Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Labelled Comp Radiopharm ; 66(3): 116-125, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36807307

RESUMEN

[18 F]FTC-146 was introduced as a very potent and selective sigma-1 receptor radioligand, which has shown promising application as an imaging agent for neuropathic pain with positron emission tomography. In line with a multi-laboratory project on animal welfare, we chose this radioligand to investigate its potential for detecting neuropathic pain and tissue damage in tumor-bearing animals. However, the radiochemical yield (RCY) of around 4-7% was not satisfactory to us, and efforts were made to improve it. Herein, we describe an improved approach for the radiosynthesis of [18 F]FTC-146 resulting in a RCY, which is sevenfold higher than that previously reported. A tosylate precursor was synthesized and radio-fluorination experiments were performed via aliphatic nucleophilic substitution reactions using either K[18 F]F-Kryptofix®222 (K2.2.2 )-carbonate system or tetra-n-butylammonium [18 F]fluoride ([18 F]TBAF). Several parameters affecting the radiolabeling reaction such as solvent, 18 F-fluorination agent with the corresponding amount of base, labeling time, and temperature were investigated. Best labeling reaction conditions were found to be [18 F]TBAF and acetonitrile as solvent at 100°C. The new protocol was then translated to an automated procedure using a FX2 N synthesis module. Finally, the radiotracer reproducibly obtained with RCYs of 41.7 ± 4.4% in high radiochemical purity (>98%) and molar activities up to 171 GBq/µmol.


Asunto(s)
Tomografía de Emisión de Positrones , Receptores sigma , Animales , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Radioisótopos de Flúor , Solventes , Receptor Sigma-1
4.
EJNMMI Res ; 12(1): 60, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36104637

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) lacks biomarkers for targeted therapy. Auger emitters display the best therapeutic effect, if delivered directly into the nucleus proximal to DNA. The nuclear protein Poly (ADP-ribose)-Polymerase 1 (PARP1) is a suitable target against which few inhibitors (PARPi) are clinically approved for treatment of breast cancer with germline BRCA mutation (BRCAmut). In this study, a theranostic approach was investigated in a TNBC xenografted mouse model by radiolabelling a close derivative of a PARPi Olaparib (termed PARPi-01) with the Auger emitters 123/125I. METHODS: TNBC cell line MDA-MB-231 was subcutaneously implanted in female NOD/SCID mice. At a tumour size of ~ 500mm3, [123I]PARPi-01 was administered intravenously, and SPECT/CT images were obtained at 4 h or 24 h post injection (p.i). A therapy study was performed with [125I]PARPi-01 in 4 doses (10 MBq/dose, 10 days apart). Tumour growth was monitored by CT scans longitudinally once per week. Upon reaching study endpoint, tissues were harvested and stained with TUNEL assay for detection of apoptosis induction. RESULTS: SPECT/CT images showed rapid hepatobiliary tracer clearance at 4 h post injection (p.i.). Retention in thyroid at 24 h p.i. suggested tracer deiodination in vivo. The tumour and liver uptake were 0.2%ID/g and 2.5%ID/g, respectively. The tumour: blood ratio was 1.3. Endogenous therapy induced a significant delay in tumour growth (doubling time increased from 8.3 to 14.2 days), but no significant survival advantage. Significantly higher apoptosis ratio was observed in [125I]PARPi-01 treated tumour tissues. No radiotoxicity was detected in the liver and thyroid. CONCLUSION: Considering the radio-cytotoxic effect in the tumour tissue and a delay on tumour doubling time, [125I]PARPi-01 presents a potential radiotherapeutics for treatment of TNBC. Improvements to overcome the suboptimal pharmacokinetics are necessary for its potential clinical application.

5.
Cancers (Basel) ; 14(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35008392

RESUMEN

PARP1 inhibitors (PARPi) are currently approved for BRCAmut metastatic breast cancer, but they have shown limited response in triple negative breast cancer (TNBC) patients. Combination of an Auger emitter with PARPis enables PARP inhibition and DNA strand break induction simultaneously. This will enhance cytotoxicity and additionally allow a theranostic approach. This study presents the radiosynthesis of the Auger emitter [125I] coupled olaparib derivative: [125I]-PARPi-01, and its therapeutic evaluation in a panel of TNBC cell lines. Specificity was tested by a blocking assay. DNA strand break induction was analysed by γH2AX immunofluorescence staining. Cell cycle analysis and apoptosis assays were studied using flow cytometry in TNBC cell lines (BRCAwt/mut). Anchorage independent growth potential was evaluated using soft agar assay. [125I]-PARPi-01 showed PARP1-specificity and higher cytotoxicity than olaparib in TNBC cell lines irrespective of BRCA their status. Cell lines harbouring DNA repair deficiency showed response to [125I]-PARPi-01 monotherapy. Combined treatment with Dox-NP further enhanced therapeutic efficiency in metastatic resistant BRCAwt cell lines. The clonogenic survival was significantly reduced after treatment with [125I]-PARPi-01 in all TNBC lines investigated. Therapeutic efficacy was further enhanced after combined treatment with chemotherapeutics. [125I]-PARPi-01 is a promising radiotherapeutic agent for low radiation dosages, and mono/combined therapies of TNBC.

6.
Pharmaceuticals (Basel) ; 14(8)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34451915

RESUMEN

Over the past 20 years, 68Ga-labelled radiopharmaceuticals have become an important part in clinical routine. However, the worldwide supply with 68Ge/68Ga generators is limited as well as the number of patient doses per batch of 68Ga radiopharmaceutical. In the recent years, a new technique appeared, making use of the ease of aqueous labelling via chelators as with 68Ga but using 18F instead. This technique takes advantage of the strong coordinative bond between aluminium and fluoride, realized in the aqueous cation [Al18F]2+. Most applications to date make use of one-pot syntheses with free Al(III) ions in the system. In contrast, we investigated the labelling approach split into two steps: generating the Al-bearing precursor in pure form and using this Al compound as a precursor in the labelling step with aqueous [18F]fluoride. Hence, no free Al3+ ions are present in the labelling step. We investigated the impact of parameters: temperature, pH, addition of organic solvent, and reaction time using the model chelator NH2-MPAA-NODA. With optimized parameters we could stably achieve a 80% radiochemical yield exerting a 30-min reaction time at 100 °C. This technique has the potential to become an important approach in radiopharmaceutical syntheses.

7.
Breast Cancer Res ; 21(1): 116, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31640747

RESUMEN

BACKGROUND: Triple-negative breast cancer has extremely high risk of relapse due to the lack of targeted therapies, intra- and inter-tumoral heterogeneity, and the inherent and acquired resistance to therapies. In this study, we evaluate the potential of prostate-specific membrane antigen (PSMA) as target for radio-ligand therapy (RLT). METHODS: Tube formation was investigated after incubation of endothelial HUVEC cells in tumor-conditioned media and monitored after staining using microscopy. A binding study with 68Ga-labeled PSMA-addressing ligand was used to indicate targeting potential of PSMA on tumor-conditioned HUVEC cells. For mimicking of the therapeutic application, tube formation potential and vitality of tumor-conditioned HUVEC cells were assessed following an incubation with radiolabeled PSMA-addressing ligand [177Lu]-PSMA-617. For in vivo experiments, NUDE mice were xenografted with triple-negative breast cancer cells MDA-MB231 or estrogen receptor expressing breast cancer cells MCF-7. Biodistribution and binding behavior of [68Ga]-PSMA-11 was investigated in both tumor models at 30 min post injection using µPET. PSMA- and CD31-specific staining was conducted to visualize PSMA expression and neovascularization in tumor tissue ex vivo. RESULTS: The triple-negative breast cancer cells MDA-MB231 showed a high pro-angiogenetic potential on tube formation of endothelial HUVEC cells. The induced endothelial expression of PSMA was efficiently addressed by radiolabeled PSMA-specific ligands. 177Lu-labeled PSMA-617 strongly impaired the vitality and angiogenic potential of HUVEC cells. In vivo, as visualized by µPET, radiolabeled PSMA-ligand accumulated specifically in the triple-negative breast cancer xenograft MDA-MB231 (T/B ratio of 43.3 ± 0.9), while no [68Ga]-PSMA-11 was detected in the estrogen-sensitive MCF-7 xenograft (T/B ratio of 1.1 ± 0.1). An ex vivo immunofluorescence analysis confirmed the localization of PSMA on MDA-MB231 xenograft-associated endothelial cells and also on TNBC cells. CONCLUSIONS: Here we demonstrate PSMA as promising target for two-compartment endogenous radio-ligand therapy of triple-negative breast cancer.


Asunto(s)
Radioisótopos de Galio/uso terapéutico , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Lutecio/uso terapéutico , Radioisótopos/uso terapéutico , Neoplasias de la Mama Triple Negativas/radioterapia , Animales , Antígenos de Superficie/metabolismo , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/fisiología , Vasos Sanguíneos/efectos de la radiación , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Dipéptidos/metabolismo , Dipéptidos/uso terapéutico , Ácido Edético/análogos & derivados , Ácido Edético/metabolismo , Ácido Edético/uso terapéutico , Isótopos de Galio , Glutamato Carboxipeptidasa II/metabolismo , Compuestos Heterocíclicos con 1 Anillo/metabolismo , Compuestos Heterocíclicos con 1 Anillo/uso terapéutico , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células Endoteliales de la Vena Umbilical Humana/efectos de la radiación , Humanos , Ligandos , Células MCF-7 , Ratones Desnudos , Oligopéptidos/metabolismo , Oligopéptidos/uso terapéutico , Antígeno Prostático Específico , Radiofármacos/uso terapéutico , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Appl Radiat Isot ; 149: 75-82, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31029937

RESUMEN

The radiochemical separation of n.c.a. arsenic on its own or for radio-labelling purposes usually involves the issue of reducing arsenic(V). Numerous approaches for reducing pentavalent arsenic have been examined. A novel HPLC method has also been presented for accessing the efficiency of the reduction in terms of *As(III)/*As(V). Labelling with trivalent radioarsenic seems to be a promising research field to access new radiopharmaceuticals, for example, using arsenic as a surrogate for phosphorus. Moreover, as a model system, the labelling reaction of *As(III) with dihydrolipoic acid has been systematically optimized.

9.
Nuklearmedizin ; 57(1): 19-25, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29536496

RESUMEN

AIM: In metastatic prostate cancer patients PSMA targeting radioligands have gained significant impact as theranostic probes. In this study a correlation between total tumor volume (TTV) and measured kidney dose as well as salivary glands (SG) uptake in 177Lu-PSMA-617 therapy was evaluated. METHODS: Eleven consecutive prostate cancer patients receiving a first cylcle of 177Lu-PSMA-617 (administered activity of approximately 6GBq) were included. The 68Ga-PSMA-11 PET/CT scan previous to therapy was used to determine TTV and SG uptake (glandulae submandibularis) employing PMOD version 3.403 with different 68Ga-PSMA-11 thresholds based on the standardized uptake value (SUV).The kidney dose was estimated with the software ULMDOS using planar whole-body scintigrams. RESULTS: Kidney dose and SG uptake was inversely correlated to TTV, indicating high kidney dose and high SG uptake in case of low tumor load and low kidney dose and low SG uptake in case of high tumor load. CONCLUSION: Our data support the hypothesis that in 177Lu-PSMA-617 therapy an individualized treatment activity based on total tumor volume could be beneficiary.


Asunto(s)
Dipéptidos/uso terapéutico , Compuestos Heterocíclicos con 1 Anillo/uso terapéutico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Anciano , Ácido Edético/análogos & derivados , Isótopos de Galio , Radioisótopos de Galio , Humanos , Riñón/diagnóstico por imagen , Riñón/metabolismo , Lutecio , Masculino , Persona de Mediana Edad , Oligopéptidos , Tomografía Computarizada por Tomografía de Emisión de Positrones , Antígeno Prostático Específico , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Radiofármacos , Glándulas Salivales/diagnóstico por imagen , Glándulas Salivales/metabolismo , Carga Tumoral
10.
FASEB J ; 32(5): 2803-2813, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29301945

RESUMEN

Triple-negative breast cancer has an extremely high rate of relapse. This is particularly due to the existence and survival of cancer stem cells (CSCs) characterized by increased amounts of glutathione (GSH). In this study, we evaluated the potential of pharmacological GSH depletion to sensitize CSCs to ionizing radiotherapy with an I-125-labeled nucleoside analog, 5-iodo-4'-thio-2'-deoxyuridine (ITdU). CSCs were isolated using CD24-- and CD44+-specific microbeads. GSH and reactive oxygen species (ROS) were evaluated by fluorescence-activated cell sorting. GSH synthesis was inhibited with buthionine sulfoximine (BSO). Apoptotic cells were identified with propidium iodide and double-strand DNA breaks were detected by γ-H2AX staining. For therapy study, BSO treated and untreated mice xenografted with breast CSCs received weekly I-125-ITdU. Therapy efficiency was monitored by fluorodeoxyglucose-18-µ-positron emission tomography. We showed that GSH modulation sensitizes CD24- and CD44+ breast cancer cells to endogenous nanoradiotherapy. BSO synergistically affects ROS generation induced by I-125-ITdU. In an in vivo study, we demonstrated a complete tumor regression as a consequence of preconditioning with a GSH-synthesis inhibitor prior to treatment with I-125-ITdU. GSH modulation in combination with an oxidative stress-generating treatment such as endogenous radiotherapy using an Auger emitter offers an extraordinary opportunity for selective and efficient eradication of drug-resistant CSCs.-Miran, T., Vogg, A. T. J., Drude, N., Mottaghy, F. M., Morgenroth, A. Modulation of glutathione promotes apoptosis in triple-negative breast cancer cells.


Asunto(s)
Roturas del ADN de Doble Cadena , Glutatión/metabolismo , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Butionina Sulfoximina/farmacología , Línea Celular Tumoral , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacología , Femenino , Fluorodesoxiglucosa F18/farmacología , Glutatión/antagonistas & inhibidores , Humanos , Ratones , Ratones Desnudos , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/radioterapia , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Med ; 6(7): 1639-1651, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28608446

RESUMEN

Here, we examined the potential of blocking the thymidine de novo synthesis pathways for sensitizing melanoma cells to the nucleoside salvage pathway targeting endogenous DNA irradiation. Expression of key nucleotide synthesis and proliferation enzymes thymidylate synthase (TS) and thymidine kinase 1 (TK1) was evaluated in differentiated (MITFhigh [microphthalmia-associated transcription factor] IGR1) and invasive (MITFmedium IGR37) melanoma cells. For inhibition of de novo pathways cells were incubated either with an irreversible TS inhibitor 5-fluoro-2'-deoxyuridine (FdUrd) or with a competitive dihydrofolate-reductase (DHFR) inhibitor methotrexate (MTX). Salvage pathway was addressed by irradiation-emitting thymidine analog [123/125 I]-5-iodo-4'-thio-2'-deoxyuridine (123/125 I-ITdU). The in vivo targeting efficiency was visualized by single-photon emission computed tomography. Pretreatment with FdUrd strongly increased the cellular uptake and the DNA incorporation of 125 I-ITdU into the mitotically active IGR37 cells. This effect was less pronounced in the differentiated IGR1 cells. In vivo, inhibition of TS led to a high and preferential accumulation of 123 I-ITdU in tumor tissue. This preclinical study presents profound rationale for development of therapeutic approach by highly efficient and selective radioactive targeting one of the crucial salvage pathways in melanomas.


Asunto(s)
Antineoplásicos/farmacología , Vías Biosintéticas/efectos de los fármacos , Melanoma/metabolismo , Timidina/biosíntesis , Animales , Antineoplásicos/uso terapéutico , Biomarcadores , Vías Biosintéticas/efectos de la radiación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Glutatión/metabolismo , Humanos , Radioisótopos de Yodo , Melanoma/diagnóstico por imagen , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Mitosis/efectos de los fármacos , Mitosis/genética , Imagen Molecular , Terapia Molecular Dirigida , Nucleósidos/metabolismo , Oxidación-Reducción , Radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
12.
Oncotarget ; 8(11): 18059-18069, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-28407689

RESUMEN

Cyclooxygenase-2 (COX-2) is an important biomarker in several tumors. Available imaging probes display relatively low tumor to background ratios (smaller than 2:1). We evaluated newly developed indomethacin (Ind) derivatives for in vivo molecular imaging of COX-2 expressing carcinoma. Radioiodinated Ind derivatives Ind-NH-(CH2)4-NH-3-[I-125]I-Bz ([I-125]5), Ind-NH-(CH2)4-NH-5-[I-124/125]I-Nic ([I-124/125]6) and Ind-NH-(CH2)4-NH-5-[I-125]I-Iphth ([I-125]7) were prepared from the respective SnBu3-precursors (45-80% radiochemical yield; > 95% radiochemical purity). The cellular uptake of [I-125]5 and [I-125]6 correlated with COX-2 expression determined by SDS page/Western blot analysis. [I-125]5 was predominantly localized in the cell membrane while [I-125]6 was internalized and displayed a diffuse and favorable cytoplasmic distribution. In contrast, [I-125]7 showed only low uptake in COX-2 positive cells. Co-incubation with the COX-2 inhibitor Celecoxib led to an almost complete suppression of cellular uptake of [I-125]5 and [I-125]6. In vivo molecular imaging using positron emission tomography (PET) in SCID mice xenografted with COX-2+ (HT29) and COX-2- (HCT116) human colorectal carcinoma cells was performed for [I-124]6. HT29 xenografts displayed a significantly higher uptake than HCT-116 xenografts (5.6 ± 1.5 vs. 0.5 ± 0.1 kBq/g, P < 0.05) with an extraordinary high tumor to muscle ratio (50.3 ± 1.5). Immunohistological staining correlated with the imaging data. In conclusion, the novel radioiodinated indomethacin derivative ([I-124/125]6) could become a valuable tool for development of molecular imaging probes for visualization of COX-2 expressing tumors.


