Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Ther ; 17(2): 348-352, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34121676

RESUMO

AIM OF STUDY: There is limited information regarding the α-emitter radiopharmaceuticals dose calculation used in the setting of men with prostate cancer (PCa). The present study investigates the α-emitter radiopharmaceuticals absorbed dose distribution in the body organs. MATERIALS AND METHODS: The α-emitter radiopharmaceuticals dose coefficient and absorbed doses biokinetics distribution, which are used for the treatment of PCa in all over the world, were performed using the "Internal Dose Assessed by Computer" (IDAC-Dose 2.1) program. The results of absorbed dose distribution in any organ of the body, were compared in studied α-emitter radiopharmaceuticals. RESULTS: The absorbed dose value of 223Ra radiopharmaceutical in the prostate organ was evaluated 9.47E-9 Gy/Bq. The maximum and minimum absorbed doses due to biokinetics distribution of 223Ra were found in the thymus (9.53E-8 Gy/Bq) and eye lenses (1.30E-10 Gy/Bq) organs, respectively. Furthermore, the 225Ac absorbed dose in the prostate organ was obtained 1.91E-9 Gy/Bq, where this value is 1% of total body dose. While the absorbed dose distribution of 225Ac in body organs shows the highest concentration in the spleen (1.47E-8 Gy/Bq) and lowest in the eye lenses (7.93E-12 Gy/Bq). CONCLUSION: The absorbed dose in the body organs due to 223Ra and 225Ac α-emitter radiopharmaceuticals which are used in metastasized castration-resistant prostate cancer (mCRPC), calculated in this study. The results of this study will assist in evaluating and analyzing human body organ doses from application of 223Ra and 225Ac that used in mCRPC patients.


Assuntos
Actínio/administração & dosagem , Neoplasias de Próstata Resistentes à Castração/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , Planejamento da Radioterapia Assistida por Computador/métodos , Rádio (Elemento)/administração & dosagem , Humanos , Masculino , Próstata/diagnóstico por imagem , Próstata/patologia , Próstata/efeitos da radiação , Neoplasias de Próstata Resistentes à Castração/secundário , Dosagem Radioterapêutica
2.
Theranostics ; 10(25): 11359-11375, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33052220

RESUMO

This is the initial report of an α-based pre-targeted radioimmunotherapy (PRIT) using 225Ac and its theranostic pair, 111In. We call our novel tumor-targeting DOTA-hapten PRIT system "proteus-DOTA" or "Pr." Herein we report the first results of radiochemistry development, radiopharmacology, and stoichiometry of tumor antigen binding, including the role of specific activity, anti-tumor efficacy, and normal tissue toxicity with the Pr-PRIT approach (as α-DOTA-PRIT). A series of α-DOTA-PRIT therapy studies were performed in three solid human cancer xenograft models of colorectal cancer (GPA33), breast cancer (HER2), and neuroblastoma (GD2), including evaluation of chronic toxicity at ~20 weeks of select survivors. Methods: Preliminary biodistribution experiments in SW1222 tumor-bearing mice revealed that 225Ac could not be efficiently pretargeted with current DOTA-Bn hapten utilized for 177Lu or 90Y, leading to poor tumor uptake in vivo. Therefore, we synthesized Pr consisting of an empty DOTA-chelate for 225Ac, tethered via a short polyethylene glycol linker to a lutetium-complexed DOTA for picomolar anti-DOTA chelate single-chain variable fragment (scFv) binding. Pr was radiolabeled with 225Ac and its imaging surrogate, 111In. In vitro studies verified anti-DOTA scFv recognition of [225Ac]Pr, and in vivo biodistribution and clearance studies were performed to evaluate hapten suitability and in vivo targeting efficiency. Results: Intravenously (i.v.) administered 225Ac- or 111In-radiolabeled Pr in mice showed rapid renal clearance and minimal normal tissue retention. In vivo pretargeting studies show high tumor accumulation of Pr (16.71 ± 5.11 %IA/g or 13.19 ± 3.88 %IA/g at 24 h p.i. for [225Ac]Pr and [111In]Pr, respectively) and relatively low uptake in normal tissues (all average ≤ 1.4 %IA/g at 24 h p.i.). Maximum tolerated dose (MTD) was not reached for either [225Ac]Pr alone or pretargeted [225Ac]Pr at administered activities up to 296 kBq/mouse. Single-cycle treatment consisting of α-DOTA-PRIT with either huA33-C825 bispecific anti-tumor/anti-DOTA-hapten antibody (BsAb), anti-HER2-C825 BsAb, or hu3F8-C825 BsAb for targeting GPA33, HER2, or GD2, respectively, was highly effective. In the GPA33 model, no complete responses (CRs) were observed but prolonged overall survival of treated animals was 42 d for α-DOTA-PRIT vs. 25 d for [225Ac]Pr only (P < 0.0001); for GD2, CRs (7/7, 100%) and histologic cures (4/7, 57%); and for HER2, CRs (7/19, 37%) and histologic cures (10/19, 56%) with no acute or chronic toxicity. Conclusions: [225Ac]Pr and its imaging biomarker [111In]Pr demonstrate optimal radiopharmacologic behavior for theranostic applications of α-DOTA-PRIT. For this initial evaluation of efficacy and toxicity, single-cycle treatment regimens were performed in all three systems. Histologic toxicity was not observed, so MTD was not observed. Prolonged overall survival, CRs, and histologic cures were observed in treated animals. In comparison to RIT with anti-tumor IgG antibodies, [225Ac]Pr has a much improved safety profile. Ultimately, these data will be used to guide clinical development of toxicity and efficacy studies of [225Ac]Pr, with the goal of delivering massive lethal doses of radiation to achieve a high probability of cure without toxicity.


