Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Chem Res Toxicol ; 31(10): 1042-1051, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30152692

RESUMEN

Acute myeloid leukemia (AML) is a rare yet deadly cancer of the blood and bone marrow. Presently, induction chemotherapy with the DNA damaging drugs cytarabine (ARA-C) and idarubicin (IDA), known as 7 + 3, is the standard of care for most AML patients. However, 7 + 3 is a relatively ineffective therapy, particularly in older patients, and has serious therapy-related toxicities. Therefore, a diagnostic test to predict which patients will respond to 7 + 3 is a critical unmet medical need. We hypothesize that a threshold level of therapy-induced 7 + 3 drug-DNA adducts determines cytotoxicity and clinical response. We further hypothesize that in vitro exposure of AML cells to nontoxic diagnostic microdoses enables prediction of the ability of AML cells to achieve that threshold during treatment. Our test involves dosing cells with very low levels of 14C-labeled drug followed by DNA isolation and quantification of drug-DNA adducts via accelerator mass spectrometry. Here, we have shown proof of principle by correlating ARA-C- and DOX-DNA adduct levels with cellular IC50 values of paired sensitive and resistant cancer cell lines and AML cell lines. Moreover, we have completed a pilot retrospective trial of diagnostic microdosing for 10 viably cryopreserved primary AML samples and observed higher ARA-C- and DOX-DNA adducts in the 7 + 3 responders than nonresponders. These initial results suggest that diagnostic microdosing may be a feasible and useful test for predicting patient response to 7 + 3 induction chemotherapy, leading to improved outcomes for AML patients and reduced treatment-related morbidity and mortality.


Asunto(s)
Citarabina/uso terapéutico , Idarrubicina/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular , Citarabina/química , Citarabina/toxicidad , ADN/química , Aductos de ADN/análisis , Resistencia a Antineoplásicos , Quimioterapia Combinada , Humanos , Idarrubicina/química , Idarrubicina/toxicidad , Leucemia Mieloide Aguda/diagnóstico , Espectrometría de Masas
2.
Chem Res Toxicol ; 31(12): 1293-1304, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30381944

RESUMEN

Platinum drugs, including carboplatin and oxaliplatin, are commonly used chemotherapy drugs that kill cancer cells by forming toxic drug-DNA adducts. These drugs have a proven, but modest, efficacy against several aggressive subtypes of breast cancer but also cause several side effects that can lead to the cessation of treatment. There is a clinical need to identify patients who will respond to platinum drugs in order to better inform clinical decision making. Diagnostic microdosing involves dosing patients or patient samples with subtherapeutic doses of radiolabeled platinum followed by measurement of platinum-DNA adducts in blood or tumor tissue and may be used to predict patient response. We exposed a panel of six breast cancer cell lines to 14C-labeled carboplatin or oxaliplatin at therapeutic and microdose (1% therapeutic dose) concentrations for a range of exposure lengths and isolated DNA from the cells. The DNA was converted to graphite, and measurement of radiocarbon due to platinum-DNA adduction was quantified via accelerator mass spectrometry (AMS). We observed a linear correlation in adduct levels between the microdose and therapeutic dose, and the level of platinum-DNA adducts corresponded to cell line drug sensitivity for both carboplatin and oxaliplatin. These results showed a clear separation in adduct levels between the sensitive and resistant groups of cell lines that could not be fully explained or predicted by changes in DNA repair rates or mutations in DNA repair genes. Further, we were able to quantitate oxaliplatin-DNA adducts in the blood and tumor tissue of a metastatic breast cancer patient. Together, these data support the use of diagnostic microdosing for predicting patient sensitivity to platinum. Future studies will be aimed at replicating this data in a clinical feasibility trial.


Asunto(s)
Complejos de Coordinación/toxicidad , Aductos de ADN/análisis , Daño del ADN/efectos de los fármacos , Platino (Metal)/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carboplatino/química , Carboplatino/toxicidad , Línea Celular Tumoral , Complejos de Coordinación/química , Reparación del ADN/efectos de los fármacos , Femenino , Humanos , Espectrometría de Masas , Oxaliplatino/química , Oxaliplatino/toxicidad
3.
Int J Cancer ; 141(3): 604-613, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28437852

RESUMEN

The platinum-based drugs cisplatin, carboplatin and oxaliplatin are often used for chemotherapy, but drug resistance is common. The prediction of resistance to these drugs via genomics is a challenging problem since hundreds of genes are involved. A possible alternative is to use mass spectrometry to determine the propensity for cells to form drug-DNA adducts-the pharmacodynamic drug-target complex for this class of drugs. The feasibility of predictive diagnostic microdosing was assessed in non-small cell lung cancer (NSCLC) cell culture and a pilot clinical trial. Accelerator mass spectrometry (AMS) was used to quantify [14 C]carboplatin-DNA monoadduct levels in the cell lines induced by microdoses and therapeutic doses of carboplatin, followed by correlation with carboplatin IC50 values for each cell line. The adduct levels in cell culture experiments were linearly proportional to dose (R2 = 0.95, p < 0.0001) and correlated with IC50 across all cell lines for microdose and therapeutically relevant carboplatin concentrations (p = 0.02 and p = 0.01, respectively). A pilot microdosing clinical trial was conducted to define protocols and gather preliminary data. Plasma pharmacokinetics (PK) and [14 C]carboplatin-DNA adducts in white blood cells and tumor tissues from six NSCLC patients were quantified via AMS. The blood plasma half-life of [14 C]carboplatin administered as a microdose was consistent with the known PK of therapeutic dosing. The optimal [14 C]carboplatin formulation for the microdose was 107 dpm/kg of body weight and 1% of the therapeutic dose for the total mass of carboplatin. No microdose-associated toxicity was observed in the patients. Additional accruals are required to significantly correlate adduct levels with response.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino , Carcinoma de Pulmón de Células no Pequeñas/patología , Aductos de ADN , Resistencia a Antineoplásicos , Neoplasias Pulmonares/patología , Anciano , Radioisótopos de Carbono/farmacocinética , Carboplatino/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cisplatino/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Estadificación de Neoplasias , Proyectos Piloto , Pronóstico , Distribución Tisular
4.
Chem Res Toxicol ; 30(1): 388-409, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-27936622

RESUMEN

Biomarker-driven drug selection plays a central role in cancer drug discovery and development, and in diagnostic strategies to improve the use of traditional chemotherapeutic drugs. DNA-modifying anticancer drugs are still used as first line medication, but drawbacks such as resistance and side effects remain an issue. Monitoring the formation and level of DNA modifications induced by anticancer drugs is a potential strategy for stratifying patients and predicting drug efficacy. In this perspective, preclinical and clinical data concerning the relationship between drug-induced DNA adducts and biological response for platinum drugs and combination therapies, nitrogen mustards and half-mustards, hypoxia-activated drugs, reductase-activated drugs, and minor groove binding agents are presented and discussed. Aspects including measurement strategies, identification of adducts, and biological factors that influence the predictive relationship between DNA modification and biological response are addressed. A positive correlation between DNA adduct levels and response was observed for the majority of the studies, demonstrating the high potential of using DNA adducts from anticancer drugs as mechanism-based biomarkers of susceptibility, especially as bioanalysis approaches with higher sensitivity and throughput emerge.


Asunto(s)
Antineoplásicos/uso terapéutico , Aductos de ADN/metabolismo , Animales , Biomarcadores/metabolismo , Humanos , Hipoxia/metabolismo , Compuestos de Mostaza Nitrogenada/uso terapéutico , Oxidorreductasas/metabolismo , Compuestos de Platino/uso terapéutico , Medicina de Precisión , Profármacos/farmacología
5.
Chem Res Toxicol ; 29(11): 1843-1848, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27657672

RESUMEN

Gemcitabine metabolites cause the termination of DNA replication and induction of apoptosis. We determined whether subtherapeutic "microdoses" of gemcitabine are incorporated into DNA at levels that correlate to drug cytotoxicity. A pair of nearly isogenic bladder cancer cell lines differing in resistance to several chemotherapy drugs were treated with various concentrations of 14C-labeled gemcitabine for 4-24 h. Drug incorporation into DNA was determined by accelerator mass spectrometry. A mechanistic analysis determined that RRM2, a DNA synthesis protein and a known resistance factor, substantially mediated gemcitabine toxicity. These results support gemcitabine levels in DNA as a potential biomarker of drug cytotoxicity.