Asunto(s)
Ciclooxigenasa 2/análisis , Indometacina , Neoplasias Experimentales/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Animales , Western Blotting , Línea Celular Tumoral , Xenoinjertos , Humanos , Inmunohistoquímica , Radioisótopos de Yodo , Ratones , Ratones SCID
13.
Adv Healthc Mater ; 5(24): 3118-3128, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27860428

RESUMEN

As the size of nanoparticles (NPs) is in the range of biological molecules and subcellular structures, they provide new perspectives in biomedicine. This work presents studies concerning the cellular uptake and distribution of phosphine-stabilized cytotoxic 1.4 nm sized AuNPs and their probable degradation during this process. Therefore, ultrasmall phosphine-stabilized AuNPs are modified by linking a fluorophore covalently to the ligand shell. Monitoring the fluorescence on a cellular level by means of flow cytometry and confocal laser scanning microscopy allows determining the fate of the ligand shell during AuNP cell internalization, due to the fact that the fluorescence of a fluorophore bound near to the AuNP surface is quenched. Cell fractionation is conducted in order to quantify the AuNP content at the cell membrane, in the cytoplasm, and the cell nucleus. The incubation of cells with the fluorophore-modified AuNPs reveals a partial loss of the ligand shell upon AuNP cell interaction, evident by the emerging fluorescence signal. This loss is the precondition to unfold high AuNP cytotoxicity. Together with their significantly different biodistribution and enhanced circulation times compared to larger AuNPs, the findings demonstrate the high potential of ultrasmall AuNPs for drug development or therapy.


Asunto(s)
Oro/metabolismo , Nanopartículas del Metal/administración & dosificación , Fosfinas/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Fluorescencia , Células HeLa , Células Hep G2 , Humanos , Tamaño de la Partícula , Propiedades de Superficie , Distribución Tisular
14.
Oncotarget ; 5(14): 5483-93, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24978848

RESUMEN

The existence of therapy resistant glioma stem cells is responsible for the high recurrence rate and incurability of glioblastomas. The Hedgehog pathway activity plays an essential role for self-renewal capacity and survival of glioma stem cells. We examined the potential of the Sonic hedgehog ligand for sensitizing of glioma stem cells to endogenous nano-irradiation. We demonstrate that the Sonic hedgehog ligand preferentially and efficiently activated glioma stem cells to enter the radiation sensitive G2/M phase. Concomitant inhibition of de novo thymidine synthesis with fluorodeoxyuridine and treatment with the Auger electron emitting thymidine analogue 5-[I-125]-Iodo-4'-thio-2'-deoxyuridine ([I-125]ITdU) leads to a fatal nano-irradiation in sensitized glioma stem cells. Targeting of proliferating glioma stem cells with DNA-incorporated [I-125]ITdU efficiently invokes the intrinsic apoptotic pathway despite active DNA repair mechanisms. Further, [I-125]ITdU completely inhibits survival of glioma stem cells in vitro. Analysis of non-stem glioblastoma cells and normal human astrocytes reveals that glioma stem cells differentially respond to Sonic hedgehog ligand. These data demonstrate a highly efficient and controllable single-cell kill therapeutic model for targeting glioma stem cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Proteínas Hedgehog/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Apoptosis/efectos de la radiación , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Desoxiuridina/administración & dosificación , Desoxiuridina/análogos & derivados , Glioblastoma/patología , Humanos , Radioisótopos de Yodo/administración & dosificación , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Tolerancia a Radiación , Radiofármacos/administración & dosificación , Transducción de Señal/efectos de la radiación
15.
Mol Cancer Ther ; 13(1): 144-53, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24174494

RESUMEN

In multiple myeloma, the presence of highly resistant cancer stem cells (CSC) that are responsible for tumor metastasis and relapse has been proven. Evidently, for achieving complete response, new therapeutic paradigms that effectively eradicate both, CSCs and bulk cancer populations, need to be developed. For achieving that goal, an innovative two-step treatment combining targeting of thymidine de novo synthesis pathway and a nanoirradiation by the Auger electron emitting thymidine analogue (123/125)I-5-iodo-4'-thio-2'-deoxyuridine ((123/125)I-ITdU) could be a promising approach. The pretreatment with thymidylate synthase inhibitor 5-fluoro-2'-deoxyuridine (FdUrd, 1 µmol/L for 1 hour) efficiently induced proliferation and terminal differentiation of isolated myeloma stem-like cells. Moreover, FdUrd stimulation led to a decreased activity of a functional CSC marker, aldehyde dehydrogenase (ALDH). The metabolic conditioning by FdUrd emerged to be essential for enhanced incorporation of (125)I-ITdU (incubation with 50 kBq/2 × 10(4) cells for 4 days) and, consequently, for the induction of irreparable DNA damage. (125)I-ITdU showed a pronounced antimyeloma effect on isolated tumor stem-like cells. More than 85% of the treated cells were apoptotic, despite activation of DNA repair mechanisms. Most important, exposure of metabolically conditioned cells to (125)I-ITdU resulted in a complete inhibition of clonogenic recovery. This is the first report showing that pretreatment with FdUrd sensitizes the stem-like cell compartment in multiple myeloma to apoptosis induced by (125)I-ITdU-mediated nanoirradiation of DNA.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Timidilato Sintasa/genética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Linaje de la Célula/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Desoxiuridina/administración & dosificación , Desoxiuridina/análogos & derivados , Inhibidores Enzimáticos/administración & dosificación , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Timidilato Sintasa/antagonistas & inhibidores
16.
Biomed Tech (Berl) ; 59(2): 165-75, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24021591

RESUMEN

Abstract Tissue engineering as a multidisciplinary field enables the development of living substitutes to replace, maintain, or restore diseased tissue and organs. Since the term was introduced in medicine in 1987, tissue engineering strategies have experienced significant progress. However, up to now, only a few substitutes were able to overcome the gap from bench to bedside and have been successfully approved for clinical use. Substantial donor variability makes it difficult to predict the quality of tissue-engineered constructs. It is essential to collect sufficient data to ensure that poor or immature constructs are not implanted into patients. The fulfillment of certain quality requirements, such as mechanical and structural properties, is crucial for a successful implantation. There is a clear need for new nondestructive and real-time online monitoring and evaluation methods for tissue-engineered constructs, which are applicable on the biomaterial, tissue, cellular, and subcellular levels. This paper reviews current established nondestructive techniques for implant monitoring including biochemical methods and noninvasive imaging.


Asunto(s)
Células Cultivadas/citología , Células Cultivadas/fisiología , Diagnóstico por Imagen/métodos , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Diseño de Equipo , Análisis de Falla de Equipo , Humanos
17.
Cancer Biother Radiopharm ; 23(6): 819-24, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19111052

RESUMEN

The monoclonal antibody anti-CD66 labeled with (99m)Tc is widely used as Scintimun granulocyte for bone marrow immunoscintigraphy. Further, recently performed clinical radioimmunotherapy studies with [(90)Y]Y-anti-CD66 proved to be suitable for the treatment of hematologic malignancies. Before radioimmunotherapy with [(90)Y]Y-anti-CD66, dosimetric estimations are required to minimize radiotoxicity and determine individual applicable activities. Planar imaging, using gamma-emitting radionuclides, is conventionally carried out to estimate the absorbed organ doses. In contrast, immuno-PET (positron emission tomography) enables the quantification of anti-CD66 accumulation and provides better spatial and temporal resolution. Therefore, in this study, a semiautomated radiosynthesis of [(18)F]F-anti-CD66 was developed, using the (18)F-acylation agent, N-succinimidyl-4-[(18)F]fluorobenzoate ([(18)F]SFB). As a proof of concept, an intraindividual comparison between PET and conventional scintigraphy, using (18)F- and (99m)Tc-labeled anti-CD66 in 1 patient with high-risk leukemia, is presented. Both labeled antibodies displayed a similar distribution pattern with high preferential uptake in bone marrow. Urinary excretion of [(18)F]F-anti-CD66 was increased and bone marrow uptake reduced, in comparison to [(99m)Tc]Tc-anti-CD66. Nevertheless, PET-based dosimetry with [(18)F]F-anti-CD66 could provide additional information to support conventional scintigraphy. Moreover, [(18)F]F-anti-CD66 is ideally suited for bone marrow imaging using PET.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antígenos CD/inmunología , Moléculas de Adhesión Celular/inmunología , Radioisótopos de Flúor/farmacocinética , Leucemia Mieloide Aguda/metabolismo , Tomografía de Emisión de Positrones , Radioinmunoterapia , Adulto , Médula Ósea/diagnóstico por imagen , Humanos , Leucemia Mieloide Aguda/terapia , Masculino , Factores de Riesgo , Distribución Tisular
18.
Clin Cancer Res ; 14(22): 7311-9, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010846

RESUMEN

PURPOSE: Auger electron emitting radiopharmaceuticals are attractive for targeted nanoirradiation therapy, provided that DNA of malignant cells is selectively addressed. Here, we examine 5-[123/125/131I]iodo-4'-thio-2'-deoxyuridine (ITdU) for targeting DNA in tumor cells in a HL60 xenograft severe combined immunodeficient mouse model. EXPERIMENTAL DESIGN: Thymidine kinase and phosphorylase assays were done to determine phosphorylation and glycosidic bond cleavage of ITdU, respectively. The biodistribution and DNA incorporation of ITdU were determined in severe combined immunodeficient mice bearing HL60 xenografts receiving pretreatment with 5-fluoro-2'-deoxyuridine (FdUrd). Organ tissues were dissected 0.5, 4, and 24 h after radioinjection and uptake of [131I]ITdU (%ID/g tissue) was determined. Cellular distribution of [125I]ITdU was imaged by microautoradiography. Apoptosis and expression of the proliferation marker Ki-67 were determined by immunohistologic staining using corresponding paraffin tissue sections. RESULTS: ITdU is phosphorylated by thymidine kinase 1 and stable toward thymidylate phosphatase-mediated glycosidic bond cleavage. Thymidylate synthase-mediated deiodination of [123/125/131I]ITdU was inhibited with FdUrd. Pretreatment with FdUrd increased preferentially tumor uptake of ITdU resulting in favorable tumor-to-normal tissue ratios and tumor selectivity. ITdU was exclusively localized within the nucleus and incorporated into DNA. In FdUrd-pretreated animals, we found in more than 90% of tumor cells apoptosis induction 24 h postinjection of ITdU, indicating a highly radiotoxic effect in tumor cells but not in cells of major proliferating tissues. CONCLUSION: ITdU preferentially targets DNA in proliferating tumor cells and leads to apoptosis provided that the thymidylate synthase is inhibited.


Asunto(s)
Desoxiuridina/análogos & derivados , Neoplasias Experimentales/tratamiento farmacológico , Radiofármacos/farmacocinética , Radiofármacos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Autorradiografía , ADN/efectos de los fármacos , Desoxiuridina/farmacocinética , Desoxiuridina/uso terapéutico , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones SCID , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/efectos de los fármacos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Eur J Nucl Med Mol Imaging ; 34(11): 1775-82, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17541585

RESUMEN

PURPOSE: The aim of this study was to determine whether the thymidine analogue 3'-deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT) is adequate for early evaluation of the response of malignant lymphoma to antiproliferative treatment in a mouse xenotransplant model. METHODS: Immunodeficient mice bearing a follicular lymphoma xenotransplant were treated with high-dose chemotherapy (cyclophosphamide, n = 10), immunotherapy (CD20 mAb, ibritumomab-tiuxetan, n = 10) or radioimmunotherapy ([(90)Y]CD20 mAb, Zevalin, n = 10). Forty-eight hours after treatment, antiproliferative effects were assessed with [(18)F]FLT. Ninety minutes after i.v. injection of 5-10 MBq [(18)F]FLT, mice were sacrificed and radioactivity within the tumour and normal organs was measured using a gamma counter and calculated as % ID/g. The proliferation fraction in tissue samples derived from treated and untreated tumours was evaluated by Ki-67 immunohistochemistry, which served as the reference for proliferative activity. RESULTS: In untreated lymphoma, the mean proliferation fraction was 83.6%. After chemotherapy, the mean proliferation fraction decreased to 39.3% (p = 0.0001), after immunotherapy to 77.6% (p = 0.0078) and after radioimmunotherapy to 78.8% (p = 0.014). In none of the animals was a significant change in tumour size observed. In untreated lymphoma, tumoural [(18)F]FLT uptake was 5.4% ID/g, after chemotherapy it was 1.5% (p = 0.0005), after immunotherapy, 3.9% (non-significant), and after radioimmunotherapy, 5.8% (non-significant). CONCLUSION: In a lymphoma xenotransplant model, [(18)F]FLT detects early antiproliferative drug activity before changes in tumour size are visible. These findings further support the use of [(18)F]FLT-PET for imaging early response to treatment in malignant lymphoma.


Asunto(s)
Didesoxinucleósidos/farmacocinética , Linfoma/diagnóstico por imagen , Linfoma/terapia , Animales , Humanos , Linfoma/metabolismo , Ratones , Cintigrafía , Radiofármacos/farmacocinética , Resultado del Tratamiento
20.
J Nucl Med ; 48(6): 1000-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17504875

RESUMEN

UNLABELLED: Resistance to radiotherapy or chemotherapy is a common cause of treatment failure in high-risk leukemias. We evaluated whether selective nanoirradiation of DNA with Auger electrons emitted by 5-(123)I-iodo-4'-thio-2'-deoxyuridine ((123)I-ITdU) can induce cell kill and break resistance to doxorubicin, beta-, and gamma-irradiation in leukemia cells. METHODS: 4'-thio-2'-deoxyuridine was radiolabeled with (123/131)I and purified by high-performance liquid chromatography. Cellular uptake, metabolic stability, DNA incorporation of (123)I-ITdU, and the effect of the thymidylate synthase (TS) inhibitor 5-fluoro-2'-deoxyuridine (FdUrd) were determined in HL60 leukemia cells. DNA damage was assessed with the comet assay and quantified by the olive tail moment. Apoptosis induction and irradiation-induced apoptosis inhibition by benzoylcarbonyl-Val-Ala-Asp-fluoromethyl ketone (z-VAD.fmk) were analyzed in leukemia cells using flow cytometry analysis. RESULTS: The radiochemical purity of ITdU was 95%. Specific activities were 900 GBq/micromol for (123)I-ITdU and 200 GBq/micromol for (131)I-ITdU. An in vitro cell metabolism study of (123)I-ITdU with wild-type HL60 cells demonstrated an uptake of 1.5% of the initial activity/10(6) cells of (123)I-ITdU. Ninety percent of absorbed activity from (123)I-ITdU in HL60 cells was specifically incorporated into DNA. (123)I-ITdU caused extensive DNA damage (olive tail moment > 12) and induced more than 90% apoptosis in wild-type HL60 cells. The broad-spectrum inhibitor of caspases zVAD-fmk reduced (123)I-ITdU-induced apoptosis from more than 90% to less than 10%, demonstrating that caspases were central for (123)I-ITdU-induced cell death. Inhibition of TS with FdUrd increased DNA uptake of (123)I-ITdU 18-fold and the efficiency of cell kill about 20-fold. In addition, (123)I-ITdU induced comparable apoptotic cell death (>90%) in sensitive parental leukemia cells and in leukemia cells resistant to beta-irradiation, gamma-irradiation, or doxorubicin at activities of 1.2, 4.1, 12.4, and 41.3 MBq/mL after 72 h. This finding indicates that (123)I-ITdU breaks resistance to beta-irradiation, gamma-irradiation, and doxorubicin in leukemia cells. CONCLUSION: (123)I-ITdU-mediated nanoirradiation of DNA efficiently induced apoptosis in sensitive and resistant leukemia cells against doxorubicin, beta-irradiation, and gamma-irradiation and may provide a novel treatment strategy for overcoming resistance to conventional radiotherapy or chemotherapy in leukemia. Cellular uptake and cell kill are highly amplified by inhibiting TS with FdUrd.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Daño del ADN , Desoxiuridina/análogos & derivados , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Tolerancia a Radiación , Radiofármacos/farmacología , Partículas beta , Línea Celular Tumoral , Desoxiuridina/uso terapéutico , Rayos gamma , Células HL-60 , Humanos , Radioisótopos de Yodo , Nanotecnología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...