Вы находитесь на странице: 1из 11

Toxicon 151 (2018) 63–73

Contents lists available at ScienceDirect

Toxicon
journal homepage: www.elsevier.com/locate/toxicon

Current technology for the industrial manufacture of snake antivenoms T



Guillermo León , Mariángela Vargas, Álvaro Segura, María Herrera, Mauren Villalta,
Andrés Sánchez, Gabriela Solano, Aarón Gómez, Melvin Sánchez, Ricardo Estrada,
José María Gutiérrez
Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica

A R T I C LE I N FO A B S T R A C T

Keywords: Snake antivenoms are formulations of animal immunoglobulins used in the treatment of snakebite envenoma-
Snake tion. The general scheme for producing snake antivenoms has undergone few changes since its development
Venoms more than a century ago; however, technological innovations have been introduced in the manufacturing pro-
Envenomation cess. These medicines must comply with identity, purity, safety and efficacy profiles, as requested by the current
Antivenom
Good Manufacturing Practices (GMPs) applied to modern biopharmaceutical drugs. Industrial production of
Industrial biotechnology
snake antivenoms comprises several stages, such as: 1) production of reference venom pools, 2) production of
hyperimmune plasma, 3) purification of the antivenom immunoglobulins, 4) formulation of the antivenom, 5)
stabilization of the formulation, and 6) quality control of in-process and final products. In this work, a general
review of the existing technology used for the industrial manufacture of snake antivenoms is presented.

1. Introduction proteins). Notwithstanding, most of the adverse reactions induced by


current antivenoms have been related to the inherent immunochemical
Snakebite envenomation is an important public health problem in characteristics of heterologous immunoglobulins, such as antic-
most tropical countries around the world (Kasturiratne et al., 2008; omplementary activity and immunogenicity (León et al., 2013).
WHO, 2016). Intravenous administration of specific antivenoms is the Pre-clinical efficacy of antivenoms is usually assessed based on the
only scientifically demonstrated therapy to control snakebite en- anti-lethal potency at which they are formulated, i.e. the capacity of
venomations (WHO, 2016). Antivenoms are preparations of im- antivenoms to neutralize venom-induced lethality (Gutiérrez et al.,
munoglobulins (i.e., whole molecules or their F(ab′)2 or Fab fragments) 2017). However, evaluation of the ability of antivenoms to recognize
purified from the plasma of animals immunized with snake venoms the venom toxins and neutralize hemorrhagic, defibrinogenating,
(Gutiérrez et al., 2011). myotoxic, necrotizing, edema-forming, and neuromuscular blocking
As pharmaceutical products, antivenoms must comply with identity, activities, depending on the venom, offer additional information that
purity, safety and efficacy profiles, according to Good Manufacturing contributes to the prediction of their clinical performance (Gutiérrez
Practices (GMPs). The identity of antivenoms is determined by the et al., 2013).
animal species used as immunoglobulin source and the venoms used as Besides the pre-clinical estimation of antivenom potency, the effi-
immunogens (e.g., equine anti-bothropic immunoglobulins; León et al., cacy of antivenoms in the clinical setting is affected by other factors
2016). Purity is determined by the combination of the neutralizing that do not depend on antivenom formulation, such as: 1) severity of
activity of hyperimmune raw plasma and the performance of the envenomation, 2) specificity of the antivenom towards the venom of the
methods used to separate the neutralizing immunoglobulins from other snake causing the envenomation, 3) time between snakebite and anti-
plasma proteins. Consequently, the potency/total protein ratio is a good venom administration, 4) route by which the antivenom is injected, and
parameter to evaluate antivenom purity (Segura et al., 2012). 5) dose of antivenom administered. Therefore, the only way to de-
On the other hand, safety refers to the assurance that antivenoms monstrate and guarantee the clinical efficacy and safety of an anti-
are free of microbial contaminants (e.g., bacteria or their endotoxins, venom formulation is through properly conducted clinical studies, such
fungi, virus or prions) and have satisfactory physicochemical char- as dose-finding trials and especially through controlled, double-blind,
acteristics (e.g., low concentration of total protein, low content of im- randomized studies (WHO, 2016).
munoglobulin aggregates and minimal content of non-immunoglobulin Large-scale production of clinically validated antivenoms requires


Corresponding author.
E-mail address: guillermo.leon@ucr.ac.cr (G. León).

https://doi.org/10.1016/j.toxicon.2018.06.084
Received 7 March 2018; Received in revised form 28 May 2018; Accepted 25 June 2018
Available online 28 June 2018
0041-0101/ © 2018 Elsevier Ltd. All rights reserved.
G. León et al. Toxicon 151 (2018) 63–73

the implementation of industrial procedures for: 1) production of re- immunization and quality control activities and the yield of venom of
ference venom pools, 2) production of hyperimmune plasma, 3) pur- each species. For this calculation, it must be also considered that venom
ification of the antivenom immunoglobulins, 4) formulation of the an- yield decreases throughout life in specimens maintained in captivity
tivenom, 5) stabilization of the formulation, and 6) quality control of (Bolaños, 1972; our unpublished observations). An example related to
in-process and final products required to verify that all antivenom antivenoms for sub-Saharan Africa is the contrast between two medi-
batches in the production line meet the product specifications. All these cally-relevant species, Echis ocellatus and Bitis arietans. The former is a
procedures should be carried out according to GMP principles (WHO, relatively small snake with a low venom yield, whereas the latter gives
2016). In this work, a general review of the current technologies used a large volume of venom. Therefore, the number of specimens that need
for the industrial manufacture of snake antivenoms is presented. Since to be kept in captivity for regular venom production is much higher in
most antivenom producers usually do not publish their industrial pro- the case of E. ocellatus than in B. arietans. A similar scenario can be
cedures, the information in this review is mainly based on the methods found with the species Micrurus nigrocinctus and Bothrops asper, where
tested and used by Instituto Clodomiro Picado. M. nigrocinctus is difficult to keep in captivity (Chacón et al., 2012) and
many snakes are needed to provide the venom quota used in antivenom
2. Production of reference venom pools manufacture. On the other hand, species such as B. asper produce large
quantities of venom; therefore, the number of snakes to be maintained
Despite the fact that snake venom toxins can be produced by re- in captivity is lower.
combinant technologies (Li et al., 2017) or from cultures of venom Venoms are obtained by mechanical massage of venom glands and
gland cells (Carneiro et al., 2006), virtually all antivenom manu- collected in vessels, which material must be validated in order to pre-
facturers still utilize venoms produced by mechanical stimulation of the vent the loss of relevant toxins owing their protein-binding properties
venom glands, i.e. ‘milking’, of specimens maintained in captivity (Goebel-Stengel et al., 2011). During this procedure, snake handling
(Chanhome et al., 2001; Chacón et al., 2012; Sasa et al., 2017), or of can be facilitated by short-acting general anesthesia (e.g., CO2 inhala-
wild specimens that, after venom collection, are released to nature (e.g., tion). However, this practice may result in a reduction of the venom
venoms produced by the Irula snake catchers cooperative in India). yield. To protect fresh venoms from auto-degradation (Sousa et al.,
Collections maintained in captivity can be composed by animals 2001) they must be frozen as soon as possible after collection, and
collected from nature or/and by ex-situ bred specimens (Corrales et al., stored at −20 °C until their stabilization.
2014). In both cases, the health of the snakes must be closely monitored Reference venom pools are used as immunogens, i.e. to stimulate
by qualified veterinarians. In addition to clinical examination, de- the immune system of the animals selected as source of antivenom
termination of normal hematological and plasma-chemistry values is antibodies (da Silva and Tambourgi, 2011; León et al., 2011), and to
important to achieve this goal (Gómez et al., 2016; Muliya and Bhat, test the ability of the raised antibodies to neutralize the toxic effects
2016). induced by the venoms (i.e., used for quality control; Gutiérrez et al.,
The selection of species used as source of venom for antivenom 2017). Therefore, reference venom pools must keep the antigenic and
manufacture is mainly based on their medical importance (WHO, toxic properties of fresh venoms of wild specimens (Farias et al., 2018)
2016). Nonetheless, intraspecific variability of venoms (i.e., individual, during long-term storage (Jesupret et al., 2014; Hatakeyama et al.,
ontogenic, wild/captive and regional variability) must be considered to 2018).
define the characteristics of the specimens (i.e., minimum number of Stabilization of venoms is achieved by desiccation or freeze-drying.
specimens, age, sex and geographical origin) used to produce reference During desiccation, the liquid venom at room temperature is submitted
venom pools which relative composition guarantees the representa- to negative pressure until all the water has evaporated, which normally
tiveness of venoms of wild snakes in the region (Chippaux et al., 1991; takes several days. In these conditions, the venom proteins experience
Gutiérrez et al., 2009, 2010; WHO, 2016). high and sustained physical stress that could produce some denatura-
Important proteomic, toxicological and/or antigenic differences tion and loss of activity and antigenicity. During freeze-drying, the solid
have been demonstrated in venoms of geographically separated popu- venom, frozen at temperatures as low as – 40 °C, is exposed to negative
lations of snakes such as the monocled cobra (Naja kaouthia) from pressure during several days. In these conditions, the water is sub-
Malaysia, Thailand and Vietnam (Tan et al., 2015a); the Russell's viper limated, exposing the venom proteins to very low physical stress.
(Daboia russelii) from different regions of India (Kalita et al., 2017); the Depending on the composition of the venom and the performance of the
Indian krait (Bungarus caeruleus) from India (Oh et al., 2017), Pakistan procedure, the toxic activities of dried venoms can differ from those of
and Sri Lanka; and the spectacled cobra (Naja naja) from India, Pakistan the original fresh venoms (Meier et al., 1991). Therefore, procedures for
and Sri Lanka (Wong et al., 2018). On the other hand, some evidence venom stabilization must be carefully selected, standardized and vali-
suggests that environmental conditions could also produce variations dated.
between venoms of wild and captive snakes (Modahl et al., 2010; Saad Frequently, venom freeze-drying is achieved under poorly con-
et al., 2012). However, recent studies show that this is not a general trolled conditions, and this is an aspect that demands improvement in
rule (Freitas-de-Sousa et al., 2015; McCleary et al., 2016). venom-producing laboratories. Usually, venoms previously frozen at
The relative composition of the reference venom pools (i.e. the −20 °C are cooled down to −70 °C for 18 h, in a laboratory freezer.
proportion of venoms of a particular number of specimens with dif- Then, venom is placed in a flask of a benchtop manifold freeze-drier,
ferent age, sex and geographical origin) must be determined based on where sublimation (primary drying) is performed at room temperature
detailed biochemical and toxicological analysis of venom variability. and 100–200 mTorr, a process that lasts for a period that depends on
Only in this way the representativeness of reference venom pools can be the volume of venom. Since heat is transferred to the product from the
guaranteed (Calvete et al., 2014). In the absence of such studies, surrounding environment, the desorption step (the secondary drying)
composition of reference venom pools could be arbitrarily defined, i.e. cannot be properly carried out with this type of equipment. The best
6–12 specimens of each age/sex group to ensure heterogeneity, as well control of the process is accomplished if a shelf freeze dryer is used.
as including similar groups from at least the northern, southern, wes- Quality control of reference venom pools must include the toxicological
tern and eastern populations of the species, since these are likely to and biochemical characterization of the venoms (e.g., quantification of
have the greatest variation. venom activities such as lethal, hemorrhagic, necrotizing, proteolytic,
After defining the relative composition of the reference venom phospholipase A2, coagulant and fibrinolytic activities, together with
pools, they must be produced on an industrial scale. The number of the analysis of SDS-PAGE and reverse phase HPLC profiles; Camey
specimens required for industrial production of antivenoms is directly et al., 2002; WHO, 2016). The characterization of many different bat-
related to the ratio between the amount of venom consumed in ches of venom is necessary to determine reference values that allow the

64
G. León et al. Toxicon 151 (2018) 63–73

identification of defective batches. emulsions (e.g., complete and incomplete Freund's adjuvants; Valverde
Characterization of reference venom pools must also include their et al., 2017) and mineral salts (e.g. aluminum hydroxide, calcium
performance in the determination of the neutralizing potency of anti- phosphate; Olmedo et al., 2013). Freund's adjuvants are the most ef-
venoms. This is perhaps the most important characteristic that a re- fective for enhancing antibody response towards snake venoms (da
ference venom pool must achieve. We have unpublished evidence in- Silva and Tambourgi, 2011; León et al., 2011). However, these ad-
dicating that two different batches of venom with similar toxicity juvants induce adverse effects such as local tissue damage, granulomas,
profiles can yield different results when used in the estimation of po- abscesses and general reactions such as arthritis and fever, and there-
tency of a single batch of antivenom. This outcome may be due to fore their use is regulated by international guidelines (Fukanoki et al.,
structural changes induced by stress experienced by toxins during 2001; Jansen et al., 2007), and should be limited to the first injections
freeze-drying (Heller et al., 1999), but this hypothesis needs to be ex- of the immunization schedule. The use of Freund's adjuvants to ad-
perimentally addressed. This example illustrates that the performance minister routine boosters could affect the general health of the animals
of reference venom pools in the determination of the neutralizing and produce unnecessary suffering that is unlike to result in better
ability of antivenoms should be controlled by using reference anti- neutralizing responses.
venoms (Fukuda et al., 2006; Araújo et al., 2008, 2017). After priming immunization, routine boosters can be administered
by using safer adjuvants such as aluminum salts and bentonite colloids,
3. Production of hyperimmune plasma which adjuvant activity is attributed to its ability to form depots from
which venoms are slowly released, producing a sustained stimulation to
It is likely that, in the future, antivenoms will be formulated in- the immune system (León et al., 2011). However, since animals im-
cluding chemical inhibitors of toxins, or human antibodies produced by munized with snake venoms coupled to aluminum salts or bentonite
recombinant technologies (Laustsen et al., 2016). However, the current develop low titers of antivenom antibodies (Sadahiro et al., 1981;
industrial production of antivenoms is based on the immunization of Sunthornandh and Ratanabanangkoon, 1994), these adjuvants are
animals and the fractionation of hyperimmune plasma as source of rarely used. In contrast, many producers immunize their animals using
antivenom immunoglobulins. no adjuvant at all, i.e. with venom dissolved in plain saline solution.
Some new adjuvants whose use is likely to increase in the next years
3.1. Immunization of animals are liposomes (León et al., 2011) and the IMS 3012 nanoparticle ad-
juvant which have shown the ability to induce high titres of neu-
Horses and sheep are the species preferred as source of antivenom tralizing antibodies towards venoms of Naja naja, Vipera ruselli, Bun-
immunoglobulins for the industrial production of antivenoms, and most garus caeruleus and Ecuis carinatus from India with minimal tissue
manufacturers use horses. In the past, it was suggested that ovine an- damage (Waghmare et al., 2009).
tivenoms are safer than equine antivenoms (Landon and Smith, 2003), Owing to their toxicity, snake venoms can produce tissue damage at
but clinical evidence has not shown major differences in the incidence the injection site. Inactivation of venom by chemical or physical
of adverse reactions induced by both types of antivenoms (Abubakar treatments is not recommended because these procedures might affect
et al., 2010). Some experimental antivenoms have been developed by the antigenicity of the toxins and consequently reduce the efficacy of
using other species such as camelids and chickens (León et al., 2011), the antivenom towards the native venom (Theakston et al., 2003).
but they have not been used in clinical trials yet. Thus, the use of native venoms is usually preferred for immunization.
Immunization can be done by using venoms of one or several spe- Traditionally, immunization with several venoms has been per-
cies, to produce monospecific or polyspecific antivenoms, respectively formed by injecting a mixture of the venoms in a single anatomical site.
(Gutiérrez et al., 2011). Use of monospecific antivenoms is suitable only However, Chotwiwatthanakun et al. (2001) have obtained higher
when the identification of the species causing the envenomation is neutralizing responses by distributing the venom injections in multiple
possible based on the clinical characteristics (Gutiérrez et al., 2011) or anatomical sites (i.e., the low dose, low volume, multi-site immuniza-
the availability of kits for venom detection (Trevett et al., 1995). tion protocol). This method has the additional advantage of reducing
However, when the identification of the offending snake is not feasible, tissue damage induced by venoms and adjuvants.
the use of polyspecific antivenoms is usually preferred (Gutiérrez et al., Using a variation of the low dose, low volume, multi-site im-
2009; WHO, 2016). munization protocol, and replacing crude venoms by ultrafiltration-
The selection of venoms used as immunogens is based on the fractions composed by toxins with low molecular weight,
medical importance of the snakes (WHO, 2016). However, raised an- Ratanabanangkoon et al. (2016) eliminated antigens with low toxicity
tibodies can neutralize not only the venoms used during immunization in some venoms and were able to increase the variety of venoms in the
(homologous venoms), but also the venoms from other species (het- immunogen mixture that they used. In this way, they induced in horses
erologous venoms), which have antigenic similarity with the homo- an antibody response with wide paraspecificity against major medically
logous venoms. Cross-neutralization studies are required to determine important elapids in Asia. This strategy could be readily adapted to
the neutralization scope of antivenoms and to identify venoms or produce antivenoms against other elapids.
venom fractions that must be included in the immunizing mixture to On the other hand, venoms can influence the antibody response
guarantee the efficacy of the antivenom in the geographical region towards other venoms used as co-immunogens. Thus, removal of the
where it is going to be used (Tan et al., 2015b; Ratanabanangkoon immunosuppressive components of the venoms, or injection of venoms
et al., 2016; Wong et al., 2016; Sánchez et al., 2017; Solano et al., at different time intervals, are strategies that can be used to improve the
2018). antibody response obtained by the traditional method (Stephano et al.,
During immunization, repeated injections of venoms are performed 2005; Arroyo et al., 2015, 2017). It is therefore relevant that manu-
to the animals, generally by the subcutaneous route, to induce the facturers assess the immunological interactions between the venoms
production of neutralizing antibodies (da Silva and Tambourgi, 2011; used for immunization to design immunization protocols that minimize
León et al., 2011). DNA immunization has been assessed as an alter- negative immunomodulatory effects.
native immunization strategy for antivenom production (Wagstaff Independently of the immunization strategy used, the rise in neu-
et al., 2006; Arce-Estrada et al., 2009). However, the titer of neu- tralizing potency in plasma due to the increment of specific antivenom
tralizing antibodies obtained by this method did not compete with that antibodies has an upper limit value, which is not possible to overcome,
obtained by directly injecting the venom. and depends on the physiological restrictions of the animal. However,
Production of antivenom antibodies is potentiated by mixing the since this upper limit is unknown for each animal, the search for better
venoms with immunological adjuvants, such as some simple or multiple immunization strategies is a permanent task. At the end of the

65
G. León et al. Toxicon 151 (2018) 63–73

immunization schedule, neutralizing potencies reached by individual used by other laboratories.


animals usually show a normal distribution. While most animals de- Throughout bleeding, a strict aseptic technique is mandatory to
velop intermediate potencies around an average value, some animals prevent microbial contamination of the blood. Before jugular veni-
develop lower or higher potencies (our unpublished observations). puncture, the skin of the horse must be shaved, washed and disinfected.
The determination of the minimum plasma potency required to The hands of the operator must be also washed and disinfected.
establish a sustainable production of effective, safe and low-cost anti- Protection of plasma by the addition of thimerosal, phenol or cresol,
venoms requires a careful analysis that must consider: 1) plasma po- and storage at 2–8 °C contribute to preserve the microbiological quality
tencies that are feasible to reach with the immunization strategy used, of the plasma. In the case that plasma must be stored during several
2) possibility of concentrating the antivenom immunoglobulins during months before process, freezing storage could be an option to prevent
the formulation process, and 3) maximal dose (i.e., volume) of anti- microbial growth.
venom that could be administered in patients to ensure a therapeutic During decades, antivenom producers used autoclaved glass bottles
effect. with an anticoagulant such as Acid Citrate Dextrose (ACD) to collect the
Some researchers have estimated the therapeutic dose of anti- blood (Levine and Broderick, 1970). Plasma can be separated, after
venoms from the relationship between the amount of venom that a sedimentation of red blood cells, by aspiration with a mechanical
snake can inject and the potency of the antivenom determined in a pump, in a clean environment. Alternatively, closed systems of sterile
murine model (Simpson and Norris, 2007). However, this approach is plastic bags containing an anticoagulant can be used. With this system,
likely to yield inaccurate results, because it does not consider the re- plasma is protected from microbial contamination, with minimal en-
lationship between the toxicokinetics of venom toxins and the phar- vironmental control. The rate of contaminated bags can be as low as
macokinetics of antivenom immunoglobulins (Gutiérrez et al., 2003), 0.1% (our unpublished observations).
nor that the potency of an antivenom varies depending on the venom Automated plasmapheresis has been suggested as a cleaner and
challenge dose used in the assay (Solano et al., 2010). The antivenom more controlled method to harvest equine plasma (Ziska et al., 2012)
dosage to be used in therapy can only be defined based on dose-finding with a lower extent of cellular contamination (Feige et al., 2003).
clinical trials. However, the yield and microbiological quality of plasma obtained by
Determination of the minimum plasma potency used for industrial this method are not necessarily better than those obtained by using
production of antivenoms has also economic implications that must be autoclaved bottles or plastic bags. Prospective comparison of manual
considered: if only high-potency plasmas are used, less vials of more and automated plasmapheresis methods regarding cost-effectiveness,
potent but more expensive antivenom will be produced, than if all the safety and animal welfare risk-benefit, is a pending task.
plasmas (including those of lower potency) are used. Once the specifi- The processing of plasmas obtained by using a poor aseptic tech-
cation of plasma potency has been established, animals that regularly nique is the principal cause of in-process and final product con-
do not comply with this standard must be separated from the group and tamination with bacterial endotoxins. Therefore, the microbiologic
replaced by new ones. quality of plasma should be carefully controlled. Specifications of en-
dotoxin content in plasma should be at least as strict as those for en-
3.2. Bleeding of animals dotoxin content in the final product (see below).

After immunization, antivenom antibodies-enriched blood is col- 4. Purification of antivenom immunoglobulins


lected. Animals subjected to bleeding must be in good physical condi-
tions, as previously determined by auscultation of cardiorespiratory and Few years after starting the clinical use of antivenoms, which in-
digestive systems, conjunctival and gingival mucosal examination, ca- itially consisted of crude hyperimmune serum (Calmette, 1894), it was
pillary filling test, overall clinical evaluation, and determination of suggested that purification of the active substance of antivenoms was
hemoglobin and hematocrit values. relevant to concentrate the neutralizing potency of the product and to
Normal ranges of hematocrit and hemoglobin change from one reduce the high incidence of adverse reactions induced by crude serum
breed of animal to another. Very often, these values are lower in horses or poorly-purified immunoglobulin preparations. Then, several techni-
subjected to snake venom immunization when compared to normal ques based on differential precipitations of proteins (Atkinson, 1900;
horses of the same breed (Angulo et al., 1997). In general terms, horses Gibson, 1906; Steinbuch and Audran, 1969) and enzyme digestion
with hematocrit and hemoglobin values lower than 30% and 10 g/dL, (Parfentjev, 1936; Pope, 1939) were gradually introduced in the pro-
respectively should not be considered for large-scale bleeding. Con- tocols of antivenom manufacture.
versely, animals with hematocrit higher than 45% may be suffering Currently, there is a diversity of methods used to purify im-
from dehydration and should not be subjected to bleeding either. munoglobulins from hyperimmune plasma at the industrial scale (e.g.,
Industrial bleeding/self-transfusion of horses can be performed pepsin digestion, salting out, caprylic acid precipitation, anionic ex-
following a variety of protocols. For example, in the protocol described change chromatography and ultrafiltration). Indeed, there is not a
by Angulo et al. (1997), horses weighing between 450 and 550 kg were standard protocol to purify antivenoms, and manufacturers use dif-
subjected to bleeding/self-transfusion during four consecutive days. ferent methodologies (WHO, 2016). Examples of protocols used to
During the first three days, volumes of blood corresponding to 1.5%, produce antivenoms based on whole Igs or F(ab′)2 fragments are shown
1.5% and 0.8% of body weight were harvested, and no blood was in Figs. 1 and 2, respectively. As a rule, the method that renders the
collected in the fourth day. In the second, third and fourth days, self- highest purification with the fewest number of steps should be pre-
transfusion of red blood cells collected the preceding day was per- ferred. However, each laboratory selects the techniques included in its
formed to prevent animals from developing anemia. It has been de- production protocols, establishes the sequence in which they are per-
monstrated that equine erythrocytes can be stored for at least 35 days formed, and defines the conditions for each procedure, according to its
before transfusion without apparent damage (Niinistö et al., 2007). The own criteria. Consequently, many producers have unique protocols to
last step of the procedure was the hydration of animals by the in- manufacture their products, and therefore there is great variation be-
travenous administration of 19 L of Ringer's lactate solution. After a 2 tween products in terms of quality and production costs.
months rest period, animals were completely recovered and ready to In general, following the purification steps, the immunoglobulin-
undergo a new immunization/bleeding cycle. Using this protocol, it is rich fractions are recovered by microfiltration or centrifugation (Rojas
possible to obtain yields of 10–12 L of plasma/horse, without producing et al., 1994; Mpandi et al., 2007), dialyzed or diafiltered for removing
adverse acute or chronic physiological alterations in the animals the low molecular mass components added (Herrera et al., 2009), and
(Angulo et al., 1997). Other strategies of bleeding/self-transfusion are concentrated by tangential flow ultrafiltration to reach the neutralizing

66
G. León et al. Toxicon 151 (2018) 63–73

Fig. 1. Scheme of a general process to produce


antivenoms composed of whole Igs purified by
caprylic acid precipitation. Green boxes at the
periphery depict quality control assays that
need to be done at various points in the pro-
duction line. (For interpretation of the refer-
ences to colour in this figure legend, the reader
is referred to the Web version of this article.)

potency specification (Rosenberg et al., 2009). In some cases, buffer responsible of the early adverse reactions induced by the intravenous
exchange is required as preparation for the next purification step. administration of equine immunoglobulins, and that F(ab’)2-based an-
tivenoms were safer than IgG-based antivenoms. However, it has been
shown that the incidence and severity of early adverse reactions in-
4.1. Enzyme digestion duced by antivenoms do not depend on whether the product is com-
posed of whole IgGs or F(ab’)2 fragments and, instead, are likely due to
Digestion of equine plasma with pepsin is performed to eliminate the physicochemical quality of the products (León et al., 2013).
non-immunoglobulin proteins (e.g., fibrinogen and albumin) and to Nevertheless, the majority of antivenom manufacturers use pepsin di-
remove the Fc fragment of immunoglobulins to obtain F(ab’)2 frag- gestion to generate F(ab′)2-based products.
ments. Previously, it was assumed that the presence of Fc fragment was

Fig. 2. Scheme of a general process to produce


antivenoms composed by (F(ab′)2 fragments
purified by salting out with ammonium sulphate.
Green boxes at the periphery depict quality
control assays that need to be done at various
points in the production line. (For interpretation
of the references to colour in this figure legend,
the reader is referred to the Web version of this
article.)

67
G. León et al. Toxicon 151 (2018) 63–73

Traditionally, pepsin digestion is performed by using whole plasma concentration of 14–16% (w/v) and the mixture is stirred for 1 h. Then
as substrate. However, digestion can be also carried out on purified the mixture is microfiltered or centrifuged to remove the precipitate,
immunoglobulins (Pépin-Covatta et al., 1997). Typical digestion with which contains mainly fibrinogen. Afterwards, ammonium sulfate
pepsin is performed by adjusting the pH and temperature of plasma at concentration in the resulting liquid fraction is adjusted to 22–24% (w/
3.0–3.3 and 30–37 °C, respectively, adding the enzyme to reach a v). Again, the mixture is stirred for 1 h and microfiltered to recover the
concentration of 1 g/L, and incubating the mixture under slow stirring precipitated immunoglobulins which are dissolved in water for injec-
for 40 min. Digestion is then halted by adjusting the pH to 7.0–7.5 tion (WFI) or saline solution (0.15 M NaCl) at pH 7.4 (Gutiérrez et al.,
(Pope, 1939). 2011). Salts are then eliminated by dialysis or diafiltration. At the end,
During pepsin digestion, the neutralizing potency of plasma de- 40–50% of the initial IgG is recovered with a purity of around 70%.
creases along with the pH at which the digestion is achieved. When the Albumin, fibrinogen, fibronectin and α2-macroglobulin are the most
digestion is performed at pH of 2.8–3.2, ELISA titer of antivenom an- abundant non-immunoglobulin contaminants in this type of formula-
tibodies is reduced to 35–70% of the initial titer (Morais and Massaldi, tions (Tan et al., 2016).
2005), and plasma potency assessed in mice is reduced by half (our On the other hand, purification of F(ab’2) fragments does not in-
unpublished results). When digestion is performed at pH higher than clude the first salting-out at 14–16% ammonium sulfate, since fi-
3.2, potency loss is reduced, with the inconvenience that there is an brinogen is digested by pepsin, and usually its fragments are removed
incomplete removal of Fc fragments. At pH values lower than 2.8, the by thermocoagulation. Thus, for the purification of F(ab’)2 fragments
loss of potency is even higher. Since pepsin digestion reduces the yield salting out is directly achieved by precipitation with 22–24% (w/v)
of antivenom, it increases the production costs. Therefore, inclusion of ammonium sulfate at pH of 7.0–7.4. Then, the resulting precipitate,
this step in the antivenom manufacture protocol should be justified by mainly consisting of F(ab’)2 fragments, is recovered by microfiltration
the demonstration of the improvement of antivenom quality. or centrifugation, dissolved in saline solution and diafiltered or dia-
On the other hand, enzymatic digestion with papain is used to lyzed to eliminate the residual ammonium sulfate (Raw et al., 1991).
produce sheep Fab fragment-based antivenoms. Since equine IgGs are
resistant to papain digestion (our unpublished results), studies of 4.4. Caprylic acid precipitation
equine derived Fab-antivenoms are not available. Papain digestion is
performed at room temperature by adjusting the pH of undiluted The addition of caprylic acid to plasma causes the irreversible
plasma to 7.0–7.2, adding 20 g/L of the enzyme powder and 1 g/L cy- precipitation of most of the non-immunoglobulin proteins, leaving most
steine, and incubating the mixture under slow stirring for 120 min. In of the immunoglobulins (or their F(ab′)2 or Fab fragments) and low
contrast to pepsin digestion, papain digestion does not reduce the po- amounts of some protein contaminants (e.g., ceruloplasmin and trans-
tency of plasma (León et al., 2000). ferrin) in solution (Segura et al., 2012). The mechanism by which
Owing to their pharmacokinetic properties, Fab fragments are ra- caprylic acid precipitation works is not fully understood. Morais and
pidly eliminated from the body by renal filtration after intravenous Massaldi (2012) proposed that caprylic acid binds to specific sites of the
administration (Gutiérrez et al., 2003). Consequently, recurrence of plasma proteins, thus increasing the hydrophobicity of the interfacial
antigenemia and envenomation is frequent in patients treated with this protein surface, which in turn increases protein-protein interactions
type of antivenom (Seifert and Boyer, 2001; Bush et al., 2015). Under and cause precipitation.
these circumstances, additional doses of antivenom are needed. Caprylic acid precipitation is performed at room temperature. The
Therefore, few laboratories have adopted the production of Fab-based pH of undiluted plasma (digested with enzymes or not) must be be-
antivenoms for commercial purposes. tween 5.5 and 7.5. Then, caprylic acid is slowly added to attain a
concentration of 5–7% (v/v), while the plasma is vigorously stirred.
4.2. Thermocoagulation Regardless of the initial pH of the plasma, at the end of the caprylic acid
addition, the final pH of the mixture is around of 5.4. Precipitation is
To denature the non-neutralizing degraded proteins, thermo- complete after 45 min of stirring. Then the precipitated material is re-
coagulation is often incorporated after pepsin digestion of plasma moved by microfiltration or centrifugation (dos Santos et al., 1989;
(Pépin-Covatta et al., 1997). Thermocoagulation is performed by Rojas et al., 1994; Raweerith and Ratanabanangkoon, 2003; Mpandi
adding 12–15% ammonium sulfate to the digested plasma, adjusting et al., 2007). Whole IgGs can be recovered with higher than 90% purity
the pH of the mixture at 4.0–4.5, and incubating at 56 °C for 60 min. At and 60–65% yield (Rojas et al., 1994). Similar protocols optimized with
the end of the step, the mixture is cooled down and the coagulated different approaches at variable experimental conditions have been
proteins are separated by filtration or centrifugation. This procedure is described (Nudel et al., 2012).
used in the preparation of F(ab′)2 antivenoms, but not when producing
whole IgG antivenoms. 4.5. Aqueous two-phase systems (ATPS)

4.3. Salting-out Recently, the development of a method for the purification of an-
tivenom immunoglobulins by differential partition in an aqueous two-
Salting-out occurs when the surface tension of water in a protein phase system (ATPS), conformed by polyethyleneglycol (PEG) 3350/
solution is enhanced by the addition of a salt, resulting in an increase of potassium phosphate, was described (Vargas et al., 2015). In this
hydrophobic interactions between proteins and water. Therefore, pro- method, NaCl (15% w/v), potassium phosphate (20% w/v) and PEG
teins are forced to reduce their surface area by folding and self-asso- 3350 (9% w/v) are sequentially added to the plasma until complete
ciating, which leads them to precipitate (Atkinson, 1900). dissolution. The IgGs precipitated in the polymeric phase (i.e., the
Usually, differential precipitation of plasma proteins by salting-out upper phase) are recovered by microfiltration, and then dissolved in
is performed with ammonium sulfate, although sodium sulfate could water. To increase the purity of the product, the IgGs can be submitted
also be used. The general procedure may vary, depending on whether to an additional purification step with caprylic acid.
the molecule to be purified is IgG or F(ab′)2. Nonetheless, in both cases Antivenoms produced by ATPS separation showed higher purity and
co-precipitation of impurities along with the active ingredient, and yield than antivenoms produced by direct caprylic acid precipitation
aggregate formation, are frequent problems associated to the salting- (Vargas et al., 2015). In addition, this method allows the recovery of
out procedure (Rojas et al., 1994). albumin from the saline phase (i.e., the bottom phase). Simultaneous
Purification of IgG from equine plasma is performed at room tem- production of antivenom and albumin from the same plasma batch may
perature and neutral pH. First, ammonium sulfate is added at a reduce the production cost of antivenoms, and consequently the price at

68
G. León et al. Toxicon 151 (2018) 63–73

which these products are offered. et al., 2004) and formulation with phenol (Caricati et al., 2013) in-
Until now, the ATPS method has been used only to produce pilot activate both enveloped and non-enveloped virus.
batches (i.e. 50 kg plasma) for preclinical evaluation. The industrial
scaling up of this method, as well as its evaluation for safety and effi- 5. Formulation
cacy at the clinical setting are still pending tasks.
After purification, immunoglobulins must be concentrated or di-
4.6. Chromatographic methods luted, to comply with potency specification. Concentration or dilution
of the final formulation depends on the relationship between the neu-
Ion exchange chromatography is used by some manufacturers to tralizing ability of the starting plasma and the potency required to
increase the purity of whole IgGs or F(ab′)2 fragments after primary control venom toxicity with a dose of antivenom, a parameter that must
purification steps, such as enzyme digestion, salting out or caprylic acid be validated in properly conducted clinical trials.
precipitation (Jones and Landon, 2002; Raweerith and After adjustment of the total protein concentration, the im-
Ratanabanangkoon, 2003). After primary purification, antibodies con- munoglobulin solution must be formulated. Sodium chloride at
stitute more than 90% of the total protein of the preparation. Therefore, 0.7–1.0% (w/v) is added to confer the ionic strength necessary to keep
anion exchange chromatography (e.g., DEAE or quaternary ammonium the immunoglobulins in solution, and pH is adjusted to physiological
gels, at pH 7.0–8.0; Gutiérrez et al., 2011), is a frequently used meth- values (7.2–7.4). Osmolytes such as sorbitol, mannitol or glycine can be
odology, since contaminants bind to the gel, whereas immunoglobulins added to protect immunoglobulins from chemical and thermal stress
are eluted in the break-through fraction. In contrast, cationic exchange (Wang, 1999; Segura et al., 2009b). In the case of freeze-dried anti-
chromatography (e.g., using exchangers such as carboxymethyl or venoms, formulation with lyoprotectant components such as 5.0% su-
sulfonic acid and pH of 4.0–5.0) bind the immunoglobulins, which are crose (m/v) is frequent (Herrera et al., 2017a, 2017b).
in higher concentration, thus being a less pragmatic methodology. Although antivenoms are not presented as multi-dose medicines,
An additional advantage of anion exchange chromatography is the most of the current products are formulated with preservatives such as
removal of endotoxins (Lee et al., 2003). Endotoxin contamination phenol 0.15–0.25% (w/v) or cresol 0.30–0.35% (w/v). However, some
comprises a potential risk in antivenom manufacture during bleeding, preservatives favor the formation of immunoglobulin aggregates, in-
plasma separation, or immunoglobulin purification, and may remain in duce turbidity (García et al., 2002; Segura et al., 2009b) and cause
the preparation after the primary purification step. However, the fea- transitory hypotension in rats when administered as bolus injections
sibility of reducing endotoxin levels by ion-exchange chromatography (García et al., 2002). Thus, the lowest concentration with proven bac-
during fractionation does not substitute the need to use non-con- teriostatic activity based on antimicrobial efficacy tests (USP, 2014a)
taminated plasma, i.e. plasma with a content of endotoxin lower than should be used (Abd-Elsalam et al., 2011). Ideally, antivenoms should
5.0 EU/mL as starting material for antivenom production. not contain preservatives. However, to attain this goal, aseptic condi-
On the other hand, affinity chromatography has been suggested as tions must be guaranteed during the aseptic filling of the product.
an alternative to purify antivenom antibodies, as it eliminates im-
munoglobulins that do not bind to venom components (Dart et al., 6. Freeze-drying stabilization
1997). In this method, an open chromatography column is packed with
snake venom immobilized on an activated Sepharose matrix or a similar Freeze-drying or lyophilization is the best method for antivenom
affinity gel. Then, diluted and microfiltered hyperimmune plasma (di- stabilization in the presence of thermal stress (Herrera et al., 2014,
gested or not), or a solution of purified IgG or IgG-fragments, is passed 2017a). Therefore, freeze-dried antivenoms are the best alternative to
through the column. Antivenom antibodies bind to the venom and the supply remote regions where the cold chain required for the storage of
rest of the material is eluted in the unbound fraction. Afterwards, an- liquid formulations is not guaranteed. When properly processed, effi-
tivenom immunoglobulins are eluted by using low pH or changes in cacy and safety of freeze antivenoms are not different from that shown
ionic strength of the loading buffer. by liquid antivenoms (Mendonça-da-Silva et al., 2017). However, the
Immunoglobulins obtained by affinity chromatography are re- physical and chemical stress induced during the freeze-drying proce-
covered with purity and yield over 97%. This is the only current dure may affect the solubility and the neutralizing ability of im-
method by which antivenom immunoglobulins can be separated from munoglobulins (Sarciaux et al., 1999; Maury et al., 2005); conse-
non-antivenom immunoglobulins, which are the main contaminant in quently, the standardization of the freeze-drying procedure must be
current antivenoms (Segura et al., 2012). Nonetheless, application of carefully addressed.
affinity chromatography is largely unpractical because it is expensive The freeze-drying process consists of three stages: freezing, primary
and high amounts of venom are required. drying or sublimation and secondary drying or desorption. During an-
tivenom freezing, the water present in the antivenom forms ice crystals,
4.7. Steps for removing or inactivating viruses leaving free spaces in which immunoglobulins are concentrated until
the formation of a solid amorphous system (Pikal, 2004). For anti-
Biotechnology products purified from animal plasma have potential venoms, the optimal freezing rate is around 1 °C/min until reaching
risk of virus transmission to the patients (EMEA, 1996). Although there −40 °C. Annealing is an alternative step to homogenize and maximize
are no reports of equine virus transmission to human patients by par- water crystallization.
enteral administration of antivenoms, protocols of antivenom manu- The following step is the drying of the frozen solid under vacuum.
facture must include validated steps effective in preventing this risk During this step, the temperature of the product should not exceed the
(i.e., three steps able to reduce the viral infectivity by at least 4 log10 in glass transition temperature (i.e., the temperature at which the anti-
the TCID50 test). venom is transformed into a solid amorphous system; Schersch et al.,
Fortunately, several steps commonly used to produce antivenoms 2010), which, in the case of an equine IgG formulated without ex-
have been shown to remove or inactivate viruses. Steps such as caprylic cipients, is around −13 °C (Herrera et al., 2014).
acid precipitation (Dichtelmüller et al., 2002; Burnouf et al., 2007; During the first phase of the drying process (primary drying or
Caricati et al., 2013); low pH treatment (Burnouf et al., 2004; Solano sublimation), frozen water is sublimated at a rate that depends on the
et al., 2012); pepsin digestion (Lazar et al., 2002; Burnouf et al., 2007; programmed shelf temperature (e.g. −15 °C). This phase ends when the
Caricati et al., 2013); and solvent/detergent treatment (Segura et al., temperature of the product equals that of the shelf. Since this is the
2009a), are effective for inactivating enveloped virus, but not against most prolonged step of the freeze-dying process, its optimization has a
non-enveloped viruses. Other steps like thermal treatment (Burnouf large impact on the process economy. The second drying phase (the

69
G. León et al. Toxicon 151 (2018) 63–73

secondary drying or desorption) is performed by exposing the dried 2014c). Alternatively, to reduce the use of animals, the pyrogen test can
product to temperatures around 30 °C during 3–4 h, to reduce the be substituted by the Limulus amoebocyte lysate assay (LAL; Solano
content of residual humidity of the product to less than 3%. et al., 2015). In this case, endotoxin limit for final antivenom can be
The stability of freeze-dried antivenoms depends on the formula- calculated as the ratio K/M, where K is the threshold pyrogenic dose of
tion, the freeze-drying process, and the storage conditions. Stability of endotoxin per kilogram of body weight (i.e., 5 EU/kg), and M is the
antivenoms must be demonstrated for each formulation, under worst- maximum total dose of antivenom administered to a 70 kg patient per
case storage conditions (WHO, 2016). Results obtained are useful for 1 h (USP, 2014d; Solano et al., 2015).
real conditions of product storage and transportation at high tempera- Physicochemical quality of in-process/final antivenoms include the
tures (EMEA, 2002). Usually, the shelf-life of freeze-dried antivenoms, determination of pH, total protein (Gornall et al., 1949) preservative
stored at temperatures lower than their glass transition temperatures, concentration (Lacoste et al., 1959), chloride concentration (USP,
can reach five years (WHO, 2016). Instability of freeze-dried anti- 2014e), turbidity (Segura et al., 2009b), electrophoretic pattern
venoms is evidenced by increases in the time of reconstitution, the (Laemmli, 1970; Tan et al., 2016), presence of aggregates (e.g. by FPLC;
turbidity of the reconstituted product, and eventually on the reduction García et al., 2002; Tan et al., 2016), residual moisture (e.g. by the Karl
of the neutralizing ability of the antivenom. Fisher titration; Herrera et al., 2014), and residual caprylic acid (e.g. by
An analysis of several products distributed in different regions of the HPLC; Herrera et al., 2009) and other substances used during fractio-
world showed that most of them were able to reconstitute in less than nation, such as ammonium sulfate.
5 min (Herrera et al., 2017b). This is particularly important in the case Neutralizing immunoglobulins or immunoglobulin fragments (i.e.
of antivenoms, because efficacy of this therapy depends on the time IgG, F(ab′)2 or Fab) are the active substance of antivenoms. Then, small
lapse between envenomation and antivenom administration, and long amounts of non-immunoglobulin proteins (i.e. albumin, fibrinogen,
reconstitution times (i.e., 40 min or longer) introduce an inconvenient transferrin and ceruloplasmin) and high amounts of non-neutralizing
delay in the start of the therapy (Hill et al., 2001; Gerring et al., 2013). immunoglobulins are the most important protein contaminants in
current antivenoms (Segura et al., 2012; Tan et al., 2016). While non-
7. Quality control of in-process and final products immunoglobulin contaminants can be determined by methods such as
FPLC and SDS-PAGE (Segura et al., 2012; Tan et al., 2016), the content
Quality control is not a separate stage of the antivenom manu- of non-neutralizing immunoglobulins only can be determined by in-
facture, but an integral part of the whole production process. It begins direct methods, such as the potency/total protein ratio (Segura et al.,
with the monitoring of: 1) clean rooms, 2) the production of water for 2012).
injection, 3) the sanitization/sterilization of the equipment line, and 4) The establishment of specifications for the minimum required con-
the quality of raw materials. However, it also includes the evaluation of tent of active molecules, or the acceptable level of non-immunoglobulin
reference venom pools, hyperimmune plasma, and in-process/final contaminants in the antivenom is a controversial issue that can be il-
antivenom. lustrated by the following example. Let's suppose that there are two
As previously mentioned, quality control of venoms includes the antivenoms formulated at the same potency towards the same venom:
determination of toxic and enzymatic activities (Camey et al., 2002), “Antivenom A”, composed of 100% immunoglobulins and formulated
the ability of the venom to estimate the potency of reference anti- at total protein of 10 g/dL, and “Antivenom B”, composed by 80%
venoms (Fukuda et al., 2006; Araújo et al., 2008, 2017), and the ver- immunoglobulins and 20% albumin, and formulated at total protein of
ification that these activities are conserved during long-term storage 5 g/dL. Which one would have the better safety profile? If it is assumed
(Jesupret et al., 2014; Hatakeyama et al., 2018). that albumin is the most dangerous contaminant in antivenoms, then
The safety and effectiveness of the immunization process is assessed “Antivenom A” would be safer, despite that it exposes the patient to a
by the clinical evaluation of hyperimmune animals used as im- higher amount of heterologous protein. In contrast, if it is assumed that
munoglobulin source (Angulo et al., 1997) and by the potency assay, the total amount of heterologous protein administered is the most im-
respectively. Usually, the potency assay is performed by mixing chal- portant factor in the manifestation of adverse reactions, then
lenge dose of venom and variable dilutions of antivenom, to achieve “Antivenom B” would be safer.
different venom/antivenom ratios. After an incubation period, aliquots Theoretically, all batches of antivenom produced by the same
of the mixtures are injected in animals and the number of deaths is master process should have the same microbiological, physicochemical
recorded and used to calculate a mathematical expression of the anti- and immunological quality. Then, the analysis of many batches of an-
venom potency (Araújo et al., 2008, 2017; Solano et al., 2010). This tivenom is required to evaluate the quality of the master process.
potency assay is also used to determine the preclinical efficacy of in- Standardization of the master process is required to guarantee that all
process/final antivenoms. batches of antivenom meet the specifications that cannot be tested
The quality of the bleeding process is determined by the clinical every batch, such as stability.
status of bleeding animals (Angulo et al., 1997) and the microbiological
profile of the hyperimmune plasma. Maximum endotoxin content al- 8. Conclusions
lowed in plasma should be calculated as the product of the endotoxin
limit for the final antivenom (see below) and the potency of the final Snakebite envenomation is an important health problem worldwide,
product, divided by the potency of the crude plasma. and its solution requires improvements in: 1) the understanding of
Immunological quality of in-process/final antivenoms is normally snakes biology and venom composition and toxicity, 2) the technology
limited to the evaluation of the ability of the antivenom to neutralize for antivenom production, 3) the strategies for antivenom distribution,
the lethality of homologous venoms, as explained above (Gutiérrez 4) the access of patients to medical attention, 5) the training of health
et al., 2017). Nevertheless, additional information can be obtained from staff in the management of envenomed patients, and 6) the strength-
the assessment of the ability of the antivenom to neutralize other toxic ening of regulatory authorities to ensure that only effective and safety
effects induced by the venoms, such as hemorrhage, defibrinogenation, antivenoms will be available in the region under their control.
myotoxicity, necrosis, edema, and neuromuscular blockade (Gutiérrez The industrial production of safe and effective antivenoms is an
et al., 2013). essential part of this multicomponent strategy. Antivenom manufacture
The microbiological quality of in-process/final antivenoms is as- is a complex process that involves different activities that must be
sessed by the sterility test (USP, 2014b) and the pyrogen test, which carefully designed and coordinately performed. These activities in-
evaluates the rise in the rectal temperature of rabbits injected in the clude: 1) production of reference venom pools, 2) production of hy-
marginal vein with a dose of antivenom, in a period of 3 h (USP, perimmune plasma, 3) purification of the antivenom immunoglobulins,

70
G. León et al. Toxicon 151 (2018) 63–73

4) formulation of the antivenom, 5) stabilization of the formulation, envenomation: a prospective, blinded, multicenter, randomized clinical trial. Clin.
and 6) quality control of in-process and final products. The conforma- Toxicol (Phila) 53, 37–45.
Calmette, A., 1894. Contribution à l'étude du venin des serpents. Immunisation des ani-
tion of multidisciplinary professional teams to perform these activities maux et traitement de l'envenimation. Annales de l'Institut Pasteur VIII, pp. 275–291.
is essential to improve the technology of antivenom production, an Calvete, J.J., Sanz, L., Pla, D., Lomonte, B., Gutiérrez, J.M., 2014. Omics meets biology:
unfinished task encompassing diverse challenges. application to the design and preclinical assessment of antivenoms. Toxins (Basel) 6
(12), 3388–3405.
Camey, K.U., Velarde, D.T., Sánchez, E.F., 2002. Pharmacological characterization and
Conflicts of interest neutralization of the venoms used in the production of Bothropic antivenom in Brazil.
Toxicon 40, 501–509.
Caricati, C., Oliveira-Nascimento, L., Yoshida, J., Stephano, M., Caricati, A., Raw, I.,
All the authors are employed at Instituto Clodomiro Picado. None of 2013. Safety of Snake Antivenom Immunoglobulins: efficacy of viral inactivation in a
the authors of this paper has a financial or personal relationship with complete downstream process. Biotechnol. Prog. 29 (4), 972–979.
other people or organizations that could inappropriately influence or Carneiro, S.M., Zablith, M.B., Kerchove, C.M., Moura-da-Silva, A.M., Quissell, D.O.,
Markus, R.P., Yamanouye, N., 2006. Venom production in long-term primary culture
bias the content of the paper. Sponsors were not involved in the study
of secretory cells of the Bothrops jararaca venom gland. Toxicon 47, 87–94.
design; in the collection, analysis and interpretation of data; in the Chacón, D., Rodríguez, S., Arias, J., Solano, G., Bonilla, F., Gómez, A., 2012. Maintaining
writing of the review; or in the decision to submit the manuscript for Coral Snakes (Micrurus nigrocinctus, Serpentes: elapidae) for venom production on an
publication. alternative fish-based diet. Toxicon 60, 249–253.
Chanhome, L., Jintakune, P., Wilde, H., Cox, M.J., 2001. Venomous snake husbandry in
Thailand. Wilderness Environ. Med. 12, 17–23.
Acknowledgements Chippaux, J.P., Williams, V., White, J., 1991. Snake venom variability: methods of study.
Toxicon 29, 1279–1303.
Chotwiwatthanakun, C., Pratapaphon, R., Akesowan, S., Sriprapat, S., Ratanabangkoon,
This study was supported by Vicerrectoría de Investigación, K., 2001. Production of potent polyvalent antivenom against three elapid venoms
Universidad de Costa Rica (project 741-B7-108). The authors thank using a low dose, low volume, multi-site immunization protocol. Toxicon 39,
Andrés Hernández and our colleagues at Instituto Clodomiro Picado for 1487–1494.
Corrales, G., Meidinger, R., Rodríguez, S., Chacón, D., Gómez, A., 2014. Reproduction in
their collaboration. captivity of the central American bushmaster (Lachesis stenophrys, serpentes: viper-
idae), in Costa Rica. Cuad. Herpetol. 28 (2), 137–139.
Transparency document da Silva, W.D., Tambourgi, D.V., 2011. The humoral immune response induced by snake
venom toxins. Inflamm. Allergy - Drug Targets 10 (5), 343–357.
Dart, R.C., Seifert, S.A., Carroll, L., Clark, R.F., Hall, E., Boyer-Hassen, L.V., Curry, S.C.,
Transparency document related to this article can be found online at Kitchens, C.S., Garcia, R.A., 1997. Affinity-purified, mixed monospecific crotalid
http://dx.doi.org/10.1016/j.toxicon.2018.06.084 antivenom ovine Fab for the treatment of crotalid venom poisoning. Ann. Emerg.
Med. 30 (1), 33–39.
Dichtelmüller, H., Rudnick, D., Kloft, M., 2002. Inactivation of lipid enveloped viruses by
References octanoic acid treatment of immunoglobulin solution. Biologicals 30, 135–142.
dos Santos, M.C., D'Império Lima, M.R., Furtado, G.C., Colletto, G.M., Kipnis, T.L., Dias da
Abd-Elsalam, M.A., Abdoon, N., Al-Ahaidib, M.S., 2011. What is the optimum con- Silva, W., 1989. Purification of F(ab’)2 anti-snake venom by caprylic acid: a fast
centration of m-cresol in antivenoms? J. Venom. Anim. Toxins Incl. Trop. Dis. 17, method for obtaining IgG fragments with high neutralization activity, purity and
12–22. yield. Toxicon 27, 297–303.
Abubakar, I.S., Abubakar, S.B., Habib, A.G., Nasidi, A., Durfa, N., Yusuf, P.O., Larnyang, EMEA (The European Agency for the Evaluation of Medicinal Products), 1996. Note for
S., Garnvwa, J., Sokomba, E., Salako, L., Theakston, R.D., Juszczak, E., Alder, N., Guidance on Virus Validation Studies: the Design, Contribution and Interpretation of
Warrell, D.A., Nigeria-UK EchiTab Study Group, 2010. Randomised controlled Studies Validating the Inactivation and Removal of Viruses. EMEA, London.
double-blind non-inferiority trial of two antivenoms for saw-scaled or carpet viper EMEA (The European Agency for the Evaluation of Medicinal Products), 2002. Note for
(Echis ocellatus) envenoming in Nigeria. PLoS Neglected Trop. Dis. 4 (7), e767. Guidance on the Production and Quality Control of Animal Immunoglobulins and
Angulo, Y., Estrada, R., Gutiérrez, J.M., 1997. Clinical and laboratory alterations in horses Immunosera for Human Use. EMEA, London.
during immunization with snake venoms for the production of polyvalent Farias, I.B., Morais-Zani, K., Serino-Silva, C., Sant'Anna, S.S., Rocha, M.M.T.D., Grego,
(Crotalinae) antivenom. Toxicon 35, 81–90. K.F., Andrade-Silva, D., Serrano, S.M.T., Tanaka-Azevedo, A.M., 2018. Functional
Araújo, H.P., Bourguignon, S.C., Boller, M.A., Dias, A.A., Lucas, E.P., Santos, I.C., and proteomic comparison of Bothrops jararaca venom from captive specimens and
Delgado, I.F., 2008. Potency evaluation of antivenoms in Brazil: the national control the Brazilian Bothropic Reference Venom. J. Proteomics 174, 36–46.
laboratory experience between 2000 and 2006. Toxicon 51 (4), 502–514. Feige, K., Ehrat, F.B., Kästner, S.B., Schwarzwald, C.C., 2003. Automated plasmapheresis
Araújo, H.P., Lucas, E.P.R., Moura, W.C., Fátima-Barbosa, C., Rodrigues, R.J., Morais, compared with other plasma collection methods in the horse. J. Vet Med. A Physiol.
J.F., Almada, J.Z., Guimarães, B.C., Almeida, A.E.C.C., Boller, M.A.A., 2017. Pathol. Clin. Med. 50, 185–189.
Interlaboratory study for the establishment of Brazilian Bothrops reference venom and Freitas-de-Sousa, L.A., Amazonas, D.R., Sousa, L.F., Sant'Anna, S.S., Nishiyama Jr., M.Y.,
antivenom for potency evaluation of Bothrops antivenom. Biologicals 49, 1–5. Serrano, S.M., Junqueira-de-Azevedo, I.L., Chalkidis, H.M., Moura-da-Silva, A.M.,
Arce-Estrada, V., Azofeifa-Cordero, G., Estrada, R., Alape-Girón, A., Flores-Díaz, M., 2009. Mourão, R.H., 2015. Comparison of venoms from wild and long-term captive Bothrops
Neutralization of venom-induced hemorrhage by equine antibodies raised by im- atrox snakes and characterization of Batroxrhagin, the predominant class PIII me-
munization with a plasmid encoding a novel P-II metalloproteinase from the lance talloproteinase from the venom of this species. Biochimie 118, 60–70.
head pitviper Bothrops asper. Vaccine 27, 460–466. Fukanoki, S., Iwakura, T., Iwaki, S., Matsumoto, K., Takeda, R., Ikeda, K., et al., 2001.
Arroyo, C., Solano, S., Herrera, M., Segura, Á., Estrada, R., Vargas, M., Villalta, M., Safety and efficacy of water-in-oil-in-water emulsion vaccines containing Newcastle
Gutiérrez, J.M., León, G., 2015. Lachesis stenophrys venom reduces the equine anti- disease virus haemagglutinin-neuraminidase glycoprotein. Avian Pathol. 30
body response towards Bothrops asper venom used as co-immunogen in the produc- 509e516.
tion of polyspecific snake antivenom. Toxicon 103, 99–105. Fukuda, T., Iwaki, M., Hong, S.H., Oh, H.J., Wei, Z., Morokuma, K., Ohkuma, K.,
Arroyo, C., Solano, S., Segura, Á., Herrera, M., Estrada, R., Villalta, M., Vargas, M., Dianliang, L., Arakawa, Y., Takahashi, M., 2006. Standardization of regional re-
Gutiérrez, J.M., León, G., 2017. Cross-reactivity and cross-immunomodulation be- ference for mamushi (Gloydius blomhoffii) antivenom in Japan, Korea, and China. Jpn.
tween venoms of the snakes Bothrops asper, Crotalus simus and Lachesis stenophrys, and J. Infect. Dis. 59 (1), 20–24.
its effect in the production of polyspecific antivenom for Central America. Toxicon García, M., Monge, M., León, G., Lizano, S., Segura, E., Solano, G., Rojas, G., Gutiérrez,
138, 43–48. J.M., 2002. Effect of preservatives on IgG aggregation, complement-activating effect
Atkinson, J.P., 1900. The fractional precipitation of the globulin and albumin of normal and hypotensive activity of horse polyvalent antivenom used in snakebite en-
horses serum and diphtheria antitoxic serum, and the anti-toxic strength of the venomation. Biologicals 30, 143–151.
precipitates. J. Exp. Med. 5, 67–76. Gerring, D., King, T.R., Branton, R., 2013. Validating a faster method for reconstitution of
Bolaños, R., 1972. Toxicity of Costa Rican snake venoms for the white mouse. Am. J. crotalidae polyvalent immune Fab (ovine). Toxicon 69, 42–49.
Trop. Med. Hyg. 21 (3), 360–363. Gibson, R.B., 1906. The concentration of antitoxin for therapeutic use. J. Biol. Chem. 1,
Burnouf, T., Griffiths, E., Padilla, A., Seddik, S., Stephano, M.A., Gutiérrez, J.M., 2004. 161–170.
Assessment of the viral safety of antivenoms fractionated from equine plasma. Goebel-Stengel, M., Stengel, A., Taché, Y., Reeve Jr., J.R., 2011. The importance of using
Biologicals 32 (3), 115–128. the optimal plasticware and glassware in studies involving peptides. Anal. Biochem.
Burnouf, T., Terpstra, F., Habib, G., Seddik, S., 2007. Assessment of viral inactivation 414, 38–46.
during pH 3.3 pepsin digestion and caprylic acid treatment of antivenoms. Biologicals Gómez, A., Arroyo, C., Astorga, W., Chacón, D., Rodríguez, S., Jiménez, M., 2016.
35, 329–334. Hematological and biochemical reference intervals for Bothrops asper and Crotalus
Bush, S.P., Ruha, A.M., Seifert, S.A., Morgan, D.L., Lewis, B.J., Arnold, T.C., Clark, R.F., simus (Serpentes: viperidae), maintained in captivity for venom extraction. Comp.
Meggs, W.J., Toschlog, E.A., Borron, S.W., Figge, G.R., Sollee, D.R., Shirazi, F.M., Clin. Pathol. 25, 615–623.
Wolk, R., de Chazal, I., Quan, D., García-Ubbelohde, W., Alagón, A., Gerkin, R.D., Gornall, A.G., Bardawill, C.J., David, M.N., 1949. Determination of serum proteins by
Boyer, L.V., 2015. Comparison of F(ab’)2 versus Fab antivenom for pit viper means of the Biüret reaction. J. Biol. Chem. 177 751e66.
Gutiérrez, J.M., León, G., Lomonte, B., 2003. Pharmacokinetic-pharmacodynamic

71
G. León et al. Toxicon 151 (2018) 63–73

relationships of immunoglobulin therapy for envenomation. Clin. Pharmacokinet. 42, Pathogenic mechanisms underlying adverse reactions induced by intravenous ad-
721–741. ministration of snake antivenoms. Toxicon 76, 63–76.
Gutiérrez, J.M., Lomonte, B., León, G., Alape-Girón, A., Flores-Díaz, M., Sanz, L., Angulo, León, G., Segura, Á., Gómez, A., Hernández, A., Navarro, D., Villalta, M., Vargas, M.,
Y., Calvete, J.J., 2009. Snake venomics and antivenomics: proteomic tools in the Herrera, M., Gutiérrez, J.M., 2016. Industrial production and quality control of snake
design and control of antivenoms for the treatment of snakebite envenoming. J. antivenoms. In: Gopalakrishnakone, P., Calvete, J.J. (Eds.), Venom Genomics and
Proteomics 72, 165–182. Proteomics. Springer, Dordrecht, pp. 1–22.
Gutiérrez, J.M., Sanz, L., Flores-Díaz, M., Figueroa, L., Madrigal, M., Herrera, M., Villalta, Levine, L., Broderick, E.J., 1970. The plasmapheresis of hyperimmunized horses. Bull.
M., León, G., Estrada, R., Borges, A., Alape-Girón, A., Calvete, J.J., 2010. Impact of World Health Organ. 42 (6), 998–1000.
regional variation in Bothrops asper snake venom on the design of antivenoms: in- Li, A., Zhang, C., Wang, J., Wang, J., Jiang, H., Li, J., Ma, X., Zhang, W., Lu, Y., 2017.
tegrating antivenomics and neutralization approaches. J. Proteome Res. 9, 564–577. Cloning, expression, purification and bioactivity evaluation of a thrombin-like en-
Gutiérrez, J.M., León, G., Lomonte, B., Angulo, Y., 2011. Antivenoms for snakebite en- zyme from Deinagkistrodon acutus venom gland library. Biotechnol. Lett. 40 (1),
venomings. Inflamm. Allergy - Drug Targets 10, 369–380. 93–102.
Gutiérrez, J.M., Solano, G., Pla, D., Herrera, M., Segura, A., Villalta, M., Vargas, M., Sanz, Maury, M., Murphy, K., Kumar, S., Mauerer, A., Lee, G., 2005. Spray-drying of proteins:
L., Lomonte, B., Calvete, J.J., León, G., 2013. Assessing the preclinical efficacy of effects of sorbitol and trehalose on aggregation and FT-IR amide I spectrum of an
antivenoms: from the lethality neutralization assay to antivenomics. Toxicon 69, immunoglobulin G. Eur. J. Pharm. Biopharm. 59 (2) 251e261.
168–179. McCleary, R.J., Sridharan, S., Dunstan, N.L., Mirtschin, P.J., Kini, R.M., 2016. Proteomic
Gutiérrez, J.M., Solano, G., Pla, D., Herrera, M., Segura, Á., Vargas, M., Villalta, M., comparisons of venoms of long-term captive and recently wild-caught Eastern brown
Sánchez, A., Sanz, L., Lomonte, B., León, G., Calvete, J.J., 2017. Preclinical evalua- snakes (Pseudonaja textilis) indicate venom does not change due to captivity. J.
tion of the efficacy of antivenoms for snakebite envenoming: state-of-the-art and Proteomics 144, 51–62.
challenges ahead. Toxins (Basel) 9 (5) pii: E163. Meier, J., Adler, C., Hösle, P., Cascio, R., 1991. The influence of three different drying
Hatakeyama, D.M., de Morais-Zani, K., Serino-Silva, C., Grego, K.F., Sant'Anna, S.S., procedures on some enzymatic activities of three Viperidae snake venoms. Mem Inst.
Fernandes, W., Aniz, P.A.E.A., Torquato, R.J.S., Tanaka, A.S., Sanz, L., Calvete, J.J., Butan. (Sao Paulo) 53 (1), 119–126.
Tanaka-Azevedo, A.M., 2018. Examination of biochemical and biological activities of Mendonça-da-Silva, I., Magela-Tavares, A., Sachett, J., Sardinha, J.F., Zaparolli, L.,
Bothrops jararaca (Serpentes: viperidae; Wied-Neuwied 1824) snake venom after up Gomes-Santos, M.F., Lacerda, M., Monteiro, W.M., 2017. Safety and efficacy of a
to 54 years of storage. Toxicon 141, 34–42. freeze-dried trivalent antivenom for snakebites in the Brazilian Amazon: an open
Heller, M.C., Carpenter, J.F., Randolph, T.W., 1999. Protein formulation and lyophili- randomized controlled phase IIb clinical trial. PLoS Neglected Trop. Dis. 11 (11),
zation cycle design: prevention of damage due to freeze-concentration induced phase e0006068.
separation. Biotechnol. Bioeng. 63, 166–174. Modahl, C.M., Doley, R., Kini, R.M., 2010. Venom analysis of long-term captive Pakistan
Herrera, M., Meneses, F., Gutiérrez, J.M., León, G., 2009. Development and validation of cobra (Naja naja) populations. Toxicon 55, 612–618.
a reverse phase HPLC method for the determination of caprylic acid in formulations Morais, V., Massaldi, H., 2005. Effect of pepsin digestion on the antivenom activity of
of therapeutic immunoglobulins and its application to antivenom production. equine immunoglobulins. Toxicon 46, 876–882.
Biologicals 37 (4), 230–234. Morais, V., Massaldi, H., 2012. A model mechanism for protein precipitation by caprylic
Herrera, M., Tattini, V., Pitombo, R., Gutiérrez, J.M., Borgognoni, C., Vega-Baudrit, J., acid: application to plasma purification. Biotechnol. Appl. Biochem. 59 (1), 50–54.
Solera, F., Cerdas, M., Segura, A., Villalta, M., Vargas, M., León, G., 2014. Freeze- Mpandi, M., Schmutz, P., Legrand, E., Duc, R., Geinoz, J., Henzelin-Nkubana, C., Giorgia,
dried snake antivenoms formulated with sorbitol, sucrose or mannitol: comparison of S., Clerc, O., Genoud, D., Weber, T., 2007. Partitioning and inactivation of viruses by
their stability in an accelerated test. Toxicon 90, 56–63. the caprylic acid precipitation followed by a terminal pasteurization in the manu-
Herrera, M., Segura, Á., Sánchez, A., Sánchez, A., Vargas, M., Villalta, M., Harrison, R.A., facturing process of horse immunoglobulins. Biologicals 35, 335–341.
Gutiérrez, J.M., León, G., 2017a. Freeze-dried EchiTAb+ICP antivenom formulated Muliya, S.K., Bhat, M.N., 2016. Hematology and serum biochemistry of Indian spectacled
with sucrose is more resistant to thermal stress than the liquid formulation stabilized cobra (Naja naja) and Indian rat snake (Ptyas mucosa). Vet. World 9, 909–914.
with sorbitol. Toxicon 133, 123–126. Niinistö, K., Raekallio, M., Sankari, S., 2007. Storage of equine red blood cells as a
Herrera, M., Solano, D., Gómez, A., Villalta, M., Vargas, M., Segura, Á., Gutiérrez, J.M., concentrate. Vet. J. 176, 227–231.
León, G., 2017b. Physicochemical characterization of commercial freeze-dried snake Nudel, B.C., Perdoménico, C., Iácono, R., Cascone, O., 2012. Optimization by factorial
antivenoms. Toxicon 126, 32–37. analysis of caprylic acid precipitation of non-immunoglobulins from hyperimmune
Hill, R.E., Bogdan, G.M., Dart, R.C., 2001. Time to reconstitution: purified Fab antivenom equine plasma for antivenom preparation. Toxicon 59, 68–73.
vs unpurified IgG antivenom. Toxicon 39, 729–731. Oh, A.M.F., Tan, C.H., Ariaranee, G.C., Quraishi, N., Tan, N.H., 2017. Venomics of
Jansen, T., Hofmans, M.P.M., Theelen, M.J.G., Manders, F.G.A., Schijns, V.E.J.C., 2007. Bungarus caeruleus (Indian krait): comparable venom profiles, variable im-
Dose and timing requirements for immunogenicity of viral poultry vaccine antigen: munoreactivities among specimens from Sri Lanka, India and Pakistan. J. Proteomics
investigations of emulsion-based depot function. Avian Pathol. 36 361e365. 164, 1–18.
Jesupret, C., Baumann, K., Jackson, T.N., Ali, S.A., Yang, D.C., Greisman, L., Kern, L., Olmedo, H., Herrera, M., Rojas, L., Villalta, M., Vargas, M., Leiguez, E., Teixeira, C.F.P.,
Steuten, J., Jouiaei, M., Casewell, N.R., Undheim, E.A., Koludarov, I., Debono, J., Estrada, R., Gutiérrez, J.M., León, G., Montero, M.L., 2013. Comparison of the ad-
Low, D.H., Rossi, S., Panagides, N., Winter, K., Ignjatovic, V., Summerhayes, R., juvant activity of aluminum hydroxide and calcium phosphate on the antibody re-
Jones, A., Nouwens, A., Dunstan, N., Hodgson, W.C., Winkel, K.D., Monagle, P., Fry, sponse towards Bothrops asper snake venom. J. Immunot. 11, 44–49.
B.G., 2014. Vintage venoms: proteomic and pharmacological stability of snake ve- Parfentjev, I.A., 1936. Method for purification of anti-toxins and the like. U.S. Patent 2,
noms stored for up to eight decades. J. Proteomics 105, 285–294. 065,196.
Jones, R.G., Landon, J., 2002. Enhanced pepsin digestion: a novel process for purifying Pépin-Covatta, S., Lutsch, C., Grandgeorge, M., Scherrmann, J.M., 1997.
antibody F(ab’)2 fragments in high yield from serum. J. Immunol. Meth. 263, 57–74. Immunoreactivity of a new generation of horse F(ab’)2 preparations against European
Kalita, B., Patra, A., Mukherjee, A.K., 2017. Unraveling the proteome composition and vipervenoms and the tetanus toxin. Toxicon 35, 411–422.
immuno-profiling of western India Russell's viper venom for in-depth understanding Pikal, M.J., 2004. Mechanism of protein stabilization during freeze-drying and storage:
of its pharmacological properties, clinical manifestations, and effective antivenom the relative importance of thermodynamic stabilization and glassy state relaxation
treatment. J. Proteome Res. 16, 583–598. dynamics. In: Rey, L., May, J.C. (Eds.), Freeze-drying/lyophilization of
Kasturiratne, A., Wickremasinghe, R., de Silva, N., Gunawardena, K., Pathmeswaran, A., Pharmaceutical and Biological Products. Marcer Dekker Inc., New York, pp. 63–107.
Premaratna, R., Savioli, L., Lalloo, D., de Silva, J., 2008. The Global Burden of Pope, C.G., 1939. The action of proteolytic enzymes on the antitoxins and proteins in
Snakebite: a literature analysis and modelling based on regional estimates of en- immune sera. I. True digestion of the proteins. Br. J. Exp. Pathol. 20, 132–149.
venoming and deaths. PLoS Med. 5, e218. Ratanabanangkoon, K., Tan, K.Y., Eursakun, S., Tan, C.H., Simsiriwong, P., Pamornsakda,
Lacoste, R.J., Venable, S.H., Stone, J.C., 1959. Modified 4-aminoantipyrine colorimetric T., Wiriyarat, W., Klinpayom, C., Tan, N.H., 2016. A simple and novel strategy for the
method for phenols. Application to an acrylic monomer. Anal. Chem. 31, 1246–1249. production of a pan-specific antiserum against elapid snakes of Asia. PLoS Neglected
Laemmli, U.K., 1970. Cleavage of structural proteins during the assembly of the head of Trop. Dis. 10 (4) e0004565.
bacteriophage T4. Nature 227, 680–685. Raw, I., Guidolin, R., Higashi, H.G., Kelen, E.M.A., 1991. Antivenins in Brazil: prepara-
Landon, J., Smith, D.C., 2003. Merits of sheep antisera for antivenom manufacture. J. tion. In: In: Tu, A.T. (Ed.), Handbook of Natural Toxins, Reptile Venoms and Toxins,
Toxicol. 22, 15–22. vol. 5. Marcel Dekker, New York, pp. 557–581.
Laustsen, A.H., Engmark, M., Milbo, C., Johannesen, J., Lomonte, B., Gutiérrez, J.M., Raweerith, R., Ratanabanangkoon, K., 2003. Fractionation of equine antivenom using
Lohse, B., 2016. From fangs to pharmacology: the future of snakebite envenoming caprylic acid precipitation in combination with cationic ion-exchange chromato-
therapy. Curr. Pharmaceut. Des. 22, 5270–5293. graphy. J. Immunol. Meth. 282, 63–72.
Lazar, A., Epstein, E., Lustig, S., Barnea, A., Silberstein, L., Reuveny, S., 2002. Inactivation Rojas, G., Jiménez, J.M., Gutiérrez, J.M., 1994. Caprylic acid fractionation of hyper-
of West-Nile virus during peptic cleavage of horse plasma IgG. Biologicals 30, immune horse plasma: description of a simple procedure for antivenom production.
163–165. Toxicon 32, 351–363.
Lee, S.H., Kim, J.S., Kim, C.W., 2003. Optimization of buffer conditions for the removal of Rosenberg, E., Hepbildikler, S., Kuhne, W., Winter, G., 2009. Ultrafiltration concentration
endotoxins using Q-sepharose. Process Biochem. 38 (7), 1091–1098. of monoclonal antibody solutions: development of an optimized method minimizing
León, G., Valverde, J.M., Rojas, G., Lomonte, B., Gutiérrez, J.M., 2000. Comparative study aggregation. J. Membr. Sci. 342, 50–59.
on the ability of IgG and Fab sheep antivenoms to neutralize local hemorrhage, Saad, E., Curtolo-Barros, L., Biscola, N., Pimenta, D.C., Barraviera, S.R., Barraviera, B.,
edema and myonecrosis by Bothrops asper (terciopelo) snake venom. Toxicon 38, Seabra-Ferreira Jr., R., 2012. Intraspecific variation of biological activities in venoms
233–244. from wild and captive Bothrops jararaca. J. Toxicol. Environ. Health A 75,
León, G., Sánchez, L., Hernández, A., Villalta, M., Herrera, M., Segura, Á., Estrada, R., 1081–1090.
Gutiérrez, J.M., 2011. Immune response towards snake venoms. Inflamm. Allergy - Sadahiro, S., Kondo, L., Yamamoto, A., 1981. Effects of adjuvants on the immunogenicity
Drug Targets 10, 381–398. of Habu-venom toxoid in the monkey and the Guinea pig. Jpn. J. Med. Sci. Biol. 34,
León, G., Herrera, M., Segura, Á., Villalta, M., Vargas, M., Gutiérrez, J.M., 2013. 169–173.

72
G. León et al. Toxicon 151 (2018) 63–73

Sánchez, A., Segura, Á., Vargas, M., Herrera, M., Villalta, M., Estrada, R., Wu, F., Litschka- of Naja kaouthia (monocled cobra) venoms from three different geographical regions
Koen, T., Perry, M.A., Alape-Girón, A., León, G., 2017. Expanding the neutralization of Southeast Asia. J. Proteomics 120, 105–125.
scope of the EchiTAb-plus-ICP antivenom to include venoms of elapids from Southern Tan, C.H., Tan, K.Y., Lim, S.E., Tan, N.H., 2015b. Venomics of the beaked sea snake,
Africa. Toxicon 125, 59–64. Hydrophis schistosus: a minimalist toxin arsenal and its cross-neutralization by het-
Sarciaux, J.M., Mansour, S., Hageman, M.J., Nail, S.L., 1999. Effects of buffer composition erologous antivenoms. J. Proteomics 126, 121–130.
and processing conditions on aggregation of bovine IgG during freeze-drying. J. Tan, C.H., Liew, J.L., Tan, K.Y., Tan, N.H., 2016. Assessing SABU (Serum Anti Bisa Ular),
Pharm. Sci. 88 (12), 1354–1361. the sole Indonesian antivenom: a proteomic analysis and neutralization efficacy
Sasa, M., Arias Ortega, J., Bonilla-Murillo, F., 2017. Assessing survival of wild-caught study. Sci. Rep. 6, 37299.
snakes in open venom production systems. Toxicon 138, 49–52. Theakston, R.D., Warrell, D.A., Griffiths, E., 2003. Report of a WHO workshop on the
Schersch, K., Betz, O., Garidel, P., Muehlau, S., Bassarab, S., Winter, G., 2010. Systematic standardization and control of antivenoms. Toxicon 41, 541–557.
investigation of the effect of lyophilizate collapse on pharmaceutically relevant Trevett, A.J., Lalloo, D.G., Nwokolo, N.C., Theakston, D.G., Naraqi, S., Warrell, D.A.,
proteins I: stability after freeze-drying. J. Pharm. Sci. 99 (5), 2256–2278. 1995. Venom detection kits in the management of snakebite in Central province,
Segura, Á., León, G., Su, C.Y., Gutiérrez, J.M., Burnouf, T., 2009a. Assessment of the Papua New Guinea. Toxicon 33 (5), 703–705.
impact of solvent/detergent treatment on the quality and potency of a whole IgG USP (United States Pharmacopeia), 2014a. < 51 > antimicrobial effectiveness test. In:
equine antivenom. Biologicals 37, 306–312. USP 37/NF 32. U. S. Pharmacopeial Convention, Rockville, pp. 58 (translated to
Segura, Á., Herrera, M., González, E., Vargas, M., Solano, G., Gutiérrez, J.M., León, G., Spanish).
2009b. Stability of equine IgG antivenoms obtained by caprylic acid precipitation: USP (United States Pharmacopeia), 2014b. < 71 > Sterility test. In: USP 37/NF 32. U. S.
towards a liquid formulation stable at tropical room temperature. Toxicon 53, Pharmacopeial Convention, Rockville, pp. 84 (translated to Spanish).
609–615. USP (United States Pharmacopeia), 2014c. Pyrogen test. In: USP 37/NF 32. U. S.
Segura, A., Herrera, M., Villalta, M., Vargas, M., Gutiérrez, J.M., León, G., 2012. Pharmacopeial Convention, Rockville, pp. 166 (translated to Spanish).
Assessment of snake antivenom purity by comparing physicochemical and im- USP (United States Pharmacopeia), 2014d. Bacterial endotoxin test. In: USP 37/NF 32. U.
munochemical methods. Biologicals 41, 93–97. S. Pharmacopeial Convention, Rockville, pp. 111 (translated to Spanish).
Seifert, S.A., Boyer, L.V., 2001. Recurrence phenomena after immunoglobulin therapy for USP (United States Pharmacopeia), 2014e. NaCl. In: USP 37/NF 32. U. S. Pharmacopeial
snake envenomations: Part 1. Pharmacokinetics and pharmacodynamics of im- Convention, Rockville, pp. 5279 (translated to Spanish).
munoglobulin antivenoms and related antibodies. Ann. Emerg. Med. 37 (2), 189–195. Valverde, J.M., Rodríguez, K., Herrera, M., Segura, Á., Vargas, M., Villalta, M., Montero,
Simpson, I.D., Norris, R.L., 2007. Snake antivenom product guidelines in India: “the devil M., Gutiérrez, J.M., León, G., 2017. Comparison of the adjuvant activity of emulsions
is in the details”. Wilderness Environ. Med. 18 (3), 163–168. with different physicochemical properties on the antibody response towards the
Solano, G., Segura, A., Herrera, M., Gómez, A., Villalta, M., Gutiérrez, J.M., León, G., venom of West African carpet viper (Echis ocellatus). Toxicon 127, 106–111.
2010. Study of the design and analytical properties of the lethality neutralization Vargas, M., Segura, Á., Villalta, M., Herrera, M., Gutiérrez, J.M., León, G., 2015.
assay used to estimate antivenom potency against Bothrops asper snake venom. Purification of equine whole IgG snake antivenom by using an aqueous two-phase
Biologicals 38, 577–585. system as a primary purification step. Biologicals 43, 37–46.
Solano, S., Segura, Á., León, G., Gutiérrez, J.M., Burnouf, T., 2012. Low pH formulation of Waghmare, A., Deopurkar, R.L., Salvi, N., Khadilkar, M., Kalolikar, M., Gade, S.K., 2009.
whole IgG antivenom: impact on quality, safety, neutralizing potency and viral in- Comparison of Montanide adjuvants, IMS 3012 (nanoparticle), ISA 206 and ISA 35
activation. Biologicals 40, 129–133. (emulsion based) along with incomplete Freund's adjuvant for hyperimmunization of
Solano, G., Gómez, A., León, G., 2015. Assessing endotoxins in equine-derived snake equines used for production of polyvalent snake antivenom. Vaccine 27, 1067–1072.
antivenoms: comparison of the USP pyrogen test and the Limulus amoebocyte lysate Wagstaff, S.C., Laing, G.D., Theakston, R.D., Papaspyridis, C., Harrison, R.A., 2006.
assay (LAL). Toxicon 105, 13–18. Bioinformatics and multiepitope DNA immunization to design rational snake anti-
Solano, G., Gómez, A., Corrales, G., Chacón, D., Estrada, R., León, G., 2018. Contributions venom. PLoS Med. 3 (6), e184.
of the snake venoms of Bothrops asper, Crotalus simus and Lachesis stenophrys to the Wang, W., 1999. Instability, stabilization and formulation of liquid protein pharmaceu-
paraspecificity of the Central American polyspecific antivenom (PoliVal-ICP). ticals. Int. J. Pharm. 185, 129–188.
Toxicon 144, 1–6. WHO (World Health Organization), 2016. Guidelines for the Production, Control and
Sousa, J.R.F., Monteiro, R.Q., Castro, H.C., Zingali, R.B., 2001. Proteolytic action of Regulation of Snake Antivenom Immunoglobulins. WHO, Geneva.
Bothrops jararaca venom upon its own constituents. Toxicon 39, 787–792. Wong, K.Y., Tan, C.H., Tan, N.H., 2016. Venom and purified toxins of the spectacled
Steinbuch, M., Audran, R., 1969. The isolation of IgG from mammalian sera with the aid cobra (Naja naja) from Pakistan: insights into toxicity and antivenom neutralization.
of caprylic acid. Arch. Biochem. Biophys. 134 (2), 279–284. Am. J. Trop. Med. Hyg. 94, 1392–1399.
Stephano, M.A., Guidolin, R., Higashi, H.G., Higashi, H.G., Tambourgi, D.V., Sant'Anna, Wong, K.Y., Tan, C.H., Tan, K.Y., Quraishi, N.H., Tan, N.H., 2018. Elucidating the bio-
O.A., 2005. The improvement of the therapeutic anti-Lachesis muta serum production geographical variation of the venom of Naja naja (spectacled cobra) from Pakistan
in horses. Toxicon 45 (4), 467–473. through a venom-decomplexing proteomic study. J. Proteomics 175, 156–173.
Sunthornandh, P., Ratanabanangkoon, K., 1994. A comparative study of three vehicles on Ziska, S.M., Schumacher, J., Duran, S.H., Brock, K.V., 2012. Development of an auto-
antibody responses against elapid snake neurotoxin immunogens. Toxicon 32, mated plasmapheresis procedure for the harvest of equine plasma in accordance with
561–571. current good manufacturing practice. Am. J. Vet. Res. 73, 762–769.
Tan, K.Y., Tan, C.H., Fung, S.Y., Tan, N.H., 2015a. Venomics, lethality and neutralization

73

Вам также может понравиться