Asunto(s)
Antineoplásicos/farmacología , Desoxicitidina/análogos & derivados , Antineoplásicos/farmacocinética , Área Bajo la Curva , Línea Celular Tumoral , Reparación del ADN , Desoxicitidina/farmacocinética , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Gemcitabina
6.
Chem Res Toxicol ; 28(12): 2250-2, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26544157

RESUMEN

This rapid report focuses on the pharmacodynamic mechanism of the carboplatin/paclitaxel combination and correlates it with its cytotoxicity. Consistent with the synergistic to additive antitumor activity (the combination index ranging from 0.53 to 0.94), cells exposed to this combination had significantly increased carboplatin-DNA adduct formation when compared to that of carboplatin alone (450 ± 30 versus 320 ± 120 adducts per 10(8) nucleotides at 2 h, p = 0.004). Removal of paclitaxel increased the repair of carboplatin-DNA adducts: 39.4 versus 33.1 adducts per 10(8) nucleotides per hour in carboplatin alone (p = 0.021). This rapid report provides the first pharmacodynamics data to support the use of carboplatin/paclitaxel combination in the clinic.


Asunto(s)
Carboplatino/metabolismo , Aductos de ADN/metabolismo , Paclitaxel/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Carboplatino/uso terapéutico , Carboplatino/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Aductos de ADN/uso terapéutico , Aductos de ADN/toxicidad , Sinergismo Farmacológico , Humanos , Paclitaxel/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
7.
Nanomedicine ; 8(7): 1116-24, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22197725

RESUMEN

Peptides featuring the LR(S/T) motif were identified that could specifically bind to the C-type lectin-like molecule-1 (CLL1), a protein preferentially expressed on acute myeloid leukemia stem cells (LSCs). Micellar nanoparticles were covalently decorated with CLL1-targeting peptides for targeted drug delivery. The resulting peptide-coated nanoparticles were 13.5 nm in diameter and could be loaded with 5 mg of daunorubicin per 20 mg of telodendrimers. These "targeting nanomicelles" transported the drug load to the interior of cells expressing CLL1 and to LSCs isolated from clinical specimens in vitro, but did not bind to normal blood or normal hematopoietic stem cells. The presence of CLL1-targeting peptides on the surface of the nanomicelles enabled the improved binding and delivery of substantially more daunorubicin into the cells expressing CLL1 and CD34(+) leukemic cells compared with unmodified nanomicelles. In conclusion, nanomicelles coated with CLL1-targeting peptides are potentially useful for eradicating LSCs and improving leukemia therapy. FROM THE CLINICAL EDITOR: Micellar nanoparticles covalently decorated with targeting peptides were used for targeted drug delivery of daunorubicin to address acute myeloid leukemia stem cells.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Daunorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Lectinas Tipo C/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de la Membrana/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Péptidos/metabolismo , Secuencia de Aminoácidos , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Daunorrubicina/uso terapéutico , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Nanopartículas/química , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Péptidos/química , Unión Proteica
8.
BMJ Open ; 12(6): e060664, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35667714

