neutropenia (70 [22%] of 315 vs 61 [20%] of 312), anaemia (68 [22%] vs 49 [16%]), fatigue (23 [7%] vs 18 [6%]), asthenia (16 [5%] vs 8 [3%]), bone pain (16 [5%] vs 5 [2%]), and febrile neutropenia (16 [5%] vs 9 [3%]). Serious adverse events were reported in 155 (49%) versus 132 (42%). 27 patients died within 30 days of treatment in the cabazitaxel and prednisone plus custirsen group, seven of which were deemed to be treatment related, versus 17 in the cabazitaxel and prednisone group, eight of which were deemed to be treatment related. Of the 21 deaths reported, 15 were reported as complications related to study treatment, either chemotherapy (eight and three, respectively) or study drug (none and four, respectively). INTERPRETATION: We noted no survival benefit in men with metastatic castration-resistant prostate cancer with the addition of custirsen to cabazitaxel and prednisone treatment. Cabazitaxel and prednisone remains the standard of care for patients with metastatic castration-resistant prostate cancer progressing after docetaxel chemotherapy. FUNDING: OncoGenex Pharmaceuticals.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Prednisona/administración & dosificación , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Taxoides/administración & dosificación , Tionucleótidos/uso terapéutico , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Supervivencia sin Enfermedad , Docetaxel , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Humanos , Internacionalidad , Estimación de Kaplan-Meier , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Neoplasias de la Próstata Resistentes a la Castración/diagnóstico por imagen , Análisis de Supervivencia , Tionucleótidos/efectos adversos , Tomografía Computarizada por Rayos X/métodos , Resultado del Tratamiento
8.
Pain ; 158(5): 822-832, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28059868

RESUMEN

Activating PKG-1α induces a long-term hyperexcitability (LTH) in nociceptive neurons. Since the LTH correlates directly with chronic pain in many animal models, we tested the hypothesis that inhibiting PKG-1α would attenuate LTH-mediated pain. We first synthesized and characterized compound N46 (N-((3R,4R)-4-(4-(2-fluoro-3-methoxy-6-propoxybenzoyl)benzamido)pyrrolidin-3-yl)-1H-indazole-5-carboxamide). N46 inhibits PKG-1α with an IC50 of 7.5 nmol, was highly selective when tested against a panel of 274 kinases, and tissue distribution studies indicate that it does not enter the CNS. To evaluate its antinociceptive potential, we used 2 animal models in which the pain involves both activated PKG-1α and LTH. Injecting complete Freund's adjuvant (CFA) into the rat hind paw causes a thermal hyperalgesia that was significantly attenuated 24 hours after a single intravenous injection of N46. Next, we used a rat model of osteoarthritic knee joint pain and found that a single intra-articular injection of N46 alleviated the pain 14 days after the pain was established and the relief lasted for 7 days. Thermal hyperalgesia and osteoarthritic pain are also associated with the activation of the capsaicin-activated transient receptor protein vanilloid-1 (TRPV1) channel. We show that capsaicin activates PKG-1α in nerves and that a subcutaneous delivery of N46 attenuated the mechanical and thermal hypersensitivity elicited by exposure to capsaicin. Thus, PKG-1α appears to be downstream of the transient receptor protein vanilloid-1. Our studies provide proof of concept in animal models that a PKG-1α antagonist has a powerful antinociceptive effect on persistent, already existing inflammatory pain. They further suggest that N46 is a valid chemotype for the further development of such antagonists.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Inflamación/complicaciones , Osteoartritis/complicaciones , Osteoartritis/enzimología , Umbral del Dolor/fisiología , Dolor/enzimología , Dolor/etiología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacocinética , Animales , Compuestos de Bifenilo/uso terapéutico , Enfermedad Crónica , GMP Cíclico/análogos & derivados , GMP Cíclico/uso terapéutico , Modelos Animales de Enfermedad , Método Doble Ciego , Inhibidores Enzimáticos/uso terapéutico , Adyuvante de Freund/toxicidad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Modelos Moleculares , Osteoartritis/tratamiento farmacológico , Dolor/tratamiento farmacológico , Umbral del Dolor/efectos de los fármacos , Piridinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Tionucleótidos/uso terapéutico , Factores de Tiempo
9.
Curr Hematol Malig Rep ; 12(1): 11-19, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28116634

RESUMEN

The BCL-2 family of proteins integrates pro- and anti-apoptotic signals within the cell and is responsible for initiation of caspase-dependent apoptosis. Chronic lymphocytic leukemia (CLL) cells are particularly dependent on the anti-apoptotic protein BCL-2 for their survival, making this an attractive therapeutic target in CLL. Several early efforts to create inhibitors of the anti-apoptotic family members faced significant challenges, but eventually, the BCL-2 specific inhibitor venetoclax moved forward in CLL. Overall and complete response rates to venetoclax monotherapy in relapsed, refractory CLL are approximately 80 and 20%, respectively, even in patients with high-risk 17p deletion. Toxicities have been manageable and include neutropenia, diarrhea, and nausea. The risk of tumor lysis syndrome (TLS), seen in early experience with the drug, has been mitigated by the use of appropriate TLS risk assessment, prophylaxis, and management. Future studies of venetoclax will focus on combination approaches, predictive biomarker discovery, and mechanisms of resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Compuestos de Anilina/uso terapéutico , Antineoplásicos/efectos adversos , Compuestos Bicíclicos Heterocíclicos con Puentes/efectos adversos , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Gosipol/química , Gosipol/uso terapéutico , Humanos , Indoles , Neutropenia/etiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirroles/uso terapéutico , Sulfonamidas/efectos adversos , Sulfonamidas/uso terapéutico , Tionucleótidos/uso terapéutico
10.
Free Radic Biol Med ; 107: 101-109, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27989755

RESUMEN

UVA accounts for about 95% of the solar ultraviolet (UV) radiation that reaches Earth and most likely contributes to human skin cancer risk. In contrast to UVB, which comprises the remaining 5% and is absorbed by DNA nucleobases to cause direct photodamage, UVA damages DNA indirectly. It does this largely through its interactions with cellular chromophores that act as photosensitisers to generate reactive oxygen species. Exogenously supplied chemicals, including some widely-prescribed medicines, may also act as photosensitisers and these drugs are associated with an increased risk of sun-related cancer. Because they amplify the effects of UVA on cells, they provide a means to investigate the mechanisms and effects of UVA-induced photodamage. Here, we describe some of the major lesions induced by two groups of UVA photosensitisers, the DNA thionucleotides and the fluoroquinolone antibiotics. In thionucleotides, replacement of the oxygen atoms of canonical nucleobases by sulfur converts them into strong UVA chromophores that can be incorporated into DNA. The fluoroquinolones are also UVA chromophores. They are not incorporated into DNA and induce a different range of DNA damages. We also draw attention to the potentially important contribution of photochemical protein damage to the cellular effects of photosensitised UVA. Proteins targeted for oxidation damage include DNA repair factors and we suggest that UVA-mediated protein damage may contribute to sunlight-induced cancer risk.


Asunto(s)
Antibacterianos/química , Daño del ADN , ADN/efectos de la radiación , Fluoroquinolonas/química , Estrés Oxidativo , Neoplasias Cutáneas/metabolismo , Tionucleótidos/química , Antibacterianos/uso terapéutico , Reparación del ADN , Fluoroquinolonas/uso terapéutico , Humanos , Oxidación-Reducción , Fármacos Fotosensibilizantes , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/patología , Tionucleótidos/uso terapéutico , Rayos Ultravioleta/efectos adversos
11.
Postepy Hig Med Dosw (Online) ; 70(0): 1182-1189, 2016 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-28026821

RESUMEN

Survivin is a member of the family of apoptosis inhibitors. It regulates several essential cellular processes, i.e. it inhibits apoptosis and promotes cell proliferation, DNA repair and autophagy. Survivin is responsible for development of the cell's resistance to chemotherapy and radiotherapy. Overexpression of survivin generally correlates with poor prognosis. Its presence has been detected in most types of human tumours. Currently much attention is paid to the possibilities of using this protein as a diagnostic marker of cancer or a prognostic factor. Survivin occurs selectively in cancer cells and is essential for their survival. These features make survivin a promising target for cancer therapy. There are some strategies for discovering survivin inhibitors. The most common strategies are antisense nucleotides, RNA interference and small molecule inhibitors of protein. Scientists are also working on using survivin to induce an immune response in cancer patients. This article discusses the potential role of survivin in the diagnosis of various types of cancer, as well as selected strategies for the inhibition of both gene expression and protein function. Detailed knowledge of the mechanisms of survivin action may therefore be crucial for effective antitumor therapy development.


Asunto(s)
Neoplasias de los Genitales Femeninos/metabolismo , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Femenino , Neoplasias de los Genitales Femeninos/diagnóstico , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Neoplasias de los Genitales Femeninos/terapia , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Proteínas Inhibidoras de la Apoptosis/genética , Naftoquinonas/farmacología , Naftoquinonas/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Pronóstico , Tratamiento con ARN de Interferencia , Survivin , Tionucleótidos/farmacología , Tionucleótidos/uso terapéutico
13.
World J Gastroenterol ; 21(27): 8262-70, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26217078

RESUMEN

Liver cancer, a large proportion of which is hepatocellular carcinoma (HCC), is diagnosed in more than 700000 people each year worldwide. Liver cancer is particularly prevalent in Asia, Sub-Saharan Africa and the South Pacific, where hepatitis B and hepatitis C infection rates are very high. However, due to resistance to chemotherapy, patients with intermediate and advanced-stage disease cannot benefit from this treatment. Clusterin, which is overexpressed in many different cancers, is a stress-induced cytoprotective protein that confers treatment resistance. Custirsen (OGX-011) is a novel 2'-methoxyethyl modified phosphorothioate antisense oligonucleotide that targets secretory clusterin protein expression and is currently in clinical trials for patients with different cancers. In recent years, a number of different clinical trials have been performed, and two phase III clinical trials of custirsen evaluating combinations with chemotherapy in patients with metastatic castration-resistant prostate cancer and metastatic non-small cell lung cancer are currently in progress. The aims of this review are to summarize the current state of research on clusterin, predict future research directions and analyze the potential of the clinical application of custirsen in HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Clusterina/metabolismo , Neoplasias Hepáticas/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Clusterina/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Oligonucleótidos Antisentido/uso terapéutico , Transducción de Señal , Tionucleótidos/uso terapéutico , Resultado del Tratamiento
14.
Semin Cutan Med Surg ; 33(2): 76-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25085666

RESUMEN

Merkel cell carcinoma is an aggressive neuroendocrine tumor with a high incidence of local recurrence, regional nodal and distant metastasis, and a high mortality rate. It has been linked to a polyomavirus in addition to immune suppression. Traditionally, treatment options have been limited to surgery and radiation therapy. Better understanding of the molecular pathways of infection and carcinogenesis has provided potential molecular targets and potential immunotherapies which are discussed in this review.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células de Merkel/terapia , Inmunoterapia , Neoplasias Cutáneas/terapia , Anticuerpos Monoclonales/uso terapéutico , Benzamidas/uso terapéutico , Biomarcadores/análisis , Antígeno CD56/inmunología , Carcinoma de Células de Merkel/diagnóstico , Carcinoma de Células de Merkel/epidemiología , Carcinoma de Células de Merkel/virología , Electroquimioterapia , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Mesilato de Imatinib , Inmunoterapia Adoptiva , Indazoles , Proteínas Inhibidoras de la Apoptosis/metabolismo , Interferones/uso terapéutico , Interleucina-12/genética , Interleucina-2/uso terapéutico , Ipilimumab , Metástasis Linfática , Proteínas de la Membrana/antagonistas & inhibidores , Poliomavirus de Células de Merkel , Oligonucleótidos Antisentido , Piperazinas/uso terapéutico , Infecciones por Polyomavirus/virología , Pronóstico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Receptores de Somatomedina/antagonistas & inhibidores , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/virología , Somatostatina/análogos & derivados , Sulfonamidas/uso terapéutico , Survivin , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tionucleótidos/uso terapéutico
15.
Int J Clin Exp Pathol ; 7(11): 7690-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25550805

RESUMEN

OBJECTIVE: To evaluate the effects of intrathecal administration p38ß antisense oligonucleotide on the development of bone cancer pain rats. METHODS: Forty female SD rats weighing 180~220 g were randomly divided into 4 groups (n = 10 each): Group A (control group): intra-tibial injection of 3 µl Hank's solution; group B (model group): intra-tibial injection of 3 µl MADB-106 mammary gland carcinoma cells of rats (4.8 × 10(3)/µl); group C (p38ß-SODN 20 µg); group D (p38ß-ASODN 20 µg). The model procedures in group C and D were same to those in the group B. From the 14th day after operation, p38ß-SODN 20 µg and p38ß-ASODN 20 µg were respectively intrathecally administrated in group C and D once daily for 6 days whereas normal saline was for group A and B. Mechanical withdrawal threshold and radiant heat threshold of rat hind paws were measured before operation and every other day until 22 d of post-operation. The lumbar 4-6 spinal cord was removed on the 22(nd) day. The expression of spinal p38ß protein was determined by Western blot. RESULTS: No significant differences in mechanical withdrawal threshold and radiant heat threshold were found at all time points in control group. During the first 6 days after operation there were obvious differences in radiant heat stimulus between control group between the other groups (P < 0.05); During 14-22 days after operation, mechanical pain threshold and radiant heat threshold between p38ß-SODN group and Model group were significantly changed compared with that in control group (P < 0.05). However, the differences were not remarkable between control group and p38ß-ASODN group (P > 0.05). The expression of p38ß protein in lumbar spinal cord was significantly higher between p38ß-SODN group and Model group than that in control group (P < 0.05). There was no significant difference in p38ß protein expression between p38ß-ASODN group and control group (P > 0.05). CONCLUSIONS: Hyperalgesia induced by bone cancer can be inhibited by intrathecal administration of p38ß antisense oligonucleotide, which is achieved by reducing expression of p38ß protein.


Asunto(s)
Neoplasias Óseas/complicaciones , Hiperalgesia/tratamiento farmacológico , Proteína Quinasa 11 Activada por Mitógenos/genética , Oligonucleótidos Antisentido/uso terapéutico , Umbral del Dolor/efectos de los fármacos , Dolor/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Hiperalgesia/etiología , Inyecciones Espinales , Oligonucleótidos Antisentido/farmacología , Dolor/etiología , Ratas , Ratas Sprague-Dawley , Tionucleótidos/farmacología , Tionucleótidos/uso terapéutico
16.
Immunol Lett ; 155(1-2): 36-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24095849

RESUMEN

The BCL2 family members play a central role in regulating programmed cell death (apoptosis) and arbitrating the cellular fate through an accurate balance between pro-apoptotic (BAX, BAK, and BH3-only proteins) and pro-survival (BCL2 and its closest homologues, BCLXL, BCLW and MCL-1) factors. Deregulation of BCL2 family proteins contributes to programmed cell death evasion, that is a hallmark of human cancers and it is often related to (chemo)therapy resistance. High BCL2 levels have been detected in most human lymphoid malignancies, not limited to follicular lymphoma (where the role of BCL2 overexpression is driven by the t[14;18] translocation) but also B-cell chronic lymphocytic leukemia (CLL) and multiple myeloma. For all these reasons, the opportunity to induce apoptosis by targeting BCL2 proteins is considered a potentially promising therapeutic approach in hematological malignancies. BCL2 family inhibition strategies currently explored in phase 1, 2 and 3 clinical trials are essentially two: (1) the use of antisense-based strategies to knockdown BCL2 or BCLXL expression (e.g. oblimersen) or (2) the use of synthetic BH3 mimetics i.e. small molecules binding to anti-apoptotic inhibitors thereby allowing the pro-apoptotic activity of BH3-only molecules (e.g. obatoclax, AT-101, ABT-737 and its derivatives ABT-263 and ABT-199). Several of these drugs demonstrated relevant clinical activity as single-agent or in combination therapy, with the most significant drawbacks in clinical use being represented by challenging pharmacokinetic profile (e.g. iv administration, high-levels of plasma proteins binding) and on-target side effects (e.g. gastrointestinal toxicity and thrombocytopenia). Further clinical development of the current compounds (e.g. ABT-199), showing high efficacy but devoid of the most threatening drug-related toxicities, is eagerly awaited. Hopefully, in the next future, BCL2 inhibitors (alone or in combination with immuno- and/or chemo-therapeutic agents) will represent target-specific drugs expanding our therapeutic armamentarium in the fight against hematologic malignancies.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Hematológicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Biomimética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Reprogramación Celular/genética , Ensayos Clínicos como Asunto , ADN sin Sentido/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Humanos , Indoles , Proteínas Proto-Oncogénicas c-bcl-2/genética , Pirroles/uso terapéutico , Tionucleótidos/uso terapéutico
18.
Proc Natl Acad Sci U S A ; 110(9): 3495-500, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23382200

RESUMEN

The refractoriness of acute promyelocytic leukemia (APL) with t(11;17)(q23;q21) to all-trans retinoic acid (ATRA)-based therapy concerns clinicians and intrigues basic researchers. By using a murine leukemic model carrying both promyelocytic leukemia zinc finger/retinoic acid receptor-α (PLZF/RARα) and RARα/PLZF fusion genes, we discovered that 8-chlorophenylthio adenosine-3', 5'-cyclic monophosphate (8-CPT-cAMP) enhances cellular differentiation and improves gene trans-activation by ATRA in leukemic blasts. Mechanistically, in combination with ATRA, 8-CPT-cAMP activates PKA, causing phosphorylation of PLZF/RARα at Ser765 and resulting in increased dissociation of the silencing mediator for retinoic acid and thyroid hormone receptors/nuclear receptor corepressor from PLZF/RARα. This process results in changes of local chromatin and transcriptional reactivation of the retinoic acid pathway in leukemic cells. Meanwhile, 8-CPT-cAMP also potentiated ATRA-induced degradation of PLZF/RARα through its Ser765 phosphorylation. In vivo treatment of the t(11;17) APL mouse model demonstrated that 8-CPT-cAMP could significantly improve the therapeutic effect of ATRA by targeting a leukemia-initiating cell activity. This combined therapy, which induces enhanced differentiation and oncoprotein degradation, may benefit t(11;17) APL patients.


Asunto(s)
Diferenciación Celular , AMP Cíclico/análogos & derivados , Leucemia Promielocítica Aguda/tratamiento farmacológico , Proteínas de Fusión Oncogénica/metabolismo , Proteolisis , Tionucleótidos/uso terapéutico , Translocación Genética , Tretinoina/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 17/genética , AMP Cíclico/farmacología , AMP Cíclico/uso terapéutico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Co-Represor 2 de Receptor Nuclear/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Tionucleótidos/farmacología , Translocación Genética/efectos de los fármacos , Tretinoina/farmacología
19.
Curr Oncol Rep ; 15(2): 113-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23266703

RESUMEN

Adenocarcinoma of the prostate is the most common cancer in men in the Western Hemisphere. This diagnosis includes a clinicopathologically diverse collection of disease entities, encompassing a spectrum from early localized disease to advanced-stage castration-sensitive and ultimately metastatic, castration-resistant states. Although early-stage disease is treatable and potentially curable, treatment options for castration-resistant prostate cancer, the common pathway to prostate cancer death, remain limited and palliative in nature. Therapeutic resistance to androgen blockade, cytotoxic chemotherapy, and radiotherapy is underpinned by a number of cellular mechanisms. The upregulation of protective, antiapoptotic chaperone proteins is one of these mechanisms, and is exemplified by the protein clusterin in castration-resistant prostate cancer. Antisense oligonucleotide technology provides the potential to inhibit specific genes in cancer cells and with this the possibility of a vast impact in oncology, but no antisense drugs have been approved for use in cancer patients to date. Custirsen (OGX-011) is a novel antisense oligonucleotide drug which targets clusterin expression, and its application in prostate cancer is reviewed in this article.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Clusterina/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Tionucleótidos/uso terapéutico , Adenocarcinoma/secundario , Ensayos Clínicos como Asunto , Humanos , Masculino , Neoplasias de la Próstata/secundario
20.
Exp Mol Pathol ; 94(1): 137-47, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23064047

RESUMEN

With the advent of advanced tools in molecular biology, understanding on cancer etiology has improved. siRNA can be considered as an effective tool in cancer therapy through silencing overexpressed genes responsible for cell proliferation or preventing apoptosis. However, some contentious issues such as stability and delivery of siRNA are to be resolved. Bcl-2, an anti-apoptotic gene, is overexpressed in a wide variety of cancers and responsible for drug resistance tumors. In our earlier studies, we developed a nanoformulation of siRNA targeting the Bcl-2 and achieved successful delivery in vitro and in vivo. To extend the scope of the study further, in the present work, we studied the role of nanoformulation of siRNA as adjuvant in chemotherapy with cisplatin. Dose dependant nephrotoxicity is a serious concern apart from other adverse effects of cisplatin. The IC(50) value for cisplatin was decreased from 9.83 µmol/l to 7.43 µmol/l in HeLa cells and from 8.54 µmol/l to 6.68 µmol/l in HEp-2 cells, when it was given with siRNA nanoformulation. Cisplatin at the dose of 1.7 mg/kg in combination with siRNA nanoformulation was effective in improving the lifespan of tumor bearing mice with significant decrease in nephrotoxicity.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/uso terapéutico , Tionucleótidos/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Adyuvante , Cisplatino/farmacología , Terapia Combinada , Células HeLa , Células Hep G2 , Humanos , Ratones , ARN Interferente Pequeño/farmacología , Tionucleótidos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA