Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Virol ; 91(3)2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27795412

RESUMEN

The envelope (Env) glycoprotein of HIV is expressed on the surface of productively infected cells and can be used as a target for cytotoxic immunoconjugates (ICs), in which cell-killing moieties, including toxins, drugs, or radionuclides, are chemically or genetically linked to monoclonal antibodies (MAbs) or other targeting ligands. Such ICs could be used to eliminate persistent reservoirs of HIV infection. We have found that MAbs which bind to the external loop of gp41, e.g., MAb 7B2, make highly effective ICs, particularly when used in combination with soluble CD4. We evaluated the toxicity, immunogenicity, and efficacy of the ICs targeted with 7B2 in mice and in simian-human immunodeficiency virus-infected macaques. In the macaques, we tested immunotoxins (ITs), consisting of protein toxins bound to the targeting agent. ITs were well tolerated and initially efficacious but were ultimately limited by their immunogenicity. In an effort to decrease immunogenicity, we tested different toxic moieties, including recombinant toxins, cytotoxic drugs, and tubulin inhibitors. ICs containing deglycosylated ricin A chain prepared from ricin toxin extracted from castor beans were the most effective in killing HIV-infected cells. Having identified immunogenicity as a major concern, we show that conjugation of IT to polyethylene glycol limits immunogenicity. These studies demonstrate that cytotoxic ICs can target virus-infected cells in vivo but also highlight potential problems to be addressed. IMPORTANCE: It is not yet possible to cure HIV infection. Even after years of fully effective antiviral therapy, a persistent reservoir of virus-infected cells remains. Here we propose that a targeted conjugate consisting of an anti-HIV antibody bound to a toxic moiety could function to kill the HIV-infected cells that constitute this reservoir. We tested this approach in HIV-infected cells grown in the lab and in animal infections. Our studies demonstrated that these immunoconjugates are effective both in vitro and in test animals. In particular, ITs constructed with the deglycosylated A chain prepared from native ricin were the most effective in killing cells, but their utility was blunted because they provoked immune reactions that interfered with the therapeutic effects. We then demonstrated that coating of the ITs with polyethylene glycol minimized the immunogenicity, as has been demonstrated with other protein therapies.


Asunto(s)
Fármacos Anti-VIH/farmacología , Diseño de Fármacos , Proteínas gp160 de Envoltorio del VIH/antagonistas & inhibidores , Inmunoconjugados/farmacología , Animales , Fármacos Anti-VIH/química , Anticuerpos Monoclonales/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas gp160 de Envoltorio del VIH/inmunología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Humanos , Inmunoconjugados/química , Inmunotoxinas/farmacología , Macaca nemestrina , Ratones , Polietilenglicoles/química
2.
Mol Cancer Ther ; 12(6): 979-91, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23493312

RESUMEN

Immunotoxins containing bacterial or plant toxins have shown promise in cancer-targeted therapy, but their long-term clinical use may be hampered by vascular leak syndrome and immunogenicity of the toxin. We incorporated human granzyme B (GrB) as an effector and generated completely human chimeric fusion proteins containing the humanized anti-Her2/neu single-chain antibody 4D5 (designated GrB/4D5). Introduction of a pH-sensitive fusogenic peptide (designated GrB/4D5/26) resulted in comparatively greater specific cytotoxicity although both constructs showed similar affinity to Her2/neu-positive tumor cells. Compared with GrB/4D5, GrB/4D5/26 showed enhanced and long-lasting cellular uptake and improved delivery of GrB to the cytosol of target cells. Treatment with nanomolar concentrations of GrB/4D5/26 resulted in specific cytotoxicity, induction of apoptosis, and efficient downregulation of PI3K/Akt and Ras/ERK pathways. The endogenous presence of the GrB proteinase inhibitor 9 did not impact the response of cells to the fusion construct. Surprisingly, tumor cells resistant to lapatinib or Herceptin, and cells expressing MDR-1 resistant to chemotherapeutic agents showed no cross-resistance to the GrB-based fusion proteins. Administration (intravenous, tail vein) of GrB/4D5/26 to mice bearing BT474 M1 breast tumors resulted in significant tumor suppression. In addition, tumor tissue excised from GrB/4D5/26-treated mice showed excellent delivery of GrB to tumors and a dramatic induction of apoptosis compared with saline treatment. This study clearly showed that the completely human, functionalized GrB construct can effectively target Her2/neu-expressing cells and displays impressive in vitro and in vivo activity. This construct should be evaluated further for clinical use.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Granzimas/administración & dosificación , Receptor ErbB-2/administración & dosificación , Anticuerpos de Cadena Única/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Terapia Molecular Dirigida , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Serina Proteasas/administración & dosificación , Serina Proteasas/genética , Serina Proteasas/inmunología , Transducción de Señal/efectos de los fármacos , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología
3.
J Invest Dermatol ; 133(4): 1052-62, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23190886

RESUMEN

Fibroblast growth factor-inducible protein 14 (Fn14), the cell surface receptor for tumor necrosis factor-like weak inducer of apoptosis (TWEAK), is overexpressed in various human solid tumor types and can be a negative prognostic indicator. We detected Fn14 expression in ∼60% of the melanoma cell lines we tested, including both B-Raf WT and B-Raf(V600E) lines. Tumor tissue microarray analysis indicated that Fn14 expression was low in normal skin, but elevated in 173/190 (92%) of primary melanoma specimens and in 86/150 (58%) of melanoma metastases tested. We generated both a chemical conjugate composed of the recombinant gelonin (rGel) toxin and the anti-Fn14 antibody ITEM-4 (designated ITEM4-rGel) and a humanized, dimeric single-chain antibody of ITEM-4 fused to rGel (designated hSGZ). Both ITEM4-rGel and hSGZ were highly cytotoxic to a panel of different melanoma cell lines. Mechanistic studies showed that both immunotoxins induced melanoma cell necrosis. In addition, these immunotoxins could upregulate the cellular expression of Fn14 and trigger cell-signaling events similar to the Fn14 ligand TWEAK. Finally, treatment of mice bearing human melanoma MDA-MB-435 xenografts with either ITEM4-rGel or hSGZ showed significant tumor growth inhibition compared with controls. We conclude that Fn14 is a therapeutic target in melanoma and the hSGZ construct appears to warrant further development as a therapeutic agent against Fn14-positive melanoma.


Asunto(s)
Factores Inmunológicos/farmacocinética , Inmunotoxinas/farmacocinética , Melanoma/tratamiento farmacológico , Receptores del Factor de Necrosis Tumoral/inmunología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Diseño de Fármacos , Femenino , Humanos , Factores Inmunológicos/genética , Factores Inmunológicos/inmunología , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Unión Proteica/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Neoplasias Cutáneas/inmunología , Receptor de TWEAK , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Biochem Pharmacol ; 84(4): 451-8, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22687624

RESUMEN

Mantle cell lymphoma (MCL) is an incurable, aggressive histo-type of B-cell non-Hodgkin lymphoma associated with both high relapsed rates and relatively short survival. Because MCL over-expresses receptors for B lymphocyte stimulator (BLyS) and displays constitutively active NF-κB, agents targeting these pathways may be of therapeutic relevance in this disease. To investigate the potential clinical use of the rGel/BLyS fusion toxin in combination with bortezomib, we evaluated this fusion toxin for its ability to inhibit MCL growth in severe combined immunodeficiency (SCID) xenograft model. Compared with PBS-treated mice, mice treated with this fusion toxin prolonged both median (84 days vs. 125 days) and overall survival (0% vs. 40%) (p=0.0027). Compared with bortezomib alone-treated mice, mice treated with rGel/BLyS plus bortezomib showed significantly increased median (91 days vs. 158 days) and overall survival (0% vs. 20%) (p=0.0127). Histopathologic analysis of peritoneal intestinal mesentery from MCL-SCID mice showed no demonstrable microscopic lymphomatous involvement at 225 days after treatment with rGel/BLyS. Combination treatment resulted in a synergistic growth inhibition, down-regulation of NF-κB DNA-binding activity, inhibition of cyclin D1, Bcl-x(L), p-Akt, Akt, p-mTOR, and p-Bad, up-regulation of Bax, and induction of cellular apoptosis. Our findings demonstrate that rGel/BLyS is an effective therapeutic agent for both primary and salvage treatment of aggressive MCL expressing constitutively active NF-κB and BLyS receptors and may be an excellent candidate for clinical development.


Asunto(s)
Antineoplásicos/farmacología , Factor Activador de Células B/genética , Linfoma de Células del Manto/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Toxinas Biológicas/farmacología , Animales , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Linfoma de Células del Manto/patología , Ratones , Ratones SCID , FN-kappa B/metabolismo , Trasplante de Neoplasias , Inhibidores de Proteasoma , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Proteínas Recombinantes de Fusión/genética , Transducción de Señal , Toxinas Biológicas/genética , Trasplante Heterólogo
5.
Cancer Lett ; 322(2): 159-68, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22388102

RESUMEN

We generated a fusion protein Bax(345)/BLyS containing the truncated form of Bax (Bax(345)) at the N-terminus followed by a 218 linker to the B lymphocyte stimulator (BLyS). Bax(345)/BLyS was cytotoxic to a panel of diffuse large B cell lymphoma and mantle cell lymphoma lines expressing the BLyS receptors. Specific delivery of Bax(345)/BLyS to malignant B cells drove cells into apoptosis by mitochondrial dysfunction and treatment of cells with Bax(345)/BLyS induced down-regulation of Mcl-1, X-IAP, and survivin. Bax(345)/BLyS represents a new class of targeted therapeutic agents with a unique mechanism of action and may have therapeutic potential for malignant B cells.


Asunto(s)
Factor Activador de Células B/genética , Linfoma de Células B/metabolismo , Mitocondrias/metabolismo , Proteínas Recombinantes de Fusión/toxicidad , Proteína X Asociada a bcl-2/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Citocromos c/metabolismo , Orden Génico , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Concentración 50 Inhibidora , Linfoma de Células B/tratamiento farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
6.
Mol Cancer Ther ; 11(1): 143-53, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22090420

RESUMEN

Recombinant immunotoxins, consisting of single-chain variable fragments (scFv) genetically fused to polypeptide toxins, represent potentially effective candidates for cancer therapeutics. We evaluated the affinity of various anti-Her2/neu scFv fused to recombinant gelonin (rGel) and its effect on antitumor efficacy and off-target toxicity. A series of rGel-based immunotoxins were created from the human anti-Her2/neu scFv C6.5 and various affinity mutants (designated ML3-9, MH3-B1, and B1D3) with affinities ranging from 10(-8) to 10(-11) mol/L. Against Her2/neu-overexpressing tumor cells, immunotoxins with increasing affinity displayed improved internalization and enhanced autophagic cytotoxicity. Targeting indices were highest for the highest affinity B1D3/rGel construct. However, the addition of free Her2/neu extracellular domain (ECD) significantly reduced the cytotoxicity of B1D3/rGel because of immune complex formation. In contrast, ECD addition had little impact on the lower affinity constructs in vitro. In vivo studies against established BT474 M1 xenografts showed growth suppression by all immunotoxins. Surprisingly, therapy with the B1D3-rGel induced significant liver toxicity because of immune complex formation with shed Her2/neu antigen in circulation. The MH3-B1/rGel construct with intermediate affinity showed effective tumor growth inhibition without inducing hepatotoxicity or complex formation. These findings show that while high-affinity constructs can be potent antitumor agents, they may also be associated with mistargeting through the facile formation of complexes with soluble antigen leading to significant off-target toxicity. Constructs composed of intermediate-affinity antibodies are also potent agents that are more resistant to immune complex formation. Therefore, affinity is an exceptionally important consideration when evaluating the design and efficacy of targeted therapeutics.


Asunto(s)
Inmunotoxinas/farmacología , Receptor ErbB-2/inmunología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Anticuerpos de Cadena Única/farmacología , Animales , Afinidad de Anticuerpos , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Humanos , Inmunotoxinas/química , Inmunotoxinas/inmunología , Ratones , Ratones Desnudos , Neoplasias/inmunología , Proteínas Inactivadoras de Ribosomas Tipo 1/inmunología , Anticuerpos de Cadena Única/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Transl Oncol ; 4(6): 350-4, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22190999

RESUMEN

Fullerene (C(60))-monoclonal antibody (mAb) immunoconjugates have been determined to internalize into target cells using water-soluble Gd(3+) ion-filled metallofullerenes (Gd@C(60)[OH](x)). Two separate conjugations of Gd@C(60)(OH)(x) with the antibody ZME-018 and a murine antibody mixture (MuIgG) were performed in a 1:5 mAb/Gd@C(60) ratio. Characterization of the immunoconjugates was established using inductively coupled plasma mass spectrometry (ICP-MS) for Gd(3+) and UV-Vis spectrometry (for Gd@C(60) + C(60)). Once conjugated, enzyme-linked immunosorbent assays showed little change in the specific binding of ZME-018. Each immunoconjugate was exposed to two cancer cell lines, A375m (antigen positive), and T24, bladder carcinoma (antigen negative). Internalization levels of the immunoconjugate were determined at various time points during 24 hours by harvesting and digesting the cells with 70% HNO(3) for Gd(3+) ion analysis by ICP-MS. These results are the first to demonstrate the practicality of a targeted cancer therapy based on fullerene immunotherapy.

8.
Mol Cancer Ther ; 10(7): 1276-88, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21586630

RESUMEN

TNF-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor (FGF)-inducible 14 (Fn14) are a TNF superfamily ligand-receptor pair involved in many cellular processes including proliferation, migration, differentiation, inflammation, and angiogenesis. The Fn14 receptor is expressed at relatively low levels in normal tissues, but it is known to be dramatically elevated in a number of tumor types, including brain and breast tumors. Thus, it seems to be an excellent candidate for therapeutic intervention. We first analyzed Fn14 expression in human tumor cell lines. Fn14 was expressed in a variety of lines including breast, brain, bladder, skin, lung, ovarian, pancreatic, colon, prostate, and cervical cancer cell lines. We then developed an immunoconjugate containing a high-affinity anti-Fn14 monoclonal antibody (ITEM-4) conjugated to recombinant gelonin (rGel), a highly cytotoxic ribosome-inactivating N-glycosidase. Both ITEM-4 and the conjugate were found to bind to cells to an equivalent extent. Confocal microscopic analysis showed that ITEM4-rGel specifically and rapidly (within 2 hours) internalized into Fn14-positive T-24 bladder cancer cells but not into Fn14-deficient mouse embryonic fibroblasts. Cytotoxicity studies against 22 different tumor cell lines showed that ITEM4-rGel was highly cytotoxic to Fn14-expressing cells and was 8- to 8 × 10(4)-fold more potent than free rGel. ITEM4-rGel was found to kill cells by inducing apoptosis with high-mobility group box 1 protein release. Finally, ITEM4-rGel immunoconjugate administration promoted long-term tumor growth suppression in nude mice bearing T-24 human bladder cancer cell xenografts. Our data support the use of an antibody-drug conjugate approach to selectively target and inhibit the growth of Fn14-expressing tumors.


Asunto(s)
Antineoplásicos/farmacología , Inmunoconjugados/metabolismo , Neoplasias/metabolismo , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Animales , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Proteína HMGB1/metabolismo , Células HT29 , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/aislamiento & purificación , Inmunoconjugados/farmacología , Inmunoconjugados/toxicidad , Inyecciones Intravenosas , Células Jurkat , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/patología , Unión Proteica/fisiología , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Receptor de TWEAK , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Biochem Pharmacol ; 80(9): 1335-42, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20654581

RESUMEN

Aberrant signal transducer and activator of transcription (STAT)3 signaling participates in the development and progress of human cancers. We previously generated a highly cytotoxic fusion toxin designated rGel/BLyS for receptor-mediated delivery of the rGel toxin to malignant B-cells. In this study, we examined this fusion toxin for its ability to impact STAT3 signaling in diffuse large B-cell lymphoma (DLBCL). The activated B cell-like DLBCL lines were found to express higher levels of interleukin-6 receptor (IL-6R) and STAT3 than did the germinal center B cell-like DLBCL lines. Treatment of DLBCL cells with rGel/BLyS resulted in down-regulation of IL-6R and inhibited STAT3 phosphorylation, STAT3-DNA binding activity, and IL-6-inducible STAT3 reporter gene activity. In agreement with these results, we additionally found that rGel/BLyS down-regulated levels of several STAT3 targets (c-Myc, p21, Mcl-1, and Bcl-x(L)) and p-SYK, a positive regulator of STAT3. Inhibition of IL-6R-mediated STAT3 signaling by rGel/BLyS led to growth inhibition, triggered accumulation of cells in the sub-G(1) phase of the cell cycle, and induced apoptosis. Our results indicate that rGel/BLyS is an excellent candidate for the treatment of aggressive DLBCL which is resistant to conventional chemotherapeutic regimens and STAT3 signaling pathway may be an attractive therapeutic target for non-Hodgkin's lymphoma.


Asunto(s)
Factor Activador de Células B/farmacología , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Receptores de Interleucina-6/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , ADN/metabolismo , Regulación hacia Abajo , Fase G1/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Quinasa Syk
10.
Neoplasia ; 12(5): 366-75, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20454508

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is an aggressive subtype of B-cell non-Hodgkin lymphoma (NHL) and accounts for 30%to 40%of NHL. Molecules targeting nuclear factor-kappaB (NF-kappaB) are expected to be of therapeutic value in those tumors where NF-kappaB seems to play a unique survival role such as activated B-cell (ABC)-subtype DLBCL. We previously generated a rGel/BLyS fusion toxin for receptor-mediated delivery of the rGel toxin specifically to malignant B cells. In this study, we examined this fusion toxin for its ability to suppress DLBCL growth in vitro and in vivo. rGel/BLyS was specifically cytotoxic to DLBCL lines expressing all three BLyS receptors and constitutively active NF-kappaB. Treatment with rGel/BLyS induced down-regulation of the phosphorylation of inhibitory subunit of NF-kappaB (IkappaB-alpha), inhibition of NF-kappaB DNA-binding activity, and accumulation of IkappaB-alpha. In agreement with these results, we additionally found that rGel/BLyS downregulated levels of several NF-kappaB targets including Bcl-xL, Mcl-1, survivin, and x-chromosome linked inhibitor-of-apoptosis. Treatment also induced up-regulation of Bax and apoptosis through caspase-3 activation and poly ADP-ribose polymerase cleavage. Importantly, rGel/BLyS significantly inhibited tumor growth (P < .05) in a DLBCL xenograft model. Thus, our results indicate that rGel/BLyS is an excellent candidate for the treatment of aggressive NHLs that are both dependent on NF-kappaB and are resistant to conventional chemotherapeutic regimens.


Asunto(s)
Antineoplásicos/administración & dosificación , Factor Activador de Células B/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Toxinas Biológicas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Factor Activador de Células B/genética , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Separación Celular , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Etiquetado Corte-Fin in Situ , Linfoma de Células B Grandes Difuso/metabolismo , Masculino , Ratones , Ratones SCID , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Toxinas Biológicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Res ; 69(23): 8987-95, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19934334

RESUMEN

The goal of this study was to characterize a series of anti-Her2/neu immunotoxin constructs to identify how different antibodies and linker choices affect the specificity and cytotoxicity of these proteins. We constructed a series of immunotoxins containing either the human single-chain antibody (scFv) C6.5 or the murine scFv e23 fused to the highly toxic recombinant gelonin (rGel) molecule. Based on the flexible GGGGS linker (L), the fusion construct C6.5-L-rGel was compared with e23-L-rGel to evaluate the specific cytotoxic effects against Her2/neu-positive and Her2/neu-negative tumor cells. Both constructs retained the specificity of the original antibody as well as the biological activity of rGel toxin. The two constructs displayed similar cytotoxicity against different carcinoma cells. We additionally introduced the modified linkers TRHRQPRGWEQL (Fpe) and AGNRVRRSVG (Fdt), which contained furin cleavage sites, to determine the effect of these design changes on stability and cell killing efficiency. The introduction of furin cleavage linkers (Fpe or Fdt) into the molecules resulted in dissimilar sensitivity to protease cleavage compared with the constructs containing the L linker, but very similar intracellular rGel release, cytotoxic kinetics, and induction of autophagic cell death in vitro. Xenograft studies with SKOV3 ovarian tumors were done using various C6.5/rGel constructs. C6.5-L-rGel was more efficient in tumor inhibition than constructs containing furin linkers, attributing to a higher stability in vivo of the L version. Therefore, our studies suggest that human C6.5-L-rGel may be an effective novel clinical agent for therapy of patients with Her2/neu-overexpressing malignancies.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Inmunotoxinas/farmacología , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Secuencia de Aminoácidos , Animales , Antineoplásicos Fitogénicos/química , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Femenino , Furina/farmacología , Humanos , Fragmentos de Inmunoglobulinas/biosíntesis , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/inmunología , Inmunotoxinas/química , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligopéptidos/química , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Receptor ErbB-2/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Cancer Ther ; 6(2): 460-70, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17267661

RESUMEN

B lymphocyte stimulator (BLyS) is crucial for B-cell survival, and the biological effects of BLyS are mediated by three cell surface receptors designated B cell-activating factor receptor (BAFF-R), transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI), and B-cell maturation antibody (BCMA). Increased expression of BLyS and its receptors has been identified in numerous B-cell malignancies. We generated a fusion toxin designated rGel/BLyS for receptor-mediated delivery of the recombinant gelonin (rGel) toxin to neoplastic B cells, and we characterized its activity against various B-cell tumor lines. Three mantle cell lymphoma (MCL) cell lines (JeKo-1, Mino, and SP53) and two diffuse large B-cell lymphoma (DLBCL) cell lines (SUDHL-6 and OCI-Ly3) expressing all three distinct BLyS receptors were found to be the most sensitive to the fusion toxin (IC(50) = 2-5 pmol/L and 0.001-5 nmol/L for MCL and DLBCL, respectively). The rGel/BLyS fusion toxin showed specific binding to cells expressing BLyS receptors and rapid internalization of the rGel component into target cells. The cytotoxic effects of rGel/BLyS were inhibited by pretreatment with free BLyS or with soluble BAFF-R, TACI, and BCMA decoy receptors. This suggests that the cytotoxic effects of the fusion toxin are mediated through BLyS receptors. The rGel/BLyS fusion toxin inhibited MCL cell growth through induction of apoptosis associated with caspase-3 activation and poly (ADP-ribose) polymerase cleavage. Our results suggest that BLyS has the potential to serve as an excellent targeting ligand for the specific delivery of cytotoxic molecules to neoplastic B cells expressing the BLyS receptors, and that the rGel/BLyS fusion toxin may be an excellent candidate for the treatment of B-cell malignancies especially MCL and DLBCL.


Asunto(s)
Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/metabolismo , Linfocitos B/efectos de los fármacos , Proteínas de Plantas/genética , Proteínas Recombinantes de Fusión/farmacología , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Apoptosis/efectos de los fármacos , Receptor del Factor Activador de Células B/genética , Antígeno de Maduración de Linfocitos B/genética , Linfocitos B/metabolismo , Western Blotting , Humanos , Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Linfoma/patología , ARN Mensajero/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1 , Toxinas Biológicas/farmacología , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Células Tumorales Cultivadas
14.
Cancer Res ; 66(12): 5989-92, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778167

RESUMEN

We have used RNA aptamer:gelonin conjugates to target and specifically destroy cells overexpressing the known cancer biomarker prostate-specific membrane antigen (PSMA). Aptamer:toxin conjugates have an IC50 of 27 nmol/L and display an increased potency of at least 600-fold relative to cells that do not express PSMA. The aptamer not only promotes uptake into target cells but also decreases the toxicity of gelonin in non-target cells. These results validate the notion that "escort aptamers" may be useful for the treatment of specific tumors expressing unique antigen targets.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Aptámeros de Nucleótidos/genética , Proteínas de Plantas/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Antineoplásicos Fitogénicos/farmacocinética , Aptámeros de Nucleótidos/farmacocinética , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Humanos , Concentración 50 Inhibidora , Masculino , Proteínas de Plantas/farmacocinética , Antígeno Prostático Específico/biosíntesis , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1
15.
J Phys Chem B ; 109(12): 5482-4, 2005 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16851586

RESUMEN

The physisorption of radiolabeled (125)I(-) ions from aqueous solution and the Brunauer-Emmett-Teller (BET) surface areas of various carbonaceous materials [HiPco single-wall carbon nanotubes (SWNTs), F-SWNTs, cut SWNTs, charcoal, graphite, F-graphite and C(60)] have been measured and compared. By far, cut SWNTs (mainly 20-50 nm lengths) displayed the largest surface area of the materials (1180 m(2).g(-1)), being approximately double that of uncut SWNT and charcoal. At low concentrations of (125)I(-), nearly all of the (125)I(-) was adsorbed from aqueous solution within 1 min at room temperature by the cut SWNTs, uncut SWNTs, and charcoal; the other materials showed much less adsorption under the same conditions. Once adsorbed, the (125)I(-) wash-off rate by pure water was highly variable but was especially slow for cut SWNTs (t(1/2) approximately 2720 h) compared to the other materials; wash-off of (125)I(-) by an aqueous H(2)O(2) solution was even slower (t(1/2) approximately 14 300 h). Taken together, these data demonstrate the greatly increased surface area and dramatically enhanced retention properties of cut SWNTs over uncut SWNTs.


Asunto(s)
Radioisótopos de Yodo/química , Nanotubos de Carbono/química , Radioisótopos/química , Adsorción , Carbón Orgánico/química , Peróxido de Hidrógeno/química , Yoduro de Sodio/química , Soluciones/química , Agua/química
16.
Mol Cancer Ther ; 3(3): 345-52, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15026555

RESUMEN

A major obstacle in the successful delivery of antibody-based therapeutics to tumor cells is the heterogeneity of target antigen expression. We reported previously that retinoic acid (RA) is a potent and selective inducer of the cell-surface antigen CD38 in myeloid leukemia cells. The purpose of this study was to determine whether the RA-induced CD38 antigen could be a target for an anti-CD38-based immunotoxin to induce selective killing of leukemia cells. The combination of RA and the anti-CD38 gelonin immunotoxin induced a synergistic killing of leukemia cells. Thus, coculture of myeloid leukemia cells and cell lines with as little as 1 nM RA in the presence of immunotoxin induced substantial killing (>90%) of leukemia cell clones. More importantly, the blasts of myeloid leukemia patients, irrespective of their morphological and phenotypic features, also responded to the RA and immunotoxin combination when cultured ex vivo. A similar synergistic effect between RA and immunotoxin was observed against a multidrug-resistant variant subline of HL-60 cells. However, another variant of HL-60 cells, HL-60R, in which the retinoid receptor function has been abrogated by a trans-dominant-negative mutation, exhibited complete resistance to the immunotoxin-induced killing effect in the presence or absence of RA. Our results suggest that RA combined with anti-CD38-based therapeutic agent may offer exciting opportunities for the treatment of myeloid leukemias despite their multiplicity of genetic and clinical varieties.


Asunto(s)
ADP-Ribosil Ciclasa/biosíntesis , Antígenos CD/biosíntesis , Antineoplásicos/farmacología , Inmunoterapia/métodos , Inmunotoxinas/metabolismo , Inmunotoxinas/uso terapéutico , Leucemia/terapia , Tretinoina/farmacología , ADP-Ribosil Ciclasa/uso terapéutico , ADP-Ribosil Ciclasa 1 , Anticuerpos Monoclonales/química , Antígenos CD/uso terapéutico , Línea Celular Tumoral , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Múltiples Medicamentos , Células HL-60 , Humanos , Inmunotoxinas/química , Glicoproteínas de Membrana , Mutación , Proteínas de Plantas/química , Proteínas Inactivadoras de Ribosomas Tipo 1 , Tretinoina/metabolismo
17.
Int J Cancer ; 108(4): 549-57, 2004 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-14696119

RESUMEN

Fusion constructs targeting tumor cells have significant potential applications against both solid tumors and hematologic malignancies. We developed a fusion construct of tumor necrosis factor (TNF) and a single-chain antibody (scFvMEL) recognizing the gp240 antigen on human melanoma cells. The scFvMEL/TNF construct, like TNF itself, was found to exist in solution primarily as a trimer of 45 kDa monomers (trimeric molecular weight = 135 kDa). The fusion construct bound specifically to gp240 antigen-positive but not to antigen-negative cells. The TNF component of the construct was biologically active (specific activity = 1 x 10(7) U/mg) compared with free TNF (specific activity = 2.6 x 10(7) U/mg) and was more cytotoxic to antigen-positive A375-M melanoma cells (IC(50) = 100 pM) than TNF alone (IC(50) = 1,000 pM) and, additionally, was active against AAB-527 melanoma cells (IC(50) = 20 nM) resistant to TNF itself (IC(50) > 1,000 nM). The augmented cytotoxicity was mediated by antibody-specific binding to the cell surface. Both A375-M and AAB-527 cells were shown to express TNFR1 and TNFR2 on the cell surface. The TNF moiety of the fusion construct was efficiently delivered into cells in time-dependent increase in cytosol as assessed by immunofluorescent staining of human melanoma cells. Radiolabeled scFvMEL/TNF localized effectively in human melanoma xenografts in nude (nu/nu) mice with a tumor:blood ratio of approximately 8 at 72 hr after administration. Our studies suggest that because of its unique biologic activity and low antigenic potential, scFvMEL/TNF makes an excellent cytotoxic protein for potential clinical treatment of human melanoma.


Asunto(s)
Anticuerpos Antineoplásicos/genética , Inmunotoxinas/administración & dosificación , Melanoma Experimental/tratamiento farmacológico , Proteoglicanos/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Factor de Necrosis Tumoral alfa/genética , Animales , Antígenos de Neoplasias/inmunología , Sistemas de Liberación de Medicamentos , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Expresión Génica , Humanos , Fragmentos de Inmunoglobulinas/genética , Inmunotoxinas/aislamiento & purificación , Melanoma Experimental/inmunología , Ratones , Ratones Desnudos , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Distribución Tisular , Células Tumorales Cultivadas
18.
Cancer Res ; 63(14): 3995-4002, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12873997

RESUMEN

We constructed a single-chain anti-gp240 antibody (designated MEL sFv) and fused this to the recombinant toxin gelonin (rGel). MEL sFv-rGel was produced in bacterial expression plasmid (pET-32), and the protein composition was confirmed by both DNA sequencing and Western analysis. Inhibition of cell-free protein synthesis by the fusion construct demonstrated an IC(50) of 100 pM, comparable with that for native gelonin (104 pM). The MEL sFv-rGel fusion toxin bound to antigen-positive but not antigen-negative cells as assessed by ELISA. Internalization into A-375 target cells was demonstrable by 1 h after exposure. Against A-375 cells, MEL sFv-rGel demonstrated an IC(50) of approximately 8 nM, which was 250-fold lower than that for free rGel (2000 nM). The cytotoxic effects of the construct did not involve apoptosis because terminal deoxynucleotidyl transferase-mediated nick end labeling assays of treated cells were negative. (125)I-labeled MEL sFv-rGel demonstrated biphasic clearance of the construct from plasma (t(1/2) alpha and t(1/2) beta were 0.46 and 7.2 h, respectively). At 72 h after administration, xenograft studies showed that the tissue:blood ratio was highest for tumor followed by spleen, kidney, and liver. Groups of tumor-bearing nude mice were treated with fusion toxin at either 2 or 20 mg/kg. Compared with saline-treated controls, for which mean tumor burden increased 6-fold, the groups treated with the high and low doses of fusion construct showed no increase or only a 2-fold increase, respectively. These studies suggest that this recombinant fusion construct has potent cytotoxic activity both in vitro and in vivo and is an excellent candidate for clinical development.


Asunto(s)
Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Inmunotoxinas/química , Inmunotoxinas/farmacología , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Proteínas de Plantas/química , Proteínas de Plantas/farmacología , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/metabolismo , Apoptosis/efectos de los fármacos , Secuencia de Bases , Citotoxicidad Inmunológica , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Humanos , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/aislamiento & purificación , Fragmentos de Inmunoglobulinas/farmacología , Inmunotoxinas/genética , Inmunotoxinas/aislamiento & purificación , Etiquetado Corte-Fin in Situ , Radioisótopos de Yodo , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plásmidos/genética , Inhibidores de la Síntesis de la Proteína/química , Inhibidores de la Síntesis de la Proteína/aislamiento & purificación , Inhibidores de la Síntesis de la Proteína/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Inactivadoras de Ribosomas Tipo 1 , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Proc Natl Acad Sci U S A ; 99(12): 7866-71, 2002 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-12060733

RESUMEN

Vascular endothelial growth factor (VEGF) plays a key role in the growth and metastasis of solid tumors. We generated a fusion protein containing VEGF(121) linked by a flexible G(4)S tether to the toxin gelonin (rGel) and expressed this as a soluble protein in bacteria. Purified VEGF(121)/rGel migrated as an 84-kDa homodimer under nonreducing conditions. VEGF(121)/rGel bound to purified, immobilized Flk-1, and the binding was competed by VEGF(121). Both VEGF(121)/rGel and VEGF(121) stimulated cellular kinase insert domain receptor (KDR) phosphorylation. The VEGF(121)/rGel fusion construct was highly cytotoxic to endothelial cells overexpressing the KDR/Flk-1 receptor. The IC(50) of the construct on dividing endothelial cells expressing 10(5) or more KDR/Flk-1 receptors per cell was 0.5-1 nM, as compared with 300 nM for rGel itself. Dividing endothelial cells overexpressing KDR were approximately 60-fold more sensitive to VEGF(121)/rGel than were nondividing cells. Endothelial cells overexpressing FLT-1 were not sensitive to the fusion protein. Human melanoma (A-375) or human prostate (PC-3) xenografts treated with the fusion construct demonstrated a reduction in tumor volume to 16% of untreated controls. The fusion construct localized selectively to PC-3 tumor vessels and caused thrombotic damage to tumor vessels with extravasation of red blood cells into the tumor bed. These studies demonstrate the successful use of VEGF(121)/rGel fusion construct for the targeted destruction of tumor vasculature in vivo.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Factores de Crecimiento Endotelial , Endotelio Vascular/fisiología , Linfocinas , Proteínas de Plantas , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Aorta Torácica , Secuencia de Bases , Bovinos , Circulación Cerebrovascular/efectos de los fármacos , Cartilla de ADN , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Ratones , Reacción en Cadena de la Polimerasa , Conejos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/aislamiento & purificación , Reticulocitos/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1 , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA