Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Biother Radiopharm ; 33(5): 182-193, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29916748

RESUMO

Refinement of treatment regimens enlisting targeted α-radiation therapy (TAT) is an ongoing effort. Among the variables to consider are the target molecule, radionuclide, dosage, and administration route. The panitumumab F(ab')2 fragment targeting epidermal growth factor receptor tolerated modification with the TCMC chelate as well as radiolabeling with 203Pb or 212Pb. Good specific activity was attained when the immunoconjugate was labeled with 212Pb (9.6 ± 1.4 mCi/mg). Targeting of LS-174T tumor xenografts with the 203Pb-panitumumab F(ab')2 demonstrated comparable amounts of uptake to the similarly radiolabeled panitumumab IgG. A dose escalation study was performed to determine an effective working dose for both intraperitoneal (i.p.) and intravenous (i.v.) injections of 212Pb-panitumumab F(ab')2. Therapeutic efficacy, with modest toxicity, was observed with 30 µCi given i.p. Results for the i.v. administration were not as definitive and the experiment was repeated with a higher dose range. From this study, 20 µCi given i.v. was selected as the effective working dose. A subsequent therapy study combined gemcitabine or paclitaxel with i.v. 212Pb-panitumumab F(ab')2, which increased the median survival (MS) of LS-174T tumor-bearing mice to 208 and 239 d, respectively. Meanwhile, the MS of mice treated with i.v. 212Pb-panitumumab F(ab')2 alone was 61 and 11 d for the untreated group of mice. In conclusion, the panitumumab F(ab')2 fragment whether given by i.p. or i.v. injection, is a viable candidate as a delivery vector for TAT of disseminated i.p. disease.


Assuntos
Partículas alfa , Anticorpos Monoclonais/administração & dosagem , Neoplasias do Colo/terapia , Receptores ErbB/antagonistas & inibidores , Radioisótopos de Chumbo/uso terapêutico , Radioimunoterapia , Administração Intravenosa , Animais , Apoptose , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Receptores ErbB/imunologia , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Injeções Intraperitoneais , Camundongos , Camundongos Nus , Panitumumabe , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Dalton Trans ; 46(42): 14591-14601, 2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-28675216

RESUMO

Identification of the appropriate combination of radionuclide, target and targeting vehicle is critical for successful radioimmunotherapy. For the treatment of disseminated peritoneal diseases such as pancreatic or ovarian cancer, α-emitting radionuclides have been proposed for targeted radiation therapy. This laboratory has taken a systematic approach investigating targeted α-radiation therapy, allowing comparisons to now be made between 211At, 227Th, 213Bi and 212Pb. Herein, trastuzumab radiolabeled with 211At and 227Th was evaluated for therapeutic efficacy in the LS-174T i.p. tumor model. A dose escalation study was conducted with each radioimmunoconjugate (RIC). Therapeutic benefit was realized with 211At-trastuzumab with doses of 20, 30 and 40 µCi. At doses >40 µCi, toxicity was observed with greater weight loss and 2-fold higher decrease in the platelet counts. Following a second study comparing the effect of 20, 30 and 40 µCi of 211At-trastuzumab, 30 µCi was selected as the dose for future studies. A parallel study was performed evaluating 0.25, 0.5, 1.0, 2.0 and 5.0 µCi of 227Th-trastuzumab. The 0.5 and 1.0 µCi injected dose resulted in a therapeutic response; a lower degree of weight loss was experienced by the mice in the 0.5 µCi cohort. When the data is normalized for comparing 211At, 227Th, 213Bi and 212Pb, the choice of radionuclide for RIT is perhaps not entirely based on simple therapeutic efficacy, other factors may play a role in choosing the "right" radionuclide.


Assuntos
Partículas alfa/uso terapêutico , Neoplasias do Colo/radioterapia , Radioimunoterapia/métodos , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Feminino , Humanos , Camundongos , Distribuição Tecidual , Trastuzumab/farmacocinética , Trastuzumab/uso terapêutico
3.
Transl Oncol ; 10(4): 535-545, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28577439

RESUMO

Identifying molecular targets and an appropriate targeting vehicle, i.e., monoclonal antibodies (mAb) and their various forms, for radioimmunotherapy (RIT) remains an active area of research. Panitumumab, a fully human and less immunogenic mAb that binds to the epidermal growth factor receptor (Erb1; HER1), was evaluated for targeted α-particle radiation therapy using 212Pb, an in vivo α generator. A single dose of 212Pb-panitumumab administered to athymic mice bearing LS-174T intraperitoneal (i.p.) tumor xenografts was found to have greater therapeutic efficacy when directly compared with 212Pb-trastuzumab, which binds to HER2. A dose escalation study determined a maximum effective working dose of 212Pb-panitumumab to be 20µCi with a median survival of 35 days versus 25 days for the untreated controls. Pretreatment of tumor-bearing mice with paclitaxel and gemcitabine 24hours prior to injection of 212Pb-pantiumumab at 10 or 20µCi resulted in the greatest enhanced therapeutic response at the higher dose with median survivals of 106 versus 192 days, respectively. The greatest therapeutic impact, however, was observed in the animals that were treated with topotecan 24hours prior to RIT and then again 24hours after RIT; the best response from this combination was also obtained with the lower 10-µCi dose of 212Pb-panitumumab (median survival >280 days). In summary, 212Pb-panitumumab is an excellent candidate for the treatment of HER1-positive disseminated i.p. disease. Furthermore, the potentiation of the therapeutic impact of 212Pb-pantiumumab by chemotherapeutics confirms and validates the importance of developing a multimodal therapy regimen.

4.
PLoS One ; 11(7): e0159904, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467592

RESUMO

In pre-clinical studies, combination therapy with gemcitabine and targeted radioimmunotherapy (RIT) using 212Pb-trastuzumab showed tremendous therapeutic potential in the LS-174T tumor xenograft model of disseminated intraperitoneal disease. To better understand the underlying molecular basis for the observed cell killing efficacy, gene expression profiling was performed after a 24 h exposure to 212Pb-trastuzumab upon gemcitabine (Gem) pre-treatment in this model. DNA damage response genes in tumors were quantified using a real time quantitative PCR array (qRT-PCR array) covering 84 genes. The combination of Gem with α-radiation resulted in the differential expression of apoptotic genes (BRCA1, CIDEA, GADD45α, GADD45γ, IP6K3, PCBP4, RAD21, and p73), cell cycle regulatory genes (BRCA1, CHK1, CHK2, FANCG, GADD45α, GTSE1, PCBP4, MAP2K6, NBN, PCBP4, and SESN1), and damaged DNA binding and repair genes (BRCA1, BTG2, DMC1, ERCC1, EXO1, FANCG, FEN1, MSH2, MSH3, NBN, NTHL1, OGG1, PRKDC, RAD18, RAD21, RAD51B, SEMA4G, p73, UNG, XPC, and XRCC2). Of these genes, the expression of CHK1, GTSE1, EXO1, FANCG, RAD18, UNG and XRCC2 were specific to Gem/212Pb-trastuzumab administration. In addition, the present study demonstrates that increased stressful growth arrest conditions induced by Gem/212Pb-trastuzumab could suppress cell proliferation possibly by up-regulating genes involved in apoptosis such as p73, by down-regulating genes involved in cell cycle check point such as CHK1, and in damaged DNA repair such as RAD51 paralogs. These events may be mediated by genes such as BRCA1/MSH2, a member of BARC (BRCA-associated genome surveillance complex). The data suggest that up-regulation of genes involved in apoptosis, perturbation of checkpoint genes, and a failure to correctly perform HR-mediated DSB repair and mismatch-mediated SSB repair may correlate with the previously observed inability to maintain the G2/M arrest, leading to cell death.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias do Colo/patologia , Desoxicitidina/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Radioisótopos de Chumbo/administração & dosagem , Trastuzumab/farmacologia , Animais , Apoptose/genética , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Dano ao DNA/genética , Reparo do DNA/genética , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Xenoenxertos , Humanos , Camundongos , Trastuzumab/uso terapêutico , Gencitabina
5.
Int J Mol Sci ; 17(5)2016 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-27196891

RESUMO

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.


Assuntos
Neoplasias do Colo/radioterapia , Imunoconjugados/farmacologia , Lutécio/farmacologia , Radioisótopos/farmacologia , Trastuzumab/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antineoplásicos/farmacologia , Apoptose/genética , Apoptose/efeitos da radiação , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Aberrações Cromossômicas/efeitos da radiação , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Immunoblotting , Injeções Intraperitoneais , Transferência Linear de Energia , Camundongos Nus , Transplante de Neoplasias/métodos , Rad51 Recombinase/metabolismo , Radioimunoterapia/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Pharmaceuticals (Basel) ; 8(3): 435-54, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26230702

RESUMO

The first-in-human phase 1 clinical radioimmunotherapy (RIT) trial with 212Pb-1,4,7,10-tetraaza-1,4,7,10-tetra-(2-carbamoylmethyl)-cyclododecane-trastuzumab (212Pb-TCMC-trastuzumab) was completed in October 2014 as a joint effort at the University of Alabama (UAB) and the University of California San Diego Moores Cancer Center. The preliminary reports indicate that after five dose-levels of intraperitoneally administered 212Pb-TCMC-trastuzumab, patients with carcinomatosis experienced minimal agent-related toxicity. This report presents the data accumulated to date on the stability of the clinical grade, produced according to current good manufacturing practices (cGMP), TCMC-trastuzumab conducted in support of that clinical trial. Of the eleven tests performed with the cGMP TCMC-trastuzumab all but one remained within specifications throughout the 5 year testing period. The protein concentration varied by 0.01 mg/mL at 48 months. Two other assays, ion-exchange high performance liquid chromatography (IEX-HPLC) and a competitive radioimmunoassay (RIA) indicated that the cGMP TCMC-trastuzumab integrity may be changing, although the change thus far is within specifications. Subsequent stability testing will confirm if a trend has truly developed. The cGMP TCMC-trastuzumab was also evaluated for tolerance to higher temperatures and the potential of storage at -80 °C. The immunoconjugate proved stable when subjected to the lower temperatures and to multiple freeze-thaw cycles. The size exclusion (SE) HPLC analysis of the 203Pb-TCMC-trastuzumab was the only indicator that cGMP TCMC-trastuzumab may be sensitive to storage at 37 °C for 3 months.

7.
Pharmaceuticals (Basel) ; 8(3): 416-34, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26213947

RESUMO

Faced with the novelty of a 212Pb-labeled monoclonal antibody (mAb) for clinical translation, concerns were expressed by the Food and Drug Administration (FDA) regarding 212Pb prematurely released from the mAb-chelate conjugate. The objective of this study was to simulate the worst case scenario of such a failure. Groups of Balb/c mice (n = 9-20) were administered 212Pb by intraperitoneal (0.0925-1.85 MBq) or intravenous (0.0925-1.11 MBq) injection and then euthanized at 7 or 90 days to assess acute or chronic effects. Weights were recorded prior to injection of the 212Pb and at the end of the observation periods. Blood samples were collected for clinical chemistry and blood cell analysis. Thirty tissues were harvested and formalin fixed for histopathological examination. Treatment related effects of the 212Pb were observed in the bone marrow, spleen, kidneys and the liver. Histological alterations in these organs were considered mild to moderate, indicating low grade toxicity, and not considered severe enough to affect function. This data was presented to the FDA and determined to be acceptable. The clinical trial with 212Pb-TCMC-trastuzumab was approved in January 2011 and the trial opened at the University of Alabama at Birmingham (UAB) in July.

8.
Bioorg Med Chem Lett ; 25(10): 2056-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25870133

RESUMO

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.


Assuntos
Meios de Contraste/síntese química , Complexos de Coordenação/síntese química , Gadolínio/química , Integrina alfaVbeta3/química , Imageamento por Ressonância Magnética , Peptidomiméticos/química , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Meios de Contraste/química , Complexos de Coordenação/química , Humanos , Concentração Inibidora 50 , Peptidomiméticos/síntese química
9.
MAbs ; 7(1): 255-64, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25587678

RESUMO

Although the epidermal growth factor receptor (EGFR), also known as HER1, has been studied for over a decade, it continues to be a molecule of great interest and focus of investigators for development of targeted therapies. The marketed monoclonal antibody cetuximab binds to HER1, and thus might serve as the basis for creation of imaging or therapies that target this receptor. The potential of cetuximab as a vehicle for the delivery of α-particle radiation was investigated in an intraperitoneal tumor mouse model. The effective working dose of 10 µCi of (212)Pb-cetuximab was determined from a dose (10-50 µCi) escalation study. Toxicity, as indicated by the lack of animal weight loss, was not evident at the 10 µCi dose of (212)Pb-cetuximab. A subsequent study demonstrated (212)Pb-cetuximab had a therapeutic efficacy similar to that of (212)Pb-trastuzumab (p = 0.588). Gemcitabine given 24 h prior to (212)Pb-cetuximab increased the median survival from 174 d to 283 d, but carboplatin suppressed the effectiveness of (212)Pb-cetuximab. Notably, concurrent treatment of tumor-bearing mice with (212)Pb-labeled cetuximab and trastuzumab provided therapeutic benefit that was greater than either antibody alone. In conclusion, cetuximab proved to be an effective vehicle for targeting HER1-expressing tumors with α-radiation for the treatment of disseminated intraperitoneal disease. These studies provide further evidence that the multimodality therapy regimens may have greater efficacy and benefit in the treatment of cancer patients.


Assuntos
Partículas alfa , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias Peritoneais/radioterapia , Radioimunoterapia/métodos , Animais , Linhagem Celular Tumoral , Cetuximab , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Humanos , Isótopos , Camundongos , Camundongos Nus , Radiossensibilizantes/farmacologia , Gencitabina
10.
PLoS One ; 9(9): e108511, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25268703

RESUMO

To better understand the molecular basis of the enhanced cell killing effected by the combined modality of paclitaxel and ²¹²Pb-trastuzumab (Pac/²¹²Pb-trastuzumab), gene expression in LS-174T i.p. xenografts was investigated 24 h after treatment. Employing a real time quantitative PCR array (qRT-PCR array), 84 DNA damage response genes were quantified. Differentially expressed genes following therapy with Pac/²¹²Pb-trastuzumab included those involved in apoptosis (BRCA1, CIDEA, GADD45α, GADD45γ, GML, IP6K3, PCBP4, PPP1R15A, RAD21, and p73), cell cycle (BRCA1, CHK1, CHK2, GADD45α, GML, GTSE1, NBN, PCBP4, PPP1R15A, RAD9A, and SESN1), and damaged DNA repair (ATRX, BTG2, EXO1, FEN1, IGHMBP2, OGG1, MSH2, MUTYH, NBN, PRKDC, RAD21, and p73). This report demonstrates that the increased stressful growth arrest conditions induced by the Pac/²¹²Pb-trastuzumab treatment suppresses cell proliferation through the regulation of genes which are involved in apoptosis and damaged DNA repair including single and double strand DNA breaks. Furthermore, the study demonstrates that ²¹²Pb-trastuzumab potentiation of cell killing efficacy results from the perturbation of genes related to the mitotic spindle checkpoint and BASC (BRCA1-associated genome surveillance complex), suggesting cross-talk between DNA damage repair and the spindle damage response.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/terapia , Regulação Neoplásica da Expressão Gênica , Paclitaxel/farmacologia , Neoplasias Peritoneais/terapia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Terapia Combinada , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , Quebras de DNA de Cadeia Simples/efeitos da radiação , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Perfilação da Expressão Gênica , Humanos , Injeções Intraperitoneais , Radioisótopos de Chumbo , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Radioimunoterapia/métodos , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Bioconjug Chem ; 25(10): 1801-10, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25243604

RESUMO

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.


Assuntos
Anticorpos Monoclonais/análise , Corantes/análise , Imunoconjugados/análise , Verde de Indocianina/análogos & derivados , Animais , Anticorpos Monoclonais/farmacocinética , Corantes/farmacocinética , Imunoconjugados/farmacocinética , Verde de Indocianina/análise , Verde de Indocianina/farmacocinética , Camundongos Nus , Imagem Óptica , Panitumumabe
12.
Cancer Biother Radiopharm ; 28(6): 441-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23758610

RESUMO

The studies described herein assess the potential of combining platinum-based chemotherapy with high-linear energy transfer (LET) α-particle-targeted radiation therapy using trastuzumab as the delivery vehicle. An initial study explored the combination of cisplatin with (213)Bi-trastuzumab in the LS-174T i.p. xenograft model. This initial study determined the administration sequence of cisplatin and (213)Bi-trastuzumab. Cisplatin coinjected with (213)Bi-trastuzumab increased the median survival (MS) to 90 days versus 65 days for (213)Bi-trastuzumab alone. Toxicity was observed with a weight loss of 17.6% in some of the combined treatment groups. Carboplatin proved to be better tolerated. Maximal therapeutic benefit, that is, a 5.1-fold increase in MS, was obtained in the group injected with (213)Bi-trastuzumab, followed by carboplatin 24 hours later. This was further improved by administration of multiple weekly doses of carboplatin. The MS achieved with administration of 3 doses of carboplatin was 180 days versus 60 days with (213)Bi-trastuzumab alone. The combination of carboplatin with (212)Pb radioimmunotherapy was also evaluated. The therapeutic efficacy of (212)Pb-trastuzumab (58-day MS) increased when the mice were pretreated with carboplatin 24 hours prior (157-day MS). These results again demonstrate the necessity of empirically determining the administration sequence when combining therapeutic modalities.


Assuntos
Partículas alfa/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/terapia , Compostos Organoplatínicos/farmacologia , Receptor ErbB-2/metabolismo , Animais , Bismuto/administração & dosagem , Linhagem Celular Tumoral , Quimiorradioterapia , Neoplasias do Colo/enzimologia , Neoplasias do Colo/imunologia , Feminino , Humanos , Radioisótopos de Chumbo/administração & dosagem , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Radioimunoterapia , Radioisótopos/administração & dosagem , Compostos Radiofarmacêuticos/farmacologia , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Nucl Med Biol ; 40(5): 592-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23602604

RESUMO

INTRODUCTION: Alpha particles possess an exquisite degree of cytotoxicity when employed for targeted α-particle therapy (TAT) or radioimmunotherapy (RIT). (212)Pb, which acts as an in vivo generator of the α-emitting nuclide (212)Bi has shown great promise in pre-clinical studies when used to label the HER2 binding antibody, trastuzumab. Currently, the first RIT clinical trial employing (212)Pb radiolabeled trastuzumab is in progress. This report provides detailed current protocol operations and steps that were generated for use in the clinical trial as well as the relevant pre-clinical experimentation, and describes in detail the labeling of proteins or peptides with (212)Pb as provided via a (224)Ra based generator system. METHODS: (212)Pb was eluted from the (224)Ra/(212)Pb generator using hydrochloric acid (2M). The generator eluate was evaporated and digested with nitric acid (8M) followed by extraction of the (212)Pb with dilute nitric acid (0.1M). The dilute nitric acid solution of (212)Pb was used to label the immunoconjugate Trastuzumab-TCMC (2-(4-isothiocyanatobenzyl-1,4,7,10-tetraaza-1,4,7,10,tetra-(2-carbamonylmethyl)-cyclododecane) at pH5.5. RESULTS: Elution of (212)Pb from the generator was efficient yielding>90% of available (212)Pb. Trastuzumab-TCMC was efficiently labeled with a radiochemical yield of 94% ± 4% (n=7) by ITLC and an isolated yield of 73% ± 3% (n=7). CONCLUSIONS: The results show the feasibility of generating radioimmunoconjugates and peptide conjugates for use as in vivo α generator systems in the clinic. The technology holds promise in applications involving the treatment of minimal disease such as micrometastases and residual tumor after surgical debulking, hematological cancers, infections, and compartmental cancers, such as ovarian cancer.


Assuntos
Marcação por Isótopo/métodos , Radioisótopos de Chumbo/química , Peptídeos/química , Proteínas/química , Radioisótopos de Chumbo/uso terapêutico
14.
Nucl Med Biol ; 40(5): 600-5, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23541026

RESUMO

INTRODUCTION: Despite the great potential of targeted α-radioimmunotherapy (RIT) as demonstrated by pre-clinical and clinical trials, limited progress has been made on the improvement of chelation chemistry for (212)Bi and (213)Bi. A new bifunctional ligand 3p-C-NETA was evaluated for targeted α RIT using (212)Bi and (213)Bi. METHODS: Radiolabeling of 3p-C-NETA with (205/6)Bi, a surrogate of (212)Bi and (213)Bi, was evaluated at pH5.5 and room temperature. In vitro stability of the (205/6)Bi-3p-C-NETA-trastuzumab conjugate was evaluated using human serum (pH7, 37 °C). Immunoreactivity and specific activity of the (205/6)Bi-3p-C-NETA-trastuzumab conjugate were measured. An in vivo biodistribution study was performed to evaluate the in vivo stability and tumor targeting properties of the (205/6)Bi-3p-C-NETA-trastuzumab conjugate in athymic mice bearing subcutaneous LS174T tumor xenografts. RESULT: The 3p-C-NETA-trastuzumab conjugate was extremely rapid in complexing with (205/6)Bi, and the corresponding (205/6)Bi-3p-C-NETA-trastuzumab was stable in human serum. (205/6)Bi-3p-C-NETA-trastuzumab was prepared with a high specific activity and retained immunoreactivity. (205/6)Bi-3p-C-NETA-trastuzumab conjugate displayed excellent in vivo stability and targeting as evidenced by low normal organ and high tumor uptake. CONCLUSION: The results of the in vitro and in vivo studies indicate that 3p-C-NETA is a promising chelator for RIT applications using (212)Bi and (213)Bi. Further detailed in vivo evaluations of 3p-C-NETA for targeted α RIT are warranted.


Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais Humanizados/uso terapêutico , Bismuto/uso terapêutico , Compostos Heterocíclicos/química , Radioimunoterapia/métodos , Radioisótopos/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Monoclonais Humanizados/farmacocinética , Transporte Biológico , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Feminino , Humanos , Marcação por Isótopo , Ligantes , Camundongos , Distribuição Tecidual , Trastuzumab
15.
Radiology ; 267(1): 173-82, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23329660

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores ErbB/metabolismo , Imageamento por Ressonância Magnética/métodos , Mesotelioma/metabolismo , Imagem Multimodal/métodos , Tomografia por Emissão de Pósitrons , Receptor ErbB-2/metabolismo , Tomografia Computadorizada por Raios X , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Área Sob a Curva , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Radioisótopos do Iodo , Radioisótopos de Irídio , Mesotelioma/diagnóstico por imagem , Mesotelioma/patologia , Camundongos , Terapia de Alvo Molecular , Panitumumabe , Derrame Pleural/diagnóstico por imagem , Derrame Pleural/patologia , Trastuzumab , Imagem Corporal Total , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Int J Radiat Oncol Biol Phys ; 85(4): 1119-26, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23200172

RESUMO

PURPOSE: To elucidate the mechanism of the therapeutic efficacy of targeted α-particle radiation therapy using (212)Pb-TCMC-trastuzumab together with gemcitabine for treatment of disseminated peritoneal cancers. METHODS AND MATERIALS: Mice bearing human colon cancer LS-174T intraperitoneal xenografts were pretreated with gemcitabine, followed by (212)Pb-TCMC-trastuzumab and compared with controls. RESULTS: Treatment with (212)Pb-TCMC-trastuzumab increased the apoptotic rate in the S-phase-arrested tumors induced by gemcitabine at earlier time points (6 to 24 hours). (212)Pb-TCMC-trastuzumab after gemcitabine pretreatment abrogated G2/M arrest at the same time points, which may be associated with the inhibition of Chk1 phosphorylation and, in turn, cell cycle perturbation, resulting in apoptosis. (212)Pb-TCMC-trastuzumab treatment after gemcitabine pretreatment caused depression of DNA synthesis, DNA double-strand breaks, accumulation of unrepaired DNA, and down-regulation of Rad51 protein, indicating that DNA damage repair was blocked. In addition, modification in the chromatin structure of p21 may be associated with transcriptionally repressed chromatin states, indicating that the open structure was delayed at earlier time points. CONCLUSION: These findings suggest that the cell-killing efficacy of (212)Pb-TCMC-trastuzumab after gemcitabine pretreatment may be associated with abrogation of the G2/M checkpoint, inhibition of DNA damage repair, and chromatin remodeling.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Morte Celular/fisiologia , Reparo do DNA/efeitos da radiação , Desoxicitidina/análogos & derivados , Compostos Heterocíclicos/farmacologia , Isotiocianatos/farmacologia , Radioisótopos de Chumbo/farmacologia , Rad51 Recombinase/antagonistas & inibidores , Radioimunoterapia/métodos , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Cromatina/efeitos dos fármacos , Cromatina/efeitos da radiação , Terapia Combinada/métodos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/farmacologia , Reparo do DNA/efeitos dos fármacos , Desoxicitidina/farmacologia , Sinergismo Farmacológico , Feminino , Fase G2/fisiologia , Fase G2/efeitos da radiação , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Transferência Linear de Energia/fisiologia , Camundongos , Camundongos Nus , Mitose/efeitos da radiação , Neoplasias Peritoneais/radioterapia , Fosforilação/efeitos da radiação , Proteínas Quinases/imunologia , Proteínas Quinases/metabolismo , Radiossensibilizantes/farmacologia , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Gencitabina
17.
Cancer Med ; 2(5): 646-53, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24403230

RESUMO

Recent studies have demonstrated that therapy with (212) Pb-TCMC-trastuzumab resulted in (1) induction of apoptosis, (2) G2/M arrest, and (3) blockage of double-strand DNA damage repair in LS-174T i.p. (intraperitoneal) xenografts. To further understand the molecular basis of the cell killing efficacy of (212) Pb-TCMC-trastuzumab, gene expression profiling was performed with LS-174T xenografts 24 h after exposure to (212) Pb-TCMC-trastuzumab. DNA damage response genes (84) were screened using a quantitative real-time polymerase chain reaction array (qRT-PCR array). Differentially regulated genes were identified following exposure to (212) Pb-TCMC-trastuzumab. These included genes involved in apoptosis (ABL, GADD45α, GADD45γ, PCBP4, and p73), cell cycle (ATM, DDIT3, GADD45α, GTSE1, MKK6, PCBP4, and SESN1), and damaged DNA binding (DDB) and repair (ATM and BTG2). The stressful growth arrest conditions provoked by (212) Pb-TCMC-trastuzumab were found to induce genes involved in apoptosis and cell cycle arrest in the G2/M phase. The expression of genes involved in DDB and single-strand DNA breaks was also enhanced by (212) Pb-TCMC-trastuzumab while no modulation of genes involved in double-strand break repair was apparent. Furthermore, the p73/GADD45 signaling pathway mediated by p38 kinase signaling may be involved in the cellular response, as evidenced by the enhanced expression of genes and proteins of this pathway. These results further support the previously described cell killing mechanism by (212) Pb-TCMC-trastuzumab in the same LS-174T i.p. xenograft. Insight into these mechanisms could lead to improved strategies for rational application of radioimmunotherapy using α-particle emitters.


Assuntos
Neoplasias do Colo/radioterapia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Radioisótopos de Chumbo/uso terapêutico , Radioimunoterapia/métodos , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , Apoptose/genética , Apoptose/efeitos da radiação , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Dano ao DNA , Reparo do DNA/genética , Reparo do DNA/efeitos da radiação , DNA de Neoplasias/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/fisiologia , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Trastuzumab , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/fisiologia , Regulação para Cima/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas GADD45
18.
J Labelled Comp Radiopharm ; 55(11): 423-426, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23162207

RESUMO

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.

19.
Bioorg Med Chem Lett ; 22(17): 5517-22, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22853992

RESUMO

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.


Assuntos
Quelantes/química , Quelantes/metabolismo , Integrina alfaVbeta3/antagonistas & inibidores , Integrina alfaVbeta3/metabolismo , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Acetatos/síntese química , Acetatos/química , Acetatos/metabolismo , Quelantes/síntese química , Radioisótopos de Gálio/química , Radioisótopos de Gálio/metabolismo , Compostos Heterocíclicos com 1 Anel/síntese química , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/metabolismo , Humanos , Radioisótopos de Chumbo/química , Radioisótopos de Chumbo/metabolismo , Peptidomiméticos/síntese química , Tomografia por Emissão de Pósitrons , Radioimunoensaio , Tomografia Computadorizada de Emissão de Fóton Único
20.
Mol Cancer Ther ; 11(3): 639-48, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22238365

RESUMO

In preclinical studies, targeted radioimmunotherapy using (212)Pb-TCMC-trastuzumab as an in vivo generator of the high-energy α-particle emitting radionuclide (212)Bi is proving an efficacious modality for the treatment of disseminated peritoneal cancers. To elucidate mechanisms associated with this therapy, mice bearing human colon cancer LS-174T intraperitoneal xenografts were treated with (212)Pb-TCMC-trastuzumab and compared with the nonspecific control (212)Pb-TCMC-HuIgG, unlabeled trastuzumab, and HuIgG, as well as untreated controls. (212)Pb-TCMC-trastuzumab treatment induced significantly more apoptosis and DNA double-strand breaks (DSB) at 24 hours. Rad51 protein expression was downregulated, indicating delayed DNA double-strand damage repair compared with (212)Pb-TCMC-HuIgG, the nonspecific control. (212)Pb-TCMC-trastuzumab treatment also caused G(2)-M arrest, depression of the S phase fraction, and depressed DNA synthesis that persisted beyond 120 hours. In contrast, the effects produced by (212)Pb-TCMC-HuIgG seemed to rebound by 120 hours. In addition, (212)Pb-TCMC-trastuzumab treatment delayed open chromatin structure and expression of p21 until 72 hours, suggesting a correlation between induction of p21 protein and modification in chromatin structure of p21 in response to (212)Pb-TCMC-trastuzumab treatment. Taken together, increased DNA DSBs, impaired DNA damage repair, persistent G(2)-M arrest, and chromatin remodeling were associated with (212)Pb-TCMC-trastuzumab treatment and may explain its increased cell killing efficacy in the LS-174T intraperitoneal xenograft model for disseminated intraperitoneal disease.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias do Colo/radioterapia , Radioimunoterapia/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Anticorpos Monoclonais Humanizados/química , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Western Blotting , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Feminino , Citometria de Fluxo , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Compostos Heterocíclicos/química , Humanos , Imunoglobulina G/química , Imunoglobulina G/uso terapêutico , Isotiocianatos/química , Radioisótopos de Chumbo/química , Camundongos , Camundongos Nus , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/radioterapia , Fatores de Tempo , Trastuzumab
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...