Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Chem Commun (Camb) ; 55(72): 10784-10787, 2019 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-31432802

RESUMO

Medical magnetic resonance imaging (MRI) produces high-resolution anatomical images of the human body, but has limited capacity to provide useful molecular information. The light-responsive, liposomal MRI contrast agent described herein could be used to provide an intrinsic theranostic aspect to MRI and enable tracking the distribution and cargo release of drug delivery systems upon light-triggered activation.


Assuntos
Meios de Contraste/química , Sistemas de Liberação de Medicamentos , Gadolínio/química , Luz , Imageamento por Ressonância Magnética , Humanos , Lipossomos/química , Estrutura Molecular
2.
Peptides ; 67: 45-54, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25797109

RESUMO

The gastrin-releasing peptide receptor (GRPR) is overexpressed in a variety of human malignancies, including prostate cancer. Bombesin (BBN) is a 14 amino acids peptide that selectively binds to GRPR. In this study, we developed two novel Al(18)F-labeled lanthionine-stabilized BBN analogs, designated Al(18)F-NOTA-4,7-lanthionine-BBN and Al(18)F-NOTA-2,6-lanthionine-BBN, for positron emission tomography (PET) imaging of GRPR expression using xenograft prostate cancer models. (Methyl)lanthionine-stabilized 4,7-lanthionine-BBN and 2,6-lanthionine-BBN analogs were conjugated with a NOTA chelator and radiolabeled with Al(18)F using the aluminum fluoride strategy. Al(18)F-NOTA-4,7-lanthionine-BBN and Al(18)F-NOTA-2,6-lanthionine-BBN was labeled with Al(18)F with good radiochemical yield and specific activity>30 GBq/µmol for both radiotracers. The logD values measured for Al(18)F-NOTA-4,7-lanthionine-BBN and Al(18)F-NOTA-2,6-lanthionine-BBN were -2.14 ± 0.14 and -2.34 ± 0.15, respectively. In athymic nude PC-3 xenografts, at 120 min post injection (p.i.), the uptake of Al(18)F-NOTA-4,7-lanthionine-BBN and Al(18)F-NOTA-2,6-lanthionine-BBN in prostate cancer (PC-3) mouse models was 0.82 ± 0.23% ID/g and 1.40 ± 0.81% ID/g, respectively. An excess of unlabeled ɛ-aminocaproic acid-BBN(7-14) (300-fold) was co-injected to assess GRPR binding specificity. Tumor uptake of Al(18)F-NOTA-4,7-lanthionine-BBN and Al(18)F-NOTA-2,6-lanthionine-BBN in PC-3 tumors was evaluated by microPET (µPET) imaging at 30, 60 and 120 min p.i. Blocking studies showed decreased uptake in PC-3 bearing mice. Stabilized 4,7-lanthionine-BBN and 2,6-lanthionine-BBN peptides were rapidly and successfully labeled with (18)F. Both tracers may have potential for GRPR-positive tumor imaging.


Assuntos
Neoplasias da Próstata/diagnóstico por imagem , Receptores da Bombesina/metabolismo , Alanina/análogos & derivados , Alanina/farmacocinética , Animais , Bombesina/farmacocinética , Linhagem Celular Tumoral , Radioisótopos de Flúor/farmacocinética , Humanos , Masculino , Camundongos Nus , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons , Neoplasias da Próstata/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Sulfetos/farmacocinética , Distribuição Tecidual , Microtomografia por Raio-X
3.
Mol Imaging Biol ; 16(6): 747-55, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24849133

RESUMO

In oncology, sensitive and reliable detection tumor tissue is crucial to prevent recurrences and to improve surgical outcome. Currently, extensive research is focused on the use of radionuclides as well as fluorophores to provide real-time guidance during surgery to aid the surgeon in the identification of malignant tissue. Particularly, dual-modality approaches combining radionuclide and near-infrared fluorescence (NIRF) imaging have shown promising results in preclinical studies. Radionuclide imaging allows sensitive intra-operative localization of tumor lesions using a gamma probe, whereas NIRF imaging allows more accurate real-time tumor delineation. Consequently, both radionuclide and NIRF imaging might complement each other, and dual-modality image-guided surgery may overcome limitations of the currently used single-modality imaging techniques. In this review, a comprehensive overview on recent preclinical advances in tumor-targeted radionuclide and fluorescence dual-modality imaging is provided. Subsequently, the clinical applicability of dual-modality image-guided surgery is discussed.


Assuntos
Corantes Fluorescentes , Imagem Multimodal/métodos , Neoplasias/diagnóstico , Radioisótopos , Animais , Corantes Fluorescentes/química , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Tomografia por Emissão de Pósitrons/métodos , Radioisótopos/química , Espectroscopia de Luz Próxima ao Infravermelho , Tomografia Computadorizada de Emissão de Fóton Único/métodos
4.
Mol Pharm ; 10(5): 1716-24, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-23590837

RESUMO

Rational-designed multimerization of targeting ligands can be used to improve kinetic and thermodynamic properties. Multimeric targeting ligands may be produced by tethering multiple identical or two or more monomeric ligands of different binding specificities. Consequently, multimeric ligands may simultaneously bind to multiple receptor molecules. Previously, multimerization has been successfully applied on radiolabeled RGD peptides, which resulted in an improved tumor targeting activity in animal models. Multimerization of peptide-based ligands may improve the binding characteristics by increasing local ligand concentration and by improving dissociation kinetics. Here, we present a preclinical study on a novel radiolabeled bombesin (BN) homodimer, designated (111)In-DOTA-[(Aca-BN(7-14)]2, that was designed for enhanced targeting of gastrin-releasing peptide receptor (GRPR)-positive prostate cancer cells. A BN homodimer was conjugated with DOTA-NHS and labeled with (111)In. After HPLC purification, the GRPR targeting ability of (111)In-DOTA-[Aca-BN(7-14)]2 was assessed by microSPECT imaging in SCID mice xenografted with the human prostate cancer cell line PC-3. (111)In labeling of DOTA-[(Aca-BN(7-14)]2 was achieved within 30 min at 85 °C with a labeling yield of >40%. High radiochemical purity (>95%) was achieved by HPLC purification. (111)InDOTA-[Aca-BN(7-14)]2 specifically bound to GRPR-positive PC-3 prostate cancer cells with favorable binding characteristics because uptake of 111In-DOTA-[Aca-BN(7-14)]2 in GRPR-positive PC-3 cells increased over time. A maximum peak with 30% radioactivity was observed after 2 h of incubation. The log D value was -1.8 ± 0.1. (111)In-DOTA-[Aca-BN(7-14)]2 was stable in vitro both in PBS and human serum for at least 4 days. In vivo biodistribution analysis and microSPECT/CT scans performed after 1, 4, and 24 h of injection showed favorable binding characteristics and tumor-to-normal tissue ratios. This study identifies (111)In-DOTA-[(Aca-BN(7-14)]2 as a promising radiotracer for nuclear imaging of GRPR in prostate cancer.


Assuntos
Bombesina , Radioisótopos de Índio , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/metabolismo , Receptores da Bombesina/metabolismo , Animais , Linhagem Celular Tumoral , Compostos Heterocíclicos com 1 Anel , Humanos , Masculino , Camundongos , Camundongos Nus , Compostos Radiofarmacêuticos , Tomografia Computadorizada de Emissão de Fóton Único
5.
Eur J Surg Oncol ; 37(1): 32-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21106329

RESUMO

PURPOSE: Breast-conserving surgery (BCS) results in tumour-positive surgical margins in up to 40% of the patients. Therefore, new imaging techniques are needed that support the surgeon with real-time feedback on tumour location and margin status. In this study, the potential of near-infrared fluorescence (NIRF) imaging in BCS for pre- and intraoperative tumour localization, margin status assessment and detection of residual disease was assessed in tissue-simulating breast phantoms. METHODS: Breast-shaped phantoms were produced with optical properties that closely match those of normal breast tissue. Fluorescent tumour-like inclusions containing indocyanine green (ICG) were positioned at predefined locations in the phantoms to allow for simulation of (i) preoperative tumour localization, (ii) real-time NIRF-guided tumour resection, and (iii) intraoperative margin assessment. Optical imaging was performed using a custom-made clinical prototype NIRF intraoperative camera. RESULTS: Tumour-like inclusions in breast phantoms could be detected up to a depth of 21 mm using a NIRF intraoperative camera system. Real-time NIRF-guided resection of tumour-like inclusions proved feasible. Moreover, intraoperative NIRF imaging reliably detected residual disease in case of inadequate resection. CONCLUSION: We evaluated the potential of NIRF imaging applications for BCS. The clinical setting was simulated by exploiting tissue-like breast phantoms with fluorescent tumour-like agarose inclusions. From this evaluation, we conclude that intraoperative NIRF imaging is feasible and may improve BCS by providing the surgeon with imaging information on tumour location, margin status, and presence of residual disease in real-time. Clinical studies are needed to further validate these results.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/cirurgia , Mastectomia Segmentar , Neoplasia Residual/diagnóstico por imagem , Neoplasia Residual/cirurgia , Imagens de Fantasmas , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Feminino , Fluorescência , Humanos , Raios Infravermelhos , Período Intraoperatório , Modelos Anatômicos , Neoplasia Residual/patologia , Radiografia , Radioterapia Adjuvante
6.
Neuropathol Appl Neurobiol ; 36(3): 168-82, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20102513

RESUMO

Glioblastoma (GBM) is a devastating cancer with a median survival of around 15 months. Significant advances in treatment have not been achieved yet, even with a host of new therapeutics under investigation. Therefore, the quest for a cure for GBM remains as intense as ever. Of particular interest for GBM therapy is the selective induction of apoptosis using the pro-apoptotic tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). TRAIL signals apoptosis via its two agonistic receptors TRAIL-R1 and TRAIL-R2. TRAIL is normally present as homotrimeric transmembrane protein, but can also be processed into a soluble trimeric form (sTRAIL). Recombinant sTRAIL has strong tumouricidal activity towards GBM cells, with no or minimal toxicity towards normal human cells. Unfortunately, GBM is a very heterogeneous tumour, with multiple genetically aberrant clones within one tumour. Consequently, any single agent therapy is likely to be not effective enough. However, the anti-GBM activity of TRAIL can be synergistically enhanced by a variety of conventional and novel targeted therapies, making TRAIL an ideal candidate for combinatorial strategies. Here we will, after briefly detailing the biology of TRAIL/TRAIL receptor signalling, focus on the promises and pitfalls of recombinant TRAIL as a therapeutic agent alone and in combinatorial therapeutic approaches for GBM.


Assuntos
Glioma/terapia , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Apoptose/genética , Apoptose/fisiologia , Glioma/metabolismo , Humanos , Modelos Neurológicos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
7.
Curr Drug Targets ; 11(1): 95-110, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20017722

RESUMO

Despite recent advances, treatment of leukemia is often not curative. New insights indicate that this may be attributable to a small population of therapy-resistant malignant cells with self-renewal capacity and the ability to generate large numbers of more differentiated leukemia cells. These leukemia-initiating cells are commonly referred to as Leukemia Stem Cells (LSCs). LSCs are regarded as the root of leukemia origin and leukemia recurrence after seemingly successful therapy. Not surprisingly therefore, contemporary leukemia research has focused on ways to specifically eliminate LSCs, leading to the identification of several promising anti-LSC strategies. Firstly, LSCs may be eliminated by antibody- or ligand-based cell surface delivery of therapeutics such as naked antibodies, immunotoxins, and immunocytokines. This approach exploits LSC-associated surface antigens, such as CD33, CD44, CD96, CD123 and CLL-1 for LSC-selective therapy and aims to spare normal hematopoietic stem cells. A second strategy aims to disrupt the interactions between LSCs and their highly specialized niche. These interactions appear to be pivotal for maintenance of the stem cell-like characteristics of LSCs. A third strategy centers on the selective modulation of aberrantly activated signaling pathways central to LSC biology. A fourth strategy, dubbed 'epigenetic reprogramming', aims to selectively reverse epigenetic alterations that are implicated in ontogeny and maintenance of LSCs. In this review, we will discuss the rationale for these LSCs-targeted strategies and highlight recent advances that may ultimately help pave the way towards selective LSCs-elimination.


Assuntos
Leucemia/patologia , Leucemia/terapia , Células-Tronco Neoplásicas/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia/tratamento farmacológico , Células-Tronco Neoplásicas/imunologia , Transdução de Sinais/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos
8.
Leukemia ; 23(8): 1389-97, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19262596

RESUMO

Gemtuzumab ozogamicin (GO, Mylotarg) is a targeted therapeutic agent in which an anti-CD33 antibody is chemically coupled to a highly cytotoxic calicheamicin derivative through a hydrolysable linker. GO has improved the treatment outcome for a subgroup of acute myeloid leukemia (AML) patients, but its use is associated with severe myelosuppression and hepatotoxicity. Here, we report on a novel anti-leukemia agent, designated scFvCD33:sTRAIL, in which an anti-CD33 single chain fragment of variable regions (scFv) antibody fragment is genetically linked to soluble tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL). Normal CD33-positive monocytes were fully resistant to prolonged treatment with scFvCD33:sTRAIL, whereas treatment with GO resulted in substantial cytotoxicity. The activity of scFvCD33:sTRAIL towards AML cells was up to 30-fold higher than GO. The CD33-restricted anti-leukemia activity of scFvCD33:sTRAIL remained stable during prolonged storage at 37 degrees C, whereas GO showed a rapid increase in CD33-independent cytotoxicity. Moreover, scFvCD33:sTRAIL showed potent anti-leukemia activity towards CD33+ CML cells when treatment was combined with the Bcr-Abl tyrosine kinase inhibitor, Gleevec. Importantly, ex vivo treatment of patient-derived CD33+ AML tumor cells with scFvCD33:sTRAIL resulted in potent apoptosis induction that was enhanced by valproic acid, mitoxantrone and 17-(Allylamino)-17-demethoxygeldanamycin (17-AAG). Taken together, scFvCD33:sTRAIL is superior to GO in terms of tumor selectivity, activity and stability, warranting its further development for the treatment of CD33-positive leukemias.


Assuntos
Antineoplásicos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Doença Aguda , Aminoglicosídeos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Efeito Espectador , Células Cultivadas/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Gemtuzumab , Humanos , Leucemia Mieloide/patologia , Leucócitos Mononucleares/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/química , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Anticorpos de Cadeia Única , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Curr Drug Targets ; 10(2): 94-103, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19199904

RESUMO

Antibody-based therapeutic approaches are yielding more and more of the promise they have held since the conception of the 'magic bullet' theory by Paul Ehrlich. The beneficial effect of antibody-based therapies is directly related to antibody-dependent functions, such as neutralization and antibody-dependent cellular cytotoxicity, but in many cases also relies on the delivery of toxic compounds to cancerous cells. However, the clinical utility of toxic antibody conjugates can be significantly hampered by side effects. Ideal effector compounds are inactive 'en route', but gain full activity once the antibody conjugate has bound to cancerous cells. Of significant potential in this respect are the pro-apoptotic ligands Tumor Necrosis Factor (TNF), fibroblast-associated cell-surface ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL). TNF ligands are normally present as homotrimeric transmembrane proteins, but can also be processed into a soluble trimeric form. Compared to their corresponding transmembrane counterpart, soluble TNF, FasL and TRAIL have a strongly reduced capacity to activate TNF receptor 2, Fas and TRAIL receptor 2. However, all sequence information required for full activation of these receptors is latently retained in these soluble ligands and can be unmasked by oligomerization or cell surface immobilization. The latter provides a clear rationale for the use of these ligands as effectors in antibody-based therapy. The antibody-targeted ligand will be in a relatively inactive soluble form while en route. However, once bound to the targeted cancer cell the soluble TNF ligand fusion proteins will be converted into fully active membrane ligand-like molecules. Here we will, after briefly detailing the biology of TNF, TRAIL and FasL, focus on the promises and pitfalls of targeted TNF ligand fusion proteins in achieving a 'magic bullet' with maximum cancer selective activity and minimal side effects.


Assuntos
Imunoterapia , Neoplasias/terapia , Fator de Necrose Tumoral alfa/metabolismo , Proteína Ligante Fas/metabolismo , Humanos , Ligantes , Proteínas Recombinantes/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
10.
Curr Pharm Des ; 14(28): 3033-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18991717

RESUMO

Prostate cancer is one of the most common causes of cancer in men. Evaluating the different stages of prostate cancer with conventional imaging techniques still proves difficult. Nuclear imaging might provide a technique that is able to evaluate prostate cancer, but clinical application has been limited due to lack of accuracy of current radiopharmaceuticals. The development of radiopharmaceuticals that can be targeted to specific antigens, overexpressed in prostate cancer, but sparse in normal tissue, is crucial. Peptides are of particular interest because of their favourable characteristics, leading to increased attention for nuclear imaging of the gastrin-releasing-peptide-receptor (GRPR) with radiolabelled bombesin-like peptides. Several derivatives of bombesin and its truncated form have been prepared for imaging with single photon emission computed tomography (SPECT) or positron emission tomography (PET), thereby delivering potent candidates for further clinical evaluation. This article provides an overview of the development and preclinical evaluation of radiolabelled bombesin analogues for in vivo targeting of GRPR in prostate cancer. The effect of the radionuclide, chelator, spacer and unnatural amino acids on affinity, metabolic stability and image quality are discussed, as well as agonistic or antagonistic properties. Potent candidates are proposed based on these selection criteria: (I) high affinity for GRPR, with rapid and specific tumour uptake (II) high hydrophilicity resulting in the preferred renal-urinary mode of excretion and low hepatobiliary excretion, (III) high stability, but relatively rapid clearance from blood. Also, a summary is made of clinical studies that report on the detection of prostate cancer with GRPR targeted radiopharmaceuticals.


Assuntos
Neoplasias da Próstata/diagnóstico por imagem , Compostos Radiofarmacêuticos , Receptores da Bombesina/metabolismo , Animais , Bombesina/farmacocinética , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Humanos , Masculino , Tomografia por Emissão de Pósitrons/métodos , Neoplasias da Próstata/diagnóstico , Compostos Radiofarmacêuticos/farmacocinética , Tomografia Computadorizada de Emissão de Fóton Único/métodos
11.
Kidney Int ; 73(12): 1364-73, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18354382

RESUMO

Ischemia-reperfusion injury is a leading cause of acute renal failure and a major determinant in the outcome of kidney transplantation. Here we explored systemic gene therapy with a modified adenovirus expressing Interleukin (IL)-13, a cytokine with strong anti-inflammatory and cytoprotective properties. When ischemia was induced we found that the IL-13 receptor is expressed in both the normal and experimental kidneys. Prior to the induction of ischemia, rats received adenovirus-IL-13, control adenovirus or saline. IL-13 plasma levels increased more than 50-fold in adenovirus-IL-13 treated animals, confirming successful IL-13 gene delivery. Histological analysis showed decreased tubular epithelial cell damage with adenovirus-IL-13 therapy, accompanied by reduced kidney injury molecule-1 expression. Interstitial infiltration by neutrophils and macrophages was reduced by half as was interstitial fibrosis and expression of alpha-smooth muscle actin. IL-13 treatment significantly diminished the expression of E-selectin, IL-8, MIP-2, TNF-alpha and MCP-1 mRNA. These results suggest that the use of systemic IL-13 gene therapy may be useful in reducing renal tubulointerstitial damage and inflammation caused by ischemia-reperfusion.


Assuntos
Terapia Genética , Interleucina-13/genética , Túbulos Renais/irrigação sanguínea , Insuficiência Renal/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , Animais , Proliferação de Células , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Regulação para Baixo , Selectina E/genética , Selectina E/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibrose , Interleucina-13/sangue , Interleucina-8/genética , Interleucina-8/metabolismo , Antígeno Ki-67/análise , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Macrófagos/imunologia , Neutrófilos/imunologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Receptores de Interleucina-13/agonistas , Insuficiência Renal/etiologia , Insuficiência Renal/patologia , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Leukemia ; 21(2): 248-52, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17122863

RESUMO

Gemtuzumab ozogamicin (GO) is a calicheamicin-conjugated antibody directed against CD33, an antigen highly expressed on acute myeloid leukemic (AML) cells. CD33-specific binding triggers internalization of GO and subsequent hydrolytic release of calicheamicin. Calicheamicin then translocates to the nucleus, intercalates in the DNA structure and subsequently induces double-strand DNA breaks. GO is part of clinical practice for AML, but is frequently associated with severe side effects. Therefore, combination of GO with other therapeutics is warranted to reduce toxicity, while maximizing therapeutic selectivity. We hypothesized that the histone deacetylase inhibitor valproic acid (VPA) sensitizes AML cells to GO. VPA-induced histone hyperacetylation opens the chromatin structure, whereby the DNA intercalation of calicheamicin should be augmented. We found that clinically relevant concentrations of VPA potently augmented the tumoricidal activity of GO towards AML cell lines and primary AML blasts. Moreover, VPA treatment indeed augmented the DNA intercalation of calicheamicin and enhanced DNA degradation. Importantly, synergy was restricted to CD33-positive AML cells and did not require caspase activation. In conclusion, the synergistic proapoptotic activity of cotreatment of AML cells with VPA and GO indicates the potential value of this strategy for AML.


Assuntos
Aminoglicosídeos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases , Leucemia Mieloide Aguda/patologia , Ácido Valproico/toxicidade , Anticorpos Monoclonais Humanizados , Anticonvulsivantes/toxicidade , Antígenos CD/sangue , Antígenos de Diferenciação Mielomonocítica/sangue , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , DNA de Neoplasias/efeitos dos fármacos , Sinergismo Farmacológico , Gemtuzumab , Humanos , Substâncias Intercalantes/farmacologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Células U937
13.
Curr Opin Mol Ther ; 3(1): 53-62, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11249732

RESUMO

Bi-specific antibodies (BsAbs) combine immune cell activation with tumor cell recognition as a result of which tumor cells are killed by pre-defined effector cells. In this review a brief introduction to monoclonal antibodies will precede a more in-depth presentation of the current status of BsAb therapy for cancer. Target molecules and effector mechanisms aimed at tumor cells or aimed at tumor vasculature, and the application of recombinant DNA technology in the construction of antibodies, will be discussed. The lessons learned from the last decade will be discussed in consideration of the potential future development of BsAbs for cancer therapy.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Animais , Anticorpos Biespecíficos/química , Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/química , Antígenos de Neoplasias/metabolismo , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Receptores Fc/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Linfócitos T/imunologia
14.
Mol Cell Biol ; 21(7): 2570-80, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11259604

RESUMO

Ep-CAM is a new type of cell adhesion molecule (CAM) which does not structurally resemble the members of the four major families (cadherins, integrins, selectins, and CAMs of the immunoglobulin superfamily) and mediates Ca(2+)-independent, homophilic adhesions. The extracellular domain of Ep-CAM consists of a cysteine-rich region, containing two type II epidermal growth factor (EGF)-like repeats, followed by a cysteine-poor region. We generated mutated Ep-CAM forms with various deletions in the extracellular domain. These deletion mutants, together with monoclonal antibodies recognizing different epitopes in the extracellular domain, were used to investigate the role of the EGF-like repeats in the formation of intercellular contacts mediated by Ep-CAM molecules. We established that both EGF-like repeats are required for the formation of Ep-CAM-mediated homophilic adhesions, including the accumulation of Ep-CAM molecules at the cell-cell boundaries, and the anchorage of the Ep-CAM adhesion complex to F-actin via alpha-actinin. Deletion of either EGF-like repeat was sufficient to inhibit the adhesion properties of the molecule. The first EGF-like repeat of Ep-CAM is required for reciprocal interactions between Ep-CAM molecules on adjacent cells, as was demonstrated with blocking antibodies. The second EGF-like repeat was mainly required for lateral interactions between Ep-CAM molecules. Lateral interactions between Ep-CAM molecules result in the formation of tetramers, which might be the first and necessary step in the formation of Ep-CAM-mediated intercellular contacts.


Assuntos
Antígenos de Neoplasias/fisiologia , Moléculas de Adesão Celular/fisiologia , Fator de Crescimento Epidérmico/genética , Adesão Celular/genética , Linhagem Celular , Molécula de Adesão da Célula Epitelial , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Humanos , Mutação , Sequências de Repetição em Tandem/genética
15.
Int J Cancer ; 85(6): 871-6, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10709110

RESUMO

The UBE1L gene isolated from the chromosome 3p21 region has an extremely reduced level of mRNA in lung cancer. Sequence analysis showed a 45% homology to the human ubiquitin-activating enzyme E1 at the amino acid level. To further characterize the protein product, we generated UBE1L protein-specific antibodies. Immunoblot analysis revealed a full-length gene product of approximately 112 kDa. Assessment of the level and distribution pattern of the UBE1L protein in normal and tumor tissue using the generated antibodies showed that the UBE1L protein was present in normal lung cells and non-lung cancer cell lines, but was undetectable in all 14 human lung cancer cell lines analyzed. This difference in expression of the UBE1L protein between normal lung tissue and lung tumor-derived cell lines suggests a possible involvement of an E1-like protein in the origin and/or progression of lung tumors.


Assuntos
Ligases/metabolismo , Neoplasias Pulmonares/enzimologia , Western Blotting , Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Ligases/genética , Ligases/imunologia , Neoplasias Pulmonares/patologia , Células Tumorais Cultivadas , Enzimas Ativadoras de Ubiquitina , Ubiquitina-Proteína Ligases
16.
J Immunol Methods ; 237(1-2): 131-45, 2000 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-10725458

RESUMO

A versatile expression vector is described for the rapid construction and evaluation of bispecific scFvs and scFv-based fusion proteins. An important feature of this vector is the presence of two multiple cloning sites (MCS) separated by an in frame linker sequence. The first MCS was specifically designed to contain unique SfiI and NotI restriction enzyme sites that can be used for directional and in frame insertion of scFvs (or potentially any molecule) selected from established phage-display systems. Using this new vector, a functional bs-(scFv)(2) (2C11-MOC31) was constructed for retargeted T-cell cytotoxicity towards EGP2 positive tumor cells. The vector was also used for grafting of a number of promising biological effector principles onto scFv MOC31, including the prodrug converting enzyme cytosine deaminase, the anti-angiogenic factor angiostatin, and the thrombogenic molecule tissue factor. We aimed at producing biologically active fusion proteins by directing them through the endoplasmic reticulum-based protein folding machinery of eukaryotic cells (COS-7) using a kappa light chain leader, thereby taking advantage of the associated quality control mechanisms that allow only fully folded and processed fusion proteins to be secreted into the medium. Supernatants derived from fusion protein transfected COS-7 cells, which were transiently transfected at low transfection rates, were directly assayed for the biological and/or targeting activity of the excreted fusion proteins without any prior purification steps. This procedure might help to identify those fusion proteins that have favourable characteristics like stability and biological activity in the presence of serum and at low protein concentrations. Targeted delivery of all effector principles was subsequently assessed in an in vitro model system. The method we devised is both rapid and versatile and can be useful to construct and identify series of new chimeric proteins with enhanced therapeutic potential in human cancer therapy.


Assuntos
Vetores Genéticos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/isolamento & purificação , Angiostatinas , Animais , Sequência de Bases , Células COS , Citosina Desaminase , Citotoxicidade Imunológica , Primers do DNA/genética , Expressão Gênica , Humanos , Fragmentos de Imunoglobulinas/biossíntese , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular , Nucleosídeo Desaminases/biossíntese , Nucleosídeo Desaminases/genética , Nucleosídeo Desaminases/isolamento & purificação , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/isolamento & purificação , Plasmídeos/genética , Plasminogênio/biossíntese , Plasminogênio/genética , Plasminogênio/isolamento & purificação , Dobramento de Proteína , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Linfócitos T Citotóxicos/imunologia , Tromboplastina/biossíntese , Tromboplastina/genética , Tromboplastina/isolamento & purificação , Transfecção , Células Tumorais Cultivadas
17.
Br J Cancer ; 82(2): 472-9, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10646907

RESUMO

Bispecific antibody (BsMAb) BIS-1 has been developed to redirect the cytolytic activity of cytotoxic T lymphocytes (CTL) to epithelial glycoprotein-2 (EGP-2) expressing tumour cells. Intravenous administration of BIS-1 F(ab')2 to carcinoma patients in a phase I/II clinical trial, caused immunomodulation as demonstrated by a rapid lymphopenia prior to a rise in plasma tumour necrosis factor-alpha and interferon-gamma levels. Yet, no lymphocyte accumulation in the tumour tissue and no anti-tumour effect could be observed. These data suggest a BsMAb-induced lymphocyte adhesion to blood vessel walls and/or generalized redistribution of the lymphocytes into tissues. In this study, we describe the effects of BIS-1 F(ab')2 binding to peripheral blood mononuclear cells (PBMC) on their capacity to interact with resting endothelial cells in vitro. Resting and pre-activated PBMC exhibited a significant increase in adhesive interaction with endothelial cells when preincubated with BIS-1 F(ab')2, followed by an increase in transendothelial migration (tem). Binding of BIS-1 F(ab')2 to PBMC affected the expression of a number of adhesion molecules involved in lymphocyte adhesion/migration. Furthermore, PBMC preincubated with BIS-1 F(ab')2 induced the expression of endothelial cell adhesion molecules E-selectin, VCAM-1 and ICAM-1 during adhesion/tem. These phenomena were related to the CD3 recognizing antibody fragment of the BsMAb and dependent on lymphocyte-endothelial cell contact. Possibly, in patients, the BIS-1 F(ab')2 infusion induced lymphopenia is a result of generalized activation of endothelial cells, leading to the formation of a temporary sink for lymphocytes. This process may distract the lymphocytes from homing to the tumour cells, and hence prevent the occurrence of BIS-1 F(ab')2 - CTL-mediated tumour cell lysis.


Assuntos
Anticorpos Biespecíficos/imunologia , Complexo CD3/imunologia , Adesão Celular/imunologia , Linfócitos/imunologia , Comunicação Celular/imunologia , Movimento Celular , Endotélio/citologia , Endotélio/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoterapia , Técnicas In Vitro , Molécula 1 de Adesão Intercelular/imunologia , Linfopenia/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia
18.
J Control Release ; 64(1-3): 229-39, 2000 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-10640660

RESUMO

To overcome dose limiting toxicities and to increase efficacy of immunotherapy of cancer, a number of strategies are under development for selectively redirecting effector cells/molecules towards tumor cells. Many of these strategies exploit the specificity of tumor associated antigen recognition by monoclonal antibodies. Using either hybridoma fusion, chemical derivatization or molecular biology technology, antibodies with dual specificity can be constructed. These so called biospecific antibodies (BsAbs) have been used to redirect the cytolytic activity of a variety of immune effector cells such as cytotoxic T lymphocytes, natural killer cells, neutrophils and monocytes/macrophages to tumor cells. Local administration of BsAbs, either alone or in combination with autologous effector cells, is highly effective in eradicating tumor cells. In contrast, systemic application of BsAb at present is only suitable for adjuvant treatment of minimal residual disease due to poor tumor cell accessibility. As an alternative, angiogenesis related determinants on tumor blood vessels can be exploited for the selective delivery of effector cells/molecules apart from being used to inhibit angiogenesis. Important advantages of this strategy is that the endothelial cell associated target epitope(s) are easy accessible. The dependence of tumor growth on the tumor's blood supply also renders tumor endothelial cells an attractive target for therapy. Although still in its infancy, attacking the tumor's blood supply for example by delivering coagulation factors or toxins, or by BsAb directed immunotherapies holds great promise for antineoplastic therapy.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Imunoterapia/métodos , Neoplasias/terapia , Neovascularização Patológica/prevenção & controle , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/administração & dosagem , Vasos Sanguíneos/efeitos dos fármacos , Ensaios Clínicos como Assunto , Epitopos/efeitos dos fármacos
19.
Nat Biotechnol ; 17(3): 276-81, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10096296

RESUMO

A single-chain Fv antibody fragment specific for the tumor-associated Ep-CAM molecule was isolated from a semisynthetic phage display library and converted into an intact, fully human IgG1 monoclonal antibody (huMab). The purified huMab had an affinity of 5 nM and effectively mediated tumor cell killing in in vitro and in vivo assays. These experiments show that nonimmunized phage antibody display libraries can be used to obtain high-affinity, functional, and clinically applicable huMabs directed against a tumor-associated antigen.


Assuntos
Anticorpos Monoclonais/química , Antígenos de Neoplasias/imunologia , Antineoplásicos/química , Moléculas de Adesão Celular/imunologia , Neoplasias do Colo/tratamento farmacológico , Fragmentos de Imunoglobulinas/química , Biologia Molecular/métodos , Animais , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Bacteriófagos/genética , Western Blotting , Contagem de Células , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Molécula de Adesão da Célula Epitelial , Citometria de Fluxo , Biblioteca Gênica , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Imuno-Histoquímica , Leucócitos Mononucleares/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neutrófilos/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
20.
Br J Cancer ; 78(11): 1407-16, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9836471

RESUMO

The tumour-associated antigen epithelial glycoprotein-2 (EGP-2) is a promising target for detection and treatment of a variety of human carcinomas. Antibodies to this antigen have been successfully used in patients for imaging of small-cell lung cancer and for adjuvant treatment of minimal residual disease of colon cancer. We describe here the isolation and complete characterization of high-affinity single-chain variable fragments (scFv) to the EGP-2 antigen. First, the binding kinetics of four murine whole antibodies directed to EGP-2 (17-1A, 323/A3, MOC-31 and MOC-161) were determined using surface plasmon resonance (SPR). The MOC-31 antibody has the lowest apparent off-rate, followed by MOC-161 and 323/A3. The V-genes of the two MOC hybridomas were cloned as scFv in a phage display vector and antigen-binding phage were selected by panning on recombinant antigen. The scFvs compete with the original hybridoma antibodies for binding to antigen and specifically bind to human carcinomas in immunohistochemistry. MOC-31 scFv has an off-rate which is better than those of the bivalent 17-1A and 323/A3 whole antibodies, providing it with an essential characteristic for tumour retention in vivo. The availability of these high-affinity anti-EGP-2 antibody fragments and of their encoding V-genes creates a variety of possibilities for their future use as tumour-targeting vehicles.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/imunologia , Moléculas de Adesão Celular/imunologia , Fragmentos de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/metabolismo , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/metabolismo , Sequência de Bases , Biomarcadores Tumorais/metabolismo , Moléculas de Adesão Celular/metabolismo , Ensaio de Imunoadsorção Enzimática , Molécula de Adesão da Célula Epitelial , Genes de Imunoglobulinas , Vetores Genéticos , Humanos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Camundongos , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...