Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Theranostics ; 9(8): 2129-2142, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31149033

RESUMO

Androgen ablating drugs increase life expectancy in men with metastatic prostate cancer, but resistance inevitably develops. In a majority of these recurrent tumors, the androgen axis is reactivated in the form of increased androgen receptor (AR) expression. Targeting proteins that are expressed as a down-stream effect of AR activity is a promising rationale for management of this disease. The humanized IgG1 antibody hu11B6 internalizes into prostate and prostate cancer (PCa) cells by binding to the catalytic cleft of human kallikrein 2 (hK2), a prostate specific enzyme governed by the AR-pathway. In a previous study, hu11B6 conjugated with Actinium-225 (225Ac), a high linear energy transfer (LET) radionuclide, was shown to generate an AR-upregulation driven feed-forward mechanism that is believed to enhance therapeutic efficacy. We assessed the efficacy of hu11B6 labeled with a low LET beta-emitter, Lutetium-177 (177Lu) and investigated whether similar tumor killing and AR-enhancement is produced. Moreover, single-photon emission computed tomography (SPECT) imaging of 177Lu is quantitatively accurate and can be used to perform treatment planning. [177Lu]hu11B6 therefore has significant potential as a theranostic agent. Materials and Methods: Subcutaneous PCa xenografts (LNCaP s.c.) were grown in male mice. Biokinetics at 4-336 h post injection and uptake as a function of the amount of hu11B6 injected at 72 h were studied. Over a 30 to 120-day treatment period the therapeutic efficacy of different activities of [177Lu]hu11B6 were assessed by volumetric tumor measurements, blood cell counts, molecular analysis of the tumor as well as SPECT/CT imaging. Organ specific mean absorbed doses were calculated, using a MIRD-scheme, based on biokinetic data and rodent specific S-factors from a modified MOBY phantom. Tumor tissues of treated xenografts were immunohistochemically (IHC) stained for Ki-67 (proliferation) and AR, SA-ß-gal activity (senescence) and analyzed by digital autoradiography (DAR). Results: Organ-to-blood and tumor-to-blood ratios were independent of hu11B6 specific activity except for the highest amount of antibody (150 µg). Tumor accumulation of [177Lu]hu11B6 peaked at 168 h with a specific uptake of 29 ± 9.1 percent injected activity per gram (%IA/g) and low accumulation in normal organs except in the submandibular gland (15 ± 4.5 %IA/g), attributed to a cross-reaction with mice kallikreins in this organ, was seen. However, SPECT imaging with therapeutic amounts of [177Lu]hu11B6 revealed no peak in tumor accumulation at 7 d, probably due to cellular retention of 177Lu and decreasing tumor volumes. For [177Lu]hu11B6 treated mice, tumor decrements of up to 4/5 of the initial tumor volume and reversible myelotoxicity with a nadir at 12 d were observed after a single injection. Tumor volume reduction correlated with injected activity and the absorbed dose. IHC revealed retained expression of AR throughout treatment and that Ki-67 staining reached a nadir at 9-14 d which coincided with high SA- ß-gal activity (14 d). Quantification of nuclei staining showed that Ki-67 expression correlated negatively with activity uptake. AR expression levels in cells surviving therapy compared to previous timepoints and to controls at 30 d were significantly increased (p = 0.017). Conclusions: This study shows that hu11B6 labeled with the low LET beta-emitting radionuclide 177Lu can deliver therapeutic absorbed doses to prostate cancer xenografts with transient hematological side-effects. The tumor response correlated with the absorbed dose both on a macro and a small scale dosimetric level. Analysis of AR staining showed that AR protein levels increased late in the study suggesting a therapeutic mechanism, a feed forward mechanism coupled to AR driven response to DNA damage or clonal lineage selection, similar to that reported in high LET alpha-particle therapy using 225Ac labeled hu11B6, however emerging at a later timepoint.


Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Lutécio/farmacologia , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/terapia , Radioimunoterapia/métodos , Radioisótopos/farmacologia , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único/métodos , Calicreínas Teciduais/metabolismo , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Autorradiografia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Lutécio/administração & dosagem , Masculino , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Ligação Proteica , Radioisótopos/administração & dosagem , Nanomedicina Teranóstica/métodos , Calicreínas Teciduais/imunologia , Transplante Heterólogo , Resultado do Tratamento
2.
Tumour Biol ; 39(4): 1010428317697550, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28381167

RESUMO

The tumor microenvironment can act so as to stimulate or reject tumor cells. Among the determining factors are cytokines produced, for example, by infiltrating immune cells, tumor cells, and fibroblasts. External radiotherapy has been shown to be able to activate an immune response against tumor cells with cytokine signaling as an important part of the activation. The aim of this study was to evaluate the cytokines present in the tumor microenvironment and whether the cytokine profile changed during tumor regression induced by radioimmunotherapy with the beta emitter 177Lu. Immunocompetent rats with colon carcinoma tumors were injected with 400 MBq/kg 177Lu-mAb, and the tumors were excised after 1, 2, 3, 4, 6, or 8 days post injection (4 rats/day on days 1-6 and 8 rats on day 8). Tumors from 10 untreated rats were used as control tissue. The tumors were divided into half: one half was prepared for cytokine analysis with a cytokine array kit and the other half was used for histological analysis. A total of 18 of the 29 cytokines evaluated were detected in this tumor model, and the majority of these act in a pro-inflammatory manner or stimulate the infiltration of immune cells. The differences between treated tumors and control tumors were small, thus the cytokine profile in the untreated tumors did not transfer to an anti-inflammatory profile during tumor regression induced by radioimmunotherapy with 177Lu. Histological evaluation demonstrated a heterogeneous pattern of ongoing cell death and the formation of granulation tissue.


Assuntos
Neoplasias do Colo/radioterapia , Citocinas/análise , Radioimunoterapia , Animais , Biomarcadores Tumorais/análise , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Imunocompetência , Ratos , Ratos Endogâmicos BN , Microambiente Tumoral
3.
EJNMMI Res ; 5(1): 47, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26374556

RESUMO

BACKGROUND: Immune cells within the tumor can act either to promote growth or rejection of tumor cells. The aim of the present study was to evaluate immune cell markers (number and localization) within the tumor before and during rejection due to radioimmunotherapy, to determine whether there is a change in markers related to rejection and/or tolerance of the tumor cells. METHODS: Thirty immunocompetent rats were inoculated with syngeneic rat colon carcinoma cells and 13-14 days later 21 of these rats were treated with 400 MBq/kg of (177)Lu-DOTA-BR96 monoclonal antibodies. The treated animals were sacrificed and dissected 1, 2, 3, 4, 6, and 8 days post-injection in groups of three animals per day (6 animals on day 8); while the nine untreated animals were sacrificed and dissected on day 0. Paraffin sections were used for immunohistochemical staining of CD2, CD3, CD8α, CD68, and CD163 antigens. Positive cells were counted within: vital tumor cell areas, necrotic areas, granulation tissue surrounding and between the tumor cell areas. The change in the number of positive cells over time in tumors treated with radioimmunotherapy in the same location was evaluated with linear regression models. The number of positive cells in various locations and the number of various antigen-positive cells within the same location were also evaluated over time using box plots. RESULTS: There were a higher number of cells expressing immune cell markers in granulation tissue compared with vital tumor cell areas. Cells expressing markers decreased during radioimmunotherapy, and T-cell markers decreased more than macrophage markers in tumors treated with radioimmunotherapy. The expression of CD8α was higher than that of the other T-cell markers evaluated (CD3 and CD2), which could be explained by the additional expression of CD8α by natural killer (NK) cells and a subset of dendritic cells (DCs). The expression of CD68 (all macrophages, DCs, and neutrophils) tended to be higher than that of CD163 (pro-tumor macrophages). CONCLUSIONS: In this model, we demonstrated a higher number of positive cells for immune cell markers related to augmenting the immune rejection than immune tolerance of tumor cells in tumors and a decrease in markers during radioimmunotherapy.

4.
EJNMMI Res ; 5: 3, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25853009

RESUMO

BACKGROUND: CD8-positive cells might play a crucial role in the therapeutic response to radiation, which has however not been investigated in radioimmunotherapy (RIT). The aim of this study was to evaluate whether cytotoxic T cells affect the response of established tumors and, above all, if they delay or prevent the development of distant metastases after RIT, using an immunocompetent syngeneic rat colon carcinoma model. METHODS: The cytotoxic T cells were depleted in 15 rats by anti-CD8 before the injection of the radioimmunoconjugate (400 MBq/kg body weight (177)Lu-BR96, which binds to the tumor-associated antigen Lewis Y). Fifteen other rats were treated with RIT only. Both groups were followed for 99 days. Blood samples were collected at least once weekly, and tumors were monitored twice weekly. RESULTS: Twenty-nine of the 30 animals exhibited local complete response. The non-responder was treated with anti-CD8 and RIT but succumbed later due to metastases. Five animals in the group given anti-CD8 + RIT were sacrificed due to metastatic disease, and 4 additional animals were found to have metastases at autopsy. In the group given RIT, 4 animals developed metastatic disease, but no metastases were found in the remaining 11 animals at autopsy. Thus, at the end of the study, 6 animals in the anti-CD8 + RIT group were free from metastases, while 11 were free from metastases in the group receiving RIT. CD3(+)CD4(-)CD8(+) lymphocytes were consistently depleted by the anti-CD8 treatment. The myelosuppression was otherwise similar in the two groups. The initial depletion of CD8-positive cells in our syngeneic rat colon carcinoma model resulted in a higher frequency of animals developing metastases. CONCLUSIONS: Depletion of CD8-positive cells during RIT in an immunocompetent rat tumor model might influence the number of animals developing metastases, indicating that the immune system may be important in the long-term outcome of RIT.

5.
Cancer Biother Radiopharm ; 29(6): 238-46, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24971673

RESUMO

UNLABELLED: Alpha-particle emitters, such as astatine-211 (211At), are generally considered suitable for the treatment of small cell clusters due to their short path length, while beta-particle emitters, for example, Lutetium-177 (177Lu), have a longer path length and are considered better for small, established tumors. A combination of such radionuclides may be successful in regimens of radioimmunotherapy. In this study, rats were treated by sequential administration of first a 177Lu-labeled antibody, followed by a 211At-labeled antibody 25 days later. METHODS: Rats bearing solid colon carcinoma tumors were treated with 400 MBq/kg body weight 177Lu-BR96. After 25 days, three groups of animals were given either 5 or 10 MBq/kg body weight of 211At-BR96 simultaneously with or without a blocking agent reducing halogen uptake in normal tissues. Control animals were not given any 211At-BR96. Myelotoxicity, body weight, tumor size, and development of metastases were monitored for 120 days. RESULTS: Tumors were undetectable in 90% of the animals on day 25, independent of treatment. Additional treatment with 211At-labeled antibodies did not reduce the proportion of animals developing metastases. The rats suffered from reversible myelotoxicity after treatment. CONCLUSIONS: Sequential administration of 177Lu-BR96 and 211At-BR96 resulted in tolerable toxicity providing halogen blocking but did not enhance the therapeutic effect.


Assuntos
Astato/administração & dosagem , Neoplasias do Colo/radioterapia , Lutécio/administração & dosagem , Radioimunoterapia/métodos , Radioisótopos/administração & dosagem , Partículas alfa , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Humanos , Compostos Radiofarmacêuticos/administração & dosagem , Ratos
6.
Cancer Biother Radiopharm ; 29(4): 143-52, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24693940

RESUMO

PURPOSE: To monitor cell death in tumors during the rejection process after treatment with an antibody radiolabeled with a ß-emitter. METHODS: Tumors during rejection after treatment with (177)Lu-labeled antibody BR96 and after administration of unlabeled BR96 were compared with untreated tumors from the same immunocompetent syngeneic rat tumor model. Cell death was monitored with the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and immunohistochemical staining of activated caspase-3 and γH2AX. These data were evaluated together with histopathological morphology, BR96-binding antigen expression, and (177)Lu radioactivity distribution imaged by digital autoradiography. RESULTS: The untreated tumors showed staining for all the markers, mainly in and around the necrotic areas. One to 2 days p.i. large areas were stained with anti-γH2AX, followed by a slight decrease. Staining of activated caspase-3 was intense and extensive 1-2 days p.i., while found in and around necrotic areas 3-8 days p.i. TUNEL staining was similar to activated caspase-3 staining 1-2 days p.i. but more extensive than activated caspase-3 staining 3-4 days p.i. Digital autoradiography revealed activity concentration in granulation tissue from 1 day p.i. CONCLUSION: Following radioimmunotherapy in an immunocompetent syngeneic colon carcinoma model, tumor cells did not only die through caspase-3-dependent apoptosis, but also by other mechanisms.


Assuntos
Apoptose/efeitos da radiação , Neoplasias do Colo/patologia , Neoplasias do Colo/radioterapia , Imunotoxinas/farmacologia , Lutécio/administração & dosagem , Radioimunoterapia/métodos , Radioisótopos/administração & dosagem , Animais , Apoptose/fisiologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Humanos , Imunotoxinas/imunologia , Masculino , Ratos
7.
J Nucl Med ; 54(8): 1404-10, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23804328

RESUMO

UNLABELLED: The therapeutic effect of radioimmunotherapy depends on the distribution of the absorbed dose in relation to viable cancer cells within the tumor, which in turn is a function of the activity distribution. The aim of this study was to investigate the distribution of (177)Lu-DOTA-BR96 monoclonal antibodies targeting the Lewis Y antigen over 7 d using a syngeneic rat model of colon carcinoma. METHODS: Thirty-eight tumor-bearing rats were intravenously given 25 or 50 MBq of (177)Lu-DOTA-BR96 per kilogram of body weight and were sacrificed 2, 8, 24, 48, 72, 96, 120, or 168 h after injection, with activity measured in blood and tumor samples. Adjacent cryosections of each tumor were analyzed in 3 ways: imaging using a silicon-strip detector for digital autoradiography, staining for histologic characterization, or staining to determine the distribution of the antigen, vasculature, and proliferating cells using immunohistochemistry. Absorbed-dose rate distribution images at the moment of sacrifice were calculated using the activity distribution and a point-dose kernel. The correlations between antigen expression and both activity uptake and absorbed-dose rate were calculated for several regions of interest in each tumor. Nine additional animals with tumors were given unlabeled antibody to evaluate possible immunologic effects. RESULTS: At 2-8 h after injection, activity was found in the tumor margins; at 24 h, in viable antigen-expressing areas within the tumor; and at 48 h and later, increasingly in antigen-negative areas of granulation tissue. The correlation between antigen expression and both the mean activity and the absorbed-dose rate in regions of interest changed from positive to negative after 24 h after injection. Antigen-negative areas also increased over time in animals injected with unlabeled BR96, compared with untreated tumors. CONCLUSION: The results indicate that viable Lewis Y-expressing tumor cells are most efficiently treated during the initial uptake period. The activity then seems to remain in these initial uptake regions after the elimination of tumor cells and formation of granulation tissue. Further studies using these techniques could aid in determining the effects of the intratumoral activity distribution on overall therapeutic efficacy.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Lutécio/uso terapêutico , Radioisótopos/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/radioterapia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoconjugados , Marcação por Isótopo , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Masculino , Neovascularização Patológica , Doses de Radiação , Ratos , Fatores de Tempo
8.
EJNMMI Res ; 3(1): 23, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23557183

RESUMO

BACKGROUND: Most carcinomas are prone to metastasize despite successful treatment of the primary tumor. One way to address this clinical challenge may be targeted therapy with α-emitting radionuclides such as astatine-211 (211At). Radioimmunotherapy utilizing α-particle emitting radionuclides is considered especially suitable for the treatment of small cell clusters and single cells, although lesions of different sizes may also be present in the patient. The aim of this study was primarily to evaluate the toxicity and secondarily in vivo efficacy of a 211At-labeled monoclonal antibody (mAb) directed against colon carcinoma with tumor diameters of approximately 10 mm. METHODS: Eighteen rats with subperitoneal syngeneic colon carcinoma were allocated to three groups of six animals together with three healthy rats in each group. The groups were injected intravenously with either 150 µg of unlabeled mAbs (controls) or 2.5 or 5 MBq 211At-mAbs directed towards the Lewis Y antigen expressed on the cell membrane of several carcinomas. Tumor volume, body weight, and blood cell counts were monitored for 100 days after treatment. RESULTS: Local tumors were non-palpable in five out of six rats after treatment with both activities of 211At-mAbs, compared to one out of six in the control group. At the study end, half of the animals in each group given 211At-BR96 and one animal in the control group were free from disease. Radioimmunotherapy resulted in dose-dependent, transient weight loss and myelotoxicity. Survival was significantly better in the groups receiving targeted alpha therapy than in those receiving unlabeled mAbs. CONCLUSIONS: This study demonstrates the possibility of treating small, solid colon carcinoma tumors with α-emitting radionuclides such as 211At bound to mAbs, with tolerable toxicity.

9.
Exp Hematol Oncol ; 1(1): 34, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23210841

RESUMO

BACKGROUND: Repeated administration of antibody-based therapies such as radioimmunotherapy depends on preserved antigen expression in tumor lesions. The purpose of this study was to evaluate whether the antigen expression in metastases observed after radioimmunotherapy differs from that of untreated primary tumors. FINDINGS: 30 of the 35 Brown Norway rats with syngeneic colon carcinoma treated with 400 MBq/kg 177Lu-DOTA-BR96 exhibited consistent complete response of the primary tumor. 13 animals developed metastases that were detected after treatment. The antigen expression was reduced in 17 of 23 metastases detected after radioimmunotherapy compared with untreated tumors. No tumors completely lacked positively stained tumor cells. CONCLUSIONS: Although it was not possible to demonstrate that the antigen reduction is triggered by the radioimmunotherapy this result stress the importance of considering the risk of reduced antigen expression in metastases after radioimmunotherapy prior to further targeted therapies.

10.
Cancer Biother Radiopharm ; 27(3): 175-82, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22417248

RESUMO

Metastatic disease after successful treatment of the primary tumor continues to be a therapeutic challenge. Enhancement of therapeutic effects by the administration of unlabeled monoclonal antibodies (mAbs) after radioimmunotherapy (RIT) may provide a means of preventing or delaying the development of metastatic disease. In the present study, Brown Norway rats with syngeneic grafted colon carcinomas were administered the minimal effective therapeutic dose of 400 MBq/kg lutetium-177 ((177)Lu)-DOTA-BR96. After 2 weeks, half of the animals were given 15 mg/kg unlabeled mAb BR96 as consolidation therapy. Treatment response and toxicity were monitored 100 days after the treatment with unlabeled BR96. The treatment with unlabeled mAb after RIT resulted in a complete response (CR) in 19 of 19 animals, while RIT alone resulted in a CR in 17 of 19 animals. The additional treatment did not affect the number of animals with metastatic disease or the time to clinical symptoms of metastases. RIT resulted in reversible myelotoxicity. The unlabeled mAb BR96 did not cause any additional toxicity, making it possible to repeat the consolidation therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias do Colo/terapia , Modelos Animais de Doenças , Radioimunoterapia , Animais , Quelantes/uso terapêutico , Neoplasias do Colo/patologia , Neoplasias do Colo/radioterapia , Ensaio de Imunoadsorção Enzimática , Compostos Heterocíclicos com 1 Anel/uso terapêutico , Técnicas Imunoenzimáticas , Lutécio/uso terapêutico , Masculino , Radioisótopos/uso terapêutico , Ratos , Ratos Endogâmicos BN
11.
Cancer Biother Radiopharm ; 27(2): 134-40, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22229635

RESUMO

AIM: Fractionation is generally used as a mean to improve radioimmunotherapy (RIT). Since RIT is considered suitable for small-volume disease, the aim of the current study was to investigate whether repeated administration of (177)Lu-labeled mAb BR96 was tolerated and could delay or prevent metastatic disease after complete remission of the tumor obtained by the first administration. METHODS: Immunocompetent rats bearing a syngeneic colon carcinoma were first treated with 400 MBq/kg (177)Lu-DOTA-BR96, an activity resulting in complete response in 29 of 30 animals. On day 21, two groups of rats were given an additional activity of 150 or 350 MBq/kg resulting in total administered activities corresponding to 0.9 and 1.3 times the maximal tolerated dose. RESULTS: The additional treatment resulted in tolerable myelotoxicity; however, the frequency of metastatic disease and survival were not affected. Immunohistochemistry demonstrated binding of the BR96 antibody to tissue sections of analyzed metastases. CONCLUSIONS: In our model, development of metastatic disease after treatment of the manifest tumor was not prevented by an additional treatment with the same radioimmunoconjugate. Therefore, the antibody should be labeled with a more suitable radionuclide for treatment of metastases. The repeated targeted therapy was well tolerated in aspects of myelotoxicity.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Carcinoma/radioterapia , Neoplasias do Colo/radioterapia , Lutécio/administração & dosagem , Radioimunoterapia/métodos , Radioisótopos/administração & dosagem , Animais , Anticorpos Monoclonais/efeitos adversos , Plaquetas/efeitos da radiação , Peso Corporal/efeitos da radiação , Carcinoma/patologia , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Relação Dose-Resposta à Radiação , Imunoconjugados/administração & dosagem , Imunoconjugados/efeitos adversos , Leucócitos/efeitos da radiação , Lutécio/efeitos adversos , Masculino , Radioimunoterapia/efeitos adversos , Radioisótopos/efeitos adversos , Ratos , Ratos Endogâmicos BN
12.
Acta Oncol ; 50(5): 711-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21214493

RESUMO

BACKGROUND: One of many approaches being evaluated in experimental models and in the clinic for the treatment of cancer is the use of antibodies conjugated to various drugs or radionuclides. The aim of the present study was to compare the toxicity profiles of radioimmunoconjugates and drug-immunoconjugates based on the same monoclonal antibody, evaluated in the same experimental model, that much resembles human studies. The pattern of dose-limiting toxicity of a monomethylauristatin-conjugated monoclonal antibody (BR96) was compared to that of the same antibody conjugated with lutetium-177, and to the same non-conjugated antibody. MATERIAL AND METHODS: Rats with established colon carcinoma were injected with monomethylauristatin-conjugated mAb-BR96, (177)Lu-BR96, or non-conjugated BR96. Liver, kidney, and myelotoxicity were assessed for 100 days by analysis of blood parameters. Body weight and therapeutic effects was also monitored. RESULTS: Myelotoxicity was found to be dose limiting for the radionuclide BR96 conjugate. The dose-limiting factor was prolonged suppression of leukocytes (>28 days) with increased risk of infections. For monomethylauristatin-conjugated BR96, liver toxicity was dose limiting, whereas no dose-limiting toxicity was observed with non-conjugated BR96. Both the drug-immunoconjugate and the radioimmunoconjugate resulted in decreased platelet counts, but the time to nadir and duration differed. CONCLUSION: The two conjugates resulted in different patterns of toxicity. By using the two conjugates of BR96 in a sequential therapeutic design it could be possible to increase the therapeutic window and hence probably the efficacy without significantly increasing the toxicity. This concept is regarded as valid regardless of conjugate or model chosen.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Carcinoma/tratamento farmacológico , Carcinoma/radioterapia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/radioterapia , Imunoconjugados/administração & dosagem , Imunoconjugados/efeitos adversos , Animais , Anticorpos Monoclonais/química , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Carcinoma/patologia , Células Cultivadas , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Imunoconjugados/química , Transplante de Neoplasias , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Radioimunoterapia , Radioisótopos/administração & dosagem , Radioisótopos/efeitos adversos , Radioisótopos/química , Ratos , Ratos Endogâmicos BN , Transplante Isogênico
13.
Cancer ; 116(4 Suppl): 1033-42, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20127954

RESUMO

BACKGROUND: Antibody-drug conjugates, comprising monoclonal antibodies (MoAbs) that bind to tumor-associated antigens, display different toxicity profiles compared with radiolabeled MoAbs. Dose-limiting toxicities may include damage to the liver and myelotoxicity. The drug component is the antimitotic agent auristatin, which is 100-1000 times more potent than doxorubicin. Consequently, auristatin antibody-drug conjugates require a high selectivity in tumor targeting to display pronounced activity at well-tolerated doses. We have evaluated the possibility of increasing the therapeutic index of BR96-auristatin by combining the administration of conjugates with subsequent extracorporeal affinity adsorption treatment. METHODS: Rats were injected with biotinylated, monomethyl auristatin F (MMAF)-conjugated monoclonal antibody BR96. The conjugate was then removed from the circulation by extracorporeal affinity adsorption treatment, 24 hours postinjection using an avidin affinity column. By analyzing blood parameters for 100 days, myelotoxicity, hepatotoxicity, and nephrotoxicity were assessed. Body weight, general status, and tumor size were also recorded. The toxicity-reducing effect of extracorporeal affinity adsorption treatment was evaluated. RESULTS: Extracorporeal affinity adsorption treatment removed 85%-90% of BR96-MMAF from the circulation. Early toxicity-related death was seen in nontumor-bearing animals that were given MMAF-conjugated BR96, in contrast to animals that were given a higher amount of BR96-MMAF with subsequent extracorporeal affinity adsorption treatment, in which all survived 100 days postinjection. Extracorporeal affinity adsorption treatment reduced the loss of body weight, myelotoxicity, and hepatotoxicity. CONCLUSIONS: Extracorporeal affinity adsorption treatment can be used to reduce the toxicity associated with administration of BR96-MMAF conjugates, making it possible to increase the amount of conjugates administered. The combined treatment will be further optimized in future studies.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Neoplasias do Colo/tratamento farmacológico , Imunoconjugados/administração & dosagem , Oligopeptídeos/administração & dosagem , Animais , Modelos Animais de Doenças , Circulação Extracorpórea , Estudos de Viabilidade , Imunoconjugados/efeitos adversos , Oligopeptídeos/química , Ratos , Ratos Endogâmicos BN , Redução de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...