Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Bioorg Med Chem ; 25(21): 5975-5980, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28964629

RESUMO

In this report we describe the development of an alternative approach to arylstannane chemistry for radiolabeling antibodies with radioiodine or astatine based on aryliodonium salts precursors. Bifunctional aryliodonium salts were designed and tested for the synthesis of 125I and 211At labeled prosthetic groups for bioconjugation. The nature of the electron rich aryl group was varied and its impact on the regioselectivity of radiohalogenation was evaluated. Unexpectedly, whereas the 2-thienyl group provided the best regioselectivity towards the radioiodination of the aryl moiety of interest (98:2), it was less selective for astatination (87:13); the anisyl group providing the best regioselectivity of astatination (94:6). Under optimized conditions, both radioiodination and astatination could be performed very efficiently in mild conditions (radiochemical yields>85%). The ionic nature of the precursors was exploited to develop an efficient purification approach: the HPLC step that is usually necessary in conventionnal approaches to optimize removal of organotin toxic precursors and side products was replaced by a filtration through a silica cartridge with a significantly reduced loss of radiolabeled product. The purified radioiodinated and astatinated prosthetic groups were then conjugated efficiently to an anti-CD138 monoclonal antibody (75-80% conjugation yield). By using this novel and simple radiohalogenation procedure, higher overall radiochemical yields of astatination were obtained in comparison with the use of an arylstannane precursor and procedures of the litterature for labeling the same antibody. Overall, due to their simplicity of use and high robustness, these new precursors should simplify the labeling of proteins of interest with iodine and astatine radioisotopes for imaging and therapeutic applications.


Assuntos
Anticorpos Monoclonais/química , Astato/química , Hidrocarbonetos Iodados/química , Hidrocarbonetos Iodados/síntese química , Radioisótopos do Iodo , Estrutura Molecular , Sais/síntese química , Sais/química , Temperatura
2.
Br J Cancer ; 108(10): 2013-20, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23632482

RESUMO

BACKGROUND: Paclitaxel has recently been reported by this laboratory to potentiate the high-LET radiation therapeutic (212)Pb-TCMC-trastuzumab, which targets HER2. To elucidate mechanisms associated with this therapy, targeted α-particle radiation therapeutic (212)Pb-TCMC-trastuzumab together with paclitaxel was investigated for the treatment of disseminated peritoneal cancers. METHODS: Mice bearing human colon cancer LS-174T intraperitoneal xenografts were pre-treated with paclitaxel, followed by treatment with (212)Pb-TCMC-trastuzumab and compared with groups treated with paclitaxel alone, (212)Pb-TCMC-HuIgG, (212)Pb-TCMC-trastuzumab and (212)Pb-TCMC-HuIgG after paclitaxel pre-treatment. RESULTS: (212)Pb-TCMC-trastuzumab with paclitaxel given 24 h earlier induced increased mitotic catastrophe and apoptosis. The combined modality of paclitaxel and (212)Pb-TCMC-trastuzumab markedly reduced DNA content in the S-phase of the cell cycle with a concomitant increase observed in the G2/M-phase. This treatment regimen also diminished phosphorylation of histone H3, accompanied by an increase in multi-micronuclei, or mitotic catastrophe in nuclear profiles and positively stained γH2AX foci. The data suggests, possible effects on the mitotic spindle checkpoint by the paclitaxel and (212)Pb-TCMC-trastuzumab treatment. Consistent with this hypothesis, (212)Pb-TCMC-trastuzumab treatment in response to paclitaxel reduced expression and phosphorylation of BubR1, which is likely attributable to disruption of a functional Aurora B, leading to impairment of the mitotic spindle checkpoint. In addition, the reduction of BubR1 expression may be mediated by the association of a repressive transcription factor, E2F4, on the promoter region of BubR1 gene. CONCLUSION: These findings suggest that the sensitisation to therapy of (212)Pb-TCMC-trastuzumab by paclitaxel may be associated with perturbation of the mitotic spindle checkpoint, leading to increased mitotic catastrophe and cell death.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/radioterapia , Radioisótopos de Chumbo/uso terapêutico , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Paclitaxel/uso terapêutico , Radioimunoterapia , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Terapia Combinada , Feminino , Humanos , Camundongos , Camundongos Nus , Paclitaxel/farmacologia , Radioimunoterapia/métodos , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Bioconjug Chem ; 22(6): 1128-35, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21604692

RESUMO

A new bifunctional ligand 3p-C-DEPA was synthesized and evaluated for use in targeted α-radioimmunotherapy. 3p-C-DEPA was efficiently prepared via regiospecific ring opening of an aziridinium ion and conjugated with trastuzumab. The 3p-C-DEPA-trastuzumab conjugate was extremely rapid in binding (205/6)Bi, and the corresponding (205/6)Bi-3p-C-DEPA-trastuzumab complex was stable in human serum. Biodistribution studies were performed to evaluate in vivo stability and tumor targeting of (205/6)Bi-3p-C-DEPA-trastuzumab conjugate in tumor bearing athymic mice. (205/6)Bi-3p-C-DEPA-trastuzumab conjugate displayed excellent in vivo stability and targeting as evidenced by low organ uptake and high tumor uptake. The results of the in vitro and in vivo studies indicate that 3p-C-DEPA is a promising chelator for radioimmunotherapy of (212)Bi and (213)Bi.


Assuntos
Bismuto/química , Glicina/análogos & derivados , Compostos Heterocíclicos com 1 Anel/química , Compostos Organometálicos/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Bismuto/sangue , Linhagem Celular Tumoral , Feminino , Glicina/sangue , Glicina/química , Compostos Heterocíclicos com 1 Anel/sangue , Humanos , Ligantes , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/sangue , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Compostos Organometálicos/sangue , Compostos Organometálicos/química , Radioimunoterapia , Radioisótopos/química , Compostos Radiofarmacêuticos/sangue , Compostos Radiofarmacêuticos/química , Estereoisomerismo , Distribuição Tecidual , Trastuzumab
4.
Nanotechnology ; 21(17): 175704, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20368682

RESUMO

Multi-modality imaging probes combine the advantages of individual imaging techniques to yield highly detailed anatomic and molecular information in living organisms. Herein, we report the synthesis and characterization of a dual-modality nanoprobe that couples the magnetic properties of ultrasmall superparamagnetic iron oxide nanoparticles (USPIOs) with the near infrared fluorescence of Cy5.5. The fluorophore is encapsulated in a biocompatible shell of silica surrounding the iron oxide core for a final diameter of approximately 17 nm. This silica-coated iron oxide nanoparticle (SCION) has been analyzed by transmission electron microscopy, dynamic light scattering, and superconducting quantum interference device (SQUID). The particle demonstrates a strong negative surface charge and maintains colloidal stability in the physiological pH range. Magnetic hysteresis analysis confirms superparamagnetic properties that could be manipulated for thermotherapy. The viability of primary human monocytes, T cells, and B cells incubated with the particle has been examined in vitro. In vivo analysis of agent leakage into subcutaneous A431 tumors in mice was also conducted. This particle has been designed for diagnostic application with magnetic resonance and fluorescence imaging, and has future potential to serve as a heat-sensitive targeted drug delivery platform.


Assuntos
Diagnóstico por Imagem/métodos , Compostos Férricos/química , Magnetismo , Nanopartículas Metálicas/química , Microscopia de Fluorescência/métodos , Técnicas de Sonda Molecular , Animais , Carbocianinas/metabolismo , Sobrevivência Celular , Células Cultivadas , Sistemas de Liberação de Medicamentos , Humanos , Leucócitos Mononucleares/metabolismo , Camundongos , Propilaminas , Silanos
5.
Bioorg Med Chem ; 18(16): 5925-31, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20663676

RESUMO

In this report we directly compare the in vivo and in vitro MRI properties of gadolinium-dendrimer conjugates of derivatized acyclic diethylenetriamine-N,N',N',N'',N''-pentaacetic acid (1B4M-DTPA) and macrocyclic 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (C-DOTA). The metal-ligand chelates were pre-formed in alcohol prior to conjugation to the generation 4 PAMAM dendrimer (G4D), and the dendrimer-based agents were purified by Sephadex(R) G-25 column. The analysis and SE-HPLC data indicated chelate to dendrimer ratios of 30:1 and 28:1, respectively. Molar relaxivity measured at pH 7.4, 22 degrees C, and 3T are comparable (29.5 vs 26.9 mM(-1)s(-1)), and both conjugates are equally viable as MRI contrast agents based on the images obtained. The macrocyclic agent however exhibits a faster rate of clearance in vivo (t(1/2)=16 vs 29 min). Our conclusion is that the macrocyclic-based agent is the more suitable agent for in vivo use for these reasons combined with kinetic inertness associated with the Gd(III) DOTA complex stability properties.


Assuntos
Meios de Contraste/química , Dendrímeros/química , Compostos Heterocíclicos/química , Imageamento por Ressonância Magnética , Compostos Organometálicos/química , Ácido Pentético/química , Animais , Meios de Contraste/farmacocinética , Dendrímeros/farmacocinética , Feminino , Compostos Heterocíclicos/farmacocinética , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Nus , Compostos Organometálicos/farmacocinética , Ácido Pentético/farmacocinética
6.
J Transl Sci ; 5(2)2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30906574

RESUMO

INTRODUCTION: Tumors over-expressing the human epithelial receptor 2 (HER2) or exhibiting amplification or mutation of its proto-oncogene have a poorer prognosis. Using trastuzumab and/or other HER2 targeted therapies can increase overall survival in patients with HER2(+) tumors making it critical to accurately identify patients who may benefit. We report on a Phase 0 study of the imaging agent, 111In-CHX-A"-DTPA trastuzumab, in patients with known HER2 status to evaluate its safety and biodistribution and to obtain preliminary data regarding its ability to provide an accurate, whole-body, non-invasive means to determine HER2 status. METHODS: 111In-CHX-A"-DTPA trastuzumab was radiolabeled on-site and slowly infused into 11 patients who underwent single (n=5) or multiple (n=6) ɣ-camera (n=6) and/or SPECT (n=8) imaging sessions. RESULTS: No safety issues were identified. Visual and semi-quantitative imaging data were concordant with tissue HER2 expression profiling in all but 1 patient. The biodistribution showed intense peak liver activity at the initial imaging timepoint (3.3h) and a single-phase clearance fit of the average time-activity curve (TAC) estimated t1/2=46.9h (R2=0.97; 95%CI 41.8 to 53h). This was followed by high gastrointestinal (GI) tract activity peaking by 52h. Linear regression predicted GI clearance by 201.2h (R2 =0.96; 95%CI 188.5 to 216.9h). Blood pool had lower activity with its maximum on the initial images. Non-linear regression fit projected a t1/2=34.2h (R2 =0.96; 95%CI 25.3 to 46.3h). Assuming linear whole-body clearance, linear regression projected complete elimination (x-intercept) at 256.5hr (R2=0.96; 95%CI 186.1 to 489.2h). CONCLUSION: 111In-CHX-A"-DTPA trastuzumab can be safely imaged in humans. The biodistribution allowed for visual and semiquantitative analysis with results concordant with tissue expression profiling in 10 of 11 patients. Advances in Knowledge and Implications for Patient Care Using readily available components and on-site radiolabeling 111In-CHX-A"-DTPA trastuzumab SPECT imaging may provide an economical, non-invasive means to detect HER2 over-expression.

7.
Dalton Trans ; 46(14): 4749-4758, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28338136

RESUMO

Three hydroxypyridinone (HOPO) positional isomers - 1,2-HOPO (L1H) and its water soluble analogue (L1'H), 3,2-HOPO (L2H) and 3,4-HOPO (L3H) have been investigated for the complexation of Zr(iv). Potentiometric and UV-Vis spectrometric studies show a higher thermodynamic stability for the formation of Zr(L1')4 in comparison with Zr(L2)4 and Zr(L3)4 as well as a higher kinetic inertness in competition studies with EDTA or Fe3+ at a radiotracer concentration with 89Zr. Besides the low pKa of L1H or L1'H (pKa = 5.01) in comparison with L2H and L3H (pKa = 8.83 and 9.55, respectively), the higher stability of Zr(L1')4 can be attributed in part to the presence of the amide group next to the chelating oxygen that induces intramolecular H-bond and amide/π interactions that were observed by X-ray crystallography and confirmed by quantum chemical calculations. The data presented here indicate that the 1,2-HOPO L1' exhibits the best characteristics for Zr(iv) complexation. However, 3,2-HOPO and 3,4-HOPO patterns, if appropriately tuned, for instance with the addition of an amide group as in the 1,2-HOPO ligand, may also become interesting alternatives for the design of Zr(iv) chelators with improved characteristics for applications in nuclear imaging with 89Zr.

8.
J Natl Cancer Inst ; 80(6): 449-52, 1988 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-3367385

RESUMO

For comparison of cytotoxicity from alpha-particle irradiation with that from conventional x-irradiation, 212Bi, an alpha-emitting radionuclide, was attached to a monoclonal antibody that recognizes a cell surface antigen on human pancreatic carcinoma cells. For a given level of survival, the 212Bi-antibody complex was found to be approximately 20 times more efficient in cell killing than x-irradiation and 5 times more cytotoxic when compared with the cytotoxicity of an antigen-negative cell line or an isotype-matched control antibody. High linear energy transfer radioimmunotherapy using alpha emitters linked to monoclonal antibodies may be useful in vivo and in vitro for selectively killing target cell populations, especially those resistant to other forms of treatment.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Bismuto/administração & dosagem , Neoplasias Pancreáticas/radioterapia , Radioisótopos/administração & dosagem , Antígenos de Neoplasias/imunologia , Antígenos de Superfície/imunologia , Sobrevivência Celular/efeitos da radiação , Transferência de Energia , Humanos , Neoplasias Pancreáticas/imunologia , Radioimunoensaio , Células Tumorais Cultivadas
9.
J Natl Cancer Inst ; 92(19): 1573-81, 2000 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-11018093

RESUMO

BACKGROUND: HER2 is a membrane receptor whose overexpression is strongly associated with poor prognosis in breast carcinomas. Inhibition of HER2 activity can reduce tumor growth, which led to the development of Herceptin, an anti-HER2 monoclonal antibody (MAb) that is already in clinical use. However, the objective response rate to Herceptin monotherapy is quite low. HER2 activity can also be inhibited by the highly cytotoxic antibiotic geldanamycin (GA). However, GA is not used clinically because of its adverse toxicity. Our purpose was to enhance the inhibitory activity of anti-HER2 MAb by coupling it to GA. METHODS: We synthesized 17-(3-aminopropylamino)GA (17-APA-GA) and conjugated it to the anti-HER2 MAb e21, to form e21 : GA. The noninternalizing anti-HER2 MAb AE1 was used as a control. Internalization assays and western blot analyses were used to determine whether the anti-HER2 MAbs and their immunoconjugates were internalized into HER2-expressing cells and reduced HER2 levels. All statistical tests were two-sided. RESULTS: The immunoconjugate e21 : GA inhibited the proliferation of HER2-overexpressing cell lines better than unconjugated e21 (concentration required for 50% inhibition = 40 versus 1650 microg/mL, respectively). At 15 microg/mL, e21 : GA reduced HER2 levels by 86% within 16 hours, whereas unconjugated e21, 17-APA-GA, or AE1 : GA reduced HER2 levels by only 20%. These effects were not caused by release of 17-APA-GA from the immunoconjugate because immunoconjugates containing [(3)H]GA were stable in serum at 37 degrees C. Furthermore, e21 : GA did not significantly inhibit proliferation of the adult T-cell leukemia cell line HuT102, which is HER2 negative yet highly sensitive to GA. CONCLUSIONS: Our findings suggest that conjugating GA to internalizing MAbs enhances the inhibitory effect of the MAbs. This approach might also be applied in cellular targeting via growth factors and may be of clinical interest.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Imunoconjugados , Quinonas/farmacologia , Receptor ErbB-2/metabolismo , Animais , Antibióticos Antineoplásicos/imunologia , Anticorpos Monoclonais/uso terapêutico , Benzoquinonas , Western Blotting , Neoplasias da Mama/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lactamas Macrocíclicas , Camundongos , Camundongos Endogâmicos BALB C , Quinonas/imunologia , Receptor ErbB-2/imunologia , Células Tumorais Cultivadas , Regulação para Cima
10.
Cancer Res ; 50(14): 4221-6, 1990 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-2364380

RESUMO

We have used a series of bifunctional chelating agents to prepare 206Bi-labeled monoclonal antibody and have assessed the in vivo stability and tumor targeting of these conjugates in the Rauscher murine erythroleukemia model. Several derivatives of diethylenetriaminepentaacetic acid [the dicyclic dianhydride of diethylenetriaminepentaacetic acid (ca-DTPA), 2-(p-isothiocyanatobenzyl)diethylenetriaminepentaacetic acid (SCNBzDTPA), and 2-(p-isothiocyanatobenzyl)-5(6)-methyl-diethylenetriaminepentaacet ic acid (MxDTPA)], as well as a macrocyclic polyazacycloalkane-N-acetic acid [2-(p-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane-N ,N',N",N"'- tetraacetic acid (DOTA)], were conjugated to monoclonal antibody 103A, which is specific for gp70 expressed on Rauscher virus-infected cells. The stability in vivo of 206Bi chelate-103A conjugates was first evaluated in normal mice by determining the levels of 206Bi in blood and kidney, since these were the organs in which free 206Bi, 206Bi-caDTPA-103A, and 35S-103A accumulated. The biodistribution of 206Bi administered as a chelate of caDTPA-103A was virtually indistinguishable from that of free 206Bi, indicating a low degree of in vivo stability of this bismuth chelate when compared to biosynthetically labeled 35S-103A. There was a progressive increase in the 206Bi levels observed in blood when the series of 103A conjugates prepared using SCNBzDTPA, MxDTPA, and DOTA was compared to 206Bi administered free or as a caDTPA-103A chelate. At 1 h after injection into normal mice, the blood level of 206Bi-DOTA-103A was 25-fold greater than that observed for 206Bi-caDTPA-103A and the level in kidney was 6-fold less, values that did not differ significantly from those observed for 35S-103A. Targeting to leukemic spleen was increased by 10-fold when the DOTA conjugate was used; the tumor level was 90% injected dose/g for DOTA, as compared to only 9% injected dose/g for caDTPA-103A at 1 h after injection. Use of the DOTA chelator also reduced by 7-fold the level of uptake by the kidney in the leukemic animals. We, therefore, conclude that the chelator DOTA is a promising reagent for the delivery of 212Bi-antibody conjugates to vascularized tumors under conditions that require targeting via the circulatory system.


Assuntos
Anticorpos Monoclonais , Bismuto , Radioisótopos , Animais , Bismuto/farmacocinética , Quelantes , Feminino , Hibridomas/imunologia , Rim/metabolismo , Leucemia Experimental/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Baço/metabolismo , Radioisótopos de Enxofre , Distribuição Tecidual
11.
Cancer Res ; 52(20): 5818-20, 1992 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-1394209

RESUMO

The specificity, toxicity, and efficacy of alpha-particle-mediated radioimmunotherapy of murine erythroleukemia was assessed by use of tumor-specific monoclonal antibody 103A labeled with 212Bi. Forty % of the injected dose/g tissue targeted to neoplastic spleens within 1 h after i.v. injection. When 212Bi-103A was injected on day 13 of disease, a dose-dependent response was achieved, as measured by a reduction in splenomegaly and absence of liver metastasis. Mice treated with 212Bi-103A on day 8 of disease showed no histological evidence of erythroleukemia on day 22 and survived significantly longer (median, 118 days) than mice treated with 212Bi-control IgG (78 days) or untreated mice (63 days), indicating successful specific radioimmunotherapy.


Assuntos
Partículas alfa/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Bismuto/uso terapêutico , Radioimunoterapia , Radioisótopos/uso terapêutico , Animais , Bismuto/farmacocinética , Bovinos , Imunoglobulina G/uso terapêutico , Camundongos , Radioisótopos/farmacocinética , Vírus Rauscher
12.
Cancer Res ; 60(17): 4804-11, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10987290

RESUMO

The biodistribution characteristics of a humanized anti-Lewis(y) antibody (hu3S193) radiolabeled to three radioisotopes, 125I, 111In, and 90Y, were examined in a BALB/c nude mouse xenograft model of breast cancer. The immunoreactivity of both 125I- and 111In-bound hu3S193 exceeded 50% and was 20% for 90Y. In vivo, labeled antibody was shown by gamma camera imaging and immunohistochemical and autoradiographic techniques to localize to Lewis(y)-expressing breast xenografts with minimal normal tissue uptake. Maximal radioisotope uptake peaked at 48 h for all three isotopes; however, the percentage of injected dose/gram and tumor retention were greater for 111In- and 90Y-bound antibody than for 125I-bound antibody. Although immunoreactivity of 111In- and 125I-labeled hu3S193 in serum was stable over a 5-day period, the amount of unlabeled 111In in serum was lower than 125I, which together with higher tumor uptake indicates better retention of 111In-labeled hu3S193 and catabolites within the tumor cells. Superior tumor uptake and retention of 111In-labeled hu3S193 and similar blood clearance compared with 125I-labeled hu3S193, suggest that radiometals are the preferred radioisotope for this antibody-antigen system. Humanized 3S193 is a promising new construct for the targeting and potential therapy of Lewis(y)-expressing tumors.


Assuntos
Adenocarcinoma/metabolismo , Anticorpos Monoclonais/farmacocinética , Neoplasias da Mama/metabolismo , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/imunologia , Animais , Anticorpos Monoclonais/imunologia , Autorradiografia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Estabilidade de Medicamentos , Feminino , Meia-Vida , Humanos , Imuno-Histoquímica , Radioisótopos de Índio/farmacocinética , Radioisótopos do Iodo/farmacocinética , Marcação por Isótopo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Cintilografia , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas , Radioisótopos de Ítrio/farmacocinética
13.
Cancer Res ; 53(23): 5683-9, 1993 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8242624

RESUMO

A major factor that is critical to the potential effectiveness of alpha-emitter 212Bi radioimmunotherapy is the design of radiometal-chelated antibodies that will be stable in vivo. The chelate should bind the radiometal firmly to minimize release of the radionuclide from the monoclonal antibody-chelate complex. The present study examines a member of a new class of polyamine carboxylate chelating compounds, the DOTA ligands, for conjugating radiometal ions to antibody. Biocompatibility and stability are assessed with the anti-Tac monoclonal antibody that is directed against the human interleukin 2 receptors. The scientific basis for the clinical use of this antibody in radioimmunotherapy is that resting normal cells do not express the interleukin 2 receptor, whereas the receptor is expressed on the surface of certain neoplasms and by activated T-cells in select autoimmune diseases and in allograft rejection. First, we examined the impact of the labeling procedure and the presence of the chelate, DOTA, on antibody bioavailability and survival. Next, we studied the capacity of the antibody-chelate complex to retain radiobismuth. Coupling DOTA to antibody or adding Bi(III) to DOTA-coupled antibody did not disturb antibody immunoreactivity in in vitro binding studies. In addition, as analyzed by in vivo studies, DOTA-antibody dummy labeled with nonradioactive bismuth showed pharmacokinetics and tissue distribution identical to those of antibody not modified with DOTA. DOTA-anti-Tac charged with radioactive bismuth showed pharmacokinetics identical to radioiodinated dummy-labeled DOTA-antibody, suggesting little premature release of radioactive bismuth from the antibody complex. Moreover, in the early, therapeutically relevant time points (2 h and 6 h), there was no significant preferential accumulation of bismuth in any organ. At the 5-day time point, beyond the range of therapeutic interest, there was delayed excretion of bismuth from reticuloendothelial tissues relative to radioiodine from catabolized antibody. Excretion of catabolized DOTA-bismuth had an apparent t1/2 of approximately 1 day without the marked renal accumulation typical of the free bismuth ion. The compatibility of DOTA conjugation with antibody bioactivity and the stability of the radioactive bismuth complex in vivo provide important preclinical validation of the potential utility of this new chelating agent for 212Bi monoclonal antibody radioimmunotherapy in humans.


Assuntos
Anticorpos Monoclonais/metabolismo , Bismuto/metabolismo , Compostos Heterocíclicos com 1 Anel , Compostos Heterocíclicos/metabolismo , Radioimunoterapia/métodos , Radioisótopos/metabolismo , Receptores de Interleucina-2/imunologia , Partículas alfa , Animais , Anticorpos Monoclonais/uso terapêutico , Bismuto/farmacocinética , Meia-Vida , Compostos Heterocíclicos/farmacocinética , Humanos , Camundongos , Camundongos Nus , Radioisótopos/farmacocinética , Distribuição Tecidual
14.
Cancer Res ; 54(16): 4362-70, 1994 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8044783

RESUMO

The efficacy, specificity, and toxicity of bismuth (212Bi) alpha particle-mediated radioimmunotherapy was evaluated in nude mice bearing a murine lymphoma transfected with the human CD25 [human Tac; interleukin 2 receptor alpha (IL-2R alpha)] gene. The therapeutic agent used was the tumor-specific humanized monoclonal antibody anti-Tac conjugated to 212Bi. The human IL-2R alpha-expressing cell line was produced by transfecting the gene encoding human Tac into the murine plasmacytoma cell line SP2/0. The resulting cell line, SP2/Tac, expressed approximately 18,000 human IL-2R alpha molecules/cell. Following s.c. or i.p. injection of 2 x 10(6) SP2/Tac cells into nude mice, rapidly growing tumors developed in all animals after a mean of 10 and 13 days, respectively. The bifunctional chelate cyclohexyldiethylenetriaminepentaacetic acid was used to couple 212Bi to the humanized anti-Tac monoclonal antibody. This immunoconjugate was shown to be stable in vivo. Specifically, in pharmacokinetic studies in nude mice, the blood clearance patterns of i.v. administered 205/206Bi-anti-Tac and coinjected 125I-anti-Tac were comparable. The toxicity and therapeutic efficacy of 212Bi-anti-Tac were evaluated in nude mouse ascites or solid tumor models wherein SP2/Tac cells were administered either i.p. or s.c., respectively. The i.p. administration of 212Bi-anti-Tac, 3 days following i.p. tumor inoculation, led to a dose-dependent, significant prolongation of tumor-free survival. Doses of 150 or 200 microCi prevented tumor occurrence in 75% (95% confidence interval, 41-93%) of the animals. In the second model, i.v. treatment with 212Bi-anti-Tac 3 days following s.c. tumor inoculation also resulted in a prolongation of the period before tumor development. However, prevention of tumor occurrence decreased to 30% (95% confidence interval, 11-60%). In both the i.p. and s.c. tumor trials, 212Bi-anti-Tac was significantly more effective for i.p. (P2 = 0.0128 50/100 microCi 212Bi-anti-Tac versus 50/100 microCi Mik beta; P2 = 0.0142 150/200 microCi anti-Tac versus 150/200 microCi Mik beta) and for s.c. tumors (P2 = 0.0018 100 microCi anti-Tac versus 100 microCi Mik beta; P2 = 0.0042 200 microCi anti-Tac versus 200 microCi Mik beta 1) than the control antibody Mik beta 1 coupled to 212Bi at comparable dose levels. In contrast to the efficacy observed in the adjuvant setting, therapy of large, established s.c. SP-2/Tac-expressing tumors with i.v. administered 212Bi-anti-Tac (at doses up to 200 microCi/animal) failed to induce tumor regression.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Anticorpos Monoclonais/uso terapêutico , Bismuto/uso terapêutico , Leucemia-Linfoma de Células T do Adulto/radioterapia , Radioimunoterapia/métodos , Radioisótopos/uso terapêutico , Receptores de Interleucina-2/imunologia , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/metabolismo , Bismuto/efeitos adversos , Bismuto/metabolismo , Relação Dose-Resposta Imunológica , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Leucemia-Linfoma de Células T do Adulto/imunologia , Leucemia-Linfoma de Células T do Adulto/metabolismo , Camundongos , Camundongos Nus , Radioimunoterapia/efeitos adversos , Radioisótopos/efeitos adversos , Radioisótopos/metabolismo , Dosagem Radioterapêutica , Receptores de Interleucina-2/metabolismo , Distribuição Tecidual
15.
Cancer Res ; 61(13): 4966-70, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431325

RESUMO

DAB-Am64-(1B4M-Gd)(64) is a newly synthesized macromolecular liver magnetic resonance imaging (MRI) contrast agent with a polypropylenimine diaminobutane (DAB) dendrimer conjugated with a bifunctional diethylenetriaminepentaacetic acid (DTPA) derivative for complexing Gd(III) atoms. The characteristics of DAB-Am64-(1B4M-Gd)(64), which quickly accumulated in the liver, have been reported recently. In the present study, the dynamic micro-MRI with DAB-Am64-(1B4M-Gd)(64) was obtained in the mouse liver metastasis model using colon carcinoma cells to evaluate the ability to visualize the micrometastatic tumors compared with that using Gd-DTPA. The dynamic micro-MRI with DAB-Am64-(1B4M-Gd)(64) was able to homogeneously enhance the normal liver parenchyma and visualize micrometastatic tumors of 0.3-mm diameter in the liver of the mice with better contrast than that with Gd-DTPA. In conclusion, DAB-Am64-(1B4M-Gd)(64) is a new liver MRI contrast agent potentially useful for diagnosis of micrometastasis in the liver.


Assuntos
Meios de Contraste , Gadolínio DTPA/análogos & derivados , Gadolínio DTPA/uso terapêutico , Neoplasias Hepáticas Experimentais/secundário , Animais , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Gadolínio DTPA/toxicidade , Humanos , Neoplasias Hepáticas Experimentais/diagnóstico , Neoplasias Hepáticas Experimentais/patologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo
16.
Cancer Res ; 53(12): 2834-9, 1993 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8504427

RESUMO

Biodistribution and imaging characteristics of monoclonal antibody B3 were evaluated in nude mice bearing A431 human epidermoid carcinoma xenografts. B3 is a murine IgG1k, recently isolated, reacting with a carbohydrate epitope abundantly and uniformly expressed by most carcinomas. B3 was conjugated to a new backbone-substituted derivative of diethylenetriaminepentaacetic acid, 2-(p-isothiocyanato benzyl)-cyclohexyl-diethylenetriaminepentaacetic acid, and labeled with 111In. Tumor-bearing mice were given i.v. injections of approximately 5 microCi of either 111In-B3 or 111In-MOPC-21, an isotype-matched control, and sacrificed in groups of five at 6 h and daily up to 168 h. Imaging was performed at 24, 72, and 144 h. Significant differences were observed in tumor uptake at all time points with peak values at 48 h (25 +/- 5.2% versus 6.3 +/- 0.4% of the injected dose/g tissue) (mean +/- SD) for 111In-B3 and 111In-MOPC-21, respectively (P < 0.001). All tumor to organ ratios increased with time for 111In-B3. In particular, tumor:liver ratios rose from 3.2 +/- 0.6 at 24 h to 6.3 +/- 1.2 at 168 h. Imaging results showed selective and progressive accumulation of 111In-B3 at the tumor site, whereas 111In-MOPC-21 did not show specific localization. In summary, 111In-labeled B3 demonstrated good and specific tumor targeting, which warrants its future clinical evaluation.


Assuntos
Anticorpos Monoclonais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Imunoglobulina G/metabolismo , Animais , Carcinoma de Células Escamosas/diagnóstico por imagem , Feminino , Humanos , Radioisótopos de Índio/farmacocinética , Camundongos , Camundongos Nus , Radioimunodetecção , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas
17.
Cancer Res ; 49(10): 2639-44, 1989 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-2785435

RESUMO

One factor that is critical to the potential effectiveness of radioimmunotherapy is the design of radiometal-chelated antibodies that will be stable in vivo. Stability in vivo depends on the condition that both the chelate linkage and radiolabeling procedures not alter antibody specificity and biodistribution. In addition, synthesis and selection of the chelating agent is critical for each radiometal in order to prevent inappropriate release of the radiometal in vivo. In the present study, we compare the in vivo stability of seven radioimmunoconjugates that use different polyaminocarboxylate chelating agents to complex yttrium-88 to the mouse anti-human interleukin-2 receptor monoclonal antibody, anti-Tac. Chelate linkage and radiolabeling procedures did not alter the immunospecificity of anti-Tac. In order to assess whether yttrium was inappropriately released from the chelate-coupled antibody in vivo, iodine-131-labeled and yttrium-88 chelate-coupled antibodies were simultaneously administered to the same animals to correlate the decline in yttrium and radioiodinated antibody activity. The four stable yttrium-88 chelate-coupled antibodies studied displayed similar iodine-131 and yttrium-88 activity, indicating minimal elution of yttrium-88 from the complex. In contrast, the unstable yttrium-88 chelate-coupled antibodies had serum yttrium-88 activities that declined much more rapidly than their iodine-131 activities, suggesting loss of the radiolabel yttrium-88 from the chelate. Furthermore, high rates of yttrium-88 elution correlated with deposition in bone. Four chelating agents emerged as promising immunotherapeutic reagents: isothiocyanate benzyl DTPA and its derivatives 1B3M, MX, and 1M3B. All four isothiocyanate agents showed prolonged yttrium-88 vascular survival which was essentially identical to that of their iodine-131 activity with only minimum accumulation (1.4-1.8%/g) of the yttrium-88 injected dose into bone. Thus, these four chelating agents were very stable in vivo and suitable for yttrium-monoclonal antibody radioimmunotherapy.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Quelantes/administração & dosagem , Reagentes de Ligações Cruzadas/administração & dosagem , Neoplasias Experimentais/terapia , Radioisótopos de Ítrio/administração & dosagem , Animais , Feminino , Imunotoxinas/uso terapêutico , Camundongos , Receptores de Interleucina-2/imunologia , Distribuição Tecidual
18.
Cancer Res ; 60(21): 6095-100, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085533

RESUMO

A novel alpha-particle emitting monoclonal antibody construct targeting the external domain of prostate-specific membrane antigen (PSMA) was prepared and evaluated in vitro and in vivo. The chelating agent, N-[2-amino-3-(p-isothiocyanatophen-yl)propyl]-trans-cyclohexane-1, 2-diamine-N,N',N',N'',N''-pentaacetic acid, was appended to J591 monoclonal antibody to stably bind the 213Bi radiometal ion. Bismuth-213 is a short-lived (t 1/2 = 46 min) radionuclide that emits high energy alpha-particles with an effective range of 0.07-0.10 mm that are ideally suited to treating single-celled neoplasms and micrometastatic carcinomas. The LNCaP prostate cancer cell line had an estimated 180,000 molecules of PSMA per cell; J591 bound to PSMA with a 3-nM affinity. After binding, the radiolabeled construct-antigen complex was rapidly internalized into the cell, carrying the radiometal inside. [213Bi]J591 was specifically cytotoxic to LNCaP. The LD50 value of [213Bi]J591 was 220 nCi/ml at a specific activity of 6.4 Ci/g. The potency and specificity of [213Bi]J591 directed against LNCaP spheroids, an in vitro model for micrometastatic cancer, also was investigated. [213Bi]J591 effectively stopped growth of LNCaP spheroids relative to an equivalent dose of the irrelevant control [213Bi]HuM195 or unlabeled J591. Cytotoxicity experiments in vivo were carried out in an athymic nude mouse model with an i.m. xenograft of LNCaP cells. [213Bi]J591 was able to significantly improve (P < 0.0031) median tumor-free survival (54 days) in these experiments relative to treatment with irrelevant control [213Bi]HuM195 (33 days), or no treatment (31 days). Prostate-specific antigen (PSA) was also specifically reduced in treated animals. At day 51, mean PSA values were 104 ng/ml +/- 54 ng/ml (n = 4, untreated animals), 66 ng/ml +/- 16 ng/ml (n = 6, animals treated with [213Bi]HuM195), and 28 ng/ml +/- 22 ng/ml (n = 6, animals treated with [213Bi]J591). The reduction of PSA levels in mice treated with [213Bi]J591 relative to mice treated with [213Bi]HuM195 and untreated control animals was significant with P < 0.007 and P < 0.0136, respectively. In conclusion, a novel [213Bi]-radiolabeled J591 has been constructed that selectively delivers alpha-particles to prostate cancer cells for potent and specific killing in vitro and in vivo.


Assuntos
Anticorpos Monoclonais/farmacologia , Bismuto/farmacologia , Imunotoxinas/farmacologia , Neoplasias da Próstata/radioterapia , Radioimunoterapia , Radioisótopos/farmacologia , Partículas alfa/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Sítios de Ligação , Morte Celular/efeitos da radiação , Humanos , Imunotoxinas/imunologia , Imunotoxinas/metabolismo , Cinética , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo , Esferoides Celulares/efeitos da radiação , Especificidade por Substrato , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Cancer Res ; 61(11): 4474-82, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389078

RESUMO

The chimeric monoclonal antibody KM871, directed against the G(D3) antigen, is under evaluation for its potential to target melanoma. To facilitate the in vivo evaluation of biodistribution properties and measurement of pharmacokinetics, KM871 was radiolabeled with (125)I via tyrosine residues and with (111)In via the bifunctional metal ion chelator C-functionalized trans-cyclohexyl diethylenetriaminepentaacetic acid (CHX-A"-DTPA) to lysine residues. Using antigen-positive SK-MEL-28 melanoma cells, immunoreactivities of 42 and 40% cell binding were obtained, respectively, for the two radioconjugates. Binding was enhanced in the presence of added unlabeled antibody. A humanized A33 antibody was similarly labeled with the two isotopes and used as a control. To determine and compare in vivo biodistribution characteristics of KM871 radiolabeled with (111)In or (125)I, mixtures of the radioconjugates were injected i.v. into BALB/c nude mice bearing G(D3)-positive-SK-MEL-28 melanoma xenografts. Gamma camera images were acquired; groups of five mice were sacrificed at various time intervals, and tumors, blood, and tissues were analyzed. (111)In-labeled CHX-A"-DTPA-KM871 showed a maximum tumor uptake of 41.9 +/- 7.0% injected dose/g at 72 h with prolonged retention over a 15-day period. The tumor:blood ratio was 3:1 by 72 h, and higher ratios were observed at later time points. No abnormal accumulation of (111)In-labeled conjugate was found in normal tissues. In contrast, there was little accumulation of (125)I-labeled KM871 in the same tumors. The specificity of antibody localization was confirmed by the low tumor uptake values for radiolabeled control antibody. Gamma camera imaging demonstrated excellent uptake of (111)In-labeled CHX-A"-DTPA-KM871 in the xenografts. Chromatographic analyses of xenograft cytosolic extracts demonstrated tumor internalization and catabolism of radiolabeled KM871 with the formation of small molecular weight metabolites. Laser scanning confocal microscopy demonstrated that the majority of intracellular KM871 is localized to lysosomes. Despite the catabolism of the radioconjugate, a dose-dependent increase in KM871 tumor localization was shown through immunohistochemical examination of xenograft biopsies. This study demonstrates for the first time the in vivo localization of a radiolabeled anti-G(D3) monoclonal antibody to G(D3)-expressing xenografts using gamma camera scanning techniques and tumor cell internalization of KM871 tagged with a green fluorescent dye, Alexa Fluor 488, through confocal microscopy. KM871 has potential for targeting tumors in patients with melanoma.


Assuntos
Anticorpos Monoclonais/farmacocinética , Gangliosídeos/imunologia , Imunoconjugados/farmacocinética , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Ácido Pentético/análogos & derivados , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos Monoclonais/imunologia , Quelantes/química , Quelantes/farmacocinética , Feminino , Câmaras gama , Gangliosídeos/biossíntese , Humanos , Imuno-Histoquímica , Radioisótopos de Índio/química , Radioisótopos do Iodo/química , Isotiocianatos/química , Isotiocianatos/farmacocinética , Marcação por Isótopo , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ácido Pentético/química , Ácido Pentético/farmacocinética , Cintilografia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Clin Oncol ; 19(19): 3976-87, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11579119

RESUMO

PURPOSE: KM871 is a chimeric monoclonal antibody against the ganglioside antigen GD3, which is highly expressed on melanoma cells. We conducted an open-label, dose escalation phase I trial of KM871 in patients with metastatic melanoma. PATIENTS AND METHODS: Seventeen patients were entered onto one of five dose levels (1, 5, 10, 20, and 40 mg/m2). Patients received three infusions of KM871 at 2-week intervals, with the first infusion of KM871 trace-labeled with indium-111 (111In) to enable assessment of biodistribution in vivo. Biopsies of metastatic melanoma sites were performed on days 7 to 10. RESULTS: Fifteen of 17 patients completed a cycle of three infusions of KM871. No dose-limiting toxicity was observed during the trial; the maximum-tolerated dose was therefore not reached. Three patients (at the 1-, 5-, and 40-mg/m2 dose levels) developed pain and/or erythema at tumor sites consistent with an inflammatory response. No normal tissue uptake of 111In-KM871 was observed, and tumor uptake of 111In-KM871 was observed in all lesions greater than 1.5 cm (tumor biopsy 111KM871 uptake results: range, 0.001% to 0.026% injected dose/g). The ratio of maximum tumor to normal tissue was 15:1. Pharmacokinetic analysis revealed a 111In-KM871 terminal half-life of 7.68 +/- 2.94 days. One patient had a clinical partial response that lasted 11 months. There was no serologic evidence of human antichimeric antibody in any patient, including one patient who received 16 infusions over a 12-month period. CONCLUSION: This study is the first to demonstrate the biodistribution and specific targeting of an anti-GD3 antibody to metastatic melanoma in patients. The long half-life and lack of immunogenicity of KM871 makes this antibody an attractive potential therapy for patients with metastatic melanoma.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Complexo CD3/imunologia , Melanoma/imunologia , Melanoma/metabolismo , Adulto , Idoso , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos , Biópsia , Feminino , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Radioisótopos de Índio , Masculino , Melanoma/diagnóstico por imagem , Melanoma/terapia , Pessoa de Meia-Idade , Cintilografia , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA