Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 27(3)2022 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-35164002

RESUMEN

Carboxylic acid is a commonly utilized functional group for covalent surface conjugation of carbon nanoparticles that is typically generated by acid oxidation. However, acid oxidation generates additional oxygen containing groups, including epoxides, ketones, aldehydes, lactones, and alcohols. We present a method to specifically enrich the carboxylic acid content on fluorescent nanodiamond (FND) surfaces. Lithium aluminum hydride is used to reduce oxygen containing surface groups to alcohols. The alcohols are then converted to carboxylic acids through a rhodium (II) acetate catalyzed carbene insertion reaction with tert-butyl diazoacetate and subsequent ester cleavage with trifluoroacetic acid. This carboxylic acid enrichment process significantly enhanced nanodiamond homogeneity and improved the efficiency of functionalizing the FND surface. Biotin functionalized fluorescent nanodiamonds were demonstrated to be robust and stable single-molecule fluorescence and optical trapping probes.

2.
Eur J Nucl Med Mol Imaging ; 49(1): 64-72, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34378064

RESUMEN

BACKGROUND: Radiopharmaceutical targeted therapy (RPT) has been studied for decades; however, recent clinical trials demonstrating efficacy have helped renewed interest in the modality. METHODS: This article reviews National Cancer Institute (NCI)'s support of RPT through communication via workshops and interest groups, through funding extramural programs in academia and small business, and through intramural research, including preclinical and clinical studies. RESULTS: NCI has co-organized workshops and organized interest groups on RPT and RPT dosimetry to encourage the community and facilitate rigorous preclinical and clinical studies. NCI has been supporting RPT research through various mechanisms. Research has been funded through peer-reviewed NCI Research and Program Grants (RPG) and NCI Small Business Innovation Research (SBIR) Development Center, which funds small business-initiated projects, some of which have led to clinical trials. The NCI Cancer Therapy Evaluation Program (CTEP)'s Radiopharmaceutical Development Initiative supports RPT in NCI-funded clinical trials, including Imaging and Radiation Oncology Core (IROC) expertise in imaging QA and dosimetry procedures. Preclinical targeted a-emitter therapy (TAT) research at the NCI's intramural program is ongoing, building on foundational work dating back to the 1980s. Ongoing "bench-to-bedside" efforts leverage the unique infrastructure of the National Institutes of Health's (NIH) Clinical Center. CONCLUSION: Given the great potential of RPT, our goal is to continue to encourage its development that will generate the high-quality evidence needed to bring this multidisciplinary treatment to patients.


Asunto(s)
Neoplasias , Humanos , National Cancer Institute (U.S.) , Neoplasias/radioterapia , Radiometría , Radiofármacos , Estados Unidos
3.
Proc Natl Acad Sci U S A ; 112(42): 13045-50, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438866

RESUMEN

Despite significant advances in the treatment of Hodgkin's lymphoma (HL), a significant proportion of patients will not respond or will subsequently relapse. We identified CD25, the IL-2 receptor alpha subunit, as a favorable target for systemic radioimmunotherapy of HL. The scientific basis for the clinical trial was that, although most normal cells with exception of Treg cells do not express CD25, it is expressed by a minority of Reed-Sternberg cells and by most polyclonal T cells rosetting around Reed-Sternberg cells. Forty-six patients with refractory and relapsed HL were evaluated with up to seven i.v. infusions of the radiolabeled anti-CD25 antibody (90)Y-daclizumab. (90)Y provides strong ß emissions that kill tumor cells at a distance by a crossfire effect. In 46 evaluable HL patients treated with (90)Y-daclizumab there were 14 complete responses and nine partial responses; 14 patients had stable disease, and nine progressed. Responses were observed both in patients whose Reed-Sternberg cells expressed CD25 and in those whose neoplastic cells were CD25(-) provided that associated rosetting T cells expressed CD25. As assessed using phosphorylated H2AX (γ-H2AX) as a bioindicator of the effects of radiation exposure, predominantly nonmalignant cells in the tumor microenvironment manifested DNA damage, as reflected by increased expression of γ-H2AX. Toxicities were transient bone-marrow suppression and myelodysplastic syndrome in six patients who had not been evaluated with bone-marrow karyotype analyses before therapy. In conclusion, repeated (90)Y-daclizumab infusions directed predominantly toward nonmalignant T cells rosetting around Reed-Sternberg cells provided meaningful therapy for select HL patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Enfermedad de Hodgkin/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Subunidad alfa del Receptor de Interleucina-2/inmunología , Radioisótopos de Itrio/química , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Daclizumab , Femenino , Enfermedad de Hodgkin/inmunología , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Fosforilación , Recurrencia , Adulto Joven
4.
Chemistry ; 22(35): 12332-9, 2016 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-27305065

RESUMEN

Aryliodonium salts have become precursors of choice for the synthesis of (18) F-labeled tracers for nuclear imaging. However, little is known on the reactivity of these compounds with heavy halides, that is, radioiodide and astatide, at the radiotracer scale. In the first comparative study of radiohalogenation of aryliodonium salts with (125) I(-) and (211) At(-) , initial experiments on a model compound highlight the higher reactivity of astatide compared to iodide, which could not be anticipated from the trends previously observed within the halogen series. Kinetic studies indicate a significant difference in activation energy (Ea =23.5 and 17.1 kcal mol(-1) with (125) I(-) and (211) At(-) , respectively). Quantum chemical calculations suggest that astatination occurs via the monomeric form of an iodonium complex whereas iodination occurs via a heterodimeric iodonium intermediate. The good to excellent regioselectivity of halogenation and high yields achieved with diversely substituted aryliodonium salts indicate that this class of compounds is a promising alternative to the stannane chemistry currently used for heavy radiohalogen labeling of tracers in nuclear medicine.

5.
Int J Mol Sci ; 17(5)2016 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-27196891

RESUMEN

Radiolabeled antibodies (mAbs) provide efficient tools for cancer therapy. The combination of low energy ß(-)-emissions (500 keVmax; 130 keVave) along with a γ-emission for imaging makes (177)Lu (T1/2 = 6.7 day) a suitable radionuclide for radioimmunotherapy (RIT) of tumor burdens possibly too large to treat with α-particle radiation. RIT with (177)Lu-trastuzumab has proven to be effective for treatment of disseminated HER2 positive peritoneal disease in a pre-clinical model. To elucidate mechanisms originating from this RIT therapy at the molecular level, tumor bearing mice (LS-174T intraperitoneal xenografts) were treated with (177)Lu-trastuzumab comparatively to animals treated with a non-specific control, (177)Lu-HuIgG, and then to prior published results obtained using (212)Pb-trastuzumab, an α-particle RIT agent. (177)Lu-trastuzumab induced cell death via DNA double strand breaks (DSB), caspase-3 apoptosis, and interfered with DNA-PK expression, which is associated with the repair of DNA non-homologous end joining damage. This contrasts to prior results, wherein (212)Pb-trastuzumab was found to down-regulate RAD51, which is involved with homologous recombination DNA damage repair. (177)Lu-trastuzumab therapy was associated with significant chromosomal disruption and up-regulation of genes in the apoptotic process. These results suggest an inhibition of the repair mechanism specific to the type of radiation damage being inflicted by either high or low linear energy transfer radiation. Understanding the mechanisms of action of ß(-)- and α-particle RIT comparatively through an in vivo tumor environment offers real information suitable to enhance combination therapy regimens involving α- and ß(-)-particle RIT for the management of intraperitoneal disease.


Asunto(s)
Neoplasias del Colon/radioterapia , Inmunoconjugados/farmacología , Lutecio/farmacología , Radioisótopos/farmacología , Trastuzumab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/farmacología , Apoptosis/genética , Apoptosis/efectos de la radiación , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Aberraciones Cromosómicas/efectos de la radiación , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Immunoblotting , Inyecciones Intraperitoneales , Transferencia Lineal de Energía , Ratones Desnudos , Trasplante de Neoplasias/métodos , Recombinasa Rad51/metabolismo , Radioinmunoterapia/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Bioorg Med Chem Lett ; 25(10): 2056-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25870133

RESUMEN

There is growing interest in small and rigid peptidomimetic αvß3 integrin antagonists that are readily synthesized and characterized and amenable to physiological conditions. Peptidomimetic 4-[2-(3,4,5,6-tetrahydropyrimidine-2-ylamino)ethyloxy]benzoyl-2-[N-(3-amino-neopenta-1-carbamyl)]-aminoethylsulfonyl-amino-ß-alanine (IAC) was successfully conjugated to DOTA, complexed with Gd(III) and radiolabeled with (153)Gd. Radioassay results demonstrated specificity of the labeled conjugate by blocking ∼95% binding with the addition of a 50-fold molar excess of cold IAC to the reaction solution. Relaxometry was used to support the hypothesis that the specificity of the Gd-peptidomimetic targeting αvß3 integrin would increase the contrast and therefore enhance the sensitivity of an MRI scan of αvß3 integrin positive tissues. Magnetic resonance imaging of cell pellets (M21 human melanoma) was also performed, and the images clearly show that cells reacted with Gd(III)-DOTA-IAC display a brighter image than cells without the Gd(III)-DOTA-IAC contrast agent. In addition, Gd(III)-DOTA-IAC and IAC, with IC50 of 300nM and 230nM, respectively, are 2.1 and 2.7 times more potent than c(RGDfK) whose IC50 is 625nM. This promising preliminary data fuels further investigation of DOTA-IAC conjugates for targeting tumor associated angiogenesis and αvß3 integrin positive tumors using magnetic resonance imaging.


Asunto(s)
Medios de Contraste/síntesis química , Complejos de Coordinación/síntesis química , Gadolinio/química , Integrina alfaVbeta3/química , Imagen por Resonancia Magnética , Peptidomiméticos/química , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Medios de Contraste/química , Complejos de Coordinación/química , Humanos , Concentración 50 Inhibidora , Peptidomiméticos/síntesis química
7.
Bioconjug Chem ; 25(10): 1801-10, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25243604

RESUMEN

Indocyanine green (IC-Green), the only FDA approved near-infrared (NIR) fluorophore for clinical use, is attractive to researchers for the development of targeted optical imaging agents by modification of its structure and conjugation to monoclonal antibodies (mAbs) or their fragments. IC-Green derivative, ICG-sulfo-OSu (ICG-sOSu), is frequently used for antibody conjugation. However, ICG-sOSu is amphiphilic and readily facilitates aggregation of mAbs that is not easily separable from the desired immunoconjugates. Complications originating from this behavior are frequently overlooked by researchers. This study examined detailed chemical and biological characteristics of an ICG-sOSu-labeled mAb, panitumumab, and provided a clinically applicable strategy to deliver a pure conjugation product. Size-exclusion high-performance liquid chromatography (SE-HPLC) analysis of conjugation reactions, performed at molar reaction ratios of ICG-sOSu: mAb of 5, 10, or 20, resulted in isolable desired ICG-sOSu-panitumumab conjugation product in 72%, 53%, and 19% yields, respectively, with the remainder consisting of high molecular weight aggregates (>150 kDa) 14%, 30%, and 51%, respectively. The HPLC-purified ICG-sOSu-panitumumab products were analyzed by native and SDS polyacrylamide gel electrophoresis (PAGE) followed by optical imaging. Results indicated that the interaction between ICG-sOSu and panitumumab was due to both covalent and noncovalent binding of the ICG-sOSu to the protein. Noncovalently bound dye in the ICG-sOSu-panitumumab conjugate products was removed by extraction with ethyl acetate to further purify the HPLC-isolated conjugation products. With conserved immunoreactivity, excellent target-specific uptake of the doubly purified bioconjugates was observed with minimal liver retention in athymic nude mice bearing HER1-expressing tumor xenografts. In summary, the preparation of well-defined bioconjugate products labeled with commercial ICG-sOSu dye is not a simple process and control of the conjugation reaction ratio and conditions is crucial. Furthermore, absolute purification and characterization of the products is necessitated prior to in vivo optical imaging. Use of validated and characterized dye conjugate products should facilitate the development of clinically viable and reproducible IC-Green derivative and other NIR dye mAb conjugates for optical imaging applications.


Asunto(s)
Anticuerpos Monoclonales/análisis , Colorantes/análisis , Inmunoconjugados/análisis , Verde de Indocianina/análogos & derivados , Animales , Anticuerpos Monoclonales/farmacocinética , Colorantes/farmacocinética , Inmunoconjugados/farmacocinética , Verde de Indocianina/análisis , Verde de Indocianina/farmacocinética , Ratones Desnudos , Imagen Óptica , Panitumumab
8.
Chemistry ; 20(19): 5584-91, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24740517

RESUMEN

Metals of interest for biomedical applications often need to be complexed and associated in a stable manner with a targeting agent before use. Whereas the fundamentals of most transition-metal complexation processes have been thoroughly studied, the complexation of Zr(IV) has been somewhat neglected. This metal has received growing attention in recent years, especially in nuclear medicine, with the use of (89) Zr, which a ß(+) -emitter with near ideal characteristics for cancer imaging. However, the best chelating agent known for this radionuclide is the trishydroxamate desferrioxamine B (DFB), the Zr(IV) complex of which exhibits suboptimal stability, resulting in the progressive release of (89) Zr in vivo. Based on a recent report demonstrating the higher thermodynamic stability of the tetrahydroxamate complexes of Zr(IV) compared with the trishydroxamate complexes analogues to DFB, we designed a series of tetrahydroxamic acids of varying geometries for improved complexation of this metal. Three macrocycles differing in their cavity size (28 to 36-membered rings) were synthesized by using a ring-closing metathesis strategy, as well as their acyclic analogues. A solution study with (89) Zr showed the complexation to be more effective with increasing cavity size. Evaluation of the kinetic inertness of these new complexes in ethylenediaminetetraacetic acid (EDTA) solution showed significantly improved stabilities of the larger chelates compared with (89) Zr-DFB, whereas the smaller complexes suffered from insufficient stabilities. These results were rationalized by a quantum chemical study. The lower stability of the smaller chelates was attributed to ring strain, whereas the better stability of the larger cyclic complexes was explained by the macrocyclic effect and by the structural rigidity. Overall, these new chelating agents open new perspectives for the safe and efficient use of (89) Zr in nuclear imaging, with the best chelators providing dramatically improved stabilities compared with the reference DFB.


Asunto(s)
Quelantes/química , Complejos de Coordinación/química , Deferoxamina/química , Ácido Edético/química , Compuestos Férricos/química , Ácidos Hidroxámicos/síntesis química , Radioisótopos/química , Circonio/química , Cristalografía por Rayos X , Ácidos Hidroxámicos/química , Cinética , Tomografía de Emisión de Positrones
9.
J Am Chem Soc ; 135(21): 7815-8, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23581827

RESUMEN

Fluorescent nanodiamonds (FNDs) emit in the near-IR and do not photobleach or photoblink. These properties make FNDs better suited for numerous imaging applications compared with commonly used fluorescence agents such as organic dyes and quantum dots. However, nanodiamonds do not form stable suspensions in aqueous buffer, are prone to aggregation, and are difficult to functionalize. Here we present a method for encapsulating nanodiamonds with silica using an innovative liposome-based encapsulation process that renders the particle surface biocompatible, stable, and readily functionalized through routine linking chemistries. Furthermore, the method selects for a desired particle size and produces a monodisperse agent. We attached biotin to the silica-coated FNDs and tracked the three-dimensional motion of a biotinylated FND tethered by a single DNA molecule with high spatial and temporal resolution.


Asunto(s)
Coloides/química , Nanodiamantes , Dióxido de Silicio/química , ADN/química , Fluorescencia , Liposomas , Espectroscopía Infrarroja Corta , Propiedades de Superficie
10.
Radiology ; 267(1): 173-82, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23329660

RESUMEN

PURPOSE: To evaluate the potential of anti-human epidermal growth factor receptor (HER)1- and anti-HER2-targeted radiolabeled antibodies and magnetic resonance (MR) imaging for imaging of orthotopic malignant pleural mesothelioma (MPM) in mouse models. MATERIALS AND METHODS: Animal studies with 165 mice were performed in accordance with National Institutes of Health guidelines for the humane use of animals, and all procedures were approved by the institutional Animal Care and Use Committee. Flow cytometry studies were performed to evaluate HER1 and HER2 expression in NCI-H226 and MSTO-211H mesothelioma cells. Biodistribution and single photon emission computed tomography (SPECT)/computed tomography (CT) imaging studies were performed in mice (four or five per group, depending on tumor growth) bearing subcutaneous and orthotopic MPM tumors by using HER1- and HER2-targeted indium 111 ((111)In)- and iodine 125 ((125)I)-labeled panitumumab and trastuzumab, respectively. Longitudinal MR imaging over 5 weeks was performed in three mice bearing orthotopic tumors to monitor tumor growth and metastases. SPECT/CT/MR imaging studies were performed at the final time point in the orthotopic models (n = 3). The standard unpaired Student t test was used to compare groups. RESULTS: Orthotopic tumors and pleural effusions were clearly visualized at MR imaging 3 weeks after tumor cell inoculation. At 2 days after injection, the mean (111)In-panitumumab uptake of 29.6% injected dose (ID) per gram ± 2.2 (standard error of the mean) was significantly greater than the (111)In-trastuzumab uptake of 13.6% ID/g ± 1.0 and the (125)I-panitumumab uptake of 7.4% ID/g ± 1.2 (P = .0006 and P = .0001, respectively). MR imaging fusion with SPECT/CT provided more accurate information about (111)In-panitumumab localization in the tumor, as the tumor was poorly visualized at CT alone. CONCLUSION: This study demonstrates the utility of radiolabeled anti-HER1 antibodies in the imaging of MPM in preclinical models. SUPPLEMENTAL MATERIAL: http://radiology.rsna.org/lookup/suppl/doi:10.1148/radiol.12121021/-/DC1.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Receptores ErbB/metabolismo , Imagen por Resonancia Magnética/métodos , Mesotelioma/metabolismo , Imagen Multimodal/métodos , Tomografía de Emisión de Positrones , Receptor ErbB-2/metabolismo , Tomografía Computarizada por Rayos X , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Área Bajo la Curva , Línea Celular Tumoral , Femenino , Citometría de Flujo , Radioisótopos de Yodo , Radioisótopos de Iridio , Mesotelioma/diagnóstico por imagen , Mesotelioma/patología , Ratones , Terapia Molecular Dirigida , Panitumumab , Derrame Pleural/diagnóstico por imagen , Derrame Pleural/patología , Trastuzumab , Imagen de Cuerpo Entero , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Nucl Med ; 64(1): 109-116, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35981897

RESUMEN

We studied the feasibility of using the α-emitting 213Bi-anti-CD20 therapy with direct bioluminescent tracking of micrometastatic human B-cell lymphoma in a SCID mouse model. Methods: A highly lethal SCID mouse model of minimal-tumor-burden disseminated non-Hodgkin lymphoma (NHL) was established using human Raji lymphoma cells transfected to express the luciferase reporter. In vitro and in vivo radioimmunotherapy experiments were conducted. Single- and multiple-dose regimens were explored, and results with 213Bi-rituximab were compared with various controls, including no treatment, free 213Bi radiometal, unlabeled rituximab, and 213Bi-labeled anti-HER2/neu (non-CD20-specific antibody). 213Bi-rituximab was also compared in vivo with the low-energy ß-emitter 131I-tositumomab and the high-energy ß-emitter 90Y-rituximab. Results: In vitro studies showed dose-dependent target-specific killing of lymphoma cells with 213Bi-rituximab. Multiple in vivo studies showed significant and specific tumor growth delays with 213Bi-rituximab versus free 213Bi, 213Bi-labeled control antibody, or unlabeled rituximab. Redosing of 213Bi-rituximab was more effective than single dosing. With a single dose of therapy given 4 d after intravenous tumor inoculation, disease in all untreated controls, and in all mice in the 925-kBq 90Y-rituximab group, progressed. With 3,700 kBq of 213Bi-rituximab, 75% of the mice survived and all but 1 survivor was cured. With 2,035 kBq of 131I-tositumomab, 75% of the mice were tumor-free by bioluminescent imaging and 62.5% survived. Conclusion: Cure of micrometastatic NHL is achieved in most animals treated 4 d after intravenous tumor inoculation using either 213Bi-rituximab or 131I-tositumomab, in contrast to the lack of cures with unlabeled rituximab or 90Y-rituximab or if there was a high tumor burden before radioimmunotherapy. α-emitter-labeled anti-CD20 antibodies are promising therapeutics for NHL, although a longer-lived α-emitter may be of greater efficacy.


Asunto(s)
Antineoplásicos , Linfoma de Células B , Linfoma no Hodgkin , Linfoma , Ratones , Humanos , Animales , Rituximab/uso terapéutico , Ratones SCID , Anticuerpos Monoclonales/uso terapéutico , Linfoma de Células B/radioterapia , Linfoma no Hodgkin/radioterapia , Linfoma no Hodgkin/tratamiento farmacológico , Radioinmunoterapia/métodos , Antígenos CD20
12.
Med Phys ; 50(3): e53-e61, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36705550

RESUMEN

Over several months, representatives from the U.S. Department of Energy (DOE) Office of Science and National Institutes of Health (NIH) had a number of meetings that lead to the conclusion that innovations in the Nation's health care could be realized by more directed interactions between NIH and DOE. It became clear that the expertise amassed and instrumentation advances developed at the DOE physical science laboratories to enable cutting-edge research in particle physics could also feed innovation in medical healthcare. To meet their scientific mission, the DOE laboratories created advances in such technologies as particle beam generation, radioisotope production, high-energy particle detection and imaging, superconducting particle accelerators, superconducting magnets, cryogenics, high-speed electronics, artificial intelligence, and big data. To move forward, NIH and DOE initiated the process of convening a joint workshop which occurred on July 12th and 13th, 2021. This Special Report presents a summary of the findings of the collaborative workshop and introduces the goals of the next one.