Assuntos
Partículas alfa/uso terapêutico , Neoplasias/terapia , Radioimunoterapia/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Nanomedicina Teranóstica/métodos , Actínio/administração & dosagem , Actínio/farmacocinética , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Feminino , Meia-Vida , Compostos Heterocíclicos com 1 Anel/administração & dosagem , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/farmacocinética , Humanos , Radioisótopos de Índio/administração & dosagem , Radioisótopos de Índio/farmacocinética , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/imunologia , Neoplasias/patologia , Radioimunoterapia/efeitos adversos , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Dosagem Radioterapêutica , Distribuição Tecidual , Testes de Toxicidade Crônica , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Pharm ; 17(11): 4270-4279, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33044830

RESUMO

Interest in the use of 225Ac for targeted alpha therapies has increased dramatically over the past few years, resulting in a multitude of new isotope production and translational research efforts. However, 225Ac radioimmunoconjugate (RIC) research is still in its infancy, with most prior experience in hematologic malignancies and only one reported preclinical solid tumor study using 225Ac RICs. In an effort to compare 225Ac RICs to other current antibody conjugates, a variety of RICs are tested against intractable small-cell lung cancer (SCLC). We directly compare, in vitro and in vivo, two promising candidates of each α or ß- category, 225Ac and 177Lu, versus pyrrolobenzodiazepine (PBD) nonradioactive benchmarks. The monoclonal antibody constructs are targeted to either delta like 3 protein (DLL3), a recently discovered SCLC target, or CD46 as a positive control. An immunocompromised maximum tolerated dose assay is performed on NOD SCID mice, along with tumor efficacy proof-of-concept studies in vivo. We overview the conjugation techniques required to create serum-stable RICs and characterize and compare in vitro cell killing with RICs conjugated to nonspecific antibodies (huIgG1) with either native or site-specific thiol loci against tumor antigen DLL3-expressing and nonexpressing cell lines. Using patient-derived xenografts of SCLC onto NOD SCID mice, solid tumor growth was controlled throughout 3 weeks before growth appeared, in comparison to PBD conjugate controls. NOD SCID mice showed lengthened survival using 225Ac compared to 177Lu RICs, and PBD dimers showed full tumor suppression with nine out of ten mice. The exploration of RICs on a variety of antibody-antigen systems is necessary to direct efforts in cancer research toward promising candidates. However, the anti-DLL3-RIC system with 225Ac and 177Lu appears to be not as effective as the anti-DLL3-PBD counterpart in SCLC therapy with matched antibodies and portrays the challenges in both SCLC therapy as well as the specialized utility of RICs in cancer treatment.


Assuntos
Actínio/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Imunoconjugados/administração & dosagem , Imunoglobulina G/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Lutécio/administração & dosagem , Radioisótopos/administração & dosagem , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Partículas alfa/uso terapêutico , Animais , Antígenos de Neoplasias/imunologia , Benzodiazepinas/administração & dosagem , Partículas beta/uso terapêutico , Feminino , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Neoplasias Pulmonares/patologia , Dose Máxima Tolerável , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pirróis/administração & dosagem , Carcinoma de Pequenas Células do Pulmão/patologia , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Biother Radiopharm ; 35(6): 459-473, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32013538

RESUMO

An α particle-emitting nanodrug that is a potent and specific antitumor agent and also prompts significant remodeling of local immunity in the tumor microenvironment (TME) has been developed and may impact the treatment of melanoma. Biocompatible ultrasmall fluorescent core-shell silica nanoparticles (C' dots, diameter ∼6.0 nm) have been engineered to target the melanocortin-1 receptor expressed on melanoma through α melanocyte-stimulating hormone peptides attached to the C' dot surface. Actinium-225 is also bound to the nanoparticle to deliver a densely ionizing dose of high-energy α particles to cancer. Nanodrug pharmacokinetic properties are optimal for targeted radionuclide therapy as they exhibit rapid blood clearance, tumor-specific accumulation, minimal off-target localization, and renal elimination. Potent and specific tumor control, arising from the α particles, was observed in a syngeneic animal model of melanoma. Surprisingly, the C' dot component of this drug initiates a favorable pseudopathogenic response in the TME generating distinct changes in the fractions of naive and activated CD8 T cells, Th1 and regulatory T cells, immature dendritic cells, monocytes, MΦ and M1 macrophages, and activated natural killer cells. Concomitant upregulation of the inflammatory cytokine genome and adaptive immune pathways each describes a macrophage-initiated pseudoresponse to a viral-shaped pathogen. This study suggests that therapeutic α-particle irradiation of melanoma using ultrasmall functionalized core-shell silica nanoparticles potently kills tumor cells, and at the same time initiates a distinct immune response in the TME.


Assuntos
Partículas alfa/uso terapêutico , Portadores de Fármacos/química , Melanoma Experimental/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , Neoplasias Cutâneas/radioterapia , Microambiente Tumoral/efeitos da radiação , Actínio/administração & dosagem , Actínio/farmacocinética , Animais , Linhagem Celular Tumoral/transplante , Biologia Computacional , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Imunidade Celular/genética , Imunidade Celular/efeitos da radiação , Masculino , Dose Máxima Tolerável , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Terapia de Alvo Molecular/métodos , Nanopartículas/química , RNA-Seq , Compostos Radiofarmacêuticos/farmacocinética , Receptor Tipo 1 de Melanocortina/antagonistas & inibidores , Receptor Tipo 1 de Melanocortina/metabolismo , Dióxido de Silício/química , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Distribuição Tecidual , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
5.
Cancer Res ; 80(4): 868-876, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31772036

RESUMO

Preclinical studies, in vivo, and in vitro studies, in combination with mathematical modeling can help optimize and guide the design of clinical trials. The design and optimization of alpha-particle emitter radiopharmaceutical therapy (αRPT) is especially important as αRPT has the potential for high efficacy but also high toxicity. We have developed a mathematical model that may be used to identify trial design parameters that will have the greatest impact on outcome. The model combines Gompertzian tumor growth with antibody-mediated pharmacokinetics and radiation-induced cell killing. It was validated using preclinical experimental data of antibody-mediated 213Bi and 225Ac delivery in a metastatic transgenic breast cancer model. In modeling simulations, tumor cell doubling time, administered antibody, antibody specific-activity, and antigen-site density most impacted median survival. The model was also used to investigate treatment fractionation. Depending upon the time-interval between injections, increasing the number of injections increased survival time. For example, two administrations of 200 nCi, 225Ac-labeled antibody, separated by 30 days, resulted in a simulated 31% increase in median survival over a single 400 nCi administration. If the time interval was 7 days or less, however, there was no improvement in survival; a one-day interval between injections led to a 10% reduction in median survival. Further model development and validation including the incorporation of normal tissue toxicity is necessary to properly balance efficacy with toxicity. The current model is, however, useful in helping understand preclinical results and in guiding preclinical and clinical trial design towards approaches that have the greatest likelihood of success. SIGNIFICANCE: Modeling is used to optimize αRPT.


Assuntos
Partículas alfa/uso terapêutico , Neoplasias da Mama/radioterapia , Modelos Biológicos , Radioimunoterapia/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Actínio/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Bismuto/administração & dosagem , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/genética , Linhagem Celular Tumoral/transplante , Imagem de Difusão por Ressonância Magnética , Gerenciamento Clínico , Fracionamento da Dose de Radiação , Esquema de Medicação , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Tomografia por Emissão de Pósitrons , Radioisótopos/administração & dosagem , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único
6.
Eur J Pharm Biopharm ; 127: 85-91, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29428791

RESUMO

Alpha emitters have great potential in targeted tumour therapy, especially in destroying micrometastases, due to their high linear energy transfer (LET). To prevent toxicity caused by recoiled daughter atoms in healthy tissue, alpha emitters like 225Ac can be encapsulated in polymeric nanocarriers (polymersomes), which are capable of retaining the daughter atoms to a large degree. In the translation to a (pre-)clinical setting, it is essential to evaluate their therapeutic potential. As multicellular tumour spheroids mimic a tumour microenvironment more closely than a two-dimensional cellular monolayer, this study has focussed on the interaction of the polymersomes with U87 human glioma spheroids. We have found that polymersomes distribute themselves throughout the spheroid after 4 days which, considering the long half-life of 225Ac (9.9 d) (Vaidyanathan and Zalutsky, 1996), allows for irradiation of the entire spheroid. A decrease in spheroidal growth has been observed upon the addition of only 0.1 kBq 225Ac, an effect which was more pronounced for the 225Ac in polymersomes than when only coupled to DTPA. At higher activities (5 kBq), the spheroids have been found to be destroyed completely after two days. We have thus demonstrated that 225Ac containing polymersomes effectively inhibit tumour spheroid growth, making them very promising candidates for future in vivo testing.


Assuntos
Actínio/administração & dosagem , Actínio/química , Glioma/tratamento farmacológico , Polímeros/administração & dosagem , Polímeros/química , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Humanos , Esferoides Celulares/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
7.
Langmuir ; 32(33): 8329-38, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27468779

RESUMO

Effective targeting by uniformly functionalized nanoparticles is limited to cancer cells expressing at least two copies of targeted receptors per nanoparticle footprint (approximately ≥2 × 10(5) receptor copies per cell); such a receptor density supports the required multivalent interaction between the neighboring receptors and the ligands from a single nanoparticle. To enable selective targeting below this receptor density, ligands on the surface of lipid vesicles were displayed in clusters that were designed to form at the acidic pH of the tumor interstitium. Vesicles with clustered HER2-targeting peptides within such sticky patches (sticky vesicles) were compared to uniformly functionalized vesicles. On HER2-negative breast cancer cells MDA-MB-231 and MCF7 {expressing (8.3 ± 0.8) × 10(4) and (5.4 ± 0.9) × 10(4) HER2 copies per cell, respectively}, only the sticky vesicles exhibited detectable specific targeting (KD ≈ 49-69 nM); dissociation (0.005-0.009 min(-1)) and endocytosis rates (0.024-0.026 min(-1)) were independent of HER2 expression for these cells. MDA-MB-231 and MCF7 were killed only by sticky vesicles encapsulating doxorubicin (32-40% viability) or α-particle emitter (225)Ac (39-58% viability) and were not affected by uniformly functionalized vesicles (>80% viability). Toxicities on cardiomyocytes and normal breast cells (expressing HER2 at considerably lower but not insignificant levels) were not observed, suggesting the potential of tunable clustered ligand display for the selective killing of cancer cells with low receptor densities.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Lipídeos/química , Nanopartículas/química , Actínio/administração & dosagem , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacocinética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Endocitose , Feminino , Humanos , Cinética , Ligantes , Células MCF-7 , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Nanotecnologia , Receptor ErbB-2/metabolismo
8.
Oncotarget ; 7(22): 33306-15, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27119227

RESUMO

The standard treatment for ductal carcinoma in situ (DCIS) of the breast is surgical resection, followed by radiation. Here, we tested localized therapy of DCIS in mice using the immunoconjugate 225Ac linked-trastuzumab delivered through the intraductal (i.duc) route. Trastuzumab targets HER-2/neu, while the alpha-emitter 225Ac (half-life, 10 days) delivers highly cytotoxic, focused doses of radiation to tumors. Systemic 225Ac, however, elicits hematologic toxicity and at high doses free 213Bi, generated by its decay, causes renal toxicity. I.duc delivery of the radioimmunoconjugate could bypass its systemic toxicity. Bioluminescent imaging showed that the therapeutic efficacy of intraductal 225Ac-trastuzumab (10-40 nCi per mammary gland; 30-120 nCi per mouse) in a DCIS model of human SUM225 cancer cells in NSG mice was significantly higher (p<0.0003) than intravenous (120 nCi per mouse) administration, with no kidney toxicity or loss of body weight. Our findings suggest that i.duc radioimmunotherapy using 225Ac-trastuzumab deserves greater attention for future clinical development as a treatment modality for early breast cancer.


Assuntos
Actínio/administração & dosagem , Partículas alfa , Neoplasias da Mama/radioterapia , Carcinoma Intraductal não Infiltrante/radioterapia , Imunoconjugados/administração & dosagem , Radioimunoterapia/métodos , Radioisótopos/administração & dosagem , Compostos Radiofarmacêuticos/administração & dosagem , Receptor ErbB-2/metabolismo , Trastuzumab/administração & dosagem , Actínio/farmacocinética , Actínio/toxicidade , Partículas alfa/efeitos adversos , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/imunologia , Carcinoma Intraductal não Infiltrante/metabolismo , Carcinoma Intraductal não Infiltrante/patologia , Feminino , Humanos , Imunoconjugados/farmacocinética , Células MCF-7 , Camundongos Endogâmicos NOD , Radioimunoterapia/efeitos adversos , Radioisótopos/farmacocinética , Radioisótopos/toxicidade , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/toxicidade , Receptor ErbB-2/imunologia , Distribuição Tecidual , Trastuzumab/farmacocinética , Carga Tumoral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nat Rev Cancer ; 15(6): 347-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25998714

RESUMO

The eradication of cancer remains a vexing problem despite recent advances in our understanding of the molecular basis of neoplasia. One therapeutic approach that has demonstrated potential involves the selective targeting of radionuclides to cancer-associated cell surface antigens using monoclonal antibodies. Such radioimmunotherapy (RIT) permits the delivery of a high dose of therapeutic radiation to cancer cells, while minimizing the exposure of normal cells. Although this approach has been investigated for several decades, the cumulative advances in cancer biology, antibody engineering and radiochemistry in the past decade have markedly enhanced the ability of RIT to produce durable remissions of multiple cancer types.


Assuntos
Neoplasias/radioterapia , Radioimunoterapia/métodos , Actínio/administração & dosagem , Actínio/uso terapêutico , Partículas alfa/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Ensaios Clínicos como Assunto , Neoplasias Hematológicas/radioterapia , Humanos , Terapia de Alvo Molecular/métodos
10.
Cancer Biother Radiopharm ; 29(1): 34-41, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24102173

RESUMO

For targeted alpha therapy (TAT) with 225Ac, daughter radioisotopes from the parent emissions should be controlled. Here, we report on a second-generation layered nanoparticle (NP) with improved daughter retention that can mediate TAT of lung tumor colonies. NPs of La3+, Gd3+, and 225Ac3+ ions were coated with additional layers of GdPO4 and then coated with gold via citrate reduction of NaAuCl4. MAb 201b, targeting thrombomodulin in lung endothelium, was added to a polyethylene glycol (dPEG)-COOH linker. The NPs:mAb ratio was quantified by labeling the mAb with 125I. NPs showed 30% injected dose/organ antibody-mediated uptake in the lung, which increased to 47% in mice pretreated with clodronate liposomes to reduce phagocytosis. Retention of daughter 213Bi in lung tissue was more than 70% at one hour and about 90% at 24 hours postinjection. Treatment of mice with lung-targeted 225Ac NP reduced EMT-6 lung colonies relative to cold antibody competition for targeting or phosphate-buffered saline injected controls. We conclude that LnPO4 NPs represent a viable solution to deliver the 225Ac as an in vivo α generator. The NPs successfully retain a large percentage of the daughter products without compromising the tumoricidal properties of the α-radiation.


Assuntos
Actínio/administração & dosagem , Partículas alfa/uso terapêutico , Elementos da Série dos Lantanídeos/administração & dosagem , Nanopartículas/administração & dosagem , Fosfatos/administração & dosagem , Actínio/química , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Feminino , Elementos da Série dos Lantanídeos/química , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Nanotecnologia/métodos , Fosfatos/química , Cintilografia , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética
11.
Appl Radiat Isot ; 85: 45-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24374072

RESUMO

Alpha radionuclide therapy is steadily gaining importance and a large number of pre-clinical and clinical studies have been carried out. However, due to the recoil effects the daughter recoil atoms, most of which are alpha emitters as well, receive energies that are much higher than the energies of chemical bonds resulting in decoupling of the radionuclide from common targeting agents. Here, we demonstrate that polymer vesicles (i.e. polymersomes) can retain recoiling daughter nuclei based on an experimental study examining the retention of (221)Fr and (213)Bi when encapsulating (225)Ac.


Assuntos
Actínio/administração & dosagem , Actínio/química , Actínio/farmacocinética , Partículas alfa/uso terapêutico , Transporte Biológico Ativo , Bismuto/administração & dosagem , Bismuto/química , Bismuto/farmacocinética , Butadienos/química , Composição de Medicamentos , Frâncio/química , Células HeLa , Humanos , Radioisótopos de Chumbo/química , Método de Monte Carlo , Tamanho da Partícula , Polietileno/química , Radioisótopos/química , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética
12.
Bioconjug Chem ; 22(4): 766-76, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21434681

RESUMO

Nanoscale materials have been envisioned as carriers for various therapeutic drugs, including radioisotopes. Inorganic nanoparticles (NPs) are particularly appealing vehicles for targeted radiotherapy because they can package several radioactive atoms into a single carrier and can potentially retain daughter radioisotopes produced by in vivo generators such as actinium-225 ((225)Ac, t(1/2) = 10 d). Decay of this radioisotope to stable bismuth-209 proceeds through a chain of short-lived daughters accompanied by the emission of four α-particles that release >27 MeV of energy. The challenge in realizing the enhanced cytotoxic potential of in vivo generators lies in retaining the daughter nuclei at the therapy site. When (225)Ac is attached to targeting agents via standard chelate conjugation methods, all of the daughter radionuclides are released after the initial α-decay occurs. In this work, (225)Ac was incorporated into lanthanum phosphate NPs to determine whether the radioisotope and its daughters would be retained within the dense mineral lattice. Further, the (225)Ac-doped NPs were conjugated to the monoclonal antibody mAb 201B, which targets mouse lung endothelium through the vasculature, to ascertain the targeting efficacy and in vivo retention of radioisotopes. Standard biodistribution techniques and microSPECT/CT imaging of (225)Ac as well as the daughter radioisotopes showed that the NPs accumulated rapidly in mouse lung after intravenous injection. By showing that excess, competing, uncoupled antibodies or NPs coupled to control mAbs are deposited primarily in the liver and spleen, specific targeting of NP-mAb 201B conjugates was demonstrated. Biodistribution analysis showed that ∼30% of the total injected dose of La((225)Ac)PO(4) NPs accumulated in mouse lungs 1 h postinjection, yielding a value of % ID/g >200. Furthermore, after 24 h, 80% of the (213)Bi daughter produced from (225)Ac decay was retained within the target organ and (213)Bi retention increased to ∼87% at 120 h. In vitro analyses, conducted over a 1 month interval, demonstrated that ∼50% of the daughters were retained within the La((225)Ac)PO(4) NPs at any point over that time frame. Although most of the γ-rays from radionuclides in the (225)Ac decay chain are too energetic to be captured efficiently by SPECT detectors, appropriate energy windows were found that provided dramatic microSPECT images of the NP distribution in vivo. We conclude that La((225)Ac)PO(4)-mAb 201B conjugates can be targeted efficiently to mouse lung while partially retaining daughter products and that targeting can be monitored by biodistribution techniques and microSPECT imaging.


Assuntos
Actínio/química , Anticorpos Monoclonais/química , Lantânio/química , Nanopartículas/química , Fosfatos/química , Actínio/administração & dosagem , Actínio/farmacocinética , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/farmacocinética , Endotélio/química , Feminino , Lantânio/administração & dosagem , Lantânio/farmacocinética , Pulmão/química , Pulmão/citologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Fosfatos/administração & dosagem , Fosfatos/farmacocinética , Radioisótopos/química , Radioisótopos/farmacocinética , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único
13.
Int J Nanomedicine ; 5: 783-802, 2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-21042424

RESUMO

Single wall carbon nanotube (SWCNT) constructs were covalently appended with radiometal-ion chelates (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid [DOTA] or desferrioxamine B [DFO]) and the tumor neovascular-targeting antibody E4G10. The E4G10 antibody specifically targeted the monomeric vascular endothelial-cadherin (VE-cad) epitope expressed in the tumor angiogenic vessels. The construct specific activity and blood compartment clearance kinetics were significantly improved relative to corresponding antibodyalone constructs. We performed targeted radioimmunotherapy with a SWCNT-([(225)Ac]DOTA) (E4G10) construct directed at the tumor vasculature in a murine xenograft model of human colon adenocarcinoma (LS174T). The specific construct reduced tumor volume and improved median survival relative to controls. We also performed positron emission tomographic (PET) radioimmunoimaging of the tumor vessels with a SWCNT-([(89)Zr]DFO)(E4G10) construct in the same murine LS174T xenograft model and compared the results to appropriate controls. Dynamic and longitudinal PET imaging of LS174T tumor-bearing mice demonstrated rapid blood clearance (<1 hour) and specific tumor accumulation of the specific construct. Incorporation of the SWCNT scaffold into the construct design permitted us to amplify the specific activity to improve the signal-to-noise ratio without detrimentally impacting the immunoreactivity of the targeting antibody moiety. Furthermore, we were able to exploit the SWCNT pharmacokinetic (PK) profile to favorably alter the blood clearance and provide an advantage for rapid imaging. Near-infrared three-dimensional fluorescent-mediated tomography was used to image the LS174T tumor model, collect antibody-alone PK data, and calculate the number of copies of VE-cad epitope per cell. All of these studies were performed as a single administration of construct and were found to be safe and well tolerated by the murine model. These data have implications that support further imaging and radiotherapy studies using a SWCNT-based platform and focusing on the tumor vessels as the target.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/radioterapia , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/radioterapia , Nanotubos de Carbono/química , Compostos Radiofarmacêuticos/uso terapêutico , Actínio/administração & dosagem , Actínio/uso terapêutico , Adenocarcinoma/irrigação sanguínea , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/irrigação sanguínea , Desferroxamina/química , Compostos Heterocíclicos com 1 Anel/química , Humanos , Masculino , Camundongos , Camundongos Nus , Nanomedicina , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/radioterapia , Tomografia por Emissão de Pósitrons/métodos , Radioimunodetecção/métodos , Radioimunoterapia/métodos , Radioisótopos/administração & dosagem , Radioisótopos/uso terapêutico , Compostos Radiofarmacêuticos/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto , Zircônio/administração & dosagem , Zircônio/uso terapêutico
14.
Cancer Res ; 65(11): 4888-95, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930310

RESUMO

Alpha-particle immunotherapy by targeted alpha-emitters or alpha-emitting isotope generators is a novel form of extraordinarily potent cancer therapy. A major impediment to the clinical use of targeted actinium-225 (225Ac) in vivo generators may be the radiotoxicity of the systemically released daughter radionuclides. The daughters, especially bismuth-213 (213Bi), tend to accumulate in the kidneys. We tested the efficacy of various pharmacologic agents and the effect of tumor burden in altering the pharmacokinetics of the 225Ac daughters to modify their renal uptake. Pharmacologic treatments in animals were started before i.v. administration of the HuM195-225Ac generator. 225Ac, francium-221 (221Fr), and 213Bi biodistributions were calculated in each animal at different time points after 225Ac generator injection. Oral metal chelation with 2,3-dimercapto-1-propanesulfonic acid (DMPS) or meso-2,3-dimercaptosuccinic acid (DMSA) caused a significant reduction (P < 0.0001) in the renal 213Bi uptake; however, DMPS was more effective than DMSA (P < 0.001). The results with DMPS were also confirmed in a monkey model. The renal 213Bi and 221Fr activities were significantly reduced by furosemide and chlorothiazide treatment (P < 0.0001). The effect on renal 213Bi activity was further enhanced by the combination of DMPS with either chlorothiazide or furosemide (P < 0.0001). Competitive antagonism by bismuth subnitrate moderately reduced the renal uptake of 213Bi. The presence of a higher target-tumor burden significantly prevented the renal 213Bi accumulation (P = 0.003), which was further reduced by DMPS treatment (P < 0.0001). Metal chelation, diuresis with furosemide or chlorothiazide, and competitive metal blockade may be used as adjuvant therapies to modify the renal accumulation of 225Ac daughters.


Assuntos
Actínio/administração & dosagem , Partículas alfa , Quelantes/farmacologia , Imunotoxinas/administração & dosagem , Rim/efeitos dos fármacos , Rim/efeitos da radiação , Radioimunoterapia/métodos , Actínio/sangue , Actínio/química , Actínio/farmacocinética , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Bismuto/química , Bismuto/farmacocinética , Feminino , Frâncio/química , Frâncio/farmacocinética , Imunotoxinas/sangue , Imunotoxinas/química , Imunotoxinas/farmacocinética , Rim/metabolismo , Linfoma/metabolismo , Linfoma/patologia , Linfoma/radioterapia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Succímero/farmacologia , Unitiol/farmacologia
15.
Nucl Med Biol ; 31(4): 441-9, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15093814

RESUMO

The alpha-particle emitting radionuclides (223)Ra (t(1/2) = 11.4 d), (224)Ra (t(1/2) = 3.6 d), and (225)Ac(t(1/2) = 10.0 d) may have a broad application in targeted radiotherapy provided that they could be linked to vehicles with tumor affinity. The potential usefulness of liposomes as carriers was studied in the present work. Radium and actinium radionuclides could be loaded in good yields into sterically stabilized liposomes. Subsequent coating of the liposomes with a folate-F(ab')(2) construct yielded a product with affinity towards tumor cells expressing folate receptors. Radionuclide loaded liposomes showed excellent stability in serum in vitro.


Assuntos
Actínio/administração & dosagem , Actínio/farmacocinética , Lipossomos/química , Neoplasias Ovarianas/metabolismo , Radioimunoterapia/métodos , Rádio (Elemento)/administração & dosagem , Rádio (Elemento)/farmacocinética , Actínio/química , Partículas alfa/uso terapêutico , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Materiais Revestidos Biocompatíveis/síntese química , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Estabilidade de Medicamentos , Feminino , Receptores de Folato com Âncoras de GPI , Ácido Fólico/química , Ácido Fólico/farmacocinética , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Lipossomos/síntese química , Lipossomos/farmacocinética , Taxa de Depuração Metabólica , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Rádio (Elemento)/química , Receptores de Superfície Celular/metabolismo , Estereoisomerismo
16.
J Nucl Med ; 45(2): 253-60, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14960644

RESUMO

UNLABELLED: Disseminated, metastatic cancer is frequently incurable. Targeted alpha-particle emitters hold great promise as therapeutic agents for disseminated disease. (225)Ac is a radionuclide generator that has a 10-d half-life and results in alpha-emitting daughter elements ((221)Fr, (217)At, (213)Bi) that lead to the emission of a total of 4 alpha-particles. The aim of this study was to develop approaches for stable and controlled targeting of (225)Ac to sites of disseminated tumor metastases. Liposomes with encapsulated (225)Ac were developed to retain the potentially toxic daughters at the tumor site. METHODS: (225)Ac was passively entrapped in liposomes. To experimentally test the retention of actinium and its daughters by the liposomes, the gamma-emissions of (213)Bi were measured in liposome fractions, which were separated from the parent liposome population and the free radionuclides, at different times. Under equilibrium conditions the decay rate of (213)Bi was used to determine the concentration of (225)Ac. Measurements of the kinetics of (213)Bi activity were performed to estimate the entrapment of (213)Bi, the last alpha-emitting daughter in the decay chain. RESULTS: Stable pegylated phosphatidylcholine-cholesterol liposomes of different sizes and charge were prepared. Multiple (more than 2) (225)Ac atoms were successfully entrapped per liposome. (225)Ac retention by zwitterionic liposomes was more than 88% over 30 d. Retention by cationic liposomes was lower. A theoretical calculation showed that for satisfactory (213)Bi retention (>50%), liposomes of relatively large sizes (>650 nm in diameter) are required. (213)Bi retention was experimentally verified to be liposome-size dependent. For large liposomes, the measured (213)Bi retention was lower than theoretically predicted (less than 10%). CONCLUSION: This work supports the hypothesis that it may be possible to develop (225)Ac-based therapies by delivering multiple (225)Ac atoms in liposomes. Improvements in the retention of (225)Ac daughters will likely be necessary to fulfill this potential. Because of the size of the liposomal structures required to contain the daughters, the approach is ideally suited for locoregional therapy (e.g., intraperitoneal, intrahepatic artery, or intrathecal).


Assuntos
Actínio/uso terapêutico , Actínio/administração & dosagem , Partículas alfa , Animais , Bismuto , Humanos , Lipossomos , Microscopia Eletrônica , Metástase Neoplásica/radioterapia , Radioisótopos
17.
J Nucl Med ; 45(1): 129-37, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14734685

RESUMO

UNLABELLED: Short-lived alpha-emitting isotopes individually conjugated to monoclonal antibodies have now reached human use, but little is still known about their toxicity. Use of antibody targetable (225)Ac nanogenerators is a new approach in the field of alpha-immunotherapy offering the advantage of a 10-d half-life (t(1/2)) and increased potency due to generation of 3 new atoms, yielding a total of 4 alpha-particles. However, the 3 alpha-emitting daughter elements generated have the potential for significant toxicity as these nuclides are no longer bound to the carrier IgG. METHODS: Cynomolgus monkeys were used to evaluate the toxicity of prototype (225)Ac nanogenerators. Monoclonal antibody HuM195 (anti-CD33) is the carrier for planned human clinical trials of (225)Ac; there are no CD33 sites in cynomolgus monkeys. In one experiment, 2 monkeys received a single intravenous dose of (225)Ac-HuM195 at 28 kBq/kg. This dose level is approximately the planned initial human dose. In another experiment, 2 animals received a dose escalation schedule of 3 increasing (225)Ac-HuM195 doses with a cumulative activity of 377 kBq/kg. The whole-blood t(1/2) of (225)Ac, ratios of (225)Ac to its ultimate alpha-emitting daughter nuclide (213)Bi, generation of monkey anti-HuM195 antibodies (MAHA), hematologic indices, serum biochemistries, and clinical parameters were measured. Monkeys were euthanized and examined histopathologically when the dose escalation reached toxicity. RESULTS: The blood t(1/2) of (225)Ac-HuM195 was 12 d, and 45% of generated (213)Bi daughters were cleared from the blood. MAHA production was not detected. Approximately 28 kBq/kg of (225)Ac caused no toxicity at 6 mo, whereas a cumulative dose of approximately 377 kBq/kg caused severe toxicity. In the cumulative dosing schedule, single doses of approximately 37 kBq/kg resulted in no toxicity at 6 wk. After approximately 130 kBq/kg were administered, no toxicity was observed for 13 wk. However, 28 wk after this second dose administration, mild anemia and increases of blood urea nitrogen and creatinine were detected. After administration of an additional 185 kBq/kg, toxicity became clinically apparent. Monkeys were euthanized 13 and 19 wk after the third dose administration (cumulative dose was 377 kBq/kg). Histopathologic evaluation revealed mainly renal tubular damage associated with interstitial fibrosis. CONCLUSION: (225)Ac nanogenerators may result in renal toxicity and anemia at high doses. The longer blood t(1/2) and the lack of target cell antigens in cynomolgus monkeys may increase toxicity compared with human application. Therefore, a dose level of at least 28 kBq/kg may be a safe starting dose in humans. Hematologic and renal function will require close surveillance during clinical trials.


Assuntos
Actínio/efeitos adversos , Actínio/farmacocinética , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Creatina/sangue , Rim/metabolismo , Rim/efeitos da radiação , Actínio/administração & dosagem , Actínio/sangue , Anemia/etiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais Humanizados , Relação Dose-Resposta à Radiação , Humanos , Inativação Metabólica , Rim/efeitos dos fármacos , Rim/patologia , Macaca fascicularis , Masculino , Doses de Radiação , Radioimunoterapia/métodos , Radiometria , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/efeitos adversos , Compostos Radiofarmacêuticos/sangue , Compostos Radiofarmacêuticos/farmacocinética , Insuficiência Renal/etiologia
18.
Science ; 294(5546): 1537-40, 2001 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11711678

RESUMO

A single, high linear energy transfer alpha particle can kill a target cell. We have developed methods to target molecular-sized generators of alpha-emitting isotope cascades to the inside of cancer cells using actinium-225 coupled to internalizing monoclonal antibodies. In vitro, these constructs specifically killed leukemia, lymphoma, breast, ovarian, neuroblastoma, and prostate cancer cells at becquerel (picocurie) levels. Injection of single doses of the constructs at kilobecquerel (nanocurie) levels into mice bearing solid prostate carcinoma or disseminated human lymphoma induced tumor regression and prolonged survival, without toxicity, in a substantial fraction of animals. Nanogenerators targeting a wide variety of cancers may be possible.


Assuntos
Actínio/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias/radioterapia , Radioimunoterapia/métodos , Actínio/administração & dosagem , Actínio/farmacocinética , Partículas alfa/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Feminino , Meia-Vida , Compostos Heterocíclicos com 1 Anel , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/farmacocinética , Transferência Linear de Energia , Linfoma/radioterapia , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/radioterapia , Taxa de Sobrevida , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...