RESUMEN

INTRODUCTION: The COVID-19 pandemic brought an urgent need to discover novel effective therapeutics for patients hospitalised with severe COVID-19. The Investigation of Serial studies to Predict Your Therapeutic Response with Imaging And moLecular Analysis (ISPY COVID-19 trial) was designed and implemented in early 2020 to evaluate investigational agents rapidly and simultaneously on a phase 2 adaptive platform. This manuscript outlines the design, rationale, implementation and challenges of the ISPY COVID-19 trial during the first phase of trial activity from April 2020 until December 2021. METHODS AND ANALYSIS: The ISPY COVID-19 Trial is a multicentre open-label phase 2 platform trial in the USA designed to evaluate therapeutics that may have a large effect on improving outcomes from severe COVID-19. The ISPY COVID-19 Trial network includes academic and community hospitals with significant geographical diversity across the country. Enrolled patients are randomised to receive one of up to four investigational agents or a control and are evaluated for a family of two primary outcomes-time to recovery and mortality. The statistical design uses a Bayesian model with 'stopping' and 'graduation' criteria designed to efficiently discard ineffective therapies and graduate promising agents for definitive efficacy trials. Each investigational agent arm enrols to a maximum of 125 patients per arm and is compared with concurrent controls. As of December 2021, 11 investigational agent arms had been activated, and 8 arms were complete. Enrolment and adaptation of the trial design are ongoing. ETHICS AND DISSEMINATION: ISPY COVID-19 operates under a central institutional review board via Wake Forest School of Medicine IRB00066805. Data generated from this trial will be reported in peer-reviewed medical journals. TRIAL REGISTRATION NUMBER: NCT04488081.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Insuficiencia Respiratoria , Teorema de Bayes , Humanos , Pandemias , SARS-CoV-2 , Resultado del Tratamiento
9.
Int J Cancer ; 129(6): 1425-34, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21128223

RESUMEN

Formation and repair of platinum (Pt)-induced DNA adducts is a critical step in Pt drug-mediated cytotoxicity. Measurement of Pt-DNA adduct kinetics in tumors may be useful for better understanding chemoresistance and therapeutic response. However, this concept has yet to be rigorously tested because of technical challenges in measuring the adducts at low concentrations and consistent access to sufficient tumor biopsy material. Ultrasensitive accelerator mass spectrometry was used to detect [(14)C]carboplatin-DNA monoadducts at the attomole level, which are the precursors to Pt-DNA crosslink formation, in six cancer cell lines as a proof-of-concept. The most resistant cells had the lowest monoadduct levels at all time points over 24 hr. [(14)C]Carboplatin "microdoses" (1/100th the pharmacologically effective concentration) had nearly identical adduct formation and repair kinetics compared to therapeutically relevant doses, suggesting that the microdosing approach can potentially be used to determine the pharmacological effects of therapeutic treatment. Some of the possible chemoresistance mechanisms were also studied, such as drug uptake/efflux, intracellular inactivation and DNA repair in selected cell lines. Intracellular inactivation and efficient DNA repair each contributed significantly to the suppression of DNA monoadduct formation in the most resistant cell line compared to the most sensitive cell line studied (p < 0.001). Nucleotide excision repair (NER)-deficient and -proficient cells showed substantial differences in carboplatin monoadduct concentrations over 24 hr that likely contributed to chemoresistance. The data support the utility of carboplatin microdosing as a translatable approach for defining carboplatin-DNA monoadduct formation and repair, possibly by NER, which may be useful for characterizing chemoresistance in vivo.


Asunto(s)
Antineoplásicos/metabolismo , Carboplatino/uso terapéutico , Aductos de ADN/metabolismo , Reparación del ADN , Carboplatino/administración & dosificación , Carboplatino/química , Carboplatino/metabolismo , Línea Celular Tumoral , Daño del ADN , Esquema de Medicación , Resistencia a Antineoplásicos , Glutatión/análisis , Humanos , Concentración 50 Inhibidora , Espectrometría de Masas
10.
Bioorg Med Chem Lett ; 20(8): 2448-51, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20335033

RESUMEN

Oxaliplatin is a third-generation platinum-based anticancer drug that is currently used in the treatment of metastatic colorectal cancer. Oxaliplatin, like other platinum-based anticancer drugs such as cisplatin and carboplatin, is known to induce apoptosis in tumor cells by binding to nuclear DNA, forming monoadducts, and intra- and interstrand diadducts. Previously, we reported an accelerator mass spectrometry (AMS) assay to measure the kinetics of oxaliplatin-induced DNA damage and repair [Hah, S. S.; Sumbad, R. A.; de Vere White, R. W.; Turteltaub, K. W.; Henderson, P. T. Chem. Res. Toxicol.2007, 20, 1745]. Here, we describe another application of AMS to the measurement of oxaliplatin-DNA adduct distribution in cultured platinum-sensitive testicular (833K) and platinum-resistant breast (MDA-MB-231) cancer cells, which resulted in elucidation of cell-dependent differentiation of oxaliplatin-DNA adduct formation, implying that differential adduction and/or accumulation of the drug in cellular DNA may be responsible for the sensitivity of cancer cells to platinum treatment. Ultimately, we hope to use this method to measure the intrinsic platinated DNA adduct repair capacity in cancer patients for use as a biomarker for diagnostics or a predictor of patient outcome.


Asunto(s)
Antineoplásicos/química , Biomarcadores de Tumor/análisis , Aductos de ADN/química , Espectrometría de Masas/métodos , Neoplasias/tratamiento farmacológico , Compuestos Organoplatinos/química , Medicina de Precisión , Antineoplásicos/uso terapéutico , Cromatografía Líquida de Alta Presión , Humanos , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino
11.
Mutat Res ; 703(1): 11-7, 2010 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-20833264

RESUMEN

Recent evidence suggests that salvage of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) and 8-oxo-7,8-dihydro-guanine (8-oxoGua) can contribute substantially to levels of 8-oxoGua in DNA and RNA. However, it remains to be determined if this mechanism contributes to mutagenesis and disease. This review covers the predominant methods for detecting 8-oxoGua and its derivatives, summarizes some of the relevant recent DNA repair studies and discusses the mechanisms for metabolism of oxidized guanine derivatives in the (2'-deoxy)ribonucleoside and (2'-deoxy)ribonucleotide pools.


Asunto(s)
Daño del ADN , Reparación del ADN , Guanina/análogos & derivados , Mutación , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Guanina/metabolismo , Humanos , Estrés Oxidativo
12.
Nucleic Acids Res ; 36(1): 228-36, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18025045

RESUMEN

7,8-Dihydro-8-oxo-2'-deoxyguanosine (8-oxodG) is a well-known marker of oxidative stress. We report a mechanistic analysis of several pathways by which 8-oxodG is converted to nucleotide triphosphates and incorporated into both DNA and RNA. Exposure of MCF-7 cells to [(14)C]8-oxodG combined with specific inhibitors of several nucleotide salvage enzymes followed with accelerator mass spectrometry provided precise quantitation of the resulting radiocarbon-labeled species. Concentrations of exogenously dosed nucleobase in RNA reached one per 10(6) nucleotides, 5-6-fold higher than the maximum observed in DNA. Radiocarbon incorporation into DNA and RNA was abrogated by Immucillin H, an inhibitor of human purine nucleoside phosphorylase (PNP). Inhibition of ribonucleotide reductase (RR) decreased the radiocarbon content of the DNA, but not in RNA, indicating an important role for RR in the formation of 8-oxodG-derived deoxyribonucleotides. Inhibition of deoxycytidine kinase had little effect on radiocarbon incorporation in DNA, which is in contrast to the known ability of mammalian cells to phosphorylate dG. Our data indicate that PNP and RR enable nucleotide salvage of 8-oxodG in MCF-7 cells, a previously unrecognized mechanism that may contribute to mutagenesis and carcinogenesis.


Asunto(s)
ADN/metabolismo , Desoxiguanosina/análogos & derivados , Purina-Nucleósido Fosforilasa/metabolismo , ARN/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Radioisótopos de Carbono , Línea Celular Tumoral , ADN/química , Desoxicitidina Quinasa/metabolismo , Desoxiguanosina/química , Desoxiguanosina/metabolismo , Humanos , ARN/química , Ribonucleótido Reductasas/metabolismo
13.
Nucleic Acids Res ; 36(16): e100, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18632763

RESUMEN

Limited sensitivity of existing assays has prevented investigation of whether Adriamycin-DNA adducts are involved in the anti-tumour potential of Adriamycin. Previous detection has achieved a sensitivity of a few Adriamycin-DNA adducts/10(4) bp DNA, but has required the use of supra-clinical drug concentrations. This work sought to measure Adriamycin-DNA adducts at sub-micromolar doses using accelerator mass spectrometry (AMS), a technique with origins in geochemistry for radiocarbon dating. We have used conditions previously validated (by less sensitive decay counting) to extract [(14)C]Adriamycin-DNA adducts from cells and adapted the methodology to AMS detection. Here we show the first direct evidence of Adriamycin-DNA adducts at clinically-relevant Adriamycin concentrations. [(14)C]Adriamycin treatment (25 nM) resulted in 4.4 +/- 1.0 adducts/10(7) bp ( approximately 1300 adducts/cell) in MCF-7 breast cancer cells, representing the best sensitivity and precision reported to date for the covalent binding of Adriamycin to DNA. The exceedingly sensitive nature of AMS has enabled over three orders of magnitude increased sensitivity of Adriamycin-DNA adduct detection and revealed adduct formation within an hour of drug treatment. This method has been shown to be highly reproducible for the measurement of Adriamycin-DNA adducts in tumour cells in culture and can now be applied to the detection of these adducts in human tissues.


Asunto(s)
Antibióticos Antineoplásicos/análisis , Aductos de ADN/análisis , Doxorrubicina/análisis , Espectrometría de Masas/métodos , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/química , Línea Celular Tumoral , Aductos de ADN/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Humanos , Espectrometría de Masas/instrumentación , Aceleradores de Partículas
14.
Mol Cancer Ther ; 19(4): 1070-1079, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32029633

RESUMEN

FOLFOX is one of the most effective treatments for advanced colorectal cancer. However, cumulative oxaliplatin neurotoxicity often results in halting the therapy. Oxaliplatin functions predominantly via the formation of toxic covalent drug-DNA adducts. We hypothesize that oxaliplatin-DNA adduct levels formed in vivo in peripheral blood mononuclear cells (PBMC) are proportional to tumor shrinkage caused by FOLFOX therapy. We further hypothesize that adducts induced by subtherapeutic "diagnostic microdoses" are proportional to those induced by therapeutic doses and are also predictive of response to FOLFOX therapy. These hypotheses were tested in colorectal cancer cell lines and a pilot clinical study. Four colorectal cancer cell lines were cultured with therapeutically relevant (100 µmol/L) or diagnostic microdose (1 µmol/L) concentrations of [14C]oxaliplatin. The C-14 label enabled quantification of oxaliplatin-DNA adduct level with accelerator mass spectrometry (AMS). Oxaliplatin-DNA adduct formation was correlated with oxaliplatin cytotoxicity for each cell line as measured by the MTT viability assay. Six colorectal cancer patients received by intravenous route a diagnostic microdose containing [14C]oxaliplatin prior to treatment, as well as a second [14C]oxaliplatin dose during FOLFOX chemotherapy, termed a "therapeutic dose." Oxaliplatin-DNA adduct levels from PBMC correlated significantly to mean tumor volume change of evaluable target lesions (5 of the 6 patients had measurable disease). Oxaliplatin-DNA adduct levels were linearly proportional between microdose and therapeutically relevant concentrations in cell culture experiments and patient samples, as was plasma pharmacokinetics, indicating potential utility of diagnostic microdosing.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/sangre , Radioisótopos de Carbono/análisis , Neoplasias Colorrectales/patología , Aductos de ADN/sangre , Neoplasias Hepáticas/secundario , Oxaliplatino/sangre , Apoptosis , Proliferación Celular , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Fluorouracilo/administración & dosificación , Humanos , Leucovorina/administración & dosificación , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/tratamiento farmacológico , Oxaliplatino/administración & dosificación , Selección de Paciente , Proyectos Piloto , Pronóstico , Células Tumorales Cultivadas
15.
J Am Chem Soc ; 131(22): 7508-9, 2009 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-19449869

RESUMEN

Hydrogenases constitute a promising class of enzymes for ex vivo hydrogen production. Implementation of such applications is currently hindered by oxygen sensitivity and, in the case of membrane-bound hydrogenases (MBHs), poor water solubility. Nanolipoprotein particles (NLPs) formed from apolipoproteins and phospholipids offer a novel means of incorporating MBHs into a well-defined water-soluble matrix that maintains the enzymatic activity and is amenable to incorporation into more complex architectures. We report the synthesis, hydrogen-evolving activity, and physical characterization of the first MBH-NLP assembly. This may ultimately lead to the development of biomimetic hydrogen-production devices.


Asunto(s)
Apolipoproteínas/química , Enzimas Inmovilizadas/química , Hidrógeno/química , Hidrogenasas/química , Nanopartículas/química , Fosfolípidos/química , Membrana Celular/enzimología , Pyrococcus furiosus/enzimología , Solubilidad , Agua/química
16.
J Biomed Sci ; 16: 54, 2009 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-19534792

RESUMEN

The use of radioisotopes has a long history in biomedical science, and the technique of accelerator mass spectrometry (AMS), an extremely sensitive nuclear physics technique for detection of very low-abundant, stable and long-lived isotopes, has now revolutionized high-sensitivity isotope detection in biomedical research, because it allows the direct determination of the amount of isotope in a sample rather than measuring its decay, and thus the quantitative analysis of the fate of the radiolabeled probes under the given conditions. Since AMS was first used in the early 90's for the analysis of biological samples containing enriched 14C for toxicology and cancer research, the biomedical applications of AMS to date range from in vitro to in vivo studies, including the studies of 1) toxicant and drug metabolism, 2) neuroscience, 3) pharmacokinetics, and 4) nutrition and metabolism of endogenous molecules such as vitamins. In addition, a new drug development concept that relies on the ultrasensitivity of AMS, known as human microdosing, is being used to obtain early human metabolism information of candidate drugs. These various aspects of AMS are reviewed and a perspective on future applications of AMS to biomedical research is provided.


Asunto(s)
Espectrometría de Masas/métodos , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Investigación Biomédica/tendencias , Radioisótopos de Carbono/química , Daño del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/farmacología , Diseño de Equipo , Humanos , Estrés Oxidativo , Aceleradores de Partículas , Farmacocinética , Radioisótopos/farmacología
17.
Methods Mol Biol ; 498: 273-96, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18988032

RESUMEN

Membrane-associated proteins and protein complexes account for approximately a third or more of the proteins in the cell (1, 2). These complexes mediate essential cellular processes; including signal transduc-tion, transport, recognition, bioenergetics and cell-cell communication. In general, membrane proteins are challenging to study because of their insolubility and tendency to aggregate when removed from their protein lipid bilayer environment. This chapter is focused on describing a novel method for producing and solubilizing membrane proteins that can be easily adapted to high-throughput expression screening. This process is based on cell-free transcription and translation technology coupled with nanolipoprotein par ticles (NLPs), which are lipid bilayers confined within a ring of amphipathic protein of defined diameter. The NLPs act as a platform for inserting, solubilizing and characterizing functional membrane proteins. NLP component proteins (apolipoproteins), as well as membrane proteins can be produced by either traditional cell-based or as discussed here, cell-free expression methodologies.


Asunto(s)
Lipoproteínas/metabolismo , Proteínas de la Membrana/aislamiento & purificación , Proteínas Recombinantes/aislamiento & purificación , Animales , Biotinilación , Fraccionamiento Celular/métodos , Escherichia coli/genética , Lipoproteínas/química , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Nanopartículas/química , Análisis por Matrices de Proteínas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Solubilidad
18.
Toxics ; 7(2)2019 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-31075884

RESUMEN

This review summarizes recent developments in radiocarbon tracer technology and applications. Technologies covered include accelerator mass spectrometry (AMS), including conversion of samples to graphite, and rapid combustion to carbon dioxide to enable direct liquid sample analysis, coupling to HPLC for real-time AMS analysis, and combined molecular mass spectrometry and AMS for analyte identification and quantitation. Laser-based alternatives, such as cavity ring down spectrometry, are emerging to enable lower cost, higher throughput measurements of biological samples. Applications covered include radiocarbon dating, use of environmental atomic bomb pulse radiocarbon content for cell and protein age determination and turnover studies, and carbon source identification. Low dose toxicology applications reviewed include studies of naphthalene-DNA adduct formation, benzo[a]pyrene pharmacokinetics in humans, and triclocarban exposure and risk assessment. Cancer-related studies covered include the use of radiocarbon-labeled cells for better defining mechanisms of metastasis and the use of drug-DNA adducts as predictive biomarkers of response to chemotherapy.

19.
Nanoscale ; 10(16): 7420-7430, 2018 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-29564446

RESUMEN

Nanolipoprotein particles (NLPs) are reconstituted high-density lipoproteins, consisting of a phospholipid bilayer stabilized by an apolipoprotein scaffold protein. This class of nanoparticle has been a vital tool in the study of membrane proteins, and in recent years has been increasingly used for in vivo applications. Previous work demonstrated that the composition of the lipid bilayer component affects the stability of these particles in serum solutions. In the current study, NLPs assembled with phosphatidylcholine lipids featuring different acyl chain structures were systematically tested to understand the effect that lipid composition has on NLP stability in both neat serum and cell culture media supplemented with 10% serum by volume. The time at which 50% of the particles dissociate, as well as the fraction of the initial population that remains resistant to dissociation, were correlated to key parameters obtained from all-atom simulations of the corresponding lipid bilayers. A significant correlation was observed between the compressibility modulus of the lipid bilayer and particle stability in these complex biological milieu. These results can be used as a reference to tune the stability of these versatile biological nanoparticles for in vitro and in vivo applications.


Asunto(s)
Apolipoproteínas/química , Membrana Dobles de Lípidos/química , Lipoproteínas HDL/química , Nanopartículas/química , Fosfatidilcolinas/química , Simulación de Dinámica Molecular , Estabilidad Proteica
20.
Mol Cancer Ther ; 17(2): 474-483, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29284644

RESUMEN

Cisplatin-based therapy is highly toxic, but moderately effective in most cancers. Concurrent inhibition of cyclooxygenase-2 (COX-2) and soluble epoxide hydrolase (sEH) results in antitumor activity and has organ-protective effects. The goal of this study was to determine the antitumor activity of PTUPB, an orally bioavailable COX-2/sEH dual inhibitor, in combination with cisplatin and gemcitabine (GC) therapy. NSG mice bearing bladder cancer patient-derived xenografts were treated with vehicle, PTUPB, cisplatin, GC, or combinations thereof. Mouse experiments were performed with two different PDX models. PTUPB potentiated cisplatin and GC therapy, resulting in significantly reduced tumor growth and prolonged survival. PTUPB plus cisplatin was no more toxic than cisplatin single-agent treatment as assessed by body weight, histochemical staining of major organs, blood counts, and chemistry. The combination of PTUPB and cisplatin increased apoptosis and decreased phosphorylation in the MAPK/ERK and PI3K/AKT/mTOR pathways compared with controls. PTUPB treatment did not alter platinum-DNA adduct levels, which is the most critical step in platinum-induced cell death. The in vitro study using the combination index method showed modest synergy between PTUPB and platinum agents only in 5637 cell line among several cell lines examined. However, PTUPB is very active in vivo by inhibiting angiogenesis. In conclusion, PTUPB potentiated the antitumor activity of cisplatin-based treatment without increasing toxicity in vivo and has potential for further development as a combination chemotherapy partner. Mol Cancer Ther; 17(2); 474-83. ©2017 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Ciclooxigenasa 2/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Ciclooxigenasa 2/farmacología , Femenino , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA