Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Language
Publication year range
1.
J Pept Sci ; 20(11): 850-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044757

ABSTRACT

Accumulation of the COMMD1 protein as a druggable pharmacology event to target cancer cells has not been evaluated so far in cancer animal models. We have previously demonstrated that a second-generation peptide, with cell-penetrating capacity, termed CIGB-552, was able to induce apoptosis mediated by stabilization of COMMD1. Here, we explore the antitumor effect by subcutaneous administration of CIGB-552 in a therapeutic schedule. Outstandingly, a significant delay of tumor growth was observed at 0.2 and 0.7 mg/kg (p < 0.01) or 1.4 mg/kg (p < 0.001) after CIGB-552 administration in both syngeneic murine tumors and patient-derived xenograft models. Furthermore, we evidenced that (131)I-CIGB-552 peptide was actually accumulated in the tumors after administration by subcutaneous route. A typical serine-proteases degradation pattern for CIGB-552 in BALB/c mice serum was identified. Further, biological characterization of the main metabolites of the peptide CIGB-552 suggests that the cell-penetrating capacity plays an important role in the cytotoxic activity. This report is the first in describing the antitumor effect induced by systemic administration of a peptide that targets COMMD1 for stabilization. Moreover, our data reinforce the perspectives of CIGB-552 for cancer targeted therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/pharmacokinetics , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Arthropod Proteins/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/chemistry , Female , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasms, Experimental/pathology , Protein Stability/drug effects , Tissue Distribution , Xenograft Model Antitumor Assays
2.
Plant Biotechnol J ; 11(1): 53-65, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23046448

ABSTRACT

A broad variety of foreign genes can be expressed in transgenic plants, which offer the opportunity for large-scale production of pharmaceutical proteins, such as therapeutic antibodies. Nimotuzumab is a humanized anti-epidermal growth factor receptor (EGFR) recombinant IgG1 antibody approved in different countries for the treatment of head and neck squamous cell carcinoma, paediatric and adult glioma, and nasopharyngeal and oesophageal cancers. Because the antitumour mechanism of nimotuzumab is mainly attributed to its ability to interrupt the signal transduction cascade triggered by EGF/EGFR interaction, we have hypothesized that an aglycosylated form of this antibody, produced by mutating the N(297) position in the IgG(1) Fc region gene, would have similar biochemical and biological properties as the mammalian-cell-produced glycosylated counterpart. In this paper, we report the production and characterization of an aglycosylated form of nimotuzumab in transgenic tobacco plants. The comparison of the plantibody and nimotuzumab in terms of recognition of human EGFR, effect on tyrosine phosphorylation and proliferation in cells in response to EGF, competition with radiolabelled EGF for EGFR, affinity measurements of Fab fragments, pharmacokinetic and biodistribution behaviours in rats and antitumour effects in nude mice bearing human A431 tumours showed that both antibody forms have very similar in vitro and in vivo properties. Our results support the idea that the production of aglycosylated forms of some therapeutic antibodies in transgenic plants is a feasible approach when facing scaling strategies for anticancer immunoglobulins.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/pharmacology , ErbB Receptors/antagonists & inhibitors , Immunoglobulins/biosynthesis , Nicotiana/genetics , Nicotiana/metabolism , Plantibodies/pharmacology , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Molecular Farming/methods , Phosphorylation/drug effects , Plantibodies/metabolism , Plants, Genetically Modified/metabolism , Rats , Rats, Wistar , Recombinant Proteins , Tyrosine/metabolism
3.
J Pept Sci ; 16(1): 40-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19908203

ABSTRACT

Novel therapeutic peptides are increasingly making their way into clinical application. The cationic and amphipathic properties of certain peptides allow them to cross biological membranes in a non-disruptive way without apparent toxicity increasing drug bioavailability. By modifying the primary structure of the Limulus-derived LALF(32-51) peptide we designed a novel peptide, L-2, with antineoplastic effect and cell-penetrating capacity. Interestingly, L-2 induced cellular cytotoxicity in a variety of tumor cell lines and systemic injection into immunocompetent and nude mice bearing established solid tumor, resulted in substantial regression of the tumor mass and apoptosis. To isolate the gene transcripts specifically regulated by L-2 in tumor cells, we conducted suppressive subtractive hybridization (SSH) analysis and identified a set of genes involved in biological processes relevant to cancer biology. Our findings describe a novel peptide that modifies the gene expression of the tumor cells and exhibits antitumor effect in vivo, indicating that peptide L-2 is a potential candidate for anticancer therapy.


Subject(s)
Alanine/chemistry , Antineoplastic Agents/pharmacology , Neoplasms, Experimental/pathology , Peptide Library , Peptides/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis , Cell Cycle , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Molecular Sequence Data , Peptides/chemistry , Peptides/isolation & purification
4.
Mol Cell Biochem ; 316(1-2): 163-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18575815

ABSTRACT

Protein Kinase CK2 is a serine-threonine kinase frequently deregulated in many human tumors. Here, we hypothesized that a peptide binder to the CK2 phosphoacceptor site could exhibit anticancer properties in vitro, in tumor animal models, and in cancer patients. By screening a random cyclic peptide phage display library, we identified the CIGB-300 (formerly P15-Tat), a cyclic peptide which abrogates the CK2 phosphorylation by blocking recombinant substrates in vitro. Interestingly, synthetic CIGB-300 led to a dose-dependent antiproliferative effect in a variety of tumor cell lines and induced apoptosis as evidenced by rapid caspase activation. Importantly, CIGB-300 elicited significant antitumor effect both by local and systemic administration in murine syngenic tumors and human tumors xenografted in nude mice. Finally, we performed a First-in-Man trial with CIGB 300 in patients with cervical malignancies. The peptide was found to be safe and well tolerated in the dose range studied. Likewise, signs of clinical benefit were clearly identified after the CIGB-300 treatment as evidenced by significant decrease of the tumor lesion area and histological examination. Our results provide an early proof-of-principle of clinical benefit by using an anti-CK2 approach in cancer. Furthermore, this is the first clinical trial where an investigational drug has been used to target the CK2 phosphorylation domain.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Casein Kinase II/metabolism , Peptides, Cyclic/pharmacology , Animals , Antineoplastic Agents/adverse effects , Biological Assay , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Peptides, Cyclic/adverse effects , Phosphorylation/drug effects , Proteome/analysis , Xenograft Model Antitumor Assays
5.
Cancer Res ; 64(19): 7127-9, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466209

ABSTRACT

Protein Kinase (casein kinase 2, CK2) is a serine-threonine kinase that is frequently dysregulated in many human tumors. Therefore we hypothesized that peptides capable of binding to the CK2 acidic domain may exhibit potential anticancer properties. By screening a random cyclic peptide phage display library, we have identified a novel peptide, P15, that abrogated CK2 phosphorylation by blocking the substrate in vitro. To verify its potential antineoplastic effect, P15 was fused to the cell-penetrating peptide derived from the HIV-Tat protein. Interestingly, P15-Tat induced apoptosis as evidenced by rapid caspase activation and cellular cytotoxicity in a variety of tumor cell lines. Furthermore, direct injection of P15-Tat into C57BL6 mice bearing day 7-established solid tumors, resulted in substantial regression of the tumor mass. Our findings describe a new proapoptotic cyclic peptide that blocks the CK2 phosphorylation and exhibits antitumor effect in vivo, indicating that the P15 peptide may potentially be used clinically to treat solid tumors or as an adjuvant for cancer therapy.


Subject(s)
Apoptosis/drug effects , Peptides, Cyclic/pharmacology , Protein Serine-Threonine Kinases/metabolism , Animals , Casein Kinase II , Cell Line, Tumor , Gene Products, tat/genetics , Gene Products, tat/pharmacokinetics , Humans , Mice , Mice, Inbred C57BL , Peptide Library , Peptides, Cyclic/genetics , Peptides, Cyclic/pharmacokinetics , Phosphorylation/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology
6.
Cancer Res ; 64(19)Oct. 2004. ilus, tab, graf
Article in English | CUMED | ID: cum-39982

ABSTRACT

Protein Kinase (casein kinase 2, CK2) is a serine-threonine kinase that is frequently dysregulated in many human tumors. Therefore we hypothesized that peptides capable of binding to the CK2 acidic domain may exhibit potential anticancer properties. By screening a random cyclic peptide phage display library, we have identified a novel peptide, P15, that abrogated CK2 phosphorylation by blocking the substrate in vitro. To verify its potential antineoplastic effect, P15 was fused to the cell-penetrating peptide derived from the HIV-Tat protein. Interestingly, P15-Tat induced apoptosis as evidenced by rapid caspase activation and cellular cytotoxicity in a variety of tumor cell lines. Furthermore, direct injection of P15-Tat into C57BL6 mice bearing day 7-established solid tumors, resulted in substantial regression of the tumor mass. Our findings describe a new proapoptotic cyclic peptide that blocks the CK2 phosphorylation and exhibits antitumor effect in vivo, indicating that the P15 peptide may potentially be used clinically to treat solid tumors or as an adjuvant for cancer therapy(AU)


Proteína quinasa (caseína kinasa 2, CK2) es una serina-treonina quinasa que es con frecuencia dysregulated en muchos tumores humanos. Por lo tanto, la hipótesis de que los péptidos capaces de unirse a los ácidos CK2 dominio pueden presentar posibles propiedades anticancerígenas. Por una selección aleatoria péptido cíclico fagos mostrar la biblioteca, hemos identificado un nuevo péptido, P15, que derogó CK2 bloqueando la fosforilación del sustrato in vitro. Para comprobar su potencial efecto antineoplásico, P15 fue fundida a la célula de penetración del péptido derivado de la proteína Tat del VIH. Curiosamente, P15 Tat-como lo demuestra la apoptosis inducida por la rápida activación de caspasas y la citotoxicidad celular en una variedad de líneas de células tumorales. Además, la inyección directa de Tat-P15 en ratones con C57BL6 día 7-establecido los tumores sólidos, dio lugar a la regresión de la masa tumoral. Nuestros resultados describen un nuevo péptido cíclico proapoptóticos que bloquea la fosforilación y exposiciones CK2 efecto antitumoral in vivo, indicando que el péptido P15 puedan ser utilizados clínicamente para el tratamiento de tumores sólidos o como terapia adyuvante para el cáncer


Subject(s)
Animals , Mice , Apoptosis , Peptides, Cyclic/genetics , Peptides, Cyclic/pharmacology , Peptides, Cyclic/pharmacokinetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...