Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Eur J Pharm Biopharm ; 167: 104-113, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34303832

RESUMEN

Serum albumin shows slow clearance from circulation due to neonatal Fc receptor (FcRn)-mediated recycling and has been used for half-life extension. We report here fusions to a high-affinity DARPin, binding to Epithelial Cell Adhesion Molecule (EpCAM). We developed a novel, efficient expression system for such fusion proteins in Pichia pastoris with titers above 300 mg/L of lab-scale shake-flask culture. Since human serum albumin (HSA) does not bind to the murine FcRn, half-lives of therapeutic candidates are frequently measured in human FcRn transgenic mice, limiting useable tumor models. Additionally, serum albumins with extended half-life have been designed. We tested HSA7, motivated by its previously claimed extraordinarily long half-life in mice, which we could not confirm. Instead, we determined a half-life of only 29 h for HSA7, comparable to MSA. The fusion of HSA7 to a DARPin showed a similar half-life. To rationalize these findings, we measured binding kinetics and affinities to murine and human FcRn. Briefly, HSA7 showed affinity to murine FcRn only in the micromolar range, comparable to MSA to its cognate murine FcRn, and an affinity in the nanomolar range only to the human FcRn. This explains the comparable half-life of MSA and HSA7 in mice, while wild-type-HSA has a half-life of only 21 h, as it does not bind the murine FcRn and is not recycled. Thus, HSA-fusions with improved FcRn-affinity, such as HSA7, can be used for preclinical experiments in mice when FcRn transgenes cannot be used, as they reflect better the complex FcRn-mediated recycling and distribution mechanisms.


Asunto(s)
Proteínas de Repetición de Anquirina Diseñadas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Albúmina Sérica/metabolismo , Animales , Femenino , Semivida , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Ratones , Ratones Transgénicos , Receptores Fc/genética , Saccharomycetales/metabolismo , Albúmina Sérica Humana/metabolismo
2.
J Control Release ; 327: 186-197, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32768630

RESUMEN

Despite some approvals of antibody-drug conjugates for cancer therapy, their clinical success rate is unsatisfactory because of very small therapeutic windows, influenced by on-target and off-target toxicities of conjugate and liberated toxin. Additional formats with systematically investigated molecular parameters must therefore be explored to increase their therapeutic window. Here we focused on the effective molecular weight. To generate conjugates with exactly defined drug loads and tunable pharmacokinetics, we used Designed Ankyrin Repeat Proteins (DARPins), fused to unstructured polypeptides of different lengths, to produce proteins with any desired half-life, to identify those with the best efficacy. We generated an EpCAM-targeting DARPin-MMAF conjugate, fused to PAS or XTEN of different lengths, and a matched series of controls of a non-binding DARPin to account for the enhanced permeability and retention (EPR) effect, covering half-lives of minutes to 20.6 h in mice. All conjugates were produced at high purity, and demonstrated high specificity and cytotoxicity in human tumor cell cultures, with IC50 values in the low nM range, independent of the polypeptide type and length. Due to their more facile purification, the PASylated conjugates were tested in nude mice bearing HT29 tumor xenografts. Independent of their size, all PASylated conjugates were very well tolerated after repeated systemic administration of 300 nmol/kg. We found that the conjugates with intermediate size and half-life showed the strongest anti-tumor effects, and deduced that this effect is a compromise of serum half-life and diffusion within the tumor, as on-rates and affinities are essentially identical, with extravasation playing only a very minor role.


Asunto(s)
Neoplasias , Preparaciones Farmacéuticas , Animales , Semivida , Ratones , Ratones Desnudos , Proteínas
3.
Int J Mol Sci ; 21(4)2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32092937

RESUMEN

Ceramide kinase (CerK) is a lipid kinase that converts the proapoptotic ceramide to ceramide 1-phosphate, which has been proposed to have pro-malignant properties and regulate cell responses such as proliferation, migration, and inflammation. We used the parental human breast cancer cell line MDA-MB-231 and two single cell progenies derived from lung and bone metastasis upon injection of the parental cells into immuno-deficient mice. The lung and the bone metastatic cell lines showed a marked upregulation of CerK mRNA and activity when compared to the parental cell line. The metastatic cells also had increased migratory and invasive activity, which was dose-dependently reduced by the selective CerK inhibitor NVP-231. A similar reduction of migration was seen when CerK was stably downregulated with small hairpin RNA (shRNA). Conversely, overexpression of CerK in parental MDA-MB-231 cells enhanced migration, and this effect was also observed in the non-metastatic cell line MCF7 upon CerK overexpression. On the molecular level, CerK overexpression increased the activation of protein kinase Akt. The increased migration of CerK overexpressing cells was mitigated by the CerK inhibitor NVP-231, by inhibition of the phosphoinositide 3-kinase (PI3K)/Akt pathway and the Rho kinase, but not by inhibition of the classical extracellular signal-regulated kinase (ERK) pathway. Altogether, our data demonstrate for the first time that CerK promotes migration and invasion of metastatic breast cancer cells and that targeting of CerK has potential to counteract metastasis in breast cancer.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Movimiento Celular/genética , Neoplasias Pulmonares/metabolismo , Invasividad Neoplásica/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Benzotiazoles/farmacología , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Hidrocarburos Aromáticos con Puentes/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Regulación hacia Arriba
4.
Methods Mol Biol ; 2033: 253-273, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31332759

RESUMEN

For biomedical applications, proteins may require conjugation to small and large molecules. Typical examples are dyes for imaging, cytotoxic effector molecules for cell killing, or half-life extension modules for optimized pharmacokinetics. Although many conjugation strategies are straightforward to apply, most of them do not enable site-specific and orthogonal conjugation, and do not yield a defined stoichiometry. Moreover, techniques offering these desirable features often rely on complex expression procedures and suffer from low production yields. A more promising manufacturing strategy for flexible, site-specific and stoichiometrically defined payloading of proteins is the combination of click chemistry and thiol-maleimide conjugation, which even enables dual labeling when used consecutively. Here, we describe as an example the production of Designed Ankyrin Repeat Proteins (DARPins), a non-IgG binding scaffold, in a specific E. coli strain to obtain high yields of protein carrying both a thiol and an azide group. We provide straightforward protocols for strain-promoted azide-alkyne cycloaddition (SPAAC) and thiol-maleimide conjugation, and furthermore compare these conjugation chemistries with existing alternatives like copper-catalyzed azide-alkyne cycloaddition (CuAAC). Finally, detailed instructions for reactivity analysis and yield estimations of the reactions are provided.


Asunto(s)
Repetición de Anquirina/genética , Química Clic/métodos , Reacción de Cicloadición/métodos , Proteínas Recombinantes/biosíntesis , Alquinos/química , Azidas/química , Catálisis , Colorantes/química , Cobre/química , Escherichia coli/química , Escherichia coli/genética , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Proteínas Recombinantes/genética
5.
J Control Release ; 307: 379-392, 2019 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-31252038

RESUMEN

Alternative non-IgG binding proteins developed for therapy are small in size and, thus, are rapidly cleared from the circulation by renal filtration. To avoid repeated injection or continuous infusion for the maintenance of therapeutic serum concentrations, extensions of unfolded polypeptides have been developed to prolong serum half-life, but systematic, comparative studies investigating the influence of their size and charge on serum half-life, extravasation, tumor localization and excretion mechanisms have so far been lacking. Here we used a high-affinity Designed Ankyrin Repeat Protein (DARPin) targeting the tumor marker epithelial cell adhesion molecule (EpCAM) in a preclinical tumor xenograft model in mice, and fused it with a series of defined unstructured polypeptides. We used three different sizes of two previously described polypeptides, an uncharged one consisting of only Pro, Ala and Ser (termed PAS) and a charged one consisting of Pro, Ala, Ser, Thr, Gly, Glu (termed XTEN) and performed for the first time a precise comparative localization, distribution and extravasation study. Pharmacokinetic analysis showed a clear linear relationship between hydrodynamic radius and serum half-life across both polypeptides, reaching a half-life of up to 21 h in mice. Tumor uptake was EpCAM-dependent and directly proportional to half-life and size, showing an even tumor penetration for all fusion proteins without unspecific accumulation in non-target tissue. Unexpectedly, charge had no influence on any parameter, neither tumor nor tissue accumulation nor kidney elimination kinetics. Thus, both polypeptide types have a very similar potential for precise half-life modification and tumor targeting.


Asunto(s)
Repetición de Anquirina , Molécula de Adhesión Celular Epitelial/metabolismo , Neoplasias/metabolismo , Péptidos/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Animales , Femenino , Ratones , Péptidos/química , Proteínas Recombinantes de Fusión/química , Distribución Tisular
6.
Eur J Pharm Biopharm ; 141: 37-50, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31103742

RESUMEN

Specific cell targeting and efficient intracellular delivery are major hurdles for the widespread therapeutic use of nucleic acid technologies, particularly siRNA mediated gene silencing. To enable receptor-mediated cell-specific targeting, we designed a synthesis scheme that can be generically used to engineer Designed Ankyrin Repeat Protein (DARPin)-siRNA bioconjugates. Different linkers, including labile disulfide-, and more stable thiol-maleimide- and triazole- (click chemistry) tethers were employed. Crosslinkers were first attached to a 3'-terminal aminohexyl chain on the siRNA sense strands. On the protein side thiols of a C-terminal cysteine were used as anchoring sites for disulfide- and thiol-maleimide conjugate formations, while strain-promoted azido-alkyne cycloadditions were carried out at a metabolically introduced N-terminal azidohomoalanine. After establishing efficient purification methods, highly pure products were obtained. Bioconjugates of EpCAM-targeted DARPins with siRNA directed at the luciferase gene were evaluated for cell-specific binding, uptake and gene silencing. As shown by flow cytometry and fluorescence microscopy, all constructs retained the highly specific and high-affinity antigen recognition properties of the native DARPin. As expected, internalization was observed only in EpCAM-positive cell lines, and predominantly endolysosomal localization was detected. Disulfide linked conjugates showed lower serum stability against cleavage at the linker and thus lower internalization into endosomes compared to thiol-maleimide- and triazole-linked conjugates, yet induced more pronounced gene silencing. This indicates that the siRNA payload needs to be liberated from the protein in the endosome. Our data confirm the promise of DARPin-siRNA bioconjugates for tumor targeting, but also identified endosomal retention and limited cytosolic escape of the siRNA as the rate-limiting step for more efficient gene silencing.


Asunto(s)
Endosomas/metabolismo , Silenciador del Gen/fisiología , Proteínas Musculares/metabolismo , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/metabolismo , Alanina/análogos & derivados , Alanina/metabolismo , Línea Celular Tumoral , Química Clic/métodos , Molécula de Adhesión Celular Epitelial/metabolismo , Células HeLa , Humanos , Células MCF-7 , Maleimidas/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Triazoles/metabolismo
7.
Int J Mol Sci ; 19(5)2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29772789

RESUMEN

Sphingosine kinase (SK) catalyses the formation of sphingosine 1-phosphate (S1P), which acts as a key regulator of inflammatory and fibrotic reactions, mainly via S1P receptor activation. Here, we show that in the human renal proximal tubular epithelial cell line HK2, the profibrotic mediator transforming growth factor ß (TGFß) induces SK-1 mRNA and protein expression, and in parallel, it also upregulates the expression of the fibrotic markers connective tissue growth factor (CTGF) and fibronectin. Stable downregulation of SK-1 by RNAi resulted in the increased expression of CTGF, suggesting a suppressive effect of SK-1-derived intracellular S1P in the fibrotic process, which is lost when SK-1 is downregulated. In a further approach, the S1P transporter Spns2, which is known to export S1P and thereby reduces intracellular S1P levels, was stably downregulated in HK2 cells by RNAi. This treatment decreased TGFß-induced CTGF and fibronectin expression, and it abolished the strong induction of the monocyte chemotactic protein 1 (MCP-1) by the pro-inflammatory cytokines tumor necrosis factor (TNF)α and interleukin (IL)-1ß. Moreover, it enhanced the expression of aquaporin 1, which is an important water channel that is expressed in the proximal tubules, and reverted aquaporin 1 downregulation induced by IL-1ß/TNFα. On the other hand, overexpression of a Spns2-GFP construct increased S1P secretion and it resulted in enhanced TGFß-induced CTGF expression. In summary, our data demonstrate that in human renal proximal tubular epithelial cells, SK-1 downregulation accelerates an inflammatory and fibrotic reaction, whereas Spns2 downregulation has an opposite effect. We conclude that Spns2 represents a promising new target for the treatment of tubulointerstitial inflammation and fibrosis.


Asunto(s)
Proteínas de Transporte de Anión/genética , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Túbulos Renales Proximales/metabolismo , Biomarcadores , Células Cultivadas , Regulación hacia Abajo , Células Epiteliales/patología , Fibrosis , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inflamación , Túbulos Renales Proximales/patología , Lisofosfolípidos/metabolismo , Podocitos/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo
8.
Pharmacol Ther ; 185: 34-49, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29127024

RESUMEN

The immunomodulatory drug fingolimod (FTY720, GilenyaR) was approved for oral treatment of relapsing-remitting multiple sclerosis, due to its impressive efficacy and good tolerability. Pharmacologically, it acts as an unselective agonist of sphingosine 1-phosphate receptors (S1PR) and as a selective functional antagonist of the S1P1 subtype by induction of receptor downregulation. Since S1P1 is crucial for the regulation of lymphocyte trafficking, its downregulation causes redistribution of the immune cells to secondary lymphoid tissues, resulting in the depletion from the circulation and hence immunosuppression. Numerous preclinical studies have since been performed with the aim to increase the spectrum of potential indications for fingolimod with emphasis on other autoimmune disorders and diseases associated with inflammation and uncontrolled cell proliferation, including cancer. As an alternative to fingolimod, novel S1PR modulators with a more selective receptor activation profile and improved pharmacokinetic performance and tolerability have also been developed. Preclinical and clinical studies are ongoing to investigate their therapeutic potential. This review discusses the most relevant preclinical and clinical findings from S1PR-targeting and from less-well defined off-target effects reported in the literature, and reveals perspectives for using fingolimod and functionally-related derivatives and new formulations in the management of an increasing number of diseases.


Asunto(s)
Clorhidrato de Fingolimod , Inmunosupresores , Receptores de Lisoesfingolípidos/metabolismo , Animales , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico
9.
PLoS One ; 12(5): e0177244, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28472127

RESUMEN

Teneurins are a family of highly conserved pair-rule proteins involved in morphogenesis and development of the central nervous system. Their function in adult tissues and in disease is largely unknown. Recent evidence suggests a role for dysregulated expression of Teneurins in human tumors, but systematic investigations are missing. Here, we investigated Teneurin-2 and Teneurin-4 expression in various cancer cell lines and in ovarian tumor tissues. Teneurin-2 and Teneurin-4 were expressed in most of the breast cancer cell lines tested. Teneurin-4 was also detected in ovarian cancer cell lines, and throughout ovarian tumors and normal ovary tissue. Ovarian tumors with low Teneurin-4 expression showed less differentiated phenotypes and these patients had shorter mean overall survival. Similarly, Teneurin-2 expression correlated with overall survival as well, especially in patients with serous tumors. In the various cell lines, 5-Aza-cytidine-induced changes in DNA methylation did not alter expression of Teneurin-2 and Teneurin-4, despite the existence of predicted CpG islands in both genes. Interestingly, however, we found evidence for the control of Teneurin-2 expression by the oncogenic growth factor FGF8. Furthermore, we identified multiple transcript splicing variants for Teneurin-2 and Teneurin-4, indicating complex gene expression patterns in malignant cells. Finally, downregulation of Teneurin-4 expression using siRNA caused a cell-type dependent increase in proliferation and resistance to cisplatin. Altogether, our data suggest that low Teneurin-4 expression provides a growth advantage to cancer cells and marks an undifferentiated state characterized by increased drug resistance and clinical aggressiveness. We conclude that Teneurin-2 and Teneurin-4 expression levels could be of prognostic value in ovarian cancer.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Ováricas/patología , Tasa de Supervivencia , Línea Celular Tumoral , Islas de CpG/genética , Metilación de ADN , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Biochim Biophys Acta ; 1861(11): 1840-1851, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27616330

RESUMEN

Breast cancer is one of the most common and devastating malignancies among women worldwide. Recent evidence suggests that malignant progression is also driven by processes involving the sphingolipid molecule sphingosine 1-phosphate (S1P) and its binding to cognate receptor subtypes on the cell surface. To investigate the effect of this interaction on the metastatic phenotype, we used the breast cancer cell line MDA-MB-231 and the sublines 4175 and 1833 derived from lung and bone metastases in nude mice, respectively. In both metastatic cell lines expression of the S1P3 receptor was strongly upregulated compared to the parental cells and correlated with higher S1P-induced intracellular calcium ([Ca2+]i), higher cyclooxygenase (COX)-2 and microsomal prostaglandin (PG) E2 synthase expression, and consequently with increased PGE2 synthesis. PGE2 synthesis was decreased by antagonists and siRNA against S1P3 and S1P2. Moreover, in parental MDA-MB-231 cells overexpression of S1P3 by cDNA transfection also increased PGE2 synthesis, but only after treatment with the DNA methyltransferase inhibitor 5-aza-2-deoxycytidine, indicating reversible silencing of the COX-2 promoter. Functionally, the metastatic sublines showed enhanced migration and Matrigel invasion in adapted Boyden chamber assays, which further increased by S1P stimulation. This response was abrogated by either S1P3 antagonism, COX-2 inhibition or PGE2 receptor 2 (EP2) and 4 (EP4) antagonism, but not by S1P2 antagonism. Our data demonstrate that in breast cancer cells overexpression of S1P3 and its activation by S1P has pro-inflammatory and pro-metastatic potential by inducing COX-2 expression and PGE2 signaling via EP2 and EP4.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Dinoprostona/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Regulación hacia Arriba , Neoplasias de la Mama/genética , Calcio/metabolismo , Celecoxib/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisofosfolípidos/farmacología , Invasividad Neoplásica , Metástasis de la Neoplasia , Prostaglandina-E Sintasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/genética , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/genética , Esfingosina/análogos & derivados , Esfingosina/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
11.
Bioconjug Chem ; 26(11): 2176-85, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26086208

RESUMEN

Antibody-drug conjugates (ADCs) have emerged as a promising class of anticancer agents, combining the specificity of antibodies for tumor targeting and the destructive potential of highly potent drugs as payload. An essential component of these immunoconjugates is a bifunctional linker capable of reacting with the antibody and the payload to assemble a functional entity. Linker design is fundamental, as it must provide high stability in the circulation to prevent premature drug release, but be capable of releasing the active drug inside the target cell upon receptor-mediated endocytosis. Although ADCs have demonstrated an increased therapeutic window, compared to conventional chemotherapy in recent clinical trials, therapeutic success rates are still far from optimal. To explore other regimes of half-life variation and drug conjugation stoichiometries, it is necessary to investigate additional binding proteins which offer access to a wide range of formats, all with molecularly defined drug conjugation. Here, we delineate recent progress with site-specific and biorthogonal conjugation chemistries, and discuss alternative, biophysically more stable protein scaffolds like Designed Ankyrin Repeat Proteins (DARPins), which may provide such additional engineering opportunities for drug conjugates with improved pharmacological performance.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Química Clic/métodos , Reacción de Cicloadición/métodos , Humanos , Modelos Moleculares
12.
Biol Chem ; 396(6-7): 813-25, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25781541

RESUMEN

Both of the sphingosine kinase (SK) subtypes SK-1 and SK-2 catalyze the production of the bioactive lipid molecule sphingosine 1-phosphate (S1P). However, the subtype-specific cellular functions are largely unknown. In this study, we investigated the cellular function of SK-2 in primary mouse renal mesangial cells (mMC) and embryonic fibroblasts (MEF) from wild-type C57BL/6 or SK-2 knockout (SK2ko) mice. We found that SK2ko cells displayed a significantly higher proliferative and migratory activity when compared to wild-type cells, with concomitant increased cellular activities of the classical extracellular signal regulated kinase (ERK) and PI3K/Akt cascades, and of the small G protein RhoA. Furthermore, we detected an upregulation of SK-1 protein and S1P3 receptor mRNA expression in SK-2ko cells. The MEK inhibitor U0126 and the S1P1/3 receptor antagonist VPC23019 blocked the increased migration of SK-2ko cells. Additionally, S1P3ko mesangial cells showed a reduced proliferative behavior and reduced migration rate upon S1P stimulation, suggesting a crucial involvement of the S1P3 receptor. In summary, our data demonstrate that SK-2 exerts suppressive effects on cell growth and migration in renal mesangial cells and fibroblasts, and that therapeutic targeting of SKs for treating proliferative diseases requires subtype-selective inhibitors.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
13.
Bioconjug Chem ; 25(12): 2144-56, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25350699

RESUMEN

Highly potent biotoxins like Pseudomonas exotoxin A (ETA) are attractive payloads for tumor targeting. However, despite replacement of the natural cell-binding domain of ETA by tumor-selective antibodies or alternative binding proteins like designed ankyrin repeat proteins (DARPins) the therapeutic window of such fusion toxins is still limited by target-independent cellular uptake, resulting in toxicity in normal tissues. Furthermore, the strong immunogenicity of the bacterial toxin precludes repeated administration in most patients. Site-specific modification to convert ETA into a prodrug-like toxin which is reactivated specifically in the tumor, and at the same time has a longer circulation half-life and is less immunogenic, is therefore appealing. To engineer a prodrug-like fusion toxin consisting of the anti-EpCAM DARPin Ec1 and a domain I-deleted variant of ETA (ETA″), we used strain-promoted azide alkyne cycloaddition for bioorthogonal conjugation of linear or branched polyethylene glycol (PEG) polymers at defined positions within the toxin moiety. Reversibility of the shielding was provided by a designed peptide linker containing the cleavage site for the rhinovirus 3C model protease. We identified two distinct sites, one within the catalytic domain and one close to the C-terminal KDEL sequence of Ec1-ETA″, simultaneous PEGylation of which resulted in up to 1000-fold lower cytotoxicity in EpCAM-positive tumor cells. Importantly, the potency of the fusion toxin was fully restored by proteolytic unveiling. Upon systemic administration in mice, PEGylated Ec1-ETA″ was much better tolerated than Ec1-ETA″; it showed a longer circulation half-life and an almost 10-fold increased area under the curve (AUC). Our strategy of engineering prodrug-like fusion toxins by bioorthogonal veiling opens new possibilities for targeting tumors with more specificity and efficacy.


Asunto(s)
ADP Ribosa Transferasas/química , Antineoplásicos/química , Antineoplásicos/farmacología , Toxinas Bacterianas/química , Exotoxinas/química , Polietilenglicoles/química , Profármacos/farmacología , Factores de Virulencia/química , Proteasas Virales 3C , Animales , Repetición de Anquirina , Antígenos de Neoplasias/química , Antineoplásicos/farmacocinética , Área Bajo la Curva , Sitios de Unión , Moléculas de Adhesión Celular/química , Línea Celular Tumoral , Química Clic , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Diseño de Fármacos , Molécula de Adhesión Celular Epitelial , Femenino , Semivida , Humanos , Ratones Endogámicos C57BL , Profármacos/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Virales/química , Proteínas Virales/metabolismo , Exotoxina A de Pseudomonas aeruginosa
14.
Br J Pharmacol ; 171(24): 5829-44, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25134723

RESUMEN

BACKGROUND AND PURPOSE: Ceramide kinase (CerK) catalyzes the generation of ceramide-1-phosphate which may regulate various cellular functions, including inflammatory reactions and cell growth. Here, we studied the effect of a recently developed CerK inhibitor, NVP-231, on cancer cell proliferation and viability and investigated the role of cell cycle regulators implicated in these responses. EXPERIMENTAL APPROACH: The breast and lung cancer cell lines MCF-7 and NCI-H358 were treated with increasing concentrations of NVP-231 and DNA synthesis, colony formation and cell death were determined. Flow cytometry was performed to analyse cell cycle distribution of cells and Western blot analysis was used to detect changes in cell cycle regulator expression and activation. KEY RESULTS: In both cell lines, NVP-231 concentration-dependently reduced cell viability, DNA synthesis and colony formation. Moreover it induced apoptosis, as measured by increased DNA fragmentation and caspase-3 and caspase-9 cleavage. Cell cycle analysis revealed that NVP-231 decreased the number of cells in S phase and induced M phase arrest with an increased mitotic index, as determined by increased histone H3 phosphorylation. The effect on the cell cycle was even more pronounced when NVP-231 treatment was combined with staurosporine. Finally, overexpression of CerK protected, whereas down-regulation of CerK with siRNA sensitized, cells for staurosporine-induced apoptosis. CONCLUSIONS AND IMPLICATIONS: Our data demonstrate for the first time a crucial role for CerK in the M phase control in cancer cells and suggest its targeted inhibition, using drugs such as NVP-231, in combination with conventional pro-apoptotic chemotherapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzotiazoles/farmacología , Neoplasias de la Mama , Hidrocarburos Aromáticos con Puentes/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Neoplasias Pulmonares , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Ensayo de Tumor de Célula Madre , Adenocarcinoma , Western Blotting , Supervivencia Celular , ADN/biosíntesis , ADN/efectos de los fármacos , Citometría de Flujo , Humanos , Células MCF-7 , Estaurosporina/farmacología
15.
Cell Physiol Biochem ; 34(1): 119-33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24977486

RESUMEN

BACKGROUND/AIMS: Ceramide kinase (CerK) catalyzes the generation of the sphingolipid ceramide-1-phosphate (C1P) which regulates various cellular functions including cell growth and death, and inflammation. Here, we used a novel catalytic inhibitor of CerK, NVP-231, and CerK knockout cells to investigate the contribution of CerK to proliferation and inflammation in renal mesangial cells and fibroblasts. METHODS: Cells were treated with NVP-231 and [(3)H]-thymidine incorporation into DNA, [(3)H]-arachidonic acid release, prostaglandin E2 (PGE2) synthesis, cell cycle distribution, and apoptosis were determined. RESULTS: Treatment of rat mesangial cells and mouse renal fibroblasts with NVP-231 decreased DNA synthesis, but not of agonist-stimulated arachidonic acid release or PGE2 synthesis. Similarly, proliferation but not arachidonic acid release or PGE2 synthesis was reduced in CERK knockout renal fibroblasts. The anti-proliferative effect of NVP-231 on mesangial cells was due to M phase arrest as determined using the mitosis markers phospho-histone H3, cdc2 and polo-like kinase-1, and induction of apoptosis. Moreover, loss of CerK sensitized cells towards stress-induced apoptosis. CONCLUSIONS: Our data demonstrate that CerK induces proliferation but not PGE2 formation of renal mesangial cells and fibroblasts, and suggest that targeted CerK inhibition has potential for treating mesangioproliferative kidney diseases.


Asunto(s)
Fibroblastos/metabolismo , Riñón/citología , Células Mesangiales/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ácido Araquidónico/metabolismo , Benzotiazoles/farmacología , Hidrocarburos Aromáticos con Puentes/farmacología , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dinoprostona/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Técnicas de Inactivación de Genes , Histonas/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Células Mesangiales/citología , Células Mesangiales/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Quinasa Tipo Polo 1
16.
Mol Cancer Ther ; 13(2): 375-85, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24184633

RESUMEN

Fusion toxins used for cancer-related therapy have demonstrated short circulation half-lives, which impairs tumor localization and, hence, efficacy. Here, we demonstrate that the pharmacokinetics of a fusion toxin composed of a designed ankyrin repeat protein (DARPin) and domain I-truncated Pseudomonas Exotoxin A (PE40/ETA″) can be significantly improved by facile bioorthogonal conjugation with a polyethylene glycol (PEG) polymer at a unique position. Fusion of the anti-EpCAM DARPin Ec1 to ETA″ and expression in methionine-auxotrophic E. coli enabled introduction of the nonnatural amino acid azidohomoalanine (Aha) at position 1 for strain-promoted click PEGylation. PEGylated Ec1-ETA″ was characterized by detailed biochemical analysis, and its potential for tumor targeting was assessed using carcinoma cell lines of various histotypes in vitro, and subcutaneous and orthotopic tumor xenografts in vivo. The mild click reaction resulted in a well-defined mono-PEGylated product, which could be readily purified to homogeneity. Despite an increased hydrodynamic radius resulting from the polymer, the fusion toxin demonstrated high EpCAM-binding activity and retained cytotoxicity in the femtomolar range. Pharmacologic analysis in mice unveiled an almost 6-fold increase in the elimination half-life (14 vs. 82 minutes) and a more than 7-fold increase in the area under the curve (AUC) compared with non-PEGylated Ec1-ETA″, which directly translated in increased and longer-lasting effects on established tumor xenografts. Our data underline the great potential of combining the inherent advantages of the DARPin format with bioorthogonal click chemistry to overcome the limitations of engineering fusion toxins with enhanced efficacy for cancer-related therapy.


Asunto(s)
ADP Ribosa Transferasas/química , Antígenos de Neoplasias/química , Antineoplásicos/química , Toxinas Bacterianas/química , Moléculas de Adhesión Celular/química , Exotoxinas/química , Factores de Virulencia/química , ADP Ribosa Transferasas/genética , Animales , Antígenos de Neoplasias/genética , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Toxinas Bacterianas/genética , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Química Clic , Electroforesis en Gel de Poliacrilamida , Molécula de Adhesión Celular Epitelial , Exotoxinas/genética , Femenino , Células HT29 , Humanos , Células MCF-7 , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Polietilenglicoles/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Resonancia por Plasmón de Superficie , Carga Tumoral/efectos de los fármacos , Factores de Virulencia/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
17.
Mol Carcinog ; 53(7): 578-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23475563

RESUMEN

AMR-Me, a C-28 methylester derivative of triterpenoid compound Amooranin isolated from Amoora rohituka stem bark and the plant has been reported to possess multitude of medicinal properties. Our previous studies have shown that AMR-Me can induce apoptosis through mitochondrial apoptotic and MAPK signaling pathways by regulating the expression of apoptosis related genes in human breast cancer MCF-7 cells. However, the molecular mechanism of AMR-Me induced apoptotic cell death remains unclear. Our results showed that AMR-Me dose-dependently inhibited the proliferation of MCF-7 and MDA-MB-231 cells under serum-free conditions supplemented with 1 nM estrogen (E2) with an IC50 value of 0.15 µM, 0.45 µM, respectively. AMR-Me had minimal effects on human normal breast epithelial MCF-10A + ras and MCF-10A cells with IC50 value of 6 and 6.5 µM, respectively. AMR-Me downregulated PI3K p85, Akt1, and p-Akt in an ERα-independent manner in MCF-7 cells and no change in expression levels of PI3K p85 and Akt were observed in MDA-MB-231 cells treated under similar conditions. The PI3K inhibitor LY294002 suppressed Akt activation similar to AMR-Me and potentiated AMR-Me induced apoptosis in MCF-7 cells. EMSA revealed that AMR-Me inhibited nuclear factor-kappaB (NF-κB) DNA binding activity in MDA-MB-231 cells in a time-dependent manner and abrogated EGF induced NF-κB activation. From these studies we conclude that AMR-Me decreased ERα expression and effectively inhibited Akt phosphorylation in MCF-7 cells and inactivate constitutive nuclear NF-κB and its regulated proteins in MDA-MB-231 cells. Due to this multifactorial effect in hormone-dependent and independent breast cancer cells AMR-Me deserves attention for use in breast cancer prevention and therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , FN-kappa B/antagonistas & inhibidores , Ácido Oleanólico/análogos & derivados , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Factor de Transcripción ReIA/antagonistas & inhibidores , Mama/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/farmacología , Inhibidores Enzimáticos/farmacología , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Mitocondrias/efectos de los fármacos , Morfolinas/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Ácido Oleanólico/farmacología , Proteínas Oncogénicas v-mos/biosíntesis , Fosfatidilinositol 3-Quinasas/biosíntesis , Fosforilación , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-rel , Factor de Transcripción ReIB/antagonistas & inhibidores
18.
Bioconjug Chem ; 24(11): 1955-66, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24168270

RESUMEN

The generation of drug conjugates for safe and effective tumor targeting requires binding proteins tolerant to functionalization by rational engineering. Here, we show that Designed Ankyrin Repeat Proteins (DARPins), a novel class of binding proteins not derived from antibodies, can be used as building blocks for facile orthogonal assembly of bioconjugates for tumor targeting with tailored properties. DARPin Ec1, which targets the Epithelial Cell Adhesion Molecule (EpCAM), was genetically modified with a C-terminal cysteine for conjugation of the small molecule cytotoxin monomethylauristatin F (MMAF). In addition, it was N-terminally functionalized by metabolic introduction of the non-natural amino acid azidohomoalanine to enable linkage of site-specifically dibenzocyclooctyne-modified mouse serum albumin (MSA) for half-life extension using Cu(I)-free click chemistry. The conjugate MSA-Ec1-MMAF was assembled to obtain high yields of a pure and stable drug conjugate as confirmed by various analytical methods and in functional assays. The orthogonality of the assembly led to a defined reaction product and preserved the functional properties of all modules, including EpCAM-specific binding and internalization, FcRn binding mediated by MSA, and cytotoxic potency. Linkage of MMAF to the DARPin increased receptor-specific uptake of the drug while decreasing nonspecific uptake, and further coupling of the conjugate to MSA enhanced this effect. In mice, albumin conjugation increased the serum half-life from 11 min to 17.4 h, resulting in a more than 22-fold increase in the area-under-the-curve (AUC). Our data demonstrate the promise of the DARPin format for facile modular assembly of drug conjugates with improved pharmacokinetic performance for tumor targeting.


Asunto(s)
Repetición de Anquirina , Ancirinas/química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Química Clic , Citotoxinas/química , Citotoxinas/farmacocinética , Albúmina Sérica/química , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Citotoxinas/sangre , Citotoxinas/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Células HT29 , Semivida , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Oligopéptidos/sangre , Oligopéptidos/química , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Relación Estructura-Actividad , Células Tumorales Cultivadas
19.
Expert Opin Drug Deliv ; 10(4): 451-68, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23316711

RESUMEN

INTRODUCTION: The epithelial cell adhesion molecule (EpCAM) is abundantly expressed in epithelial tumors, on cancer stem cells and circulating tumor cells. Together with its role in oncogenic signaling, this has sparked interest in its potential for tumor targeting with antibodies and drug conjugates for safe and effective cancer therapy. Recent advances in protein engineering, linker design and drug formulations have provided a multitude of EpCAM-targeting anticancer agents, several of them with good perspectives for clinical development. AREAS COVERED: This article reviews the biological, therapeutic and technical aspects of EpCAM-targeted drug delivery for cancer therapy. The authors discuss seminal findings, which distinguish EpCAM as a target with oncogenic function and abundant expression in epithelial tumors. Moreover, recent trends in engineering improved anti-EpCAM antibodies, binding proteins that are not derived from immunoglobulins and drug conjugates derived from them are highlighted and their therapeutic potential based on reported preclinical and clinical data, originality of design and perspectives are critically assessed. EXPERT OPINION: EpCAM has shown promise for safe and efficient targeting of solid tumors using antibodies, alternative binding molecules and novel drug conjugates. Among the myriad of EpCAM-targeting drug delivery systems investigated so far, several could demonstrate therapeutic benefit, other formulations engineered to become tailor-made missiles are on the brink.


Asunto(s)
Antineoplásicos/administración & dosificación , Inmunotoxinas/administración & dosificación , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Animales , Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Química Farmacéutica , Sistemas de Liberación de Medicamentos , Molécula de Adhesión Celular Epitelial , Humanos , Neoplasias/metabolismo , Células Neoplásicas Circulantes/metabolismo , Células Madre Neoplásicas/metabolismo
20.
Clin Cancer Res ; 19(1): 96-105, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23172887

RESUMEN

PURPOSE: The phosphoinositide 3-kinase (PI3K) pathway is fundamental for cell proliferation and survival and is frequently altered and activated in neoplasia, including carcinomas of the lung. In this study, we investigated the potential of targeting the catalytic class I(A) PI3K isoforms in small cell lung cancer (SCLC), which is the most aggressive of all lung cancer types. EXPERIMENTAL DESIGN: The expression of PI3K isoforms in patient specimens was analyzed. The effects on SCLC cell survival and downstream signaling were determined following PI3K isoform inhibition by selective inhibitors or downregulation by siRNA. RESULTS: Overexpression of the PI3K isoforms p110-α and p110-ß and the antiapoptotic protein Bcl-2 was shown by immunohistochemistry in primary SCLC tissue samples. Targeting the PI3K p110-α with RNA interference or selective pharmacologic inhibitors resulted in strongly affected cell proliferation of SCLC cells in vitro and in vivo, whereas targeting p110-ß was less effective. Inhibition of p110-α also resulted in increased apoptosis and autophagy, which was accompanied by decreased phosphorylation of Akt and components of the mTOR pathway, such as the ribosomal S6 protein, and the eukaryotic translation initiation factor 4E-binding protein 1. A DNA microarray analysis revealed that p110-α inhibition profoundly affected the balance of pro- and antiapoptotic Bcl-2 family proteins. Finally, p110-α inhibition led to impaired SCLC tumor formation and vascularization in vivo. CONCLUSION: Together our data show the key involvement of the PI3K isoform p110-α in the regulation of multiple tumor-promoting processes in SCLC.


Asunto(s)
Neoplasias Pulmonares/enzimología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Carcinoma Pulmonar de Células Pequeñas/enzimología , Animales , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Embrión de Pollo , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Perfilación de la Expresión Génica , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neovascularización Patológica/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...