Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Hum Vaccin Immunother ; 13(11): 2548-2560, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29083947

ABSTRACT

The protein composition of an Outer Membrane Vesicle (OMV) preparation that constitutes the active pharmaceutical ingredient of VA-MENGOC-BC®, an effective vaccine against Neisseria meningitidis serogroups B, and C is presented. This preparation has a high lipid content and five abundant membrane proteins (FetA, PorA, PorB, RmpM, and Opc), constituting approximately 70% of the total protein mass. The protein composition was determined by combining the use of the Hexapeptide Ligand Library and an orthogonal tandem fractionation of tryptic peptides by reverse-phase chromatography at alkaline and acid pH. This approach equalizes the concentration of tryptic peptides derived from low- and high-abundance proteins as well as considerably simplifying the number of peptides analyzed by LC-MS/MS, enhancing the possibility of identifying low-abundance species. Fifty-one percent of the proteins originally annotated as membrane proteins in the genome of the MC58 strain were identified. One hundred and sixty-eight low-abundance cytosolic proteins presumably occluded within OMV were also identified. Four (NadA, NUbp, GNA2091, and fHbp), out of the five antigens constituting the Bexsero® vaccine, were detected in this OMV preparation. In particular, fHbp is also the active principle of the Trumenba® vaccine developed by Pfizer. The HpuA and HpuB gene products (not annotated in the MC58 genome) were identified in the CU385 strain, a clinical isolate that is used to produce this OMV. Considering the proteins identified here and previous work done by our group, the protein catalogue of this OMV preparation was extended to 266 different protein species.


Subject(s)
Meningococcal Vaccines/chemistry , Neisseria meningitidis, Serogroup B/immunology , Neisseria meningitidis/immunology , Africa/epidemiology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Brazil/epidemiology , Chromatography, Liquid , Congresses as Topic , Disease Outbreaks/prevention & control , Epidemiological Monitoring , Health Planning Guidelines , Humans , Meningococcal Infections/epidemiology , Meningococcal Infections/immunology , Meningococcal Infections/prevention & control , Neisseria meningitidis/genetics , Neisseria meningitidis, Serogroup B/genetics , Serogroup , Tandem Mass Spectrometry , Vaccination , Vaccine Potency , Whole Genome Sequencing
2.
MEDICC Rev ; 15(1): 11-5, 2013 01.
Article in English | MEDLINE | ID: mdl-23396236

ABSTRACT

Diabetic foot ulcer is a principal diabetic complication. It has been shown that diabetic patients have decreased growth factor concentrations in their tissues, particularly epidermal growth factor. Growth factor shortage impairs wound healing, which leads to chronic nonhealing wounds and sometimes eventual amputation. Ischemic diabetic foot ulcer is the most difficult to treat and confers the highest amputation risk. Injecting epidermal growth factor deep into the wound bottom and contours encourages a more effective pharmacodynamic response in terms of granulation tissue growth and wound closure. Epidermal growth factor injected into the ulcer matrix may also result in association with extracellular matrix proteins, thus enhancing cell proliferation and migration. Heberprot-P is an innovative Cuban product containing recombinant human epidermal growth factor for peri- and intra-lesional infiltration; evidence reveals it accelerates healing of deep and complex ulcers, both ischemic and neuropathic, and reduces diabetes-related amputations. Clinical trials of Heberprot-P in patients with diabetic foot ulcers have shown that repeated local infiltration of this product can enhance healing of chronic wounds safely and efficaciously. As a result, Heberprot-P was registered in Cuba in 2006, and in 2007 was included in the National Basic Medications List and approved for marketing. It has been registered in 15 other countries, enabling treatment of more than 100,000 patients. Heberprot-P is a unique therapy for the most complicated and recalcitrant chronic wounds usually associated with high amputation risk. Local injection in complex diabetic wounds has demonstrated a favorable risk-benefit ratio by speeding healing, reducing recurrences and attenuating amputation risk. Further testing and deployment worldwide of Heberprot-P would provide an opportunity to assess the product's potential to address an important unmet medical need.


Subject(s)
Diabetic Foot/drug therapy , Epidermal Growth Factor/administration & dosage , Amputation, Surgical , Clinical Trials as Topic , Cuba , Diabetic Foot/pathology , Humans , Recombinant Proteins/administration & dosage , Severity of Illness Index , Wound Healing/drug effects
3.
J Pept Sci ; 18(4): 215-23, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22407768

ABSTRACT

CIGB-300 is a novel anticancer peptide that impairs the casein kinase 2-mediated phosphorylation by direct binding to the conserved phosphoacceptor site on their substrates. Previous findings indicated that CIGB-300 inhibits tumor cell proliferation in vitro and induces tumor growth delay in vivo in cancer animal models. Interestingly, we had previously demonstrated that the putative oncogene B23/nucleophosmin (NPM) is the major intracellular target for CIGB-300 in a sensitive human lung cancer cell line. However, the ability of this peptide to target B23/NPM in cancer cells with differential CIGB-300 response phenotype remained to be determined. Interestingly, in this work, we evidenced that CIGB-300's antiproliferative activity on tumor cells strongly correlates with its nucleolar localization, the main subcellular localization of the previously identified B23/NPM target. Likewise, using CIGB-300 equipotent doses (concentration that inhibits 50% of proliferation), we demonstrated that this peptide interacts and inhibits B23/NPM phosphorylation in different cancer cell lines as evidenced by in vivo pull-down and metabolic labeling experiments. Moreover, such inhibition was followed by a fast apoptosis on CIGB-300-treated cells and also an impairment of cell cycle progression mainly after 5 h of treatment. Altogether, our data not only validates B23/NPM as a main target for CIGB-300 in cancer cells but also provides the first experimental clues to explain their differential antiproliferative response. Importantly, our findings suggest that further improvements to this cell penetrating peptide-based drug should entail its more efficient intracellular delivery at such subcellular localization.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Nucleolus/drug effects , Peptides, Cyclic/pharmacology , Antineoplastic Agents/metabolism , Apoptosis , Casein Kinase II/antagonists & inhibitors , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Nuclear Proteins/metabolism , Nucleophosmin , Peptides, Cyclic/metabolism , Phosphorylation , Protein Processing, Post-Translational/drug effects
4.
Mol Cell Biochem ; 356(1-2): 45-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21735096

ABSTRACT

CK2 represents an oncology target scientifically validated. However, clinical research with inhibitors of the CK2-mediated phosphorylation event is still insufficient to recognize it as a clinically validated target. CIGB-300, an investigational peptide-based drug that targets the phosphoaceptor site, binds to a CK2 substrate array in vitro but mainly to B23/nucleophosmin in vivo. The CIGB-300 proapoptotic effect is preceded by its nucleolar localization, inhibition of the CK2-mediated phosphorylation on B23/nucleophosmin and nucleolar disassembly. Importantly, CIGB-300 shifted a protein array linked to apoptosis, ribosome biogenesis, cell proliferation, glycolisis, and cell motility in proteomic studies which helped to understand its mechanism of action. In the clinical ground, CIGB-300 has proved to be safe and well tolerated in a First-in-Human trial in women with cervical malignancies who also experienced signs of clinical benefit. In a second Phase 1 clinical trial in women with cervical cancer stage IB2/II, the MTD and DLT have been also identified in the clinical setting. Interestingly, in cervical tumors the B23/nucleophosmin protein levels were significantly reduced after CIGB-300 treatment at the nucleus compartment. In addition, expanded use of CIGB-300 in case studies has evidenced antitumor activity when administered as compassional option. Collectively, our data outline important clues on translational and clinical research from this novel peptide-based drug reinforcing its perspectives to treat cancer and paving the way to validate CK2 as a promising target in oncology.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Casein Kinase II/chemistry , Peptides, Cyclic/pharmacology , Translational Research, Biomedical , Casein Kinase II/metabolism , Cell Line, Tumor , Female , Humans , Magnetic Resonance Imaging , Male , Nuclear Proteins/metabolism , Nucleophosmin , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary
5.
J Proteome Res ; 9(10): 5473-83, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20804217

ABSTRACT

CIGB-300 is a proapoptotic peptide-based drug that abrogates the CK2-mediated phosphorylation. This peptide has antineoplastic effect on lung cancer cells in vitro and in vivo. To understand the mechanisms involved on such anticancer activity, the NCI-H125 cell line proteomic profile after short-term incubation (45 min) with CIGB-300 was investigated. As determined by 2-DE or 2D-LC-MS/MS, 137 proteins changed their abundances more than 2-fold in response to the CIGB-300 treatment. The expression levels of proteins related to ribosome biogenesis, metastasis, cell survival and proliferation, apoptosis, and drug resistance were significantly modulated by the presence of CIGB-300. The protein translation process was the most affected (23% of the identified proteins). From the proteome analysis of the NCI-H125 cell line, novel potentialities for CIGB-300 as anticancer agent were evidenced.


Subject(s)
Peptides, Cyclic/pharmacology , Protein Biosynthesis/drug effects , Proteome/analysis , Proteomics/methods , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatography, Liquid , Cluster Analysis , Electrophoresis, Gel, Two-Dimensional , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mass Spectrometry , Proteome/classification
6.
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
7.
J Gene Med ; 12(1): 107-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19866482

ABSTRACT

BACKGROUND: In the present study, we evaluated the safety of CIGB-230, a novel vaccine candidate based on the mixture of a plasmid for DNA immunization, expressing hepatitis C virus (HCV) structural antigens, with a recombinant HCV Core protein. METHODS: Fifteen HCV chronically-infected volunteers with detectable levels of HCV RNA genotype 1b, who were nonresponders to previous treatment with interferon plus ribavirin, were intramuscularly injected with CIGB-230 on weeks 0, 4, 8, 12, 16 and 20. Individuals were also immunized at weeks 28, 32 and 36 with a recombinant vaccine against hepatitis B. Adverse events were recorded and analyzed. Blood samples were taken every 4 weeks up to month 12 for hematological, biochemical, virological and immunological analysis. RESULTS: All patients completed the treatment with CIGB-230. Adverse events were only slight (83.6%) or moderate (16.4%). No significant differences in hematological and biochemical parameters, including serum aminotransferases, were detected between the baseline and post-treatment state. Induction of a CD4+ T lymphocyte response against a particular region in HCV E1, spanning amino acids 230-312 in HCV polyprotein, was detected in 42.8% of patients during treatment with CIGB-230. The ability of T cells to proliferate in response to mitogenic stimulation was not weakened. Most individuals (78.6%) were seroprotected after anti-hepatitis B vaccination and 42.8% were hyper-responders (antibody titers > 100 UI/ml). No anti-mitochondrial, anti-nuclear and anti-extractable nuclear antigen antibodies were generated during immunization with CIGB-230. CONCLUSIONS: Vaccination with CIGB-230 in HCV chronically-infected individuals was safe, well tolerated and did not impair the ability to respond to non-HCV antigens.


Subject(s)
Hepatitis B Vaccines/immunology , Hepatitis B/prevention & control , Hepatitis C, Chronic/prevention & control , Immunity/immunology , Vaccination/adverse effects , Vaccines, DNA/immunology , Adult , Antibodies, Antinuclear/immunology , Antibody Formation/drug effects , Antibody Formation/immunology , Cell Proliferation/drug effects , Epitopes/immunology , Female , Hepatitis B/immunology , Hepatitis B Antibodies/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis C, Chronic/immunology , Humans , Immunity/drug effects , Male , Middle Aged , Mitogens/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology
8.
Mol Cancer Ther ; 8(5): 1189-96, 2009 May.
Article in English | MEDLINE | ID: mdl-19417160

ABSTRACT

CIGB-300, formerly known as P15-tat, is a proapoptotic peptide with established antiproliferative activity in vitro and antitumoral activity in vivo. This hypothesis-driven peptide was initially selected for its ability to impair the in vitro CK2-mediated phosphorylation in one of its substrates through direct binding to the conserved acidic phosphoaceptor domain. However, the actual in vivo target(s) on human cancer cells among the hundreds of CK2 substrates as well as the subsequent events that lead to apoptosis on tumor cells remains to be determined. In this work, we identified the multifunctional oncoprotein nucleophosmin/B23 as a major target for CIGB-300. In vivo, the CIGB-300-B23 interaction was shown by pull-down experiments and confirmed by the early in situ colocalization of both molecules in the cell nucleolus. Moreover, CIGB-300 inhibits the CK2-mediated phosphorylation of B23 in a dose-dependent fashion both in vitro and in vivo as shown using the recombinant GST fusion protein and the metabolic labeling approach, respectively. Such phosphorylation impairment was correlated with the ability of CIGB-300 to induce nucleolar disassembly as documented by the use of established markers for nucleolar structure. Finally, we showed that such a sequence of events leads to the rapid and massive onset of apoptosis both at the molecular and cellular levels. Collectively, these findings provide important clues by which the CIGB-300 peptide exerts its proapoptotic effect on tumor cells and highlights the suitability of the B23/CK2 pathway for cancer-targeted therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Casein Kinase II/metabolism , Cell Nucleolus/metabolism , Nuclear Proteins/metabolism , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Biomarkers, Tumor , Cell Line, Tumor , Humans , Intracellular Space , Molecular Targeted Therapy , Nucleophosmin , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Transport , Small Cell Lung Carcinoma/pathology
9.
BMC Cancer ; 9: 146, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19439079

ABSTRACT

BACKGROUND: Cervical cancer is now considered the second leading cause of death among women worldwide, and its incidence has reached alarming levels, especially in developing countries. Similarly, high grade squamous intraepithelial lesion (HSIL), the precursor stage for cervical cancer, represents a growing health problem among younger women as the HSIL management regimes that have been developed are not fully effective. From the etiological point of view, the presence of Human Papillomavirus (HPV) has been demonstrated to play a crucial role for developing cervical malignancies, and viral DNA has been detected in 99.7% of cervical tumors at the later stages. CIGB-300 is a novel cyclic synthetic peptide that induces apoptosis in malignant cells and elicits antitumor activity in cancer animal models. CIGB-300 impairs the Casein Kinase (CK2) phosphorylation, by targeting the substrate's phosphoaceptor domain. Based on the perspectives of CIGB-300 to treat cancer, this "first-in-human" study investigated its safety and tolerability in patients with cervical malignancies. METHODS: Thirty-one women with colposcopically and histologically diagnosed microinvasive or pre-invasive cervical cancer were enrolled in a dose escalating study. CIGB-300 was administered sequentially at 14, 70, 245 and 490 mg by intralesional injections during 5 consecutive days to groups of 7 - 10 patients. Toxicity was monitored daily until fifteen days after the end of treatment, when patients underwent conization. Digital colposcopy, histology, and HPV status were also evaluated. RESULTS: No maximum-tolerated dose or dose-limiting toxicity was achieved. The most frequent local events were pain, bleeding, hematoma and erythema at the injection site. The systemic adverse events were rash, facial edema, itching, hot flashes, and localized cramps. 75% of the patients experienced a significant lesion reduction at colposcopy and 19% exhibited full histological regression. HPV DNA was negative in 48% of the previously positive patients. Long term follow-up did not reveal recurrences or adverse events. CONCLUSION: CIGB 300 was safe and well tolerated. This is the first clinical trial where a drug has been used to target the CK2 phosphoaceptor domain providing an early proof-of-principle of a possible clinical benefit.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Drug-Related Side Effects and Adverse Reactions , Peptides, Cyclic/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Uterine Cervical Dysplasia/drug therapy , Uterine Cervical Neoplasms/drug therapy , Adolescent , Adult , Aged , Alphapapillomavirus/genetics , Alphapapillomavirus/isolation & purification , Drug Administration Routes , Drug Administration Schedule , Female , Humans , Middle Aged , Peptides, Cyclic/adverse effects , Protein Kinase Inhibitors/adverse effects , Uterine Cervical Neoplasms/virology , Young Adult , Uterine Cervical Dysplasia/virology
10.
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
11.
Int J Cancer ; 122(1): 57-62, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17847034

ABSTRACT

The antitumor efficacy of the CK2 inhibitors so far described has not been extensively evaluated in cancer animal models. We have previously demonstrated that a proapoptotic cyclic peptide termed P15 delivered into the cells by the Tat Cell Penetrating Peptide was able to abrogate the CK2-mediated phosphorylation and induce tumor regression when injected directly into solid tumors in mice. Here we explored the antitumor effect by systemic administration of P15-Tat in a consecutive 5-day schedule through either intraperitoneal or intravenous route. Importantly, significant delay of tumor growth was observed at 2 mg/kg (p < 0.05), 10 mg/kg (p < 0.01) or 40 mg/kg (p < 0.001) after P15-Tat administration both in syngeneic murine tumors and human tumors xenografted in nude mice. In line with this, the systemic administration of P15-Tat induced apoptosis in the tumor as evidenced by in situ DNA fragmentation. Furthermore, we evidenced that 99mTc-labeled P15-Tat peptide was certainly accumulated on the tumors after administration by both routes. This report becomes the first describing the antitumor effect induced by systemic administration of a peptide that targets the acidic phosphorylation domain for CK2 substrates. Also, our data reinforces the perspectives of P15-Tat for the cancer targeted therapy.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Casein Kinase II/metabolism , Cyclin-Dependent Kinase Inhibitor p15/pharmacology , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Female , Gene Products, tat/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Mice, Nude , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Tissue Distribution , Transplantation, Heterologous
13.
Angiogenesis ; 10(1): 23-34, 2007.
Article in English | MEDLINE | ID: mdl-17273909

ABSTRACT

Passive immunotherapy against soluble pro-angiogenic factors and/or their receptors in endothelial cells has become a promising approach in cancer therapeutics. There is also experimental evidence indicating that an active immunotherapy strategy directed towards these target molecules could also be effective. In this paper we show that it is possible to reduce tumor growth or increase the survival of tumor-bearing C57Bl/6 mice when animals are vaccinated with the human vascular endothelial growth factor (VEGF) isoform 121 gene (hVEGF(121)), and later challenged with melanoma or lung carcinoma tumor cells. Immunization was done with 10 microg DNA doses of the hVEGF121 gene, which is highly homologous to its mouse counterpart, administered on a weekly basis using a plasmid bearing 5 CpG bacterial motifs. Histopathology analyses of tumors of hVEGF(121) immunized animals showed a decrease in tumor cell density around vessels and in mitotic figures, as well as an increase in apoptotic tumor cells. A statistically significant cell cytotoxic response was found when spleen cells of immunized mice were co-cultured in vitro with mouse tumor VEGF-producing cells. Vaccination with an hVEGF121 gene mutated to make it deficient for VEGF receptor binding, produced similar in vitro and in vivo results, and significantly reduced the number of spontaneous metastases produced by the mouse Lewis lung carcinoma. Our results indicate that human VEGF DNA can be employed for anti-angiogenic active immunotherapy in mice, and that direct cell cytotoxicity is a contributor mechanism to the overall anti-tumor effects seen in immunized animals.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Lewis Lung/prevention & control , Melanoma, Experimental/prevention & control , Vaccines, DNA/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use , Angiogenesis Inhibitors/immunology , Animals , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/secondary , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Immunohistochemistry , Interferon-gamma/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/blood supply , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/physiopathology , Neovascularization, Pathologic/prevention & control , Receptors, Vascular Endothelial Growth Factor/metabolism , Vaccines, DNA/immunology
14.
Prostate ; 66(10): 1029-36, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16598740

ABSTRACT

BACKGROUND: Prostate specific antigen (PSA) is a relevant antigen in diagnosis; follow-up, and therapeutic approaches for fighting the prostate cancer. Several methods have been published previously to obtain a high purity preparation of PSA. In general, these methods are expensive, time-consuming, laborious, and in some cases produce low yields. METHODS: Based on a panel of 7 anti-PSA Mab's we carried on binding and elution experiments of PSA antigen in 96-well plates. The selected Mab were immobilized in a Sepharose CL-4B activated matrix with the purpose of purify PSA from human seminal fluid. In order to optimize the purification procedure, we test several washing and elution conditions (chaotropic agents, high ionic strength solution, and extreme pH). RESULTS: We selected a high ionic strength solution (2 M MgCl2) as elution condition, and a previous washing step with a mix of two ionic solutions (2.5 M NaCl pH 8/1 M MgCl2 pH 5.5) in order to purify PSA. Using such conditions we obtained a PSA preparation with 90% of purity and 50% of recovery. CONCLUSION: In this article, we report a simple, quickly, and non-expensive procedure to obtain free-PSA from human seminal plasma at high purity levels.


Subject(s)
Chromatography, Affinity/methods , Immunoassay/methods , Prostate-Specific Antigen/isolation & purification , Semen/chemistry , Animals , Antibodies, Monoclonal/immunology , Antigens/immunology , Chromatography, Affinity/economics , Humans , Hybridomas/immunology , Immunoassay/economics , Ligands , Male , Mice , Prostate-Specific Antigen/analysis , Prostate-Specific Antigen/immunology , Semen/immunology
15.
Biotechnol Appl Biochem ; 43(Pt 1): 39-48, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16101587

ABSTRACT

In the present paper we report the development of a bivalent scFv (single-chain Fv) antibody fragment, starting from a mouse mAb (monoclonal antibody) specific for CEA (carcinoembryonic antigen) that has received approval for in vivo radioimmunodiagnosis in humans. The diabody is well expressed in Escherichia coli, is easily purified by a combination of immobilized metal ion affinity chromatography and gel filtration and exhibits high affinity and specificity for CEA, comparable with those of the original mAb. Biodistribution experiments in athymic nude mice transplanted with human CEA+ cancer cells showed that the 125I-labelled diabody preferentially localizes in the tumour tissue and that retention is still high 48 h after injection. The diabody can be advantageous for some in vivo tumour targeting applications, due to the faster clearance derived from its smaller molecular mass.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoembryonic Antigen/immunology , Immunoglobulin Fragments/pharmacology , Neoplasms/metabolism , Recombinant Proteins/pharmacokinetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Cloning, Molecular , Escherichia coli/genetics , Female , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/immunology , Iodine Radioisotopes , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Neoplasms/diagnostic imaging , Neoplasms/immunology , Radioimmunodetection , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
16.
Curr Alzheimer Res ; 2(4): 409-17, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16248846

ABSTRACT

In order to have more specific tools available to approach amyloidogenesis in Alzheimer's disease (AD), we have produced several polyclonal and monoclonal antibodies that recognize specific sequences of the amyloid beta (Abeta) peptide. Here we present results that demonstrate that our monoclonal antibody EM5 recognizes an internal sequence (residues 11-16) of the Abeta peptide. This strategic localization of the epitope allowed us to employ this antibody, together with two previously reported polyclonal antibodies (EM2 and EM3, specific for AbetaX-40 and AbetaX-42, respectively), in an immunohistochemical study aimed at exploring the differential distribution of longer (AbetaX-40/42) and shorter (Abeta17-X) peptides along the various types of amyloid deposits of AD. This antibody panel was used in six AD brains, on sections from associative neocortex, striatum and cerebellar cortex. Single and double immunostaining revealed specific staining of vascular amyloid deposits and neuritic plaques by EM5 antibody, with high co-localization of EM2. Our results suggest that EM5 antibody recognizes pathogenic forms of Abeta deposits (amyloid angiopathy and neuritic plaques) and reveals the existence of a subset of plaques with a profile similar to vascular deposits. Additionally, our results show that diffuse plaques in AD brains may contain Abeta17-X peptides as its principal component. EM5 may be a useful tool in research both on human and transgenic mice tissue that may aid in the study of molecular heterogeneity of plaques in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal , Brain Chemistry/immunology , Peptide Fragments/immunology , Plaque, Amyloid/chemistry , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Antibody Specificity , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Peptide Fragments/chemistry , Peptide Fragments/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
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
18.
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
19.
Lancet ; 359(9310): 945-6, 2002 Mar 16.
Article in English | MEDLINE | ID: mdl-11918916

ABSTRACT

Tissue transglutaminase is the autoantigen that elicits endomysial antibodies, which are the serological hallmarks of coeliac disease. We describe a simple, rapid immunochromatographic assay for IgA and IgG antibodies to transglutaminase, which is highly accurate for diagnosis of this disease. Results were positive for all samples from 50 untreated coeliac patients, and negative for 40 non-coeliac patients with gastrointestinal disorders. The assay seems to be a useful alternative to biopsy for mass screening for coeliac disease.


Subject(s)
Antibodies, Monoclonal/blood , Celiac Disease/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Transglutaminases/blood , Antibodies, Monoclonal/isolation & purification , Celiac Disease/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Transglutaminases/immunology
20.
J Biotechnol ; 94(3): 287-98, 2002 Apr 11.
Article in English | MEDLINE | ID: mdl-11861087

ABSTRACT

We describe the construction of a phage antibody fragments library which combines, in a single cloning step, a synthetic human light chain variable region (V(L)) with a diverse set of heavy chain variable regions, from a mouse immunized with the prostate specific antigen (PSA). Despite V(L) restriction, selection from this library rendered two different single chain Fv antibody fragments, specifically recognizing PSA. The human V(L), used as a general partner for mouse heavy chains, was constructed by linking the germline A27 gene and the J(K)1 minigene segment, both of which are prominently involved in human antibody responses. Our approach offers a fast and simple way to produce half-human molecules, while keeping the advantage of immunizing animals for high affinity antibodies.


Subject(s)
Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulin Variable Region/genetics , Peptide Library , Prostate-Specific Antigen/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Base Sequence , Cloning, Molecular , Escherichia coli/genetics , Genes, Immunoglobulin , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Recombinant Fusion Proteins/immunology , Sequence Alignment , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...