Asunto(s)
Investigación Biomédica , Disciplinas de las Ciencias Naturales , Estados Unidos , Inteligencia Artificial , National Institutes of Health (U.S.) , Laboratorios
13.
Tumour Biol ; 33(3): 573-90, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22143940

RESUMEN

The effectiveness of targeted α-therapy (TAT) can be explained by the properties of α-particles. Alpha particles are helium nuclei and are ~8,000 times larger than ß(-)-particles (electrons). When emitted from radionuclides that decay via an α-decay pathway, they release enormous amounts of energy over a very short distance. Typically, the range of α-particles in tissue is 50-100 µm and they have high linear energy transfer (LET) with a mean energy deposition of 100 keV/µm, providing a more specific tumor cell killing ability without damage to the surrounding normal tissues than ß(-)-emitters. Due to these properties, the majority of pre-clinical and clinical trials have demonstrated that α-emitters such as (225)Ac, (211)At, (212)Bi, (213)Bi, (212)Pb, (223)Ra, and (227)Th are ideal for the treatment of smaller tumor burdens, micrometastatic disease, and disseminated disease. Even though these α-emitters have favorable properties, the development of TAT has been limited by high costs, unresolved chemistry, and limited availability of the radionuclides. To overcome these limitations, more potent isotopes, additional sources, and more efficient isotope production methods should be addressed. Furthermore, better chelation and labeling methods with the improvements of isotope delivery, targeting vehicles, molecular targets, and identification of appropriate clinical applications are still required.


Asunto(s)
Partículas alfa/uso terapéutico , Neoplasias/radioterapia , Radioinmunoterapia , Radioisótopos/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Radioisótopos/química
14.
Mol Pharm ; 9(3): 374-81, 2012 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-21882823

RESUMEN

Herein we report the preparation along with the in vivo and in vitro MRI characterization of two generation four and five cystamine core dendrimers loaded with thirty and fifty-eight derivatized Gd-DOTA (G4SS30, G5SS58) respectively. Likewise the development and characterization of two half-dendrimers conjugated to the F(ab')(2) fragment of the monoclonal antibody (mAb) panitumumab functionalized with a maleimide conjugation functional group site (Ab-(G4S15)(4), Ab-(G5S29)(4)) are also described. The in vitro molar relaxivity of the Ab-(G4S15)(4) conjugate, measured at pH 7.4, 22 °C, and 3T showed a moderate increase in relaxivity as compared to Magnevist (6.7 vs 4.0 mM(-1) s(-1)) while the Ab-(G5S29)(4) conjugate was 2-fold higher (9.1 vs 4.0 mM(-1) s(-1)). The data showed that only a high injection dose (0.050 mmol Gd(3+)/kg) produced a detectable contrast enhanced contrast for the Ab-(G4S15)(4) conjugate while a lower dose (0.035 mmol Gd(3+)/kg) was sufficient for the Ab-(G5S29)(4) conjugate. The antibody-SMCC conjugate was purified by a Sephadex G-100 column, and the antibody-dendrimer-based agents were purified by spin filtration using a Centricon filter (50,000 MCO). The protein assay coupled with cysteine and Ellman's assay indicated an antibody to dendrimer ratio of 1:4. The in vivo blood clearance half-lives of the four agents measured at the jugular vein were ~12-22 min.


Asunto(s)
Anticuerpos Monoclonales/química , Cistamina/química , Dendrímeros/química , Angiografía por Resonancia Magnética/métodos , Animales , Medios de Contraste/química , Ratones
15.
Bioorg Med Chem Lett ; 22(17): 5517-22, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22853992

RESUMEN

There is growing interest in small peptidomimetic α(v)ß(3) integrin antagonists that are readily synthesized and characterized and can be easily handled using physiological conditions. Peptidomimetic 4-[2-(3,4,5,6-tetrahydropyrimidine-2-ylamino)ethyloxy]benzoyl-2-[N-(3-amino-neopenta-1-carbamyl)]-aminoethylsulfonyl-amino-ß-alanine (IAC) was successfully conjugated to 1-(1-carboxy-3-carbo-t-butoxypropyl)-4,7-(carbo-tert-butoxymethyl)-1,4,7-triazacyclononane (NODA-GA(tBu)(3)) and 1-(1-carboxy-3-carbotertbutoxymethyl)-1,4,7,10-tetraazacyclododecane (DOTA-GA(tBu)(4)) and radiolabeled with (111)In, (67)Ga and (203)Pb. Results of a radioimmunoassay demonstrated binding to purified α(v)ß(3) integrin when 1-4equiv of integrin were added to the reaction. Based on this promising result, investigations are moving forward to evaluate the NODA-GA-IAC and DOTA-GA-IAC conjugates for targeting tumor associated angiogenesis and α(v)ß(3) integrin positive tumors to define their PET and SPECT imaging qualities as well as their potential for delivery of therapeutic radionuclides.


Asunto(s)
Quelantes/química , Quelantes/metabolismo , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/metabolismo , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Acetatos/síntesis química , Acetatos/química , Acetatos/metabolismo , Quelantes/síntesis química , Radioisótopos de Galio/química , Radioisótopos de Galio/metabolismo , Compuestos Heterocíclicos con 1 Anillo/síntesis química , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/metabolismo , Humanos , Radioisótopos de Plomo/química , Radioisótopos de Plomo/metabolismo , Peptidomiméticos/síntesis química , Tomografía de Emisión de Positrones , Radioinmunoensayo , Tomografía Computarizada de Emisión de Fotón Único
16.
J Labelled Comp Radiopharm ; 55(11): 423-426, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23162207

RESUMEN

Methodology for site-specific modification and chelate conjugation of a cyclic RGD (cRGD) peptide for the preparation of a radiotracer molecular imaging agent suitable for detecting α(v)ß(3) integrin is described. The method involves functionalizing the peptide with an aldehyde moiety and conjugation to a 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid (DOTA) derivative that possesses an aldehyde reactive aminooxy group. The binding assay of the (111)In-labeled peptide conjugate with α(v)ß(3) integrin showed 60% bound when four equivalents of the integrin was added, a reasonable binding affinity for a mono-valent modified RGD peptide.

17.
Int J Cancer ; 128(4): 920-6, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473899

RESUMEN

Bevacizumab is a humanized monoclonal antibody that binds to tumor-secreted vascular endothelial growth factor (VEGF)-A and inhibits tumor angiogenesis. In 2004, the antibody was approved by the US Food and Drug Administration (FDA) for the treatment of metastatic colorectal carcinoma in combination with chemotherapy. This report describes the preclinical evaluation of a radioimmunoconjugate, (86)Y-CHX-A″-DTPA-bevacizumab, for potential use in Positron Emission Tomography (PET) imaging of VEGF-A tumor angiogenesis and as a surrogate marker for (90)Y-based radioimmunotherapy. Bevacizumab was conjugated to CHX-A″-DTPA and radiolabeled with (86)Y. In vivo biodistribution and PET imaging studies were performed on mice bearing VEGF-A-secreting human colorectal (LS-174T), human ovarian (SKOV-3) and VEGF-A-negative human mesothelioma (MSTO-211H) xenografts. Biodistribution and PET imaging studies demonstrated highly specific tumor uptake of the radioimmunoconjugate. In mice bearing VEGF-A-secreting LS-174T, SKOV-3 and VEGF-A-negative MSTO-211H tumors, the tumor uptake at 3 days postinjection was 13.6 ± 1.5, 17.4 ± 1.7 and 6.8 ± 0.7 % ID/g, respectively. The corresponding tumor uptake in mice coinjected with 0.05 mg cold bevacizumab were 5.8 ± 1.3, 8.9 ± 1.9 and 7.4 ± 1.0 % ID/g, respectively at the same time point, demonstrating specific blockage of the target in VEGF-A-secreting tumors. The LS-174T and SKOV3 tumors were clearly visualized by PET imaging after injecting 1.8-2.0 MBq (86)Y-CHX-A″-DTPA-bevacizumab. Organ uptake quantified by PET closely correlated (r(2) = 0.87, p = 0.64, n = 18) to values determined by biodistribution studies. This preclinical study demonstrates the potential of the radioimmunoconjugate, (86)Y-CHX-A″-DTPA-bevacizumab, for noninvasive assessment of the VEGF-A tumor angiogenesis status and as a surrogate marker for (90)Y-CHX-A″-DTPA-bevacizumab radioimmunotherapy.


Asunto(s)
Anticuerpos Monoclonales , Inmunoconjugados , Isotiocianatos , Neovascularización Patológica/diagnóstico por imagen , Ácido Pentético/análogos & derivados , Tomografía de Emisión de Positrones , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Radioisótopos de Itrio , Inhibidores de la Angiogénesis/farmacocinética , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Bevacizumab , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/diagnóstico , Femenino , Humanos , Inmunoconjugados/farmacocinética , Isotiocianatos/farmacocinética , Ratones , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/diagnóstico , Ácido Pentético/farmacocinética , Radioinmunoensayo , Radiofármacos/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas , Radioisótopos de Itrio/farmacocinética
18.
Bioorg Med Chem Lett ; 21(24): 7513-5, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22047687

RESUMEN

A new bifunctional ligand C-DEPA was designed and synthesized as a component for antibody-targeted radiation therapy (radioimmunotherapy, RIT) of cancer. C-DEPA was conjugated to a tumor targeting antibody, trastuzumab, and the corresponding C-DEPA-trastuzumab conjugate was evaluated for radiolabeling kinetics with (205/6)Bi. C-DEPA-trastuzumab conjugate rapidly bound (205/6)Bi, and (205/6)Bi-C-DEPA-trastuzumab conjugate was stable in human serum for 72 h. The in vitro radiolabeling kinetics and serum stability data suggest that C-DEPA is a potential chelate for preclinical RIT applications using (212)Bi and (213)Bi.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Bismuto/química , Glicina/análogos & derivados , Compuestos Heterocíclicos con 1 Anillo/química , Inmunoconjugados/química , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Glicina/química , Humanos , Inmunoconjugados/metabolismo , Inmunoconjugados/uso terapéutico , Marcaje Isotópico , Cinética , Neoplasias/radioterapia , Trastuzumab
19.
Bioconjug Chem ; 21(6): 1014-7, 2010 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-20462240

RESUMEN

This report presents the preparation and characterization of three [Gd-C-DOTA](-1)-dendrimer assemblies by way of analysis, NMRD spectroscopy, and photon correlation spectroscopy (PCS). The metal-ligand chelates were preformed in alcohol media prior to conjugation to generation 4, 5, and 6 PAMAM dendrimers. The dendrimer-based agents were purified by Sephadex G-25 column chromatography. The combustion analysis, SE-HPLC, and UV-vis data indicated chelate to dendrimer ratios of 28:1, 61:1 and 115:1, respectively. Molar relaxivity measured at pH 7.4, 22 degrees C, and 3 T (29.6, 49.8, and 89.1 mM(-1) s(-1)) indicated the viability of conjugates as MRI contrast agents. 1/T(1) NMRD profiles were measured at 23 degrees C and indicated that at 22 MHz the 1/T(1) reached a plateau at 60, 85, and 140 mM(-1) s(-1) for the generation 4, 5, and 6 dendrimer conjugates, respectively. The PCS data showed the respective sizes of 5.2, 6.5, and 7.8 nm for G-4, 5, and 6 conjugates.


Asunto(s)
Medios de Contraste/síntesis química , Dendrímeros/síntesis química , Gadolinio/química , Imagen por Resonancia Magnética/métodos , Poliaminas/síntesis química , Quelantes/síntesis química , Quelantes/química , Cromatografía Líquida de Alta Presión , Medios de Contraste/química , Dendrímeros/química , Compuestos Heterocíclicos/síntesis química , Compuestos Heterocíclicos/química , Concentración de Iones de Hidrógeno , Ligandos , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Poliaminas/química , Espectrofotometría Ultravioleta , Temperatura
20.
Bioconjug Chem ; 21(12): 2305-12, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21073171

RESUMEN

A water-soluble amine-reactive PEGylated analogue of near-infrared emitting dye indocyanine green (5) was synthesized and used to label the anti-HER1 antibody panitumumab (Vectibix) at various equivalents. These conjugates were compared with non-PEGylated analogue conjugate products and the solution speciation analyzed with UV-vis spectrophotometry, size exclusion HPLC, and SDS-PAGE. PEGylation of the bioconjugates was successful in preventing aggregation in solution, a phenomenon observed with the non-PEGylated bioconjugates presumably due to the hydrophobicity of indocyanine green. Competitive radioimmunoassay demonstrated that the targeting moiety of the PEGylated bioconjugates was conserved. Fluorescence microscopy also demonstrated membrane binding of the bioconjugate to HER1-expressing A431 cells. Hence, these bioconjugates are suitable candidates for the in vivo optical imaging of HER1-expressing tumors.


Asunto(s)
Anticuerpos Monoclonales , Receptores ErbB/metabolismo , Colorantes Fluorescentes/química , Inmunoconjugados , Verde de Indocianina/química , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Unión Competitiva , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Cromatografía en Gel , Diagnóstico por Imagen/métodos , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inmunoconjugados/química , Inmunoconjugados/metabolismo , Verde de Indocianina/análogos & derivados , Microscopía Fluorescente , Panitumumab , Polietilenglicoles/química , Radioinmunoensayo , Solubilidad , Soluciones , Espectroscopía Infrarroja Corta , Neoplasias de la Vulva/diagnóstico , Neoplasias de la Vulva/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA