Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 149(6): 1313-1321, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34019700

ABSTRACT

CIGB-552 is a synthetic peptide that interacts with COMMD1 and upregulates its protein levels. The objectives of this phase I study were safety, pharmacokinetic profile, evaluation of the lymphocytes CD4+ and CD8+ and preliminary activity in patients with advanced tumors. A 3 + 3 dose-escalation design with seven dose levels was implemented. Patients were included until a grade 3 related adverse event occurred and the maximum tolerated dose was reached. The patients received subcutaneous administration of CIGB-552 three times per week for 2 weeks. Single-dose plasma pharmacokinetics was characterized at two dose levels, and tumor responses were classified by RECIST 1.1. Twenty-four patients received CIGB-552. Dose-limiting toxicity was associated with a transient grade 3 pruritic maculopapular rash at a dose of 7.0 mg. The maximum tolerated dose was defined as 4.7 mg. Ten patients were assessable for immunological status. Seven patients had significant changes in the ratio CD4/CD8 in response to CIGB-552 treatment; three patients did not modify the immunological status. Stable disease was observed in five patients, including two metastatic soft sarcomas. We conclude that CIGB-552 at dose 4.7 mg was well tolerated with no significant adverse events and appeared to provide some clinical benefits.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/administration & dosage , NF-kappa B/drug effects , Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Research Design , Treatment Outcome
2.
J Biotechnol ; 301: 79-87, 2019 Aug 10.
Article in English | MEDLINE | ID: mdl-31145935

ABSTRACT

Nuclear Factor kappa B (NF-κB) is a conserved transcription factor involved in the expression of genes that are critical to inflammation and cell survival. Exposure to particular signals results in phosphorylation of NF-κB inhibitor proteins, which in turn allows NF-κB dimers to translocate to the nucleus and induce gene expression. Pathologic consequences of NF-κB activation are vast, mainly because of the pleiotropic roles that NF-κB-induced genes have on inflammation, cell proliferation and apoptosis. Thus, experimental models assessing NF-κB activation have direct screening applications for drug discovery. In this scenario, pathway-specific reporter cell systems become valuable tools to identify and elucidate the mechanism of action of novel compounds. Here, we describe the generation, characterization, and validation of human cancer epithelial reporter cell lines for functional studies of NF-κB activation by different pro- and anti-inflammatory mediators. Human lung (H460) and breast (T-47D) cancer cell lines were transfected with a pNF-κB-hrGFP plasmid which contains the GFP gene under the control of NF-κB binding elements. The pro-inflammatory cytokine TNF-α was able to activate the reporter systems in a concentration-response manner, correlating to the activation of the NF-κB signaling pathway. Finally, the reporter cell lines were validated using dexamethasone, a potent anti-inflammatory drug, a synthetic inhibitor of NF-κB (BAY 11-7082) and a new anti-cancer peptide (CIGB-552). We have established robust H460-NF-κB-hrGFP and T-47D-NF-κB-hrGFP reporter cell lines which represent a useful cancer model for primary screening and identification of compounds with anti-inflammatory activity.


Subject(s)
Breast Neoplasms/metabolism , Cell Line, Tumor/metabolism , Drug Discovery/methods , Lung Neoplasms/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Female , Humans , Models, Biological , Reproducibility of Results
3.
Molecules ; 23(4)2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29601540

ABSTRACT

CIGB-552 is a twenty-amino-acid novel synthetic peptide that has proven to be effective in reducing tumor size and increasing lifespan in tumor-bearing mice. Such capability is conferred by its cell-penetrating peptide character, which allows it to enter cells and elicit a pro-apoptotic effect through its major mediator, COMMD1 protein. Cell-penetrating peptides are able to use different internalization mechanisms, such as endocytosis or direct transduction through the plasma membrane. Although CIGB-552 cytotoxicity has been evaluated in several non-tumor- and tumor-derived cell lines, no data regarding the relationship between cell line sensitivity, cell penetrating capacity, the internalization mechanisms involved, COMMD1 expression levels, or its subcellular localization has yet been produced. Here, we present the results obtained from a comparative analysis of CIGB-552 sensitivity, internalization capacity and the mechanisms involved in three human tumor-derived cell lines from different origins: mammary gland, colon and lung (MCF-7, HT-29 and H460, respectively). Furthermore, cell surface markers relevant for internalization processes such as phosphatidylserine, as well as CIGB-552 target COMMD1 expression/localization, were also evaluated. We found that both endocytosis and transduction are involved in CIGB-552 internalization in the three cell lines evaluated. However, CIGB-552 incorporation efficiency and contribution of each mechanism is cell-line dependent. Finally, sensitivity was directly correlated with high internalization capacity in those cell lines where endocytosis had a major contribution on CIGB-552 internalization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents , Cell-Penetrating Peptides , Endocytosis/drug effects , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Humans , MCF-7 Cells , Neoplasms/metabolism , Neoplasms/pathology
4.
Res Vet Sci ; 114: 502-510, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28987957

ABSTRACT

The absence of an effective therapy against human solid tumors has fostered the development of promising antineoplastic therapeutic candidates, as the CIGB-552 peptide. This synthetic peptide has shown to be effective in reducing tumor size and increasing the lifespan in tumor-bearing mice. Therefore, this work was aimed to explore the safety profile and preliminary assessment of antitumor activity of the CIGB-552 peptide therapeutic candidate in a small population of dogs (n=9) having malignant spontaneously-arising solid tumors. The peptide was administered by subcutaneous (s.c.) route, at three dosage levels (0.075, 0.15 and 0.3mg/kg). The results showed no dose-limiting toxicities in any dogs. The antitumor activity observed in dogs receiving CIGB-552 was associated with the reduction in the tumor volume. Given the antitumor effects of CIGB-552 as mediated by COMMD1 protein, which function is highly conserved among eukaryotic organisms, and the similarities of canine and human types of cancer with respect to tumor biology, it is likely that CIGB-552 could demonstrate comparable anti-cancer activity in human patients. Synthetic peptide, COMMD1, Tumor, Dog, CIGB-552.


Subject(s)
Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Dog Diseases/drug therapy , Neoplasms/veterinary , Animals , Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/administration & dosage , Dogs , Drug Evaluation, Preclinical/veterinary , Female , Male , Neoplasms/drug therapy
5.
J Pept Sci ; 22(11-12): 711-722, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27933724

ABSTRACT

Because of resistance development by cancer cells against current anticancer drugs, there is a considerable interest in developing novel antitumor agents. We have previously demonstrated that CIGB-552, a novel cell-penetrating synthetic peptide, was effective in reducing tumor size and increasing lifespan in tumor-bearing mice. Studies of protein-peptide interactions have shown that COMMD1 protein is a major mediator of CIGB-552 antitumor activity. Furthermore, a typical serine-protease degradation pattern for CIGB-552 in BALB/c mice serum was identified, yielding peptides which differ from CIGB-552 in size and physical properties. In the present study, we show the results obtained from a comparative analysis between CIGB-552 and its main metabolites regarding physicochemical properties, cellular internalization, and their capability to elicit apoptosis in MCF-7 cells. None of the analyzed metabolites proved to be as effective as CIGB-552 in promoting apoptosis in MCF-7. Taking into account these results, it seemed important to examine their cell-penetrating capacity and interaction with COMMD1. We show that internalization, a lipid binding-dependent process, is impaired as well as metabolite-COMMD1 interaction, key component of the apoptotic mechanism. Altogether, our results suggest that features conferred by the amino acid sequence are decisive for CIGB-552 biological activity, turning it into the minimal functional unit. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Amino Acids/chemistry , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biotransformation , Caspase 7/genetics , Caspase 7/metabolism , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Gene Expression , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship
6.
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
7.
J Amino Acids ; 2013: 251398, 2013.
Article in English | MEDLINE | ID: mdl-23401744

ABSTRACT

We have demonstrated that the peptide L-2 designed from an alanine scanning of the Limulus-derived LALF32-51 region is a potential candidate for the anticancer therapy and its cell-penetrating capacity is an associated useful property. By the modification in the primary structure of L-2, a second-generation peptide (CIGB-552) was developed. However, the molecular mechanism underlying its cytotoxic activity remains partially unknown. In this study, it was shown that CIGB-552 increases the levels of COMMD1, a protein involved in copper homeostasis, sodium transport, and the NF-κB signaling pathway. We found that CIGB-552 induces ubiquitination of RelA and inhibits the antiapoptotic activity regulated by NF-κB, whereas the knockdown of COMMD1 blocks this effect. We also found that CIGB-552 decreases the antioxidant capacity and induces the peroxidation of proteins and lipids in the tumor cells. Altogether, this study provides new insights into the mechanism of action of the peptide CIGB-552, which could be relevant in the design of future anticancer therapies.

8.
Vaccine ; 29(5): 920-30, 2011 Jan 29.
Article in English | MEDLINE | ID: mdl-21145912

ABSTRACT

The ultimate success of cancer vaccination is dependent upon the generation of tumor-specific CTLs. In this study, we designed and evaluated a novel fusion protein comprising a cell penetrating and immunostimulatory peptide corresponding to residues 32-51 of the Limulus polyphemus protein (LALF(32-51)) linked to human papillomavirus (HPV) 16 E7 antigen (LALF(32-51)-E7). We demonstrated that LALF(32-51) penetrates the cell membrane and delivers E7 into cells. In a preclinical model of HPV16-induced cervical carcinoma we showed that vaccination with adjuvant-free LALF(32-51)-E7 fusion protein significantly improves the presentation of E7-derived peptides to T-cells in vitro and induces suppression of tumor growth.


Subject(s)
Carcinoma/prevention & control , Horseshoe Crabs/immunology , Human papillomavirus 16/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/prevention & control , Animals , Carcinoma/immunology , Disease Models, Animal , Female , Horseshoe Crabs/genetics , Human papillomavirus 16/genetics , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/administration & dosage , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Uterine Cervical Neoplasms/immunology
9.
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
10.
Int Immunopharmacol ; 4(10-11): 1343-51, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15313432

ABSTRACT

Severe sepsis and septic shock are important causes of death in intensive care units. Although Gram-negative infections were predominant in the 1960s, Gram-positive infections have increased in the past two decades and now account for about half of the cases of severe sepsis. In this study, we examined the effect of a Limulus anti-LPS factor (LALF)-derived peptide on lung and liver Th1/Th2 cytokine mRNA levels during a Gram-positive sepsis. We also examined the morphopathological changes observed in these organs during the disease. Mice challenged with a high dose of Staphylococcus haemolyticus showed severe damage in lung. In contrast, the liver of challenged mice showed an accumulation of bacterial particles in the sinusoids, associated with a severe inflammatory response due to high levels of tissue mRNA proinflammatory cytokines. Treatment with the peptide LALF(32-51) ameliorated the sepsis-induced effects in the lung and liver and increased the survival of mice in a dose- and time-dependent manner. Pretreatment with the peptide LALF(32-51) differentially regulates TNF-alpha, IFN-gamma, IL-12p40, IL-2 and IL-10 mRNA levels in lung and liver of peptide-treated mice, and limits the systemic inflammatory response. These findings support for the first time the effectiveness of an LALF-derived peptide in the treatment of a Gram-positive sepsis. Modulation of the Th1/Th2 pattern in tissues relevant for sepsis correlates with an improved outcome of the disease as denoted by increased survival.


Subject(s)
Anti-Infective Agents/pharmacology , Invertebrate Hormones/chemistry , Peptide Fragments/pharmacology , Sepsis/prevention & control , Staphylococcal Infections/prevention & control , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Anti-Infective Agents/therapeutic use , Antimicrobial Cationic Peptides , Arthropod Proteins , Cytokines/blood , Cytokines/genetics , Gene Expression Regulation , Liver/pathology , Lung/pathology , Mice , Mice, Inbred BALB C , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , RNA, Messenger/blood , Sepsis/immunology , Sepsis/mortality , Staphylococcal Infections/immunology , Staphylococcal Infections/mortality , Staphylococcus haemolyticus , Survival Rate , Time Factors
11.
J Endotoxin Res ; 9(5): 281-91, 2003.
Article in English | MEDLINE | ID: mdl-14577844

ABSTRACT

The ability of LPS-binding protein (LBP) to greatly potentiate cell responses to lipopolysaccharide (LPS) may largely contribute to LPS toxicity in sepsis. The study of agents with the capacity to block the interaction between LBP and LPS might improve the understanding of the role of LBP in Gram-negative infections as well as offering new therapeutic tools for septic disorders. Here we confirm the ability of synthetic peptides comprising the human LBP amino acid region 86-108 to interfere with the LBP-LPS interaction. The analysis of selected alanine mutants of a blocking peptide corresponding to the LBP region 86-99 suggests the importance of peptide amphipathicity for the inhibitory activity. The potency of the native peptide and a selected analogue at inhibiting in vitro and in vivo LPS-induced responses was associated with their relative activity in blocking LBP-LPS interaction. It was remarkable that these peptides were at least 500-fold more active in vivo than in vitro. Also, the inhibitory activity of peptides LBP86-99 and LBPK95A seems to be independent of LBP concentrations, a behavior that may be relevant for the potential use of these peptides in septic disorders where LBP serum concentrations are considerably elevated.


Subject(s)
Acute-Phase Proteins , Carrier Proteins/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins , Peptides/pharmacology , Amino Acid Sequence , Animals , Binding Sites/drug effects , Binding Sites/immunology , Binding, Competitive/immunology , Carrier Proteins/chemistry , Dose-Response Relationship, Drug , Escherichia coli/immunology , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Longevity/drug effects , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemistry , Peptides/therapeutic use , Protein Binding/immunology , Shock, Septic/drug therapy , Shock, Septic/immunology , Shock, Septic/mortality , Structure-Activity Relationship
12.
Int Immunopharmacol ; 3(2): 247-56, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12586605

ABSTRACT

Sepsis in experimental animals and humans has been associated with perturbed immune response. A major event contributing to the decrease in immune functions in septic disorders seems to be the inadequate balance of cytokines mediating the interactions between the innate and adaptive immune systems. We previously observed that a cyclic peptide derived from the Limulus anti-LPS factor (LALF), which partially protect mice from endotoxic shock lethality, has the ability to modulate cytokine secretion in vitro. We herein examined the effects of the LALF(31-52) peptide in an experimental model of Gram-negative peritoneal sepsis and analyzed the cytokine gene expression in the spleen and liver of peptide-treated mice. The prophylactic administration of LALF(31-52) abrogated the systemic TNF-alpha response, reduced organ damage and increased the survival of infected mice. Histological examination of spleen and liver in peptide-treated mice showed prevention of tissue damage induced by the high dose of Pseudomonas aeruginosa. This treatment modulates the cytokine gene expression in these tissues, stimulating IL-2, IL-12 and IL-13 mRNA synthesis, while IL-4 and IL-10 mRNA expression was not modified. This cytokine profile induced by the LALF-derived peptide seems to be favorable for host resistance against Gram-negative bacteria acute infection. In addition, peptide treatment was effective after the initiation of the systemic inflammatory response, promoting a significant increase in mice survival. These results further demonstrate the immunomodulatory potential of LALF(31-52) and are relevant for the design of prophylactic and therapeutic strategies for acute bacteria infection and sepsis, especially for preventing or ameliorating host immunity defects in these disorders.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Cytokines/genetics , Gene Expression Regulation/drug effects , Invertebrate Hormones/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Peptide Fragments/therapeutic use , Acute Disease , Animals , Antimicrobial Cationic Peptides , Arthropod Proteins , Interleukin-12/biosynthesis , Interleukin-13/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...