ArticlePDF Available

Abstract and Figures

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.
This content is subject to copyright. Terms and conditions apply.
Research Article
Herpes Murine Model as a Biological Assay to Test
Dialyzable Leukocyte Extracts Activity
Nohemí Salinas-Jazmín,1Sergio Estrada-Parra,2Miguel Angel Becerril-García,1
Alberto Yairh Limón-Flores,1Said Vázquez-Leyva,1Emilio Medina-Rivero,1Lenin Pavón,3
Marco Antonio Velasco-Velázquez,4and Sonia Mayra Pérez-Tapia1,2,5
1Unidad de Desarrollo e Investigaci´
on en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biol´
ogicas, IPN,
Prolongaci´
on de Carpio y Plan de Ayala s/n, Col. Sto. Tom´
as, 11340 M´
exico, DF, Mexico
2Departamento de Inmunolog´
ıa, Escuela Nacional de Ciencias Biol´
ogicas, IPN, Prolongaci´
on de Carpio y Plan de Ayala s/n,
Col. Sto. Tom´
as, 11340 M´
exico, DF, Mexico
3Instituto Nacional de Psiquiatr´
ıa “Ram´
on De la Fuente Mu˜
niz”, Calzada M´
exicoXochimilco101,Col.SanLorenzoHuipulco,
14370 M´
exico, DF, Mexico
4Facultad de Medicina, Universidad Nacional Aut´
onoma de M´
exico, Ciudad Universitaria, 04510 M´
exico, DF, Mexico
5Unidad de Investigaci´
on Desarrollo e Innovaci´
on M´
edica y Biotecnol´
ogica (UDIMEB), Escuela Nacional de Ciencias Biol´
ogicas,
IPN, Prolongaci´
on de Carpio y Plan de Ayala s/n, Col. Sto. Tom ´
as, 11340 M´
exico, DF, Mexico
Correspondence should be addressed to Marco Antonio Velasco-Vel´
azquez; marcovelasco@unam.mx
and Sonia Mayra P´
erez-Tapia; smpt.@hotmail.com
Received  July ; Revised  September ; Accepted  September 
Academic Editor: Oscar Bottasso
Copyright ©  Nohem´
ı Salinas-Jazm´
ın et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
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 immunodeciencies. 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- infection. Using this model, we evaluated the activity of 
Transferon batches. All batches improved the survival of HSV--infected mice, wherein average survival rose from .% in control
mice to .% in Transferon-treated mice. e activity of Transferon correlated with increased serum levels of IFN-𝛾and reduced
IL- and TNF-𝛼concentrations. 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 eects;
and(v)theprotectiveeectofTransferonin vivo correlates with changes in serum cytokines.
1. Introduction
Governments worldwide have established standards for the
production and release of biotechnological and biological
drugs. For example, the US Food and Drug Administration
(FDA) and Japanese Ministry of Health & Welfare (MOHW)
modied their guidelines regarding biologics in , and
the European Medicines Agency (EMA) did so in  [].
In , the Sanitary Risk Authority of Mexico (COFEPRIS)
released the Ocial Mexican Standard NOM-EM--SSA-
.
A key element in the quality control of the production
of biological drugs is the demonstration of their activity, and
theirecacymustbepreservedbetweencommercialbatches.
Hindawi Publishing Corporation
Journal of Immunology Research
Volume 2015, Article ID 146305, 9 pages
http://dx.doi.org/10.1155/2015/146305
Journal of Immunology Research
Due to the unique nature and production of biological drugs,
specic methods must be developed to evaluate their quality
and attributes, including ecacy.
Human dialyzable leukocyte extracts (DLEs) are
heterogeneous mixtures of low-molecular-weight peptides
(< kDa) that are released on disruption of peripheral blood
leukocytes from healthy donors [].DLEsareproducedand
commercialized worldwide. In certain countries, such as
M´
exico, China, Cuba, and the Czech Republic, DLEs are
registered as drugs []becausetheyimproveclinical
responses in infections, allergies, cancer, and immunode-
ciencies (see Berr´
on-P´
erez et al. []andVizaetal.[]for
extensive reviews). eir complexity, however, has impeded
an extensive characterization of their components, active
substances, and biological activities.
Transferon is a human nonspecic DLE that is man-
ufactured by the National School of Biological Sciences
(ENCB), National Polytechnic Institute (IPN), Mexico, at
GMP facilities. Transferon is registered by Mexican health
authorities as a drug and is commercialized nationally. To
establish an assay that could be used routinely as a quality
control test for Transferon, we aimed to standardize and
validated a method to determine its ecacy in animals.
Other studies have demonstrated that the activity of DLEs
can be measured by assessing the induction of delayed type
hypersensitivity (DTH) in mice []andin vitro by analyzing
their eects on leukocyte migration []orIFN-𝛾secretion
[].
DLEs are eective for treating parasitic infections (acute
leishmaniasis [] and alveolar echinococcosis []) and
viral infections (herpes simplex virus- (HSV-) and herpes
zoster). Clinical trials have shown that DLEs mitigate the
duration of the acute phase, the frequency of recurrences, and
pain in herpes zoster patients better than acyclovir [,].
ese eects correlate with increased IFN-𝛾levels and CD+
cell counts []. Considering the clinical eects of DLEs
against herpetic infections and because animal models reect
thecomplexityofadrugresponseinanentireorganism,
we selected a murine model that has been reported to
emulate the natural form of HSV- infection []. is murine
model was standardized, validated, and used to evaluate the
biological activity of  batches of Transferon.
2. Materials and Methods
2.1. Quality Control of Transferon. We t e s t e d Tr a n s feron
batches that were produced between  and  using a
modied version according to Borkowsky et al. []. Briey,
leukocytes from  healthy donors were lysed with freeze-
thaw cycles and dialyzed against a -kDa membrane to
obtain low-molecular-weight peptides.
e quality control of Transferon comprised (A) endo-
toxin content, quantied using the Endosafe-Portable Test
(Charles River Laboratories, Charleston SC, USA) accord-
ing to the manufacturer’s instructions; the specication for
endotoxin was established in Mexican Pharmacopeia, Section
MGA- (. EU/mL) []; (B) microbiological tests,
according to Mexican Pharmacopeia, Section MGA-
T : Quality attributes of evaluated batches of Transferon.
Batch Protein
(mg/mL)
MAB
(CFU/mL)
FF&Y&
(CFU/mL)
Endotoxin
(EU/mL)
A . <<<.
C . << .
C . <<<.
C . <<<.
C . << .
C . <<<.
C . <<<.
D . <<<.
F . <<.
G . <<<.
H . << .
H . <<<.
K . <<<.
K . <<<.
L . <<<.
L . <<<.
A . <<<.
B . <<<.
C . <<<.
M . << .
M . <<<.
A . <<<.
C . <<<.
A . <<.
C . <<<.
B . <<<.
B . <<<.
#Mesophilic aerobic bacteria; &Filamentous fungus and yeast.
[]; (C) physicochemical characterization by a validated
ultraperformance liquid chromatography (UPLC) method
[] that analyzes molecular weights and the time of retention
of the main peaks compared with those of an internal batch
pattern. (D) Peptide content per nal dose was measured by
bicinchoninic acid (BCA) method using the Pierce BCA kit
(ermo Fisher Scientic, Waltham MA, USA) according to
the manufacturer’s instructions.
2.2. Herpes Simplex Murine Model. HSV- (KOS strain) was
obtained from American Type Culture Collection (ATCC;
Manassas VA, USA). e virus was propagated in African
green monkey kidney (Vero) cells (ATCC CCL-) that
were cultured in Eagles minimal essential medium (EMEM;
ATCC) supplemented with % fetal bovine serum (FBS;
Life Technologies, Carlsbad CA, USA). Cutaneous infection
of herpes was performed by inoculating -week-old male
BALB/c mice (Ferandelh, Mexico City, Mexico) with HSV-
, as reported [,,]. Briey, mice were anesthetized, and
 𝜇L of a viral suspension that contained ×6plaque-
forming units (PFU)/mL was administered by cutaneous
Journal of Immunology Research
0 2 4 6 8 101214161820
0
20
40
60
80
100
Time (d)
Survival (%)
(a)
0
10
20
30
Frequency of deaths (% total)
0 2 4 6 8 10 12 14 16 18 20
Time (d)
(b)
F : Standardization of mouse cutaneous herpes model. (a) ree dierent experiments performed with . ×4PFU of HSV- (closed
symbols). In each experiment, infected mice showed signicantly lower survival than controls (open circles; log-rank Mantel-Cox test; 𝑃<
0.001). (b) Frequency of deaths from the experiments shown in (a).
scarication on cm2of plucked dorsal skin []. Mice
were monitored daily for days to identify infection-
associated symptoms, such as paralysis of the lower extrem-
ities, reduced mobility, and weight loss. When the animals
experienced total loss of mobility, they were euthanized and
counted as deaths for the survival analysis.
Mice had free access to food (Harlan Laboratories, Indi-
anapolis IN, USA) and water during the experiment. All
experiments were performed blindly by trained researchers.
e administration of Transferon began on day aer
infection and continued every other day until day . Doses
employed were .ng, . 𝜇g, . 𝜇g, . 𝜇g, . 𝜇g,
. 𝜇g, or . 𝜇gpermouse(eachweighinggatday
) and perorally administered in  𝜇L. Each experimental
group comprised  mice. All experiments included a control
group of mice that received placebo (pyrogen-free water;
PISA Pharmaceutical, Mexico City, Mexico). Survival per
group was plotted as Kaplan-Meier graphs and analyzed by
Mantel-Cox test (𝛼=.)usingPrismProject,V.
(GraphPad Soware Inc, San Diego CA, USA).
2.2.1. In Vitro Eects on HSV-1. We studied the possible
direct antiviral eect of Transferon by mixing equal volumes
( 𝜇L) of Transferon (. mg peptide/mL) and HSV- (. ×
7PFUmL) and incubating the mixture for  minutes at
C. e infectivity of Transferon-treated virus was deter-
minedbyanalyzingtheinductionofavisiblecytopathiceect
(CPE) on Vero cells (4cells/well) at  h, as reported [].
Fiy percent tissue infective dose (TCID50)wascalculated
by Spearman-K¨
arber method []. Cytopathology induced
by HSV- ( PFU) in presence of Transferon ( pg/mL–
 𝜇g/mL) or acyclovir (Zoviraz, GlaxoSmithKline, Mex-
ico City, Mexico) was determined using MTS (CellTiter
 Aqueous One Solution cell proliferation assay reagent;
Promega, Madison WI, USA) as previously reported [,].
e cytoprotective eect of Transferon on target cells was
evaluated by preincubating Vero cells (4cells/well) with
Transferon ( 𝜇g/mL) for  hours before addition of HSV-
(viral stock . ×7PFU/mL). CPE was evaluated  h aer
infection []; TCID50 calculation was performed as reported
[]. To assess the eect of Transferon on virus replication, ×
5Vero cells were infected with HSV- ( PFU) and incu-
bated with Transferon ( 𝜇g/mL) or Acyclovir ( 𝜇g/mL)
for  h at C. Subsequently, viruses were recuperated from
cultures through one thaw/freeze cycle and sonication. Aer
removing cell debris by centrifugation ( g), the total
virus yield on each well was titrated by diluting samples and
incubating them with Vero cells for  h. We analyzed the
presence of CPE at the microscope and by MTS assay. As
reported by Heldt et al. [], the TCID50 was dened as the
concentration at which absorbance was % of the average
absorbance from uninfected cells and was determined by
nonlinear regression using Prism for Mac OS X (GraphPad
Soware Inc).
2.2.2. Cytometric Bead Array. To me a s u r e c y tokine c o n c e n -
trations, blood samples from the orbital sinus were collected
from anesthetized mice on days and aer infection.
Serum was obtained, and samples were stored at Cuntil
analysis. Cytokine concentrations were determined using a
cytometric bead array (CBA) mouse inammation kit (BD
Journal of Immunology Research
024681012141618
0
20
40
60
80
100
Time (d)
Survival (%)
(a)
0 3 6 9 12 15 18
0
2
4
Time (d)
−6
−4
−2
Weight dierence ±SEM
(b)
0
25
50
75
100
Survival (%)
Uninfected Placebo 0.5 𝜇g0.75 𝜇g1𝜇g1.5 𝜇g
∗∗∗ ∗∗∗
∗∗∗
(c)
F : Validation of the murine herpes model for the evaluation of Transferon activity. (a) Survival curves for HSV- infected mice.
Tre a tmen ts wit h 𝜇g (open triangles) or . 𝜇g (open squares) of Transferon improved survival over placebo (open circles; Log-rank Mantel-
Cox test; 𝑃 < 0.01). In contrast, . ng of Transferon (open diamonds) had no signicant eect. A group of mice was le uninfected (closed
circles) as control. (b) Weight changes in HSV- infected mice. Transferon partially protects mice from the weight loss induced by HSV-
(symbols are as in (a)). (c) Survival induced by ve dierent Transferon batches was evaluated during a validation protocol. All batches
improved survival versus placebo (Bonferroni ttest; 𝑃 < 0.05,∗∗𝑃 < 0.01;∗∗∗𝑃 < 0.001).
Biosciences, San Diego CA, USA) according to the manufac-
turer’s instructions. We used a FACS Aria ow cytometer and
BD CBA soware (BD Biosciences) for data acquisition and
analysis.
2.2.3. Ethics Statement. All experimental procedures with
animals were performed according to the Mexican Guidelines
for the Production, Care, and Use of Laboratory Animals
(NOM--ZOO-) and the International Guide for the
Care and Use of Laboratory Animals []. All eorts were
made to minimize animal suering and reduce the number
of animals that were used. e experimental procedures were
approved by the Ethical Committee of the Transfer Factor
Project in “Escuela Nacional de Ciencias Biol´
ogicas, Instituto
Polit´
ecnico Nacional” (protocol FTU/IB///PRO).
Journal of Immunology Research
3. Results
3.1. Quality Control of Batches. All batches had endotoxin
levels below . EU/mL and met the microbiological speci-
cations in Mexican Pharmacopeia (aerobic mesophile bacte-
ria < colony-forming units (CFU)/mL, lamentous fungi
< CFU/mL, and yeasts < CFU/mL (Ta b l e )). By UPLC,
we performed a physicochemical characterization of the
batches. In the chromatographic proles of the Transferon
batches, we noted peptidic fractions between  to . kDa,
as previously reported []. e retention times of the main
peaks corresponded with those of an internal pattern, dened
as a batch of Transferon that satised all quality control
tests for this product, as previously reported []. We also
measured peptide concentrations in the Transferon sam-
ples. All batches had peptide concentrations that met the
established specication of 0.400 ± 0.06mg/mL. e average
concentration in the  batches was 0.416 ± 0.023mg/mL
(Table ).
3.2. Herpes Murine Model. To develop a murine model of
cutaneous herpes, we rst studied the eect of an HSV-
inoculum on mouse survival. We inoculated . ×4,.×
4,and.×5PFU of HSV- and analyzed mouse survival
(Figure ). In subsequent experiments, we used . ×4PFU
of HSV-, which was the minimum inoculum that produced
a decrease in survival within % to %, as reported
[].
en, we determined the reproducibility of the eect of
the infection by repeating the assay times and measuring
survival.esurvivalofinfectedanimalswas%to%
(Figure (a)). Using data from the same experiments, we
calculated the frequency of deaths (euthanizations) over time,
noting that % of deaths occurred before day  aer infec-
tion, rising to % by day (Figure (b)). is information
allowed us to establish an endpoint for subsequent assays.
3.3. Biological Evaluation of Transferon. To determine the
doses of Transferon that were to be used to evaluate the
commercial batches, we examined a -fold range of doses
(. ng–. 𝜇g per mouse). Doses above . 𝜇g/mouse
were equally ecacious in reducing HSV--induced mortality
and weight loss. In contrast, . ng was ineective, indi-
cating that the activity of Transferon is dose-dependent. As
expected, uninfected mice gained weight and showed %
survival (Figures (a) and (b)).
e model was validated using batches that were pro-
ducedinandtestedatdoses(Figure (c)). We evaluated
(i) system specicity, by analyzing responses in placebo- and
Transferon-treated animals; the average survival in controls
was .% (range % to %), whereas all Transferon doses
induced a signicant increase in survival (average increase
versus placebo (Δsurvival) .%); (ii) system precision, by
calculating the relative standard deviation (RSD) percentage
for the results with each Transferon dose; % RSD was below
% in all cases (range: .% to .%); (iii) system suitability,
by corroborating that at least dose produced a Δsurvival
0.125 0.25 0.5 0.75 1 1.5
0
10
20
30
40
50
60
Dose (𝜇g)
Survival over placebo (Δ)±SEM
F : Evaluation of biological activity of Transferon batches
in the validated murine model of herpes. Statistical analysis
(ANOVA, Bonferroni’s Multiple Comparison Test) showed no
dierences between doses.
%. All parameters were within the limits that were
establishedinthevalidationprotocol.
Eighteen batches that were produced in  and that
weregeneratedinwereexaminedinthevalidated
murine HSV- model. In certain batches, we evaluated
additional doses (. and . 𝜇g per mouse). All batches
improved the survival of HSV--infected mice by an average
of .% (range: .% to .%) (Figure ). ere were no
dierences in average survival between doses, indicating that
any of these doses could be used in future quality control
assays.
3.4. Eects of Transferon on HSV-1 Infectivity. We me a s u r e d
the antiviral activity of Transferon by (i) preincubating HSV-
with the drug before adding it to the target (Vero) cells
(Figure (a)) and (ii) simultaneously incubating target
cells with HSV- and various Transferon concentrations
( pg/mL– 𝜇g/mL) (Figure (b)). Transferon did not
show direct antiviral eect on either experimental system.
Preincubation of target cells with Transferon for  h
before the viral challenge did not prevent HSV- infection
(Figure (c)), suggesting that Transferon does not modify
the target cell-susceptibility to infection. Additionally, we
evaluated the eect of Transferon on virus replication. e
titration of HSV- on Transferon-treated samples showed
that the TCID50 is not dierent from that of control samples.
Values obtained from nonlinear regression t were log10 =
. versus log10 =.,respectively(Figure (d)). ese
results, obtained by a colorimetric assay, correlate with the
visual analysis of CPE (Supplemental Figure available
online at http://dx.doi.org/.//), as reported
for others viruses [], and demonstrate that Transferon has
no eect on HSV- replication.
3.5. Eects of Transferon on Systemic Cytokines. Blood
(serum)cytokinelevelsweremeasuredatdaysand,
beforetheperiodofhighmortality.TNF-𝛼,IL-,andIFN-𝛾
levels rose in HSV--infected and placebo-treated mice versus
Journal of Immunology Research
ns
100
101
102
105
106
107
108
t0control Medium Transferon
TCID50/mL ±SD
60 min 60 min
(a)
0
0
20
40
60
Tra nsf e ro n
****
ns
Uninfected control ±SEM (%)
10 pg/mL 10 ng/mL 10 𝜇g/mL ACV
(b)
ns
Medium Transferon
100
101
102
105
106
107
108
TCID50/mL ±SD
(c)
1234567
0
25
50
75
100
log10 dilution factor
Optical density (%) ±SEM
(d)
F : In vitro evaluation of antiviral activity of Transferon. (a) HSV- was preincubated for  min with Transferon ( 𝜇g/mL) or
medium before evaluation of visible cytopathic eect (CPE) on Vero cells. Viruses with no preincubation (t0)wereusedascontrol(ANOVA,
Bonferroni’s Multiple Comparison Test; ns: nonsignicant). (b) Eect of Transferon ( pg/mL– 𝜇g/mL) on HSV--induced cytopathology,
evaluated by MTS assay. Acyclovir (ACV;  𝜇g/mL) was included as a positive control. e graph represents data from independent
experiments (ANOVA, Bonferroni’s Multiple Comparison Test; 𝑃 < 0.0001). (c) TCID50 obtained from Vero cells preincubated for  h
with Transferon (𝜇g/mL) or medium prior to HSV- infection (Students ttest). (d) Titration of samples obtained from Vero cells infected
 h with HSV-and simultaneously treated with medium (open circles) or 𝜇g/mL of Transferon (closed circles). ACV ( 𝜇g/mL; triangles)
was included as a positive control. Sum-of-squares Ftest showed no dierences between the TCID50 from medium- and Transferon-treated
cells (𝑃 = 0.7527). e graph represents data from independent experiments.
Journal of Immunology Research
0.0
0.5
1.0
1.5
2.0
2.5
Uninfected HSV-1
Placebo Transferon Placebo Transferon
∗∗
∗∗
Normalized TNF-𝛼 ± SD
(a)
0
2
4
6
8
10
Uninfected HSV-1
Placebo Transferon Placebo Transferon
∗∗∗∗
∗∗∗∗
Normalized IL-SD
(b)
0
5
10
15
20
25
Uninfected HSV-1
Placebo Transferon Placebo Transferon
∗∗
∗∗∗∗
∗∗∗∗
Normalized IFN-𝛾 ± SD
(c)
F : Quantitative analysis of blood (serum) cytokine levels. Transferon (Batch C; . 𝜇g/mouse) signicantly reduced the
concentrations of TNF-𝛼(a) and IL- (b) at day postinfection, but it increased that of IFN-𝛾at day (c). Transferon administration had
no eect on uninfected mice. All measurements were normalized to placebo-treated, uninfected controls. e graphs represent data of
independent experiments (ANOVA, Bonferroni’s Multiple Comparison Test; 𝑃 < 0.05;∗∗𝑃 < 0.01;∗∗∗𝑃 < 0.001;∗∗∗∗𝑃 < 0.0001).
uninfected mice (Figures (a)(c)). In contrast, Transferon
did not change the cytokines levels in uninfected mice.
Treatment of HSV--infected mice with Transferon signi-
cantly decreased TNF-𝛼and IL- levels at day , compared
with placebo-treated animals (Figures (a) and (b).In
contrast, Transferon further increased IFN-𝛾levels at day
(Figure (c)).
4. Discussion
All  batches of Transferon in this study complied with
quality standards with regard to their attributes. As shown for
batchesthatwereproducedin-[], we noted high
homogeneity in microbial content, peptide concentration,
molecular weight, and time of retention of the main peaks by
Journal of Immunology Research
UPLC. ese results demonstrate that is possible to produce
a mixture of peptides that have been extracted from complex
rawmaterials,suchaslysedhumanleukocytes.
To evaluate the activity of these batches, a murine
model of herpes was standardized and validated. Transferon
partially protected animals from the HSV--induced weight
loss, which correlated with greater survival. is model was
chosen because DLEs are eective in clinical studies of
herpetic infections. DLEs signicantly reduce the average
duration of the acute phase and the frequency of recurrences
in herpetic infections as successfully as antiviral drugs [,
]. All batches of Transferon signicantly increased survival
dose-independently, indicating that it is biologically active
over a wide range of doses (-fold). us, our murine model
of herpes can be used to measure ecacy—a fundamental
attribute of biological drugs—in DLEs.
However, the herpes murine model has several disadvan-
tages. e variability that is intrinsic to a whole-animal model
is signicant; such a model requires the use of many animals.
Also, more time is needed to evaluate activity than for an in
vitro experiment.
Transferon has no direct virucidal or antireplicative
eects on HSV- nor cytoprotective eects on target cells,
suggesting that the in vivo activity of Transferon is mediated
by its eects on the immune system. We found that HSV-
-infected mice had higher serum concentrations of TNF-
𝛼,IL-,andIFN-𝛾compared with uninfected controls.
Consistent with these ndings, in astrocyte cultures, HSV-
infection upregulates TNF-𝛼,IL-,andNF-𝜅B in a Toll-like
receptor (TLR)--dependent manner []. Administration
of Transferon to HSV--infected mice downregulates TNF-
𝛼and IL-, suggesting that it suppresses innate immune
responses. e modulation of proinammatory cytokines
by DLEs has been associated with reduced inammation-
associated tissue damage by pathogens [,].
Conversely, Transferon increased IFN-𝛾,acytokinethatis
producedduringtheadaptivephaseofimmunity,suggesting
that Transferon indirectly stimulates the activation of T lym-
phocytes that are specic for HSV-. IFN-𝛾eects resistance
against HSV- infection [], and its upregulation in DLE-
treated herpes patients favors a positive clinical response
[] and limits relapses []. Although the mechanisms of
action of DLEs have not been determined completely, our
results indicate that they have dierential eects on innate
andadaptiveimmunity.
5. Conclusions
Our analysis of  batches of Transferon demonstrate that (i)
the cutaneous model of herpes can be used to evaluate the
biological 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, meet-
ing the standards that are required for hemoderivatives that
are intended for clinical use; (iv) Transferon does not have
direct virucidal, cytoprotective, or antireplicative eects; and
(v) the protective eects of Transferon in our murine model
correlate with changes in serum cytokine levels.
Conflict of Interests
Nohem´
ı Salinas-Jazm´
ın, Sergio Estrada-Parra, Miguel Angel
Becerril-Garc´
ıa, Alberto Yairh Lim´
on-Flores, Said V´
azquez-
Leyva, and Sonia Mayra P´
erez-Tapia are employees or have
been compensated for their work at “UDIMEB” the producer
of Transferon. All other authors declare no competing inter-
ests.
Acknowledgments
is work was partially supported by FTU/IB///PRO
(Nohem´
ı Salinas-Jazm´
ın) and CONACYT INFR--
 (MAV-V). Authors are grateful for the valuable help
of Natanael Valtierra-Botello and Leonardo L´
opez-Ju´
arez
(animal housing), Emiliano Hisaki-Itaya (ow cytometry
data acquisition and analysis), Gilberto P´
erez-S´
anchez (val-
idation protocol), and Antonio Ortega-Roque (cell and virus
culture).
References
[] J. Woodcock, J. Grin, R. Behrman et al., “e FDAs assess-
ment of follow-on protein products: a historical perspective,
Nature Reviews Drug Discovery,vol.,no.,pp.,.
[] D. Niederwieser and S. Schmitz, “Biosimilar agents in oncol-
ogy/haematology: from approval to practice, European Journal
of Haematology, vol. , no. , pp. –, .
[]T.J.Giezen,A.K.Mantel-Teeuwisse,S.M.J.M.Straus,H.
Schellekens, H. G. M. Leuens, and A. C. G. Egberts, “Safety-
related regulatory actions for biologicals approved in the United
States and the European Union, Journal of the American
Medical Association,vol.,no.,pp.,.
[] H. H. Fudenberg and G. Pizza, “Transfer factor : new
frontiers, Progress in Drug Research,vol.,pp.,.
[] J. Zhou, C. Kong, Z. Yuan et al., “Preparation, characterization,
and determination of immunologicalac tivities of transfer factor
specic to human sperm antigen, BioMed Research Interna-
tional, vol. , Article ID , pages, .
[]M.A.C.Barrios,B.N.R.Montiel,J.A.F.Mourrelle,A.D.
M.delaRiva,L.M.G.Su
´
arez, and A. T. P. P´
erez, “Patrones
de prescripci´
on de factor de transferencia en  hospitales de
Ciudad de La Habana, , Revista Cubana de Salud P´
ublica,
vol. , pp. –, .
[] V. ˇ
Sr´
amek, L. Dad´
ak, M. ˇ
Stouraˇ
cov´
a, P. ˇ
Stˇ
etka, L. Komol´
ıkov´
a,
and P. Kukl´
ınek, “Immodin in the treatment of immunoparal-
ysis in intensive care patients, Vnitr ni Lek ars tvi,vol.,no.,
pp. –, .
[] E. Medina-Rivero, G. Merchand-Reyes, L. Pav´
on et al., “Batch-
to-batch reproducibility of Transferon, Journal of Pharmaceu-
tical and Biomedical Analysis, vol. , pp. –, .
[] R. Berr´
on-P´
erez, R. Ch´
avez-S´
anchez, I. Estrada-Garc´
ıa et al.,
“Indications, usage, and dosage of the transfer factor, Revista
Alergia Mexico,vol.,no.,pp.,.
[] D. Viza, H. H. Fudenberg, A. Palareti, D. Ablashi, C. De Vinci,
and G. Pizza, “Transfer factor: an overlooked potential for the
prevention and tre atment of infectious dis eases, Folia Biologica,
vol. , no. , pp. –, .
[] C. H. Kirkpatrick, A. R. Hamad, and L. C. Morton, Murine
transfer factors: Dose-response relationships and routes of
Journal of Immunology Research
administration, Cellular Immunology,vol.,no.,pp.
, .
[] G. Pizza, C. de Vinci, V. Fornarola, A. Palareti, O. Baricordi, and
D. Viza, “In vitro studies during long term oral administration
of specic t ransfer factor, Biotherapy,vol.,no.,pp.,
.
[]O.Delgado,E.L.Romano,E.Belfort,F.Pifano,J.V.Scorza,
and Z. Rojas, “Dialyzable leukocyte extract therapy in immun-
odepressed patients with cutaneous leishmaniasis, Clinical
Immunology and Immunopathology, vol. , no. , pp. –,
.
[] E. Dvoroznakova, J. Porubcov´
a, and Z. ˇ
Sevˇ
c´
ıkov´
a, “Immune
response of mice with alveolar echinococcosis to therapy with
transfer factor, alone and in combination with albendazole,
Parasitology Research,vol.,no.,pp.,.
[] S. Estrada-Parra, R. Chavez-Sanchez, R. Ondarza-Aguilera et
al., “Immunotherapy with transfer factor of recurrent herpes
simplex type I, Archives of Medical Research,vol.,pp.S
S, .
[] J. Byston, K. Cech, J. Pekarek, and J. Jilkova, “Eect of anti-
herpes specic transfer factor, Biotherapy,vol.,no.,pp.
–, .
[] S. Estrada-Parra, A. Nagaya, E. Serrano et al., “Comparative
study of transfer factor and acyclovir in the treatment of herpes
zoster, International Journal of Immunopharmacology,vol.,
no. , pp. –, .
[] A. Simmons and A. A. Nash, “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,vol.,no.,pp.,.
[] W. Borkowsky, P. Suleski, N. Bhardwaj, and H. S. Lawrence,
Antigen-specic activity of murine leukocyte dialysates con-
taining transfer factor on human leukocytes in the leukocyte
migration inhibition (LMI) assay, Journal of Immunology,vol.
,no.,pp.,.
[] Farmacopea de los Estados Unidos Mexicanos, M´
etodos Gen-
erales de An´
alisis (MGA), .
[] “Farmacopea de los Estados Unidos Mexicanos, , M´
etodos
para Productos Biol´
ogicos (MPB).
[] D. C. Lobe, T. Spector, and M. N. Ellis, “Synergistic topical
therapy by acyclovir and AU for herpes simplex virus
induced zosteriform rash in mice, Antiviral Research,vol.,
no.,pp.,.
[] M. Kurokawa, K. Nagasaka, T. Hirabayashi et al., “Ecacy
of traditional herbal medicines in combination with acyclovir
against herpes simplex virus type infection in vitro and in
vivo, Antiviral Research,vol.,no.-,pp.,.
[] A. Kristoerson, A.-C. Ericson, A. Sohl-Akerlund, and R.
Datema, “Limited ecacy of inhibitors of herpes simplex virus
DNA synthesis in murine models of recrudescent disease,
Journal of General Virology, vol. , no. , pp. –, .
[] W. A. Blyth, D. A. Harbour, and T. J. Hill, “Pathogenesis of
zosteriform spread of herpes simplex virus in the mouse,
Journal of General Virology,vol.,no.,pp.,.
[] A. Luganini, S. F. Nicoletto, L. Pizzuto et al., “Inhibition of
herpes simplex virus type and type infections by peptide-
derivatized dendrimers, Antimicrobial Agents and Chemother-
apy,vol.,no.,pp.,.
[] N. H. Wul, M. Tzatzaris, and P. J. Young, “Monte Carlo sim-
ulation of the Spearman-Kaerber TCID, Journal of Clinical
Bioinformatics,vol.,no.,article,.
[] C. L. Heldt, R. Hernandez, U. Mudiganti, P. V. Gurgel, D. T.
Brown, and R. G. Carbonell, A colorimetric assay for viral
agents that produce cytopathic eects, Journal of Virological
Methods,vol.,no.,pp.,.
[] R.Akkarawongsa,N.E.Pocaro,G.Case,A.W.Kolb,andC.
R. Brandt, “Multiple peptides homologous to herpes simplex
virus type glycoprotein B inhibit viral infection radeekorn
akkarawongsa, Antimicrobial Agents and Chemotherapy,vol.
,no.,pp.,.
[] Committee for the Update of the Guide for the Care and Use of
Laboratory Animals, Guide for the Care and Use of Laboratory
Animals, e National Academies Press, Washington, DC, USA,
.
[] Z. Liu, Y. Guan, X. Sun et al., “HSV- activates NF-kappaB in
mouse astrocytes and increases TNF-alpha and IL- expression
via Toll-like receptor , Neurological Research,vol.,no.,pp.
–, .
[] F. Robledo- ´
Avila, M. P´
erez-Tapia, A. Lim´
on-Flores et al., “Low-
dose amphotericin B and murine dialyzable spleen extracts
protect against systemic candida infection in mice, Clinical
and Developmental Immunology,vol.,ArticleID,
pages,.
[] R. A. Fabre, T. M. P´
erez,L.D.Aguilaretal.,“Transferfactors
as immunotherapy and supplement of chemotherapy in exper-
imental pulmonary tuberculosis, Clinical and Experimental
Immunology,vol.,no.,pp.,.
[] T. H. Mogensen and S. R. Paludan, “Molecular pathways in
virus-induced cytokine production, Microbiology and Molecu-
lar Biology Reviews,vol.,no.,pp.,.
... Regarding in vitro models, "Transferon Oral" increases the expression of CD80/CD86 and IL-6 levels in LPS-stimulated macrophage-like THP-1 cells, whereas it induces the differentiation of IFN-g-producing NK CD56 +CD16+CD11c+ cells from CD34+ progenitor cells obtained from human umbilical cord (Ramirez-Ramirez et al., 2016;Jimenez-Uribe et al., 2019). On the other hand, "Transferon Oral" (1-25 mg/kg) reduces tumour growth and metastasis in a murine prostate cancer model, while in Herpes Simplex Virus type 1 (HSV-1) infection murine model "Transferon Oral" decreases the bloodstream levels of TNF-a and IL-6 and increases IFN-g levels and the percent of survival when oropharyngeal administered (ORO) (Salinas-Jazmin et al., 2015;Hernandez-Esquivel et al., 2018). Nevertheless, the information provided by these models is no enough to explain how a peptide extract can modulate the immune system when administered by an enteral route. ...
... The relevance of the Ub content in Transferon was evaluated using an HSV-1 infection murine model, as previously described by Salinas-Jazmin and cols. (Salinas-Jazmin et al., 2015); trained researchers blindly performed all procedures. Briefly, 4-week-old and 14-18 g weigh male BALB/c mice (Ferandelh; Mexico City, Mexico) were shaved on the back and anaesthetized with 10 mL/g of 6.4 mg/mL sodium pentobarbital (Pisa Laboratories) via intraperitoneal. ...
... Animals were euthanized if they experienced a total loss of mobility; these events were counted as deaths. All procedures were performed according to Mexican and International Guides for Care and Use of Laboratory Animals (Secretarıá de Agricultura, 1999;Animals, 2011) and approved by the Ethical Committee of the Transfer Factor Project (protocol FTU/IB/012/010/PRO) (Salinas-Jazmin et al., 2015). All efforts were made to minimize animal suffering. ...
Article
Full-text available
“Transferon Oral” is a peptide-derived product with immunomodulatory properties obtained from the lysis and dialysis of human buffy coat. Its active pharmaceutical ingredient, generically known as Dialyzable Leucocyte Extract, is a mixture of peptide populations with reproducible proportions among batches. “Transferon Oral” modulates IFN-γ, TNF-α, and IL-6 and increases the survival rate in a herpes infection murine model when oropharyngeally (ORO) administered, which correlate with clinical observations where “Transferon Oral” is used as a therapeutic auxiliary in inflammatory diseases. Notwithstanding, how a peptide-derived product elicits systemic modulation of cytokines when ORO administered remains unclear. To shed light on the pharmacology of “Transferon Oral” its peptide components must be known. Ten “Transferon Oral” batches were sequenced by mass spectrometry and the intact peptides were identified. The most abundant peptides were the monomeric human Ubiquitin (Ub), a globular low-molecular mass protein, and an Ub variant which lacks the two-terminal Gly (Ub-GG). Recombinant Ub prevented murine death when ORO administered in a herpes infection murine model. Besides, the percentage of survival increased in groups treated with Transferon Oral+Ub and decreased in groups treated with Ub-depleted “Transferon Oral” respect to the group treated with “Transferon Oral” only. Our findings indicate that the biological properties of “Transferon Oral” are partially associated to the Ub content. They suggest that Ub may activate its extracellular receptor (CXCR-4) in the stomach eliciting systemic immunomodulatory effects via vagus nerve. This is the first report that identifies an active component of “Transferon Oral” with the potential for the development of oral peptide immunomodulators.
... Transferon ® , a drug product made of DLEs, has been tested in several models. Whereas the in vitro assays allow for obtaining basic knowledge [2,10], the in vivo animal models allow for the evaluation of its effects on pathologies such as viral diseases and neoplasia [1,11]. In humans, it has been specifically proved to be effective as a coadjuvant to conventional therapies of certain diseases involving immunological dysregulation, such as major depressive disorders [12], hypersensitivities (allergic rhinitis, atopic dermatitis, allergic asthma) [13], and some infectious diseases such as herpes zoster and sepsis [14,15]. ...
... So far, the batch release tests of Transferon ® include an in vivo assay, which evaluates the effect of Transferon ® on the survival of mice infected with herpes simplex virus 1 [11], and an in vitro assay, which analyzes the ability of Transferon ® to induce interferon-γ expression in Jurkat cells [3]. Although the two methods are convenient alternatives used to determine the biological activity of Transferon ® , both also present some disadvantages. ...
... These results are evidence of a favorable effect of Transferon ® on cell proliferation ( Figure 1). Due to the complex composition of the product, it was difficult to elucidate its mechanism of action; however, the biological responses evidenced a modulation of cytokine production, such as interleukine 6, and the ability to promote early differentiation of CD11c + NK cells [2,10,11]. It allowed for the development and validation of in vitro models to contribute toward biological characterization of the product. ...
Article
Full-text available
Transferon® is a blood product with immunomodulatory properties constituted by a complex mixture of peptides obtained from a human dialyzable leukocyte extract (DLE). Due to its complex nature, it is necessary to demonstrate batch consistency in its biological activity. Potency is the quantitative measure of biological activity and is also a quality attribute of drugs. Here we developed and validated a proliferation assay using Jurkat cells exposed to azathioprine, which is intended to determine the potency of Transferon® according to international guidelines for pharmaceuticals. The assay showed a linear response (2.5 to 40 µg/mL), coefficients of variation from 0.7 to 13.6% demonstrated that the method is precise, while r2 = 0.97 between the nominal and measured values obtained from dilutional linearity showed that the method is accurate. We also demonstrated that the cell proliferation response was specific for Transferon® and was not induced by its vehicle nor by other peptide complex mixtures (glatiramer acetate or hydrolyzed collagen). The bioassay validated here was used to assess the relative potency of eight released batches of Transferon® with respect to a reference standard, showing consistent results. The collective information from the validation and the assessment of several batches indicate that the bioassay is suitable for the release of Transferon®.
... Due to the complexity of the DLE mixture, several methods identifying the individual components have been tested and validated. Additionally, the batch-to-batch reproducibility of this biological drug product was verified and established [4][5][6][7][8][9][10]. The size-exclusion HPLC method revealed individual peaks corresponding to DLE's components and their particular molecular ...
Article
Full-text available
IMUNOR is an oral biotherapeutic drug that had been developed, registered, and approved in 1997 in the Czech Republic and Slovakia. IMUNOR is a dialyzable leukocyte extract (DLE) prepared from swine leukocytes. It is characterized as a mixture of small peptides with molecular weights smaller than 12 kDa and a specific portion of nucleotides. The medical uses of IMUNOR include therapeutic applications within its registered range of indications, primarily for the treatment of immunodeficiencies, allergies, and certain acute or relapsing bacterial infections in adults and children. Despite the long-term clinical application of DLE, with strong evidence of positive therapeutic effects and no serious side effects, a detailed physicochemical specification of this mixture was lacking. We developed several methods for more in-depth physicochemical characterization of IMUNOR, including a spectrophotometric method for quantification of the total protein concentration and total DNA concentration in a mixture, several chromatographic methods for identification of individual components present in significant concentrations in IMUNOR, such as HPLC methods and the Sodium Dodecyl Sulphate Polyacrylamide Gel Electrophoresis method, and characterization of amino acid composition of this mixture. For the investigation of the variability among different batches of IMUNOR, five to nine representative batches from a standard manufacturing process on an industrial scale were utilized. Using the analytical methods, we verified and confirmed the batch-to-batch reproducibility of the biological product IMUNOR.
... The hDLE Transferon Oral has been demonstrated to increase IFN-γ levels and CD4+ cells in people with herpes zoster, reducing refractory pain compared to patients who only receive acyclovir [134], which suggests that this immunomodulator could be used in diseases with inflammatory imbalances. In a herpes simplex virus type I (HSV-1)-infected mouse model, it was observed that the hDLE increases IFN-γ and reduces TNF-α and IL-6, improving survival [135]. Furthermore, the effect of the subcutaneous administration of the immunomodulator on the conventional pharmacological treatment of puppies infected with the Canine Parvovirus (CPV) type II was evaluated. ...
Article
Full-text available
Major depressive disorder (MDD) is a mood disorder that has become a global health emergency according to the World Health Organization (WHO). It affects 280 million people worldwide and is a leading cause of disability and financial loss. Patients with MDD present immunoendocrine alterations like cortisol resistance and inflammation, which are associated with alterations in neurotransmitter metabolism. There are currently numerous therapeutic options for patients with MDD; however, some studies suggest a high rate of therapeutic failure. There are multiple hypotheses explaining the pathophysiological mechanisms of MDD, in which several systems are involved, including the neuroendocrine and immune systems. In recent years, inflammation has become an important target for the development of new therapeutic options. Extracellular monomeric ubiquitin (emUb) is a molecule that has been shown to have immunomodulatory properties through several mechanisms including cholinergic modulation and the generation of regulatory T cells. In this perspective article, we highlight the influence of the inflammatory response in MDD. In addition, we review and discuss the evidence for the use of emUb contained in Transferon as a concomitant treatment with selective serotonin reuptake inhibitors (SSRIs).
... DLEs have been used as co-adjuvants in the treatment of viral, parasitic, fungal, and mycobacterial infections, as well as primary immunodeficiencies, and allergies [157,158]. Transferon Oral ® , a human DLE (hDLE) manufactured under good manufacturing compliance, regulates the production of the inflammatory cytokines TNF-α, IL-6 e, and IFN-γ and increases the percent of survival when orally administrated in a murine model of cutaneous herpes simplex virus-1 (HSV-1) infection [159]. In addition, this hDLE increases the percent of survival of puppies infected with canine parvovirus when subcutaneously administrated by decreasing circulating levels of cortisol and catecholamines and increasing plasma levels of norepinephrine and serotonin [160,161]. ...
Article
Full-text available
Monomeric ubiquitin (Ub) is a 76-amino-acid highly conserved protein found in eukaryotes. The biological activity of Ub first described in the 1970s was extracellular, but it quickly gained relevance due to its intracellular role, i.e., post-translational modification of intracellular proteins (ubiquitination) that regulate numerous eukaryotic cellular processes. In the following years, the extracellular role of Ub was relegated to the background, until a correlation between higher survival rate and increased serum Ub concentrations in patients with sepsis and burns was observed. Although the mechanism of action (MoA) of extracellular ubiquitin (eUb) is not yet well understood, further studies have shown that it may ameliorate the inflammatory response in tissue injury and multiple sclerosis diseases. These observations, compounded with the high stability and low immunogenicity of eUb due to its high conservation in eukaryotes, have made this small protein a relevant candidate for biotherapeutic development. Here, we review the in vitro and in vivo effects of eUb on immunologic, cardiovascular, and nervous systems, and discuss the potential MoAs of eUb as an anti-inflammatory, antimicrobial, and cardio- and brain-protective agent.
... In a murine model of infection with Herpes virus type 1, the administration of the immunomodulator used in this work reduced the serum concentrations of TNF-α and IL-6, leading to an increase in survival [34]. These data suggest that the immunomodulator can reduce the circulatory levels of in ammatory cytokines, activating VN by binding mUb and Ub∆GG to CXCR4. ...
Preprint
Full-text available
Background: The canine parvoviral enteritis (CPE) promotes sepsis and systemic inflammatory response syndrome (SIRS). Mortality in this disease is usually registered within 48–72 h post-hospitalization, the critical period of the disease. It has been recently described that the use of an immunomodulator whose major component is monomeric ubiquitin (mUb) without the last two glycine residues (Ub∆GG) in pediatric patients with sepsis improves survival. It is known that CXCR4 is the cell receptor of extracellular ubiquitin in humans. The aim of this work was to explore the effect of one immunomodulator (hDLE) as a therapeutic auxiliary in CPE puppies with sepsis and SIRS. Results: We studied two groups of puppies with CPE confirmed by polymerase chain reaction. The first group received conventional treatment (CT) and vehicle (V), while the second group received CT plus an immunomodulator (I). In both groups, we assessed the survival, clinical condition, number of erythrocytes, neutrophils, and lymphocytes, and values of hematocrit, hemoglobin, plasma proteins, cortisol, and norepinephrine epinephrine, and serotonin, during hospitalization. Puppies treated with CT+I showed 81% survival and milder clinical signs and a significant decrease in circulating neutrophils and lymphocytes in the critical period of the treatment. In contrast, the CT+V group presented a survival of 42%, severe clinical status, and no improvement of the parameters evaluated in the critical period of the disease. We determined in silico that human Ub∆GG can stimulate dog CXCR4. Conclusions: The administration of an immunomodulator (5 mg/day x 5 days) to puppies with CPE under 6 months of age reduces the severity of clinical signs, increases survival, and modulates inflammatory cell parameters. Further studies are necessary to take full advantage of these clinical findings which might be mediated by the human Ub∆GG to canine CXCR4 interaction.
... Mice treated with DLE (0.1 U) exhibited a suppressed development of CIA at a level that was comparable to that observed with DEX, a potent anti-inflammatory compound used in the treatment of RA. The mechanism of action of DLEs is not known; however, some in vitro studies in humans and in small animals suggest that the TF in DLE promotes the release of IFNγ, IL-2 and TNF, as well as IL-10 and other cytokines, 31,32 while some others suggest that TF downregulates NF-κB, a critical transcription factor for several proinflammatory cytokines. 33 This observation is in agreement with our present results, where the inhibition of inflammation as a result of the treatment with DLE was correlated with the virtual absence of NF-κB activity. ...
Article
Rheumatoid arthritis is a disabling autoimmune disease with a high global prevalence. Treatment with disease‐modifying anti‐arthritic drugs (DIMARDs) has been routinely used with beneficial effects but with adverse long‐term consequences; novel targeted biologics and small‐molecule inhibitors are promising options. In this study, we investigated whether purified omega unsaturated fatty acids (ω‐UFAs) and dialysable leukocyte extracts (DLEs) prevented the development of arthritis in a model of collagen‐induced arthritis (CIA) in mice. We also investigated whether the transcription factor NF‐κB and the NLRP3 inflammasome were involved in the process and whether their activity was modulated by treatment. The development of arthritis was evaluated for 84 days following treatment with nothing, dexamethasone, DLEs, docosahexaenoic acid, arachidonic acid, and oleic acid. Progression of CIA was monitored by evaluating clinical manifestations, inflammatory changes, and histological alterations in the pads’ articular tissues. Both DLEs and ω‐UFAs led to an almost complete inhibition of the inflammatory histopathology of CIA and this was concomitant with the inhibition of NF‐kB and the inhibition of the activation of NLRP3. These data suggest that ω‐UFAs and DLEs might have NF‐κB as a common target and that they might be used as ancillary medicines in the treatment of arthritis.
... remodelamiento del microambiente hematopoyético de soporte(Ramírez-Ramírez et al., 2016). Los modelos descritos aportan al conocimiento de la farmacodinamia del producto al demostrar efectos inmunomoduladores y de estimulación de hematopoyesis temprana.Estudios de seguridad y eficacia de Transferon ® en modelos animalesExisten cuatro estudios relevantes en modelos animales que comprenden el establecimiento de una prueba para liberación de lotes hasta la generación de información de soporte de la seguridad y usos clínicos de Transferon ® .El primero de ellos fue conducido por Salinas-Jazmín y colaboradores(Salinas-Jazmín et al., 2015) en el cual desarrolló un modelo murino de infección con el virus Herpes simplex (HSV-1, cepa KOS). Este virus es capaz de inducir un efecto neuropático en los ratones que se manifiesta clínicamente con reducción de motilidad, parálisis, pérdida de peso y muerte. ...
Article
Full-text available
Canine parvovirus type II (CPV-2) infection induces canine parvoviral enteritis (CPE), which in turn promotes sepsis and systemic inflammatory response syndrome (SIRS). Mortality in this disease is usually registered within 48–72 h post-hospitalization, the critical period of the illness. It has been recently described that the use of an immunomodulator, whose major component is monomeric ubiquitin (mUb) without the last two glycine residues (Ub∆GG), in pediatric human patients with sepsis augments survival. It is known that CXCR4 is the cell receptor of extracellular ubiquitin in humans. This work aimed to explore the effect of one immunomodulator (human Dialyzable Leukocyte Extract-hDLE) as a therapeutic auxiliary in puppies with sepsis and SIRS induced by CPE. We studied two groups of puppies with CPV-2 infection confirmed by polymerase chain reaction. The first group received conventional treatment (CT) and vehicle (V), while the second group received CT plus the immunomodulator (I). We assessed both groups' survival, clinical condition, number of erythrocytes, neutrophils, and lymphocytes during the hospitalization period. In addition, hematocrit, hemoglobin, plasma proteins and cortisol values, as well as norepinephrine/epinephrine and serotonin concentration were determined. Puppies treated with CT + I showed 81% survival, mild clinical signs, and a significant decrease in circulating neutrophils and lymphocytes in the critical period of the treatment. In contrast, the CT + V group presented a survival of 42%, severe clinical status, and no improvement of the parameters evaluated in the critical period of the disease. We determined in silico that human Ub∆GG can bind to dog CXCR4. In conclusion, the administration of a human immunomodulator (0.5 mg/day × 5 days) to puppies with CPE under six months of age reduces the severity of clinical signs, increases survival, and modulates inflammatory cell parameters. Further studies are necessary to take full advantage of these clinical findings, which might be mediated by the human Ub∆GG to canine CXCR4 interaction.
Article
Full-text available
The transfer factor is an immunostimulant that is used in a wide array of diseases. Despite the fact that indications for this product are regulated, offer is generally insufficient and it is suspected that this drug is overused in daily clinical practice. Therefore, a drug utilization study was conducted in 11 hospitals located in the City of Havana to characterize the after-market prescription of this pharmaceutical. Prescribing practices were analyzed in 425 patients, which yielded that 94.1% was given the drug by the outpatient service, 70.8% of prescriptions were not included in those approved by the health registry; 56.7% was directed to treat recurrent infectious diseases and 34 different therapeutic schemes were followed. It was concluded that the use of TF in the present clinical practice does not comply with the regulations passed for this product
Article
Full-text available
Candida albicans causes opportunistic systemic infections with high mortality (30%-50%). Despite significant nephrotoxicity, amphotericin (AmB) is still used for the treatment of this serious fungal infection. Therefore, alternative treatments are urgently needed. Dialyzable leukocyte extracts have been used successfully to treat patients with mucocutaneous candidiasis, but their effectiveness in systemic candidiasis has not been evaluated. In this study, low-dose AmB (0.1 mg/kg) plus 10 pg of murine dialyzable spleen extracts (mDSE) were tested in a systemic candidiasis mouse model. Survival, tissue fungal burden, kidney damage, kidney cytokines, and serum levels of IL-6 and hepcidin were evaluated. Our results showed that the combined treatment of low-dose AmB plus mDSE improved survival and reduced kidney fungal burden and histopathology; these effects correlated with increased kidney concentration of IFN- γ and TGF- β 1, decreased levels of TNF- α , IL-6, and IL-10, as well as high levels of systemic IL-6 and hepcidin. Low-dose AmB and mDSE synergized to clear the infectious agent and reduced tissue damage, confirming the efficacy of a low dose of AmB, which might decrease the risk of drug toxicity. Further studies are necessary to explore these findings and its implications in future therapeutic approaches.
Article
Full-text available
Human dialyzable leukocyte extracts (DLEs) are heterogeneous mixtures of low-molecular-weight peptides that modulate immune responses in various diseases. Due their complexity, standardized methods to identify their physicochemical properties and determine that production batches are biologically active must be established. We aimed to develop and validate a size exclusion ultra performance chromatographic (SE-UPLC) method to characterize Transferon™, a DLE that is produced under good manufacturing practices (GMPs). We analyzed an internal human DLE standard and 10 representative batches of Transferon™, all of which had a chromatographic profile characterized by 8 main peaks and a molecular weight range between 17.0 and 0.2kDa. There was high homogeneity between batches with regard to retention times and area percentages, varying by less than 0.2% and 30%, respectively, and the control chart was within 3 standard deviations. To analyze the biological activity of the batches, we studied the ability of Transferon™ to stimulate IFN-γ production in vitro. Transferon™ consistently induced IFN-γ production in Jurkat cells, demonstrating that this method can be included as a quality control step in releasing Transferon™ batches. Because all analyzed batches complied with the quality attributes that were evaluated, we conclude that the DLE Transferon™ is produced with high homogeneity.
Article
Full-text available
To investigate the effect of HSV-1 infection via TLR3 on the transcriptional activity of NF-kappaB and the expression of cytokines TNF-alpha and IL-6 in astrocytes. HSV-1-infected primary astrocytes were cultured until the third passage and the mRNA and protein levels of TLR3, NF-kappaB, TNF-alpha, and IL-6 were assessed by immunofluorescence, RT-PCR, and Western blot. The effects of the NF-kappaB inhibitor pyrrolidine dithiocarbamate (PDTC) and TLR3-neutralizing antibody on the expression of NF-kappaB, TNF-alpha, and IL-6 were investigated. Uninfected astrocytes expressed TLR3 and NF-kappaB at the mRNA and protein levels. After infection with HSV-1, the TLR3 mRNA and protein levels were up-regulated and NF-kappaB protein was highly expressed. Also, the mRNA and protein levels of TNF-alpha and IL-6 were up-regulated. Pyrrolidine dithiocarbamate inhibited NF-kappaB activation, resulting in the down-regulation of nuclear NF-kappaB protein, which led to the down-regulation of the mRNA and protein levels of TNF-alpha and IL-6. After blocking astrocyte membrane TLR3, the nuclear NF-kappaB protein expression was down-regulated and the mRNA and protein levels of TNF-alpha and IL-6 were increased. The antiviral functions of astrocytes were weaker, as reflected by higher HSV-1 glycoprotein D (gD) mRNA expression and increased HSV-1 titers. Astrocytes infected with HSV-1 can activate NF-kappaB via TLR3 so as to up-regulate the expression of TNF-alpha and IL-6 that have antiviral functions.
Article
Full-text available
Objective. The objective of this study was to prepare, characterize, and determine immunological activities of specific transfer factor (STF) specific to human sperm antigen (HSA) for the preparation of antisperm contraceptive vaccine that can be used as an immunocontraceptive. Methods. HSA-STF was prepared using the spleens of rabbits vaccinated with HSA. The specific immunological activities were examined by lymphocyte proliferation test (LPT), leukocyte adhesion inhibition test (LAIT), and by determining the concentrations of IL-4, γ -IFN, and IL-21. HSA-STF was a helveolous substance, having a pH value of 7.0 ± 0.4 and UV absorption maxima at 258 ± 6 nm. It contained seventeen amino acids; glycine and glutamic acids were the highest in terms of concentrations (38.8 μ g/mL and 36.3 μ g/mL, resp.). Results. The concentration of polypeptide was 2.34 ± 0.31 mg/mL, and ribose was 0.717 ± 0.043 mg/mL. The stimulation index for lymphocyte proliferation test was 1.84, and the leukocyte adhesion inhibition rate was 37.7%. There was a statistically significant difference between the cultural lymphocytes with HSA-STF and non-HSA-STF for γ -IFN and IL-21 (P < 0.05), but there was no statistical significance for IL-4 (P > 0.05). Conclusion. HSA-STF was prepared and characterized successfully. It had immunological activity which could transfer the immune response specific to HSA and prove to be a potential candidate for the development of male immunocontraceptive agents.
Article
Full-text available
In the biological sciences the TCID50 (median tissue culture infective dose) assay is often used to determine the strength of a virus. When the so-called Spearman-Kaerber calculation is used, the ratio between the pfu (the number of plaque forming units, the effective number of virus particles) and the TCID50, theoretically approaches a simple function of Eulers constant. Further, the standard deviation of the logarithm of the TCID50 approaches a simple function of the dilution factor and the number of wells used for determining the ratios in the assay. However, these theoretical calculations assume that the dilutions of the assay are independent, and in practice this is not completely correct. The assay was simulated using Monte Carlo techniques. Our simulation studies show that the theoretical results actually hold true for practical implementations of the assay. Furthermore, the simulation studies show that the distribution of the (the log of) TCID50, although discrete in nature, has a close relationship to the normal distribution. The pfu is proportional to the TCID50 titre with a factor of about 0.56 when using the Spearman-Kaerber calculation method. The normal distribution can be used for statistical inferences and ANOVA on the (the log of) TCID50 values is meaningful with group sizes of 5 and above.
Article
Full-text available
In response to the need for new antiviral agents, dendrimer-based molecules have been recognized as having a large number of potential therapeutic applications. They include peptide-derivatized dendrimers, which are hyperbranched synthetic well-defined molecules which consist of a peptidyl branching core and covalently attached surface functional peptides. However, few studies have addressed their applications as direct-acting antiviral agents. Here, we report on the ability of the peptide dendrimer SB105 and its derivative, SB105_A10, to directly inhibit herpes simplex virus 1 (HSV-1) and HSV-2 in vitro replication, with favorable selective indexes discerned for both compounds. An analysis of their mode of action revealed that SB105 and SB105_A10 prevent HSV-1 and HSV-2 attachment to target cells, whereas SB104, a dendrimer with a different amino acid sequence within the functional group and minimal antiviral activity, was ineffective in blocking HSV attachment. Moreover, both SB105 and SB105_A10 retained their ability to inhibit HSV adsorption at pH 3.0 and 4.0 and in the presence of 10% human serum proteins, conditions mimicking the physiological properties of the vagina, a potential therapeutic location for such compounds. The inhibition of HSV adsorption is likely to stem from the ability of SB105_A10 to bind to the glycosaminoglycan moiety of cell surface heparan sulfate proteoglycans, thereby blocking virion attachment to target cells. Finally, when combined with acyclovir in checkerboard experiments SB105_A10 exhibited highly synergistic activity. Taken together, these findings suggest that SB105 and SB105_A10 are promising candidates for the development of novel topical microbicides for the prevention of HSV infections.
Article
Full-text available
The regulation of biosimilars is a process that is still developing. In Europe, guidance regarding the approval and use of biosimilars has evolved with the products under consideration. It is now more than 3 years since the first biosimilar agents in oncology support, erythropoiesis-stimulating agents, were approved in the EU. More recently, biosimilar granulocyte colony-stimulating factors have received marketing approval in Europe. This review considers general issues surrounding the introduction of biosimilars and highlights current specific issues pertinent to their use in clinical practice in oncology. Information on marketing approval, extrapolation, labelling, substitution, immunogenicity and traceability of each biosimilar product is important, especially in oncology where patients are treated in repeated therapy courses, often with complicated protocols, and where biosimilars are not used as a unique therapy for replacement of e.g. growth hormone or insulin. While future developments in the regulation of biosimilars will need to address multiple issues, in the interim physicians should remain aware of the inherent differences between biosimilar and innovator products.
Article
Full-text available
The effect of dialysable leucocyte extract (transfer factor TF) on immune response of mice infected with Echinococcus multilocularis and treated with albendazole (ABZ) was observed. TF administration increased the parasite-suppressed proliferative response of T and B lymphocytes of infected mice from weeks 8 to 12 or 14 post infection (p.i.), respectively, with the most stimulative effect after TF+ABZ therapy. The CD4 T cell presence in the spleen of infected mice with TF or TF+ABZ therapy was increased from weeks 6 to 12 or 14 p.i., respectively. The production of IFN-γ (Th1 cytokine) after TF or TF+ABZ therapy was significantly higher from weeks 6 to 12 p.i., and during this time, the significantly inhibited IL-5 synthesis (Th2 cytokine) was detected, particularly after TF+ABZ therapy. The superoxide anion (O 2−) production in peritoneal macrophages of infected mice treated with TF or TF+ABZ was stimulated from weeks 8 to 18 p.i. The immunomodulative effect of TF reduced the growth of larval cysts till week 14 p.i. with a comparable intensity to the anthelmintic drug ABZ. Combined therapy TF+ABZ resulted in the greatest parasite restriction and reduced the cyst development till the end of the experiment.
Article
Transfer factor (TF) is a low-molecularweight lymphocyte extract capable of transferring antigen-specific cell-mediated immunity (CMI) to T lymphocytes. It has been used successfully as an adjuvant or primary therapy for viral, parasitic, fungal, and some bacterial infections, as well as immunodeficiencies, neoplasias, allergies and autoimmune diseases. From the list of infections that seem to respond noticeably to transfer factor, those due to viruses of the herpes family are particularly remarkable. Indeed, for these viruses it was shown that TF can prevent infection or relapse, acting as a CMI vaccine. Data also suggest its possible use for adjuvant treatment and probably prevention of two currently widespread infections: tuberculosis and AIDS. Furthermore, TF has an interesting potential: answering the challenge from unknown pathogenic agents, a black box effect permitting production of antigen-specific TF to a new pathogen, even before its identification. It thus seems that the preventative potential of transfer factor is as important as its therapeutic one, both discussed in this review.