Herpes murine model as a biological assay to test dialyzable leukocyte extracts activity

Nohemí Salinas-Jazmín, Sergio Estrada-Parra, Miguel Angel Becerril-García, Alberto Yairh Limón-Flores, Said Vázquez-Leyva, Emilio Medina-Rivero, Lenin Pavón, Marco Antonio Velasco-Velázquez, Sonia Mayra Pérez-Tapia, Nohemí Salinas-Jazmín, Sergio Estrada-Parra, Miguel Angel Becerril-García, Alberto Yairh Limón-Flores, Said Vázquez-Leyva, Emilio Medina-Rivero, Lenin Pavón, Marco Antonio Velasco-Velázquez, Sonia Mayra Pérez-Tapia

Abstract

Human dialyzable leukocyte extracts (DLEs) are heterogeneous mixtures of low-molecular-weight peptides that are released on disruption of peripheral blood leukocytes from healthy donors. DLEs improve clinical responses in infections, allergies, cancer, and immunodeficiencies. Transferon is a human DLE that has been registered as a hemoderivate by Mexican health authorities and commercialized nationally. To develop an animal model that could be used routinely as a quality control assay for Transferon, we standardized and validated a murine model of cutaneous HSV-1 infection. Using this model, we evaluated the activity of 27 Transferon batches. All batches improved the survival of HSV-1-infected mice, wherein average survival rose from 20.9% in control mice to 59.6% in Transferon-treated mice. The activity of Transferon correlated with increased serum levels of IFN-γ and reduced IL-6 and TNF-α concentrations. Our results demonstrate that (i) this mouse model of cutaneous herpes can be used to examine the activity of DLEs, such as Transferon; (ii) the assay can be used as a routine test for batch release; (iii) Transferon is produced with high homogeneity between batches; (iv) Transferon does not have direct virucidal, cytoprotective, or antireplicative effects; and (v) the protective effect of Transferon in vivo correlates with changes in serum cytokines.

Figures

Figure 1
Figure 1
Standardization of mouse cutaneous herpes model. (a) Three different experiments performed with 5.0 × 104 PFU of HSV-1 (closed symbols). In each experiment, infected mice showed significantly lower survival than controls (open circles; log-rank Mantel-Cox test; P < 0.001). (b) Frequency of deaths from the experiments shown in (a).
Figure 2
Figure 2
Validation of the murine herpes model for the evaluation of Transferon activity. (a) Survival curves for HSV-1 infected mice. Treatments with 1 μg (open triangles) or 0.125 μg (open squares) of Transferon improved survival over placebo (open circles; Log-rank Mantel-Cox test; P < 0.01). In contrast, 12.5 ng of Transferon (open diamonds) had no significant effect. A group of mice was left uninfected (closed circles) as control. (b) Weight changes in HSV-1 infected mice. Transferon partially protects mice from the weight loss induced by HSV-1 (symbols are as in (a)). (c) Survival induced by five different Transferon batches was evaluated during a validation protocol. All batches improved survival versus placebo (Bonferroni t test; ∗P < 0.05, ∗∗P < 0.01; ∗∗∗P < 0.001).
Figure 3
Figure 3
Evaluation of biological activity of 27 Transferon batches in the validated murine model of herpes. Statistical analysis (ANOVA, Bonferroni's Multiple Comparison Test) showed no differences between doses.
Figure 4
Figure 4
In vitro evaluation of antiviral activity of Transferon. (a) HSV-1 was preincubated for 60 min with Transferon (20 μg/mL) or medium before evaluation of visible cytopathic effect (CPE) on Vero cells. Viruses with no preincubation (t0) were used as control (ANOVA, Bonferroni's Multiple Comparison Test; ns: nonsignificant). (b) Effect of Transferon (10 pg/mL–10 μg/mL) on HSV-1-induced cytopathology, evaluated by MTS assay. Acyclovir (ACV; 10 μg/mL) was included as a positive control. The graph represents data from 2 independent experiments (ANOVA, Bonferroni's Multiple Comparison Test; ∗P < 0.0001). (c) TCID50 obtained from Vero cells preincubated for 24 h with Transferon (20 μg/mL) or medium prior to HSV-1 infection (Student's t test). (d) Titration of samples obtained from Vero cells infected 24 h with HSV-1 and simultaneously treated with medium (open circles) or 20 μg/mL of Transferon (closed circles). ACV (10 μg/mL; triangles) was included as a positive control. Sum-of-squares F test showed no differences between the TCID50 from medium- and Transferon-treated cells (P = 0.7527). The graph represents data from 2 independent experiments.
Figure 5
Figure 5
Quantitative analysis of blood (serum) cytokine levels. Transferon (Batch 12C04; 0.75 μg/mouse) significantly reduced the concentrations of TNF-α (a) and IL-6 (b) at day 9 postinfection, but it increased that of IFN-γ at day 7 (c). Transferon administration had no effect on uninfected mice. All measurements were normalized to placebo-treated, uninfected controls. The graphs represent data of 2 independent experiments (ANOVA, Bonferroni's Multiple Comparison Test; ∗P < 0.05; ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001).

References

    1. Woodcock J., Griffin J., Behrman R., et al. The FDA's assessment of follow-on protein products: a historical perspective. Nature Reviews Drug Discovery. 2007;6(6):437–442. doi: 10.1038/nrd2307.
    1. Niederwieser D., Schmitz S. Biosimilar agents in oncology/haematology: from approval to practice. European Journal of Haematology. 2011;86(4):277–288. doi: 10.1111/j.1600-0609.2010.01566.x.
    1. Giezen T. J., Mantel-Teeuwisse A. K., Straus S. M. J. M., Schellekens H., Leufkens H. G. M., Egberts A. C. G. Safety-related regulatory actions for biologicals approved in the United States and the European Union. Journal of the American Medical Association. 2008;300(16):1887–1896. doi: 10.1001/jama.300.16.1887.
    1. Fudenberg H. H., Pizza G. Transfer factor 1993: new frontiers. Progress in Drug Research. 1994;42:309–400.
    1. Zhou J., Kong C., Yuan Z., et al. Preparation, characterization, and determination of immunological activities of transfer factor specific to human sperm antigen. BioMed Research International. 2013;2013:7. doi: 10.1155/2013/126923.126923
    1. Barrios M. A. C., Montiel B. N. R., Mourrelle J. A. F., de la Riva A. D. M., Suárez L. M. G., Pérez A. T. P. Patrones de prescripción de factor de transferencia en 11 hospitales de Ciudad de La Habana, 2002. Revista Cubana de Salud Pública. 2005;31:291–295.
    1. Šrámek V., Dadák L., Štouračová M., Štětka P., Komolíková L., Kuklínek P. Immodin in the treatment of immunoparalysis in intensive care patients. Vnitrni Lekarstvi. 2007;53(9):954–959.
    1. Medina-Rivero E., Merchand-Reyes G., Pavón L., et al. Batch-to-batch reproducibility of Transferon. Journal of Pharmaceutical and Biomedical Analysis. 2014;88:289–294. doi: 10.1016/j.jpba.2013.09.004.
    1. Berrón-Pérez R., Chávez-Sánchez R., Estrada-García I., et al. Indications, usage, and dosage of the transfer factor. Revista Alergia Mexico. 2007;54(4):134–139.
    1. Viza D., Fudenberg H. H., Palareti A., Ablashi D., De Vinci C., Pizza G. Transfer factor: an overlooked potential for the prevention and treatment of infectious diseases. Folia Biologica. 2013;59(2):53–67.
    1. Kirkpatrick C. H., Hamad A. R., Morton L. C. Murine transfer factors: Dose-response relationships and routes of administration. Cellular Immunology. 1995;164(2):203–206. doi: 10.1006/cimm.1995.1162.
    1. Pizza G., de Vinci C., Fornarola V., Palareti A., Baricordi O., Viza D. In vitro studies during long term oral administration of specific transfer factor. Biotherapy. 1996;9(1–3):175–185. doi: 10.1007/BF02628677.
    1. Delgado O., Romano E. L., Belfort E., Pifano F., Scorza J. V., Rojas Z. Dialyzable leukocyte extract therapy in immunodepressed patients with cutaneous leishmaniasis. Clinical Immunology and Immunopathology. 1981;19(3):351–359. doi: 10.1016/0090-1229(81)90078-7.
    1. Dvoroznakova E., Porubcová J., Ševčíková Z. Immune response of mice with alveolar echinococcosis to therapy with transfer factor, alone and in combination with albendazole. Parasitology Research. 2009;105(4):1067–1076. doi: 10.1007/s00436-009-1520-z.
    1. Estrada-Parra S., Chavez-Sanchez R., Ondarza-Aguilera R., et al. Immunotherapy with transfer factor of recurrent herpes simplex type I. Archives of Medical Research. 1995;26:S87–S92.
    1. Byston J., Cech K., Pekarek J., Jilkova J. Effect of anti-herpes specific transfer factor. Biotherapy. 1996;9(1–3):73–75. doi: 10.1007/BF02628660.
    1. Estrada-Parra S., Nagaya A., Serrano E., et al. Comparative study of transfer factor and acyclovir in the treatment of herpes zoster. International Journal of Immunopharmacology. 1998;20(10):521–535. doi: 10.1016/S0192-0561(98)00031-9.
    1. Simmons A., Nash A. A. Zosteriform spread of herpes simplex virus as a model of recrudescence and its use to investigate the role of immune cells in prevention of recurrent disease. Journal of Virology. 1984;52(3):816–821.
    1. Borkowsky W., Suleski P., Bhardwaj N., Lawrence H. S. Antigen-specific activity of murine leukocyte dialysates containing transfer factor on human leukocytes in the leukocyte migration inhibition (LMI) assay. Journal of Immunology. 1981;126(1):80–82.
    1. Farmacopea de los Estados Unidos Mexicanos. Métodos Generales de Análisis (MGA) 2011.
    1. Farmacopea de los Estados Unidos Mexicanos. 2011, Métodos para Productos Biológicos (MPB)
    1. Lobe D. C., Spector T., Ellis M. N. Synergistic topical therapy by acyclovir and A1110U for herpes simplex virus induced zosteriform rash in mice. Antiviral Research. 1991;15(2):87–100.
    1. Kurokawa M., Nagasaka K., Hirabayashi T., et al. Efficacy of traditional herbal medicines in combination with acyclovir against herpes simplex virus type 1 infection in vitro and in vivo. Antiviral Research. 1995;27(1-2):19–37. doi: 10.1016/0166-3542(94)00076-K.
    1. Kristofferson A., Ericson A.-C., Sohl-Akerlund A., Datema R. Limited efficacy of inhibitors of herpes simplex virus DNA synthesis in murine models of recrudescent disease. Journal of General Virology. 1988;69(6):1157–1166. doi: 10.1099/0022-1317-69-6-1157.
    1. Blyth W. A., Harbour D. A., Hill T. J. Pathogenesis of zosteriform spread of herpes simplex virus in the mouse. Journal of General Virology. 1984;65(9):1477–1486. doi: 10.1099/0022-1317-65-9-1477.
    1. Luganini A., Nicoletto S. F., Pizzuto L., et al. Inhibition of herpes simplex virus type 1 and type 2 infections by peptide-derivatized dendrimers. Antimicrobial Agents and Chemotherapy. 2011;55(7):3231–3239. doi: 10.1128/AAC.00149-11.
    1. Wulff N. H., Tzatzaris M., Young P. J. Monte Carlo simulation of the Spearman-Kaerber TCID50. Journal of Clinical Bioinformatics. 2012;2(1, article 5) doi: 10.1186/2043-9113-2-5.
    1. Heldt C. L., Hernandez R., Mudiganti U., Gurgel P. V., Brown D. T., Carbonell R. G. A colorimetric assay for viral agents that produce cytopathic effects. Journal of Virological Methods. 2006;135(1):56–65. doi: 10.1016/j.jviromet.2006.01.022.
    1. Akkarawongsa R., Pocaro N. E., Case G., Kolb A. W., Brandt C. R. Multiple peptides homologous to herpes simplex virus type 1 glycoprotein B inhibit viral infection radeekorn akkarawongsa. Antimicrobial Agents and Chemotherapy. 2009;53(3):987–996. doi: 10.1128/AAC.00793-08.
    1. Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. Washington, DC, USA: The National Academies Press; 2011.
    1. Liu Z., Guan Y., Sun X., et al. HSV-1 activates NF-kappaB in mouse astrocytes and increases TNF-alpha and IL-6 expression via Toll-like receptor 3. Neurological Research. 2013;35(7):755–762. doi: 10.1179/016164113X13703372991516.
    1. Robledo-Ávila F., Pérez-Tapia M., Limón-Flores A., et al. Low-dose amphotericin B and murine dialyzable spleen extracts protect against systemic candida infection in mice. Clinical and Developmental Immunology. 2013;2013:7. doi: 10.1155/2013/194064.194064
    1. Fabre R. A., Pérez T. M., Aguilar L. D., et al. Transfer factors as immunotherapy and supplement of chemotherapy in experimental pulmonary tuberculosis. Clinical and Experimental Immunology. 2004;136(2):215–223. doi: 10.1111/j.1365-2249.2004.02454.x.
    1. Mogensen T. H., Paludan S. R. Molecular pathways in virus-induced cytokine production. Microbiology and Molecular Biology Reviews. 2001;65(1):131–150. doi: 10.1128/MMBR.65.1.131-150.2001.

Source: PubMed

3
Se inscrever