Buscar

2020-art-apsantos

Prévia do material em texto

C
ur
re
nt
 P
ha
rm
ac
eu
tic
al
 B
io
te
ch
no
lo
gy
�������
�	
�����
��
����������	��
�//�4��A"+!����
��//�4��"BA!CA�D
������	�������	
��
�	�������	
��
��
����
��
����
���
���	��
��
���	��
�
	�����������	����	�	�����������	����	�
����������	��
����������	��
�	
�����
��
�'
�()
*�()$�4
��E�D
�������
	
���
�
 
Ana P. dos Santos
1
, Tamara G. de Araújo
2
 and Gandhi Rádis-Baptista
3,*
 
1Program of Post-graduation in Pharmaceutical Sciences (FFEO/UFC), Federal University of Ceara, Ceara, Brazil; 
2Department of Pharmacy, Federal University of Ceara, Ceara, Brazil; 3Institute of Marine Sciences, Federal Universi-
ty of Ceara, Ceara, Brazil 
 Abstract: Venom-derived peptides display diverse biological and pharmacological activities, making 
them useful in drug discovery platforms and for a wide range of applications in medicine and pharma-
ceutical biotechnology. Due to their target specificities, venom peptides have the potential to be devel-
oped into biopharmaceuticals to treat various health conditions such as diabetes mellitus, hypertension, 
and chronic pain. Despite the high potential for drug development, several limitations preclude the di-
rect use of peptides as therapeutics and hamper the process of converting venom peptides into pharma-
ceuticals. These limitations include, for instance, chemical instability, poor oral absorption, short half-
life, and off-target cytotoxicity. One strategy to overcome these disadvantages relies on the formula-
tion of bioactive peptides with nanocarriers. A range of biocompatible materials are now available that 
can serve as nanocarriers and can improve the bioavailability of therapeutic and venom-derived pep-
tides for clinical and diagnostic application. Examples of isolated venom peptides and crude animal 
venoms that have been encapsulated and formulated with different types of nanomaterials with promis-
ing results are increasingly reported. Based on the current data, a wealth of information can be collect-
ed regarding the utilization of nanocarriers to encapsulate venom peptides and render them bioavaila-
ble for pharmaceutical use. Overall, nanomaterials arise as essential components in the preparation of 
biopharmaceuticals that are based on biological and pharmacological active venom-derived peptides. 
 
A R T I C L E H I S T O R Y 
 
 
Received: January 25, 2019 
Revised: April 17, 2019 
Accepted: May 08, 2019 
 
 
DOI: 
10.2174/1389201020666190621104624 
 
Keywords: Venom-derived peptides, therapeutic peptides, biopharmaceuticals, nanotechnology, drug delivery system, toxins. 
1. INTRODUCTION 
 Venomous animals are specialized predators that have 
developed over millions of years of evolution, the ability to 
produce venoms, and secretions composed of an incredible 
structural and functional diversity of toxins for their biologi-
cal purposes [1]. These compounds have evolved to deter, 
immobilize or kill a range of prey, predators, or competitors, 
in which the toxins can affect essential and vulnerable physi-
ological processes of the affected organism, such as neuro-
muscular signaling, hemostasis, and cardiovascular function, 
among others [2]. When injected into animals or human vic-
tims, usually in small amounts, these compounds can cause 
harmful disorders and even death. Venoms are composed of 
complex mixtures of salts, low molecular weight organic 
molecules such as polyamines, amino acids, venom en-
zymes, and, importantly, pharmacologically active peptides 
and polypeptides. A wide diversity of animals has developed 
the ability to produce these cocktails of compounds, such as 
snakes, lizards, scorpions, spiders, centipedes, ticks, leeches, 
 
*Address correspondence to this author at the Institute of Marine Sciences, 
Federal University of Ceara, Ceara, Brazil; Tel: +55 85 3366 7001; 
Fax: +55 85 3366 7002; E-mails: gandhi.radis@ufc.br; 
g.radis_baptista@yahoo.com 
cone snails, sea anemones, jellyfish, fish, cephalopods, echi-
noderms, several insect orders (such as ants, bees, wasps), 
and even some mammals (the platypus, shrews, vampire 
bats) [3]. Venoms are delivered to prey or predators through 
diverse types of specialized structures such as barbs, beaks, 
fangs, or modified teeth, harpoons, nematocysts, pinchers, 
proboscises, spines, sprays, spurs, and stingers [4]. Some of 
these animals can produce venoms in special glands, while 
others may contain toxic substances that spread throughout 
their body tissues [5]. 
 Over time, the biological diversity and potency of ven-
oms have become more refined, so that they display high 
specificity to their pharmacologically active targets, render-
ing animal toxin compounds extremely valuable for the de-
velopment of new therapeutics and adjuvant strategies in 
clinics [6, 7]. Venom-derived peptides act with excellent 
selectivity and potency on several biological targets, includ-
ing ligand- and voltage-dependent ion channels, G-protein 
coupled receptors, membrane transporters, and enzymes. 
When used in a suitable dosage, these peptides may be useful 
therapeutics [8, 9]. In the pharmaceutical field, peptide tox-
ins can be used in their original form (natural or synthetic) or 
1873-4316/20 $65.00+.00 © 2020 Bentham Science Publishers 
Send Orders for Reprints to reprints@benthamscience.net 
 Current Pharmaceutical Biotechnology, 2020, 21, 97-109 
97 
REVIEW ARTICLE 
Nanoparticles Functionalized with Venom-Derived Peptides and Toxins 
for Pharmaceutical Applications 
http://crossmark.crossref.org/dialog/?doi=10.2174/1389201020666190621104624&domain=pdf
98 Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 dos Santos et al. 
a chemically modified form, mimicking the desired action of 
the original peptide [2]. 
 In this context, drugs derived from venom peptides or 
protein toxins were approved by the Food and Drug Admin-
istration (FDA, U.S. Department of Health and Human Ser-
vices) for treatment of diverse clinical conditions, including 
diabetes, hypertension, and chronic pain. Several venom-
derived peptides are also in preclinical development, while 
others are under clinical studies that have the potential to 
treat a range of diseases, including cancer and autoimmune 
diseases [10]. Examples of venom-derived peptides and 
pharmaceuticals prepared from venom-peptide leads ap-
proved by the FDA that are in current clinical use are sum-
marized in Table 1. Although a relatively high number of 
peptides and proteins have been isolated from animal ven-
oms, such numbers reflect only a small part of the huge 
chemical diversity found in marine and venomous terrestrial 
organisms. It has been argued that there exists a practically 
infinite potentiality to be explored in the field of venom pep-
tides and derivatives [9, 10]. 
 Considering that many current therapies are deficient, the 
importance of animal venoms as innovative resources for 
drug discovery becomes clear when noting therapeutic pep-
tides are highly selective pharmacological agents. Despite 
the exceptional potential for drug discovery and develop-
ment, several limitations have been reported for this class of 
compounds, hampering the process of converting venom 
peptides into therapeutic agents. The low stability, short half-
life, and poor oral bioavailability are some of these barriers. 
Toxicity can often also be a limiting factor, capable of re-
stricting the development of new drugs. A promising strategy 
that has been developed to overcome these drawbacks is the 
use of nanocarriers for delivery and improve the bioavaila-
bility of venom peptides [11, 12]. Previous reports have em-
phasized the importance of nanostructures to deliver toxins 
and animal venoms for diverse biological applications [13, 
14]. Herein, examples of this technological approach are 
presented and discussed, from which information can be 
gleaned to select materials and conditions to encapsulate 
venom-derived peptides for pharmaceuticalapplications. In 
this scenario, the use of nanotechnology appears to be an 
advanced methodology that promises to overcome the intrin-
sic limitations of the bioactive venom-derived peptides in the 
development of biopharmaceuticals. 
2. LIMITATIONS AND CHALLENGES OF PEPTIDES 
FOR PHARMACEUTICAL APPLICATIONS 
 Despite the potentially useful pharmacological properties 
of peptide toxins, their therapeutic application is often ham-
pered by inconvenient aspects inherent to their structures. 
Similar to other peptide drugs, venom peptides generally 
exhibit varying degrees of chemical instability in vivo, since 
they have poor intestinal absorption, and are rapidly elimi-
nated when they reach the bloodstream. In addition, the tox-
icity associated with venom peptides has also contributed to 
restricting the development of new drugs [15-17]. 
 The intestinal uptake of peptides is generally limited by 
their high molecular weights and polarities, in a way that 
such characteristics, in this case, negatively interfere with the 
ability of these compounds to cross the intestinal membranes 
in an unmodified and active form. Moreover, the presence of 
proteinases in the digestive system rapidly causes the amide 
bonds of peptides to break down, contributing to their low 
stability in vivo. All these facts preclude the oral administra-
tion of peptides, which is the most favorable route acceptable 
by patients, in detriment to much less convenient invasive 
alternatives [18]. Even if peptides can reach the bloodstream 
in certain cases, they are degraded rapidly by various circu-
lating proteases, or even eliminated from the body by the 
hepatic and renal systems. This chemical instability is related 
to the individual structure of each peptide, such as stabiliza-
tion by post-translational modification. Linear peptides 
(without disulfide bonds or other covalent constraints) are 
more susceptible to proteolysis and more rapidly degraded 
in vivo [17]. These characteristics could decrease the half-life 
of peptides in the bloodstream and, consequently, require 
repetitive administration to obtain their therapeutic effects 
when in use. Repetitive drug administration is an undesirable 
regimen for patients and contributes to decreased compliance 
with a given treatment [19]. Apart from these barriers, toxici-
ty may also be associated with the use of venom peptides as 
pharmaceuticals, especially the antimicrobial and cytolytic 
peptides. These classes of peptides, expressed in the venom 
and venom glands of snakes, scorpions, bees, wasps and oth-
er animals can display an off-target effect causing cytotoxici-
ty and damage to diverse biological systems [17, 20-24]. 
High toxicity to humans imposes, in some circumstances, the 
discontinuation of venom peptides in clinical trials of drug 
development platforms [25]. 
 Considering these facts, some strategies have been de-
vised to overcome these drawbacks, to advance the devel-
opment of venom peptides for therapeutic purposes. Cycliza-
tion, for example, is a technique that has been tested to in-
crease the resistance of peptides to proteolysis. Alternatively, 
PEGylation and covalent binding to larger proteins, as well 
as the incorporation of peptides into slow release polymer 
matrices have been studied to increase peptide half-life in the 
bloodstream [9]. Additionally, to increase peptide half-life, 
platforms for controlled release with nanocarriers that can be 
optimized to deliver the peptides and circumvent peptide 
structural susceptibilities and the intrinsic toxic side-effects 
have emerged as a promising strategy for the development of 
peptides as pharmaceuticals [26]. Fig. (1) schematically il-
lustrates the main points involved in the development of bio-
pharmaceuticals from venom peptides with the application of 
nanocarriers and nanotechnology. 
3. THERAPEUTIC APPLICATIONS OF VENOM-
DERIVED PEPTIDES 
 Venom-derived peptides have a range of biological activ-
ities useful for treating various clinical conditions, and some 
are already in clinical use (Table 1). Apart from these ap-
proved drugs, listed in Table 1, studies have shown for in-
stance, venom bioactive peptides with antitumor effects are 
attractive compounds for the treatment of cancer. As exam-
ples, peptides TsAP-1 and TsAP-2 from the venom of the 
Brazilian yellow scorpion Tityus serrulatus showed an anti-
proliferative activity against human cancer cell lines [23]. 
Lycosin-1 and latarcin-2a, derived from spider venoms [27, 
28], and the recently characterized vipericidin peptides [20], 
from rattlesnake venom glands, as well as crotamine and 
Nanoparticles Functionalized with Venom-Derived Peptides Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 99 
Table 1. Approved drugs from animal venom-derived peptides and leads that are in current clinical use. 
FDA* 
Approval 
Drug Name 
(Active Ingredient) 
Source 
(Species Origin) Mechanism of Action Indication 
Dosage 
Form/Route Dosage and Administration 
1981 Captopril 
Venom of the snake 
Bothrops jararaca 
Angiotensin-converting 
enzyme inhibitor. 
Hypertension, 
Heart Failure 
Tablet/ oral 
25 mg to 150 mg two or three 
times a day. 
1998 
AGGRASTAT® 
(Tirofiban) 
Venom of the snake 
Echis carinatus 
An antagonist of the platelet 
glycoprotein (GP) IIb/IIIa 
receptor, inhibits platelet 
aggregation. 
Acute coronary 
syndrome 
Injectable/ 
Intravenous 
Initial rate of 0.4 µg/kg/min for 30 
minutes and then continued at 0.1 
µg/kg/min. 
1998 
INTEGRILIN™ 
(Eptifibatide) 
Venom of the snake 
Sistrurus miliarius 
barbouri 
Prevents binding of fibrino-
gen, von Willebrand factor, 
and other adhesive ligands 
to GPIIb/IIIa receptors; 
inhibits platelet aggregation. 
Acute coronary 
syndrome 
Injectable/ 
intravenous 
An intravenous bolus dose of 180 
µg/kg as soon as possible follow-
ing diagnosis, followed by a con-
tinuous infusion of 2 µg/kg/min 
until hospital discharge or the 
initiation of CABG surgery, up to 
72 hours. 
2000 
ANGIOMAX™ 
(Bivalirudin) 
Saliva of the medici-
nal leech Hirudo 
medicinalis 
A reversible direct thrombin 
inhibitor exhibits anticoagu-
lant effects. 
An anticoagu-
lant in percuta-
neous coronary 
intervention 
Injectable/ 
Intravenous 
An intravenous bolus dose of 1.0 
mg/kg followed by a 4-hour infu-
sion at a rate of 2.5 mg/kg/h. If 
needed, an additional infusion may 
be initiated at a rate of 0.2 mg/kg/h 
for up to 20 hours. 
2004 
PRIALT® 
(ziconotide) 
Venom of the cone 
snail Conus magus 
N-type calcium channel 
(Cav2.2) antagonist; blocks 
excitatory neurotransmitter 
release in primary afferent 
nerve terminals and antino-
ciception. 
Severe chronic 
pain 
Injectable/ 
Intrathecal 
Initiated at no more than 2.4 
µg/day (0.1 µg/h) and titrated to 
patient response. Doses may be 
titrated upward by up to 2.4 
mcg/day (0.1 µg/h) at intervals of 
no more than 2-3 times per week, 
up to a recommended maximum of 
19.2 µg/day (0.8 µg/h) by Day 21. 
2005 
BYETTA™ 
 (Exenatide) 
Venom of the Gila 
monster Heloderma 
suspectum 
An incretin-mimetic agent 
enhances glucose-dependent 
insulin secretion and several 
other anti-hyperglycemic 
actions of incretins. 
Type 2 diabe-
tes mellitus 
Injectable/ 
subcutaneous 
Initiated at 5 µg per dose twice 
daily at any time within 60-
minutes before morning and even-
ing meals. Based on clinical re-
sponses, the dose can be increased 
to 10 µg twice daily after 1 month 
of therapy 
*U.S. Food and Drug Administration, Department of Health and Human Services (http://www.fda.gov). 
crotamine-derivatives [29, 30], have shown promising anti-
tumor effects. In more mature stages, bioconjugates of chlo-
rotoxin, a disulfide-containing peptide derived from the ven-
om of the scorpion Leiurus quinquestriatus, is in phase I and 
II clinical trials to target and treat glioma tumors [31]. Cyto-
toxic activity against certain typesof cancer cells is also ex-
hibited by bee and wasp venoms, from which the peptides 
melittin and mastoparan were isolated, respectively [24, 32]. 
Melittin has been extensively studied as an antitumor agent, 
in addition to its antibacterial, antiviral, antifungal, and an-
tiparasitic effects. Such biological activities are due to mem-
brane destabilization by the peptide, forming pores or acting 
as a surfactant, causing membrane disruption. Thus, cell 
permeability is increased, and cell lysis invariably occurs 
[33, 34]. These membrane effects are common to other ven-
om peptides and are potentiated in cancer cells because of 
greater electrostatic interactions due to the small size and net 
positive charge of these compounds [35]. 
 Perturbation and disruption of the cytoplasmic mem-
branes are also the main mechanisms of antimicrobial pep-
tides that can be found in diverse animal venoms. These 
compounds have attracted special attention as a source of 
new anti-infective agents, especially with the emergence of 
drug resistance of microorganisms to conventional therapies. 
Such peptides are naturally present as part of the innate or 
adaptive immunity of animals and may cause destabilization 
of membranes and cell lysis, as well as interaction with the 
intracellular components, like DNA, contributing to the 
mechanism of toxicity in the targeted microorganism. Sever-
al antimicrobial peptides have already been characterized 
from the venom of bees, ants, scorpions, cone snails, snakes, 
spiders, centipedes, and wasps, and collectively a library 
with hundreds of active compounds against bacteria, fungi, 
protozoa, and viruses is available. These venom-derived an-
timicrobial peptides have considerable potency at minimal 
inhibitory concentrations (in the micromolar range). A limiting 
100 Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 dos Santos et al. 
 
 
Fig. (1). Nanocarriers and nanotechnology to improve the bioavailability and delivery of venom-derived peptides. Venom peptides can be devel-
oped into target-specific drugs by circumventing some intrinsic drawbacks of peptides used as therapeutics, such as chemical instability, poor 
intestinal absorption, toxicity, and short half-lives. The improvement of these peptide limitations to turn peptides into biopharmaceuticals can be 
achieved by employing nanocarriers. (A higher resolution/colour version of this figure is available in the electronic copy of the article). 
 
factor for many of these peptides is the recurrent nonspecific 
action on non-target cells and tissues, and a variable level of 
hemolytic activity, requiring additional strategies to render 
them viable for antimicrobial therapy [3, 16]. 
 Venom peptides have been discovered and developed as 
therapeutic tools for diabetes mellitus [36]. Some of these 
peptides may act by mimicking endogenous metabolic hor-
mones such as glucagon-like peptide-1 (GLP-1), stimulating the 
release of glucose. This is the case for exendin-4, derived from 
the Gila monster Heloderma suspectum, which is currently on 
the market in its synthetic form Exenatide (Byetta ™) for 
treatment of type 2 diabetes mellitus. Other GLP-1 analogs 
have recently been reported in monotremes species including 
the platypus Ornithorhynchus anatinus and four species of 
echidna. Another peptide, an analog of human insulin (Con-
Ins G1), was found in the venom of the cone snail Conus 
geographus and showed significant agonist activity at the 
human insulin receptor with rapid release of insulin, but with 
a lower potency compared to insulin. Peptides can also act 
on ion channels expressed in pancreatic beta cells stimulat-
ing insulin secretion. Hanatoxin (κ-theraphotoxin-GR1A) 
from the venom of the tarantula Grammostola rosea and 
guangxitoxin-1 (κ-theraphotoxin-Pg1a) from the tarantula 
Chilobrachys guangxiensis may act as voltage-gated potassi-
um-ion channel (subtype 2.1, KV2.1) blockers. The peptide 
Conkunitzin-S1, from the venom of the cone snail Conus 
striatus, can act as a blocker of the KV1.7 channel. Iberiotox-
in (IbTx), isolated from the venom of the East Indian red scor-
pion Buthus tamulus, blocks BK channels. Despite their bene-
ficial effects, some of these channels could be present in other 
cells and tissues of the human body, limiting the use of these 
peptides in therapy [36]. 
 Other classes of peptides, derived from animal venoms, 
include examples with specificity for the cardiovascular sys-
tem. For example, bradykinin-potentiating peptides (BPPs) 
potentiate the antihypertensive effects of bioactive kinins, 
inhibiting their degradation by angiotensin-converting en-
zyme (ACE) in the bloodstream. BPPs were originally found 
in the venom of the Brazilian pit viper Bothrops jararaca 
[37]. In this group, teprotide (Bj-PRO-9a) was the most 
promising peptide, and its most effective analog, captopril, 
was approved in the 1980s for the treatment of hypertension 
and congestive heart failure (Table 1). This class of peptides 
has also been found in the venoms of other species of snakes, 
such as Crotalus and Lachesis, as well as in the venom of 
other animals such as scorpions and spiders. Besides BPPs, 
natriuretic peptide (DNP-CNP Bj, TNP-a, b-TNP, TNP-c 
TsNP), antiarrhythmic peptides (Jingzhaotoxin -I, PhKv, 
GsMTx -4), peptides similar to vasopressin (conopressins), 
and the selective inhibitors of the α1 adrenergic receptor (ρ-
TIA conopeptides) from sea snails of the genus Conus are 
examples of other relevant animal venom toxins that have 
potential for the regulation of blood pressure and other car-
diovascular disorders [38]. 
Nanoparticles Functionalized with Venom-Derived Peptides Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 101 
 With regard to the cardiovascular system, blood hemo-
stasis is another condition that can be affected and modulated 
by animal toxins, especially those derived from snakes. In-
terfering with platelet function, these compounds can act by 
several mechanisms involving disintegrins, C-type lectin-like 
toxins, three-finger toxins, phospholipase A2, serine protein-
ases, metalloproteinases, nucleotidases and L-amino oxidas-
es [39]. The disintegrins block platelet aggregation by bind-
ing to the αIIbβ3 receptor, preventing the binding of fibrino-
gen. Despite disturbing hemostasis, peptides in this class of 
venom toxins were utilized for the development of two ther-
apeutic and commercially available antiplatelet agents, tiro-
fiban (Aggrastat™), based on an isolated venom polypeptide 
from the saw-scaled viper Echis carinatus, and eptifibatide 
(Integrillin™), a cyclic heptapeptide analog derived from the 
venom of Barbour's pygmy rattlesnake Sistrurus miliarius 
barbouri [40, 41]. Other venom peptide components may act 
as antagonists of thrombin, a blood clotting enzyme respon-
sible for cleaving fibrinogen into fibrin and leading to the 
formation of a blood clot. This is the case of bivalirudin 
(Hirulog™), a linear 20-residue peptide approved in 2009 for 
use as an anticoagulant drug. Bivalirudin was derived from 
hirudin, a mini-protein found in the saliva of the medical 
leech Hirudo medicinalis [15]. 
 Animal toxins have also been shown to be active on pain 
receptors, especially ion channels such as sodium and calci-
um, among others. A variety of animals, including bees, 
wasps, spiders, ants, centipedes, scorpions, snakes, and cone 
snails have neurotoxin peptides that act on ion channels and 
may be potential candidates for the development of new an-
algesic drugs. Ziconotide (Prialt) was approved over a dec-
ade ago by the FDA for the treatment of chronic pain 
through the blockade of N-type calcium channels. This drug 
is the ω-conotoxin MVIIA, a synthetic peptide derived from 
the venom of the cone snail Conus magus [42, 43]. 
 These few examples, summarized in Table 1, give a 
glimpse of the potential of venom-derived peptides for clini-
cal and diagnosticapplications. Further information regard-
ing the research and development of toxins as biopharmaceu-
ticals can be found elsewhere [44, 45]. 
4. NANOPARTICLES FORMULATED FOR DELIV-
ERY OF VENOM-DERIVED PEPTIDES 
4.1. Overview 
 The incorporation of drugs, and particularly, active pep-
tides into nanoparticles can potentially solve several prob-
lems concerning the delivery of therapeutic agents in con-
ventional formulations and, consequently, boost the devel-
opment of new pharmaceuticals. Desirable properties include 
improvements in solubility and bioavailability of active 
compounds or improvement in the ability to cross biological 
barriers [46]. Nanoparticles can improve the stability and 
half-life of substances as well as facilitate the absorption of 
compounds with low solubility and hydrophilicity. Pro-
longed release of active agents, which reduces the frequency 
of drug administration and decreases toxic side-effects, and 
improved patient safety are also attributes of drug and poly-
peptide nanoencapsulation. Another positive aspect concerns 
drug administration by non-invasive routes that otherwise 
might be impractical for free substances, thus compromising 
patient treatment compliance [47]. All these features have 
stimulated the incorporation of drugs into nanomaterials to 
progress the development of effective drugs and biopharma-
ceuticals. 
 Studies have reported the utility of nanoparticles for a 
diverse number of clinical applications, such as improve-
ments in the efficacy of anticancer therapies. This improve-
ment is achieved by targeting therapeutic agents directly to 
the tumor [48]. Generally, chemotherapeutics specifically or 
indiscriminately target normal and tumorous cells, causing 
significant side-effects to the patient. In these cases, the 
strategy of directing therapy is highly attractive. By enhanc-
ing drug bioavailability and chemosensitivity, nanoparticle-
mediated drug delivery may enhance the therapeutic efficacy 
against cancer cells and reduce damage to healthy cells and 
consequently attenuate side-effects, as seen with venom-
loaded nanoparticles [49]. Other promising applications of 
nanocarriers include increased efficacy and safety of antimi-
crobial therapy. Nanocarriers and nanoparticles may also 
improve oral bioavailability and permeation of medicinal 
agents through the skin and the blood-brain barrier, as well 
as be useful as systemic transporters for gene therapy [50-
55]. 
 Specifically, regarding protein loading, nanoparticles 
may confer additional and beneficial properties to the use of 
these molecules as therapeutic agents. Nanostructures can 
confer protection against degradation in vivo, increase half-
life, control release, and increase the selectivity and specific-
ity of biological targets, thus improving the safety and effi-
cacy of the therapy. This technology has enabled the delivery 
of proteins by alternative and more attractive routes, such as 
oral, nasal, pulmonary, and transdermal. Several polymeric, 
lipid, and inorganic nanocarriers have been developed for 
these purposes, and they are available for application [56]. 
Some formulations are already in the market or pharmaceuti-
cal development, such as a cyclosporine nanoemulsion (Ne-
oral™) for oral prevention of organ rejection after transplan-
tation and insulin liposomes for oral treatment of diabetes 
[57]. 
 It is noteworthy that, despite nanotechnology-based 
methodologies demonstrating excellent potential for the for-
mulation of therapeutic peptides and proteins, the clinical 
application of nanoparticles with this class of compounds is 
still in its early stages [56]. This scientific progress has led to 
the discovery of therapeutic proteins capable of effectively 
treating numerous diseases; however, the administration of 
peptide therapeutics is often hampered by several intrinsic 
limitations as mentioned above. These drawbacks have stim-
ulated research in nanotechnology to promote the administra-
tion of peptides and proteins in more efficient modes. Fur-
thermore, nanotechnology offers the possibility of incorpo-
rating peptides and proteins into nanocarriers that, among 
other advantages, protect cargo therapeutic polypeptides 
from the external environment and render them available in 
the respective sites of action [57, 58]. The use of nanostruc-
tured particles to deliver peptides, in addition to improving 
release kinetics, has the potential to minimize the effects of 
low selectivity, given that nanoparticles can be functional-
ized and can retain molecules in the nanostructure until the 
complete release at the desired site [59]. 
102 Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 dos Santos et al. 
4.2. Nanocarriers and Nanoparticles 
 Several nanoscale systems have been used in research 
and development of new drug delivery platforms. Nanocarri-
ers include inorganic nanoparticles, polymeric nanoparticles, 
solid lipid nanoparticles, nanostructured lipid carriers, lipo-
somes, and dendrimers, among other materials [60]. These 
nanocarriers have attracted great attention because they pre-
sent an exceptional surface area in relation to other systems, 
which confers excellent properties for the delivery of drugs 
such as high transport load capacity and controlled slow-
release [61]. Nanocarriers or nanoparticles are small struc-
tures, between 1 and 100 nanometers in diameter, with the 
ability to transport other substances as a single unit [62]. 
Biologically and pharmacologically active substances can be 
loaded within these nanostructures, entrapped in the matrix 
or fixed to the surface and therefore can be formulated from 
the most diverse types of materials and methodologies, prefer-
ably those that are biocompatible with biological systems 
[63]. Polymeric nanoparticles are usually formed from syn-
thetic polymers such as Poly(Lactide) (PLA), Poly(Lactide-
Co-Glycolide) (PLGA), Poly(Ε-Caprolactone) (PCL), and 
poly(amino acids), or natural polymers, such as chitosan, 
alginate, gelatin, or albumin [64]. These nanoparticles can be 
distinguished into two main types: nanocapsules and nano-
spheres. 
 Nanocapsules are characterized by a vesicular structure 
that acts as a container for active substances to be carried, by 
which entrapped compounds remain in the aqueous or non-
aqueous liquid core, surrounded by the solidified polymeric 
outer layer. Conversely, nanospheres appear as a spherical 
solid polymer mass in which the active substances are dis-
persed throughout the polymer matrix, both in the particle 
core and adsorbed to its surface [65]. Among the materials 
used to make these nanoparticles, PLGA is the most promi-
nent in drug development. It is a biodegradable and biocom-
patible material approved by the FDA and the European 
Medicines Agency for drug delivery systems [66]. Its degra-
dation by hydrolysis produces lactic acid and glycolic acid, 
is also biodegradable and its metabolites non-toxic, making 
this material an attractive resource for use in nanomedicine 
[11]. Chitosan nanoparticles have also received much atten-
tion for delivery of peptides, proteins, antigens, oligonucleo-
tides, and genes for intravenous and oral administration. 
Similarly, chitosan is widely used in pharmaceutical research 
and industry as a vehicle for drug release and as a biomedical 
material. Chitosan is a bio-adhesive polysaccharide, is bio-
degradable, non-toxic, and compatible with biological tissues 
in numerous toxicity tests and research [67]. 
 Physiologically tolerated, biocompatible, and biode-
gradable lipids also comprise nanocarriers that form 
nanostructures. If a nanostructured matrix is composed of 
solid lipids, at room temperature they form Solid Lipid Na-
noparticles (SLN) [68], whereas if the mixture is composed 
of solid and liquid lipids they are called Nanostructured Li-
pid Carriers (NLC). NLCs represent a new generation of 
lipid-based carriers,with greater capacity to incorporate 
drugs in their structures [69]. Liposomes are another type of 
nanocarriers composed of lipids, more precisely phospholip-
ids, forming one or more bilayers around an aqueous core 
[70]. In liposomes, organic drugs and polypeptides can be 
encapsulated within the aqueous core or the surface lipid 
bilayer. They have a structure similar to the cell membrane 
and are highly flexible, but their use is still limited by their 
low stability in the human body. Perfluorooctyl bromide 
nanoparticles form another type of nanostructure, consisting 
of a perfluorooctyl bromide (PFOB) core encapsulated with-
in a phospholipid monolayer or a polymeric shell [71]. 
PFOB is a linear perfluorocarbon well known for its bio-
compatibility and good tolerability in humans and has been 
approved by the FDA. Such nanoparticles may be useful as 
delivery vehicles for therapeutic agents, which may be dis-
solved in the lipid or polymeric outer layer. Lipid-coated 
particles are preferred because they can facilitate the delivery 
of active substances through a lipid exchange or lipid mix-
ture between the monolayer of the delivery system and the 
target cell membrane [71]. In addition, other materials such 
as cyclodextrins (cyclic oligosaccharides) [72], and inorganic 
nanoparticles, including gold, iron, silver, and silica nanopar-
ticles, have been used to incorporate substances for therapeu-
tic purposes [73, 74]. 
 Nanostructured particles can be synthesized from differ-
ent methods that are usually divided into two main catego-
ries, namely, bottom-up and top-down. The bottom-up meth-
odology is a constructive approach, in which nanoparticles 
are formed from the union of smaller and simpler substances. 
In contrast, the top-down method is based on a destructive 
technique, in which larger substances are broken down into 
pieces and converted into nanoparticles [75]. An essential 
consideration to prepare nanoparticles is knowledge about 
the physicochemical characteristics of nanocarriers, such as 
size, surface characteristics, loading capacity and capacity of 
drug release, as well as their stability, given that these prop-
erties can directly influence bioavailability [76, 77]. By 
changing the synthesis method or by functionalizing the sur-
face, for example, it is possible to design optimized nanocar-
riers to achieve specific purposes of drug administration 
[78], keeping in mind Paul Ehrlich´s principle of a drug act-
ing on the right target. For instance, polymeric nanocapsules 
can be formed around a protein, generating a polymer coat-
ing, combining the robustness of inorganic nanoparticles 
with the flexibility of liposomes [79], while nanoconjugation 
(bio-nanoconjugation), the covalent linkage between the 
biomolecule and the nanocarrier, can create cell- or receptor-
targeted nanoparticles [80]. Furthermore, nanoconjugation 
has the potential to reduce the toxicity of therapeutically 
active molecules, providing a mechanism to increase its ef-
fectiveness [12, 80]. Complete information regarding the 
types of nanocarriers, their structures, methods of prepara-
tion of nanoparticles, and indicated applications can be re-
trieved from specialized chapters in thematic publications 
[81-83]. 
4.3. Nanocarriers in the Cargo-Delivery of Venom-
Derived Peptides 
 Advances in nanocarriers for the formulation of venom 
peptides include, especially, an improved ability to bypass 
membranes, better absorption by alternative routes such as 
oral and nasal, increased selective cytotoxicity of peptides 
(e.g., antitumor peptides), reduced toxicity toward normal 
cells, and increased half-life. In Table 2 [84-98], examples 
from the literature of venom-derived peptides formulated with 
Nanoparticles Functionalized with Venom-Derived Peptides Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 103 
Table 2. Venom-derived peptide-loaded nanocarriers as biopharmaceuticals. 
Peptide Biological Activity Nanocarrier Application Refs. 
Exendin-4 GLP-1 agonist 
Chitosan-coated PLGA 
nanoparticles 
Increased transmembrane permeation in vitro and in vivo. 
Potential system for oral administration. 
[84] 
Lipid-based nanoparticles 
Increased transport and cellular uptake in vitro, and transport 
through the intestinal epithelium in vivo. Hypoglycemic effects in 
diabetic KKAy mice. A potential system for oral administration. 
[85] 
Solid lipid nanoparticles 
Maintenance of the positive effects on insulin secretion without 
apparent cytotoxic effects. 
[86] 
Neurotoxin Analgesic effect 
PLA nanoparticles modified 
with chitosan 
System for intranasal administration. Improved transport of the 
peptide to the brain through the blood-brain barrier. 
[87] 
Melittin 
Cytolytic/antitumor 
activity 
Perfluorocarbon 
nanoparticles 
Potential for attenuating HIV infectivity, reducing toxicity against 
spermatozoa and vaginal epithelial cells in vitro. 
[88, 89] 
Preserves antitumor effect, reduced hemolysis in vitro and 
reduced signs of toxicity in vivo. Increased melittin half-life. 
[90] 
Nano-liposomes with 
poloxamer 188 
Maintenance of antitumor activity. Reduction of toxicity in vivo. [91] 
Zeolitic imidazolate frame-
work-8 nanoparticles 
Increased antitumor activity and reduced toxicity, in vitro and 
in vivo. 
[92] 
Nanoassemblies of poly-
meric rigid core micelles 
Increased antitumor activity. [58] 
TsAP-1 
Cytolytic/antitumor 
activity 
Nanoassemblies of poly-
meric rigid core micelles 
Increased antitumor activity. [58] 
Melittin/sodium 
dodecyl sulfate 
complex 
Cytolytic/antitumoral 
activity 
PLGA nanoparticles Maintenance of antitumor effect in vitro. [93] 
NKCT1 Antitumor activity 
Gold nanoparticles 
Reduction of toxicity in vitro and in vivo. 
Potentiation of antitumor effects. 
[12, 94] 
PEGylated gold 
nanoparticles 
Potentiation of antitumor effects. [95] 
ICD-85 Antitumor activity 
Sodium alginate 
nanoparticles 
Increased toxicity in tumor cells and decreased toxicity in normal 
cells in vitro. 
[96, 97] 
Bothrops toxin-II Leishmanicidal activity Liposomes 
Selective toxicity against promastigotes and amastigotes of 
Leishmania amazonensis. 
Reduced toxicity to the host cell. 
[98] 
 
nanocarriers are listed with regard to the type of nanostruc-
ture and the improved properties that were achieved with this 
special, nanoscale formulation technology. 
 Exenatide (Byetta™) is a synthetic version of exendin-4. 
When launched in 2005 for the treatment of diabetes mellitus 
type 2, two injections per day were required [99]. Recently, a 
new version of exenatide encapsulated in PLGA micro-
spheres (Bydureon™) was approved for clinical use; in this 
version, the gradual release of exenatide allows desirable 
therapeutic effects with injections administered weekly 
[100]. Despite this improvement, formulations for subcuta-
neous injections continue to be limited in their therapeutic 
applications, which is responsible for low adherence to the 
drug regimen, especially in the treatment of chronic diseases. 
In this sense, some studies have already reported delivery 
systems based on nanotechnology capable of improving the 
oral absorption of exendin-4. 
 Uncoated and chitosan-coated PLGA nanoparticles were 
designed to increase the transmembrane transport of exen-
din-4, reaching 8.9 and 16.5-fold increases, respectively, 
compared with non-encapsulated peptides, using a Madin-
Darby canine kidney (MDCK) cell model [84]. The in-
creased transport into the cells of this kind of functionalized 
nanoparticle is attributed to the net-positive charge of chi-
tosan-coated PLGA nanoparticles that promote interaction 
with the negatively charged membrane of a target cell and 
facilitates the cellular uptake of the carrier peptide. Thus, this 
delivery strategy is useful not only to improve the oral ad-
104 Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 dos Santos et al. 
ministration ofexenatide but also of other drug cargoes. 
Similar results were found with exendin-4 encapsulated in 
lipid-based nanoparticles composed of an aqueous core with 
drug-loaded sodium cholate micelles and a mixed solid lipid 
shell [85]. The cellular uptake and transport of peptides were 
significantly increased in vitro. Exendin-4 was also readily 
transported through the epithelium, achieving a bioavailabil-
ity of 12.7% in vivo, following intestinal administration in 
rats. The use of SLN may also to be a promising alternative 
for the administration of exendin-4, as the encapsulation in 
these nanostructures was readily accomplished and main-
tained the effectiveness of the original peptide to stimulate 
glucose-dependent insulin secretion (incretin effect) in INS-1 
cells in vitro, without inducing cytotoxic effects [86]. 
 In relation to the oral administration of nanoparticles, 
size is one of the most important characteristics for absorp-
tion, distribution, and behavior in vivo. In vitro and in vivo 
cellular uptake of nanocarriers with a particle size around 
100 nm is much more expressive than larger particles, such 
as microparticles. This observation can be explained by the 
mechanisms of nanoparticle translocation through the intes-
tinal epithelium, which involves more efficient absorption in 
the Peyer’s patch [101]. 
 Another example of peptide-loaded nanocarriers de-
signed to overcome barriers has been ascribed to the encap-
sulation of an analgesic peptide neurotoxin derived from the 
venom of the Asian cobra, Naja atra. Such analgesic pep-
tides can effectively block the acetylcholine receptor in the 
target membrane of host (or patient) cells, preventing neuro-
transmission, despite the low permeability of the peptide to 
blood-brain barrier translocation, limiting its bioactivity. 
Indeed, chitosan-modified PLA nanoparticles were formulat-
ed by the double emulsification solvent evaporation method 
for nasal administration, and studies in rats showed that neu-
rotoxin transport in the brain was improved [87]. The physi-
cochemical properties of chitosan allow enhanced transmu-
cosal absorption, especially in the case of nasal delivery of 
polar drugs, including peptides and proteins. The use of 
nanocarriers has made possible the reduction of toxicity of 
many peptides toward non-target cells and tissues of biologi-
cal systems. Concomitantly, nanotechnological formulations 
have also potentiated the therapeutic effect of compounds 
already in use. 
 A remarkable application of nanotechnology for toxicity 
reduction involves the encapsulation of melittin, a cytolytic 
peptide derived from the venom of the honey bee, Apis mel-
lifera, that displays diverse biological effects in vivo, includ-
ing anticancer effects. Melittin can insert into lipid bilayers 
to form pores that result in cell lysis. The load and transport 
of melittin into perfluorocarbon nanoparticles has been re-
ported. PFOB-melittin was formulated by an oil-in-water 
emulsion, composed of a liquid perfluorooctyl bromide core 
and a phospholipid monolayer, forming a stabilizing inter-
face with the aqueous medium. Perfluorocarbon nanoparti-
cles were initially synthesized by sonication and microfluidi-
zation, and melittin was later incorporated into the outer lipid 
monolayer of this nanosystem. Melittin delivery was suc-
cessfully achieved as confirmed by the ability to form pores 
in a dual-membrane model in vitro [102]. The encapsulation 
of melittin is justified by the non-selective cytotoxicity 
caused by this peptide against normal, non-targeted cells and 
erythrocytes, which produces hemolysis and precludes the 
use of melittin in therapy. 
 Using nanocarriers, the incorporation of melittin was 
reported to reduce its cytotoxicity against human spermato-
zoa and vaginal epithelial cells (VK2/E6E7) at concentra-
tions shown to be adequate to restrain HIV infectivity [88-
89]. Perfluorocarbon nanoparticles also reduced the hemolyt-
ic activity of melittin in vitro, and no apparent toxicity was 
demonstrated in mice after injection of a dose of nanoparti-
cles equivalent to four times the LD50 of free melittin. No 
significant changes in serum enzyme or electrolyte levels 
and no evidence of hemolysis or tissue damage in the liver, 
lung, kidney, and heart were observed. Nanoparticle-
formulated melittin maintains toxicity against B16F10 mela-
noma cells in vitro, but at a much lower magnitude than free 
melittin, reflecting the slow release of the formulated 
nanosystem. In addition, nanoparticle incorporation reduced 
the clearance of melittin when administered intravenously to 
mice, resulting in a 10-fold increase in the amount of circu-
lating melittin in the blood 2 hours after injection. Interest-
ingly, the anticancer efficacy of melittin-loaded target-driven 
nanoparticles was similar to the non-targeted composites, 
indicating that such a loaded carrier may exert their anti-
tumor effects by a nonspecific uptake mechanism by the 
tumor vasculature or by binding to an overexpressed integrin 
receptor in tumor cells [90]. Other examples of melittin-
nanocarriers include the inclusion in Poloxamer 188 nano-
liposomes that also reduced toxicity in vivo, which produced 
less tissue damage and toxicity to liver cells, reduced neutro-
phil-mediated inflammatory responses, and reduced allergic 
responses in mice. At the same time, nano-liposomes main-
tained the therapeutic activity of melittin, as demonstrated by 
the induced apoptosis observed in liver carcinoma cells 
in vitro and in vivo, and the inhibition of hepatocellular car-
cinoma in the LM-3 xenograft tumor model [91]. Melittin-
loaded zeolitic imidazolate framework-8 (ZIF-8) nanoparti-
cles were also reported [92]. In another strategy, melittin was 
modified with an anionic agent, sodium dodecyl sulfate, by a 
hydrophobic ion-pairing technique and the complex was then 
incorporated into PLGA nanoparticles, which was active 
against MCF-7 breast cancer cells [93]. 
 The formulation of melittin with different nanocarriers 
illustrates the versatility of this technology concerning the 
numerous possibilities of producing biopharmaceuticals with 
venom-derived peptides and nanoparticles. In addition, 
nanostructured melittin has been prepared in association with 
a second peptide. Incorporation of polymeric rigid core mi-
celles into nano-assemblies enhanced the anticancer effects 
of melittin and the cytolytic peptide, TsAP-1, improving the 
therapeutic potential of the associated peptides in nanoparti-
cles [58]. Another example of peptide association and prepa-
ration of a delivery nanosystem includes sodium alginate 
nanoparticles loaded with ICD-85. ICD-85 nanoparticles 
were developed by an ionic gelation method and a combina-
tion of three peptides with antiproliferative activities that are 
derived from the venom of the Iranian brown snake 
Agkistrodon halys and of the yellow scorpion Hemiscorpius 
lepturus [96]. The ICD-85 nanoparticle increased cytotoxici-
ty against HeLa human cervical carcinoma cells in vitro 
compared with free ICD-85, with a significant inhibitory 
Nanoparticles Functionalized with Venom-Derived Peptides Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 105 
effect on the growth of these cells. Moreover, in vitro assays 
showed that ICD-85 nanoparticles could significantly de-
crease the cytotoxicity toward healthy primary lamb kidney 
cells compared to the unencapsulated peptides, again demon-
strating reduced off-target toxicity of peptide-loaded nano-
particles [97]. 
 Liposome nanocarriers have been used to encapsulate 
venom peptides. Bothrops toxin-II (BthTX-II), an ASP49 
phospholipase A2 derived from B. jararacussu venom, was 
formulated with lipid vesicles prepared by the extrusion 
method using dipalmitoylphosphatidylcholine, dipalmito-
ylphosphatidylserine, and cholesterol as a matrix. As an ex-
perimental therapy to treat cutaneous leishmaniasis, Asp49 
liposomes showeda high selectivity index compared with 
free Asp49, indicating a selectivity against L. amazonensis 
promastigotes, and preserving the viability of J774 macro-
phages in vitro. Liposomes also reduced the growth of 
amastigote forms within peritoneal macrophages of mice, 
demonstrating their selective leishmanicidal activity inside 
cells. This seems to be a promising armamentarium against 
leishmaniasis, in which the parasites are killed whereas host 
cells are undamaged [98]. From the venom of the monocled 
cobra Naja kaouthia, a 6.76 kDa toxin (NKCT1) that dis-
plays an anticancer effect, was conjugated with gold nano-
particles and showed increased antitumor effect compared to 
free toxin [12]. Besides the expected improvement in anti-
cancer effects, the nanoconjugation of NKCT1 reduced pep-
tide intrinsic toxicity by approximately three-fold in rat lym-
phocytes, as well as reduced the cardiotoxicity, nephrotoxici-
ty, and mast cell degranulation. Predictive outcomes of acute 
and sub-chronic toxicity were also reduced in vivo [12, 94]. 
Similarly, NKCT1 protein conjugated with PEGylated gold 
nanoparticles, using the sodium boron reduction technique, 
increased the anticancer property of NKCT1 [95]. 
4.4. Delivery of Crude Animal Venom Using Nanoparti-
cles 
 In addition to isolated, pure venom-derived peptides, 
nanoparticles have also been used to encapsulate non-
fractionated, crude venoms, as shown in Table 3. A combi-
nation of silica nanoparticles with the venom of the snake 
Walterinnesia aegyptia potentiated the growth arrest and 
apoptosis of human breast carcinoma cells (MDA-MB-231 
and MCF-7) and prostate cancer cells (PC3 and LNCaP), in 
comparison with the non-formulated venom, without affect-
ing normal cells. Similarly, these nanoparticles also reduced 
the volume of breast and prostate tumors in experimental 
animal tumor models. This reduction was twice as pro-
nounced as that seen with the crude venom, without, howev-
er, causing significant side-effects on body weight - a predic-
tive parameter of low toxicity in vivo [49]. In another 
nanosystem, chitosan tripolyphosphate nanoparticles con-
taining Russell's viper venom were formulated using the ion-
ic gelation method. No significant morphological alterations 
were produced in normal cardiac cells treated with these 
nanoparticles. These venom-loaded nanoparticles displayed 
low cytotoxicity and caused less than 10% mortality of 
healthy cardiac cells [103]. The same type of nanoparticles 
was studied for encapsulation of the crude venom of the In-
dian cobra Naja naja oxiana to be used as an alternative to 
traditional adjuvant systems for future production of biologi-
cal pharmaceuticals [67]. 
CONCLUSION 
 Venom-derived peptides have emerged as promising re-
sources for the development of new biopharmaceuticals and 
diagnostic tools, particularly due to their ability to interact 
selectively and specifically with therapeutically relevant mo-
lecular targets. As with most peptide-based drugs, stability 
and bioavailability are critical conditions for achieving a 
high efficacy of peptides as therapeutics. Moreover, off-
target effects may cause adverse reactions that are a matter 
of caution, particularly, in the case of venom peptides. The 
use of nanocarriers for drug delivery has shown to be advan-
tageous for pharmaceutical formulation, allowing fine ad-
justments in the composition of nanocarriers to transport and 
release loaded compounds that are usually hydrophilic, struc-
turally labile, and potentially toxic. The union of these two 
areas of knowledge, that is, toxinology and pharmaceutical 
manufacturing, can boost the discovery of new therapeutic 
agents and advance how peptides are used therapeutically 
and diagnostically, translating this class of compounds, from 
the bench into final products with clinical applications. The 
theme of the present work enlightens the use of nanotechnol-
ogy and nanocarriers to overcome some disadvantages of 
peptide administration, particularly venom-derived peptides 
with unique biological activities and pharmacological ef-
fects. Despite that, the majority of examples in the literature 
concern experimental studies, some formulations have al-
ready achieved clinical use, by which nanoparticles loaded or 
even functionalized with venom peptides have great potential 
Table 3. Crude animal venom-loaded nanocarriers for pharmaceutical applications. 
Venom Biological Activity Nanocarrier Application Refs. 
Walterinnesia aegyptia snake 
venom 
Antitumor activity Silica nanoparticles 
Increased antitumor activity and reduced toxicity, 
in vitro and in vivo. 
[49] 
Russell's viper venom Antigen 
Chitosan tripolyphosphate 
nanoparticles 
Low toxicity. Potential adjuvant system for future 
medicines. 
[103] 
Naja naja oxiana snake venom Antigen 
Chitosan tripolyphosphate 
nanoparticles 
Potential adjuvant system for future medicines. [67] 
 
 
106 Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 dos Santos et al. 
to move forward in drug development platforms, as well as 
in the production of biopharmaceuticals for effective applica-
tions in pharmaceutical biotechnology. 
CONSENT FOR PUBLICATION 
 Not applicable. 
FUNDING 
 Ana Paula dos Santos is the recipient of a master fellow-
ship from the State Funding Agency (FUNCAP/CE), granted 
to the Postgraduate Program in Pharmaceutical Sciences by 
the Faculty of Pharmacy, Dentistry, and Nursing, the Federal 
University of Ceara. 
CONFLICT OF INTEREST 
 The authors declare no conflict of interest, financial or 
otherwise. 
ACKNOWLEDGEMENTS 
 Gandhi Rádis-Baptista is grateful to The National Coun-
cil for Scientific and Technological Development (CNPq), 
and the Ministry of Science, Technology, and Innovation of 
the Federal Government of Brazil for the endorsement of 
research projects and financial support. 
REFERENCES 
[1] Peigneur, S.; Tytgat, J. Toxins in drug discovery and pharmacolo-
gy. Toxins (Basel), 2018, 10(3), E126. 
[http://dx.doi.org/10.3390/toxins10030126] [PMID: 29547537] 
[2] Takacs, Z.; Nathan, S. Animal Venoms in Medicine. Encyclopedia 
of Toxicology, 3rd ed; Wexler, P., Ed.; Academic Press: Oxford, 
2014, pp. 252-259. [http://dx.doi.org/10.1016/B978-0-12-386454-
3.01241-0] 
[3] Primon-Barros, M.; José Macedo, A. Animal venom peptides: 
Potential for new antimicrobial agents. Curr. Top. Med. Chem., 
2017, 17(10), 1119-1156. 
[http://dx.doi.org/10.2174/1568026616666160930151242] 
[PMID: 27697042] 
[4] Fry, B.G.; Roelants, K.; Champagne, D.E.; Scheib, H.; Tyndall, 
J.D.; King, G.F.; Nevalainen, T.J.; Norman, J.A.; Lewis, R.J.; Nor-
ton, R.S.; Renjifo, C.; de la Vega, R.C. The toxicogenomic multi-
verse: convergent recruitment of proteins into animal venoms. An-
nu. Rev. Genomics Hum. Genet., 2009, 10, 483-511. 
[http://dx.doi.org/10.1146/annurev.genom.9.081307.164356] 
[PMID: 19640225] 
[5] Utkin, Y.N. Animal venom studies: Current benefits and future 
developments. World J. Biol. Chem., 2015, 6(2), 28-33. 
[http://dx.doi.org/10.4331/wjbc.v6.i2.28] [PMID: 26009701] 
[6] Escoubas, P.; King, G.F. Venomics as a drug discovery platform. 
Expert Rev. Proteomics, 2009, 6(3), 221-224. 
[http://dx.doi.org/10.1586/epr.09.45] [PMID: 19489692] 
[7] Zambelli, V.O.; Pasqualoto, K.F.; Picolo, G.; Chudzinski-Tavassi, 
A.M.; Cury, Y. Harnessing the knowledge of animal toxins to gen-
erate drugs. Pharmacol. Res., 2016, 112, 30-36. 
[http://dx.doi.org/10.1016/j.phrs.2016.01.009] [PMID: 26826284] 
[8] Adams, D.J.; Lewis, R.J. Neuropharmacology of venom peptides. 
Neuropharmacology, 2017, 127, 1-3. 
[http://dx.doi.org/10.1016/j.neuropharm.2017.11.025] 
[PMID: 29154773] 
[9] King, G.F. Venoms as a platform for human drugs: Translating 
toxins into therapeutics. Expert Opin. Biol. Ther., 2011, 11(11), 
1469-1484. [http://dx.doi.org/10.1517/14712598.2011.621940] 
[PMID: 21939428] 
[10] Robinson, S.D.; Undheim, E.A.B.; Ueberheide, B.; King, G.F. 
Venom peptidesas therapeutics: Advances, challenges and the fu-
ture of venom-peptide discovery. Expert Rev. Proteomics, 2017, 
14(10), 931-939. 
[http://dx.doi.org/10.1080/14789450.2017.1377613] 
[PMID: 28879805] 
[11] Ye, C.; Chi, H. A review of recent progress in drug and protein 
encapsulation: Approaches, applications and challenges. Mater. 
Sci. Eng. C, 2018, 83, 233-246. 
[http://dx.doi.org/10.1016/j.msec.2017.10.003] [PMID: 29208283] 
[12] Saha, P.P.; Bhowmik, T.; Dasgupta, A.K.; Gomes, A. In vivo and 
in vitro toxicity of nanogold conjugated snake venom protein toxin 
GNP-NKCT1. Toxicol. Rep., 2014, 1, 74-84. 
[http://dx.doi.org/10.1016/j.toxrep.2014.04.007] 
[PMID: 28962228] 
[13] Utkin, Y.N. Modern trends in animal venom research - omics and 
nanomaterials. World J. Biol. Chem., 2017, 8(1), 4-12. 
[http://dx.doi.org/10.4331/wjbc.v8.i1.4] [PMID: 28289514] 
[14] Biswas, A.; Gomes, A.; Sengupta, J.; Datta, P.; Singha, S.; Dasgup-
ta, A.K.; Gomes, A. Nanoparticle-conjugated animal venom-toxins 
and their possible therapeutic potential. J. Venom Res., 2012, 3, 15-
21. [PMID: 23236583] 
[15] Harvey, A.L. Toxins and drug discovery. Toxicon, 2014, 92, 193-
200. [http://dx.doi.org/10.1016/j.toxicon.2014.10.020] 
[16] Perumal Samy, R.; Stiles, B.G.; Franco, O.L.; Sethi, G.; Lim, 
L.H.K. Animal venoms as antimicrobial agents. Biochem. Pharma-
col., 2017, 134, 127-138. 
[http://dx.doi.org/10.1016/j.bcp.2017.03.005] [PMID: 28288817] 
[17] Norton, R.S. Enhancing the therapeutic potential of peptide toxins. 
Expert Opin. Drug Discov., 2017, 12(6), 611-623. 
[http://dx.doi.org/10.1080/17460441.2017.1317243] 
[PMID: 28398099] 
[18] Lau, J.L.; Dunn, M.K. Therapeutic peptides: Historical perspec-
tives, current development trends, and future directions. Bioorg. 
Med. Chem., 2018, 26(10), 2700-2707. 
[http://dx.doi.org/10.1016/j.bmc.2017.06.052] [PMID: 28720325] 
[19] Uhlig, T.; Kyprianou, T.; Martinelli, F.G.; Oppici, C.A.; Heiligers, 
D.; Hills, D.; Calvo, X.R.; Verhaert, P. The emergence of peptides 
in the pharmaceutical business: From exploration to exploitation. 
EuPA Open Proteom., 2014, 4, 58-69. 
[http://dx.doi.org/10.1016/j.euprot.2014.05.003] 
[20] Falcao, C.B.; Pérez-Peinado, C.; de la Torre, B.G.; Mayol, X.; 
Zamora-Carreras, H.; Jiménez, M.A.; Rádis-Baptista, G.; Andreu, 
D. Structural dissection of crotalicidin, a rattlesnake venom catheli-
cidin, retrieves a fragment with antimicrobial and antitumor activi-
ty. J. Med. Chem., 2015, 58(21), 8553-8563. 
[http://dx.doi.org/10.1021/acs.jmedchem.5b01142] 
[PMID: 26465972] 
[21] Gao, B.; Zhu, S. Mesobuthus venom-derived antimicrobial peptides 
possess intrinsic multifunctionality and differential potential as 
drugs. Front. Microbiol., 2018, 9, 320. 
[http://dx.doi.org/10.3389/fmicb.2018.00320] [PMID: 29599756] 
[22] Almaaytah, A.; Zhou, M.; Wang, L.; Chen, T.; Walker, B.; Shaw, 
C. Antimicrobial/cytolytic peptides from the venom of the North 
African scorpion, Androctonus amoreuxi: Biochemical and func-
tional characterization of natural peptides and a single site-
substituted analog. Peptides, 2012, 35(2), 291-299. 
[http://dx.doi.org/10.1016/j.peptides.2012.03.016] 
[PMID: 22484288] 
[23] Guo, X.; Ma, C.; Du, Q.; Wei, R.; Wang, L.; Zhou, M.; Chen, T.; 
Shaw, C. Two peptides, TsAP-1 and TsAP-2, from the venom of 
the Brazilian yellow scorpion, Tityus serrulatus: Evaluation of their 
antimicrobial and anticancer activities. Biochimie, 2013, 95(9), 
1784-1794. [http://dx.doi.org/10.1016/j.biochi.2013.06.003] 
[PMID: 23770440] 
[24] Chen, X.; Zhang, L.; Wu, Y.; Wang, L.; Ma, C.; Xi, X.; Bininda-
Emonds, O.R.P.; Shaw, C.; Chen, T.; Zhou, M. Evaluation of the 
bioactivity of a mastoparan peptide from wasp venom and of its 
analogues designed through targeted engineering. Int. J. Biol. Sci., 
2018, 14(6), 599-607. [http://dx.doi.org/10.7150/ijbs.23419] 
[PMID: 29904274] 
[25] Pennington, M.W.; Czerwinski, A.; Norton, R.S. Peptide therapeu-
tics from venom: Current status and potential. Bioorg. Med. Chem., 
2018, 26(10), 2738-2758. 
[http://dx.doi.org/10.1016/j.bmc.2017.09.029] [PMID: 28988749] 
[26] Patel, A.; Patel, M.; Yang, X.; Mitra, A.K. Recent advances in 
protein and peptide drug delivery: A special emphasis on polymeric 
nanoparticles. Protein Pept. Lett., 2014, 21(11), 1102-1120. 
Nanoparticles Functionalized with Venom-Derived Peptides Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 107 
[http://dx.doi.org/10.2174/0929866521666140807114240] 
[PMID: 25106908] 
[27] Dubovskii, P.V.; Vassilevski, A.A.; Kozlov, S.A.; Feofanov, A.V.; 
Grishin, E.V.; Efremov, R.G. Latarcins: Versatile spider venom 
peptides. Cell. Mol. Life Sci., 2015, 72(23), 4501-4522. 
[http://dx.doi.org/10.1007/s00018-015-2016-x] [PMID: 26286896] 
[28] Tan, H.; Huang, Y.; Xu, J.; Chen, B.; Zhang, P.; Ye, Z.; Liang, S.; 
Xiao, L.; Liu, Z. Spider toxin peptide lycosin-i functionalized gold 
nanoparticles for in vivo tumor targeting and therapy. Theranostics, 
2017, 7(12), 3168-3178. 
[http://dx.doi.org/10.7150/thno.19780] [PMID: 28839471] 
[29] Mambelli-Lisboa, N.C.; Sciani, J.M.; Brandão Prieto da Silva, 
A.R.; Kerkis, I. Co-localization of crotamine with internal mem-
branes and accentuated accumulation in tumor cells. Molecules, 
(Basel, Switzerland), 2018, 23(4), E968. 
[http://dx.doi.org/10.3390/molecules23040968] [PMID: 29693555] 
[30] Tansi, F.L.; Filatova, M.P.; Koroev, D.O.; Volpina, O.M.; Lange, 
S.; Schumann, C.; Teichgräber, U.K.; Reissmann, S.; Hilger, I. 
New generation CPPs show distinct selectivity for cancer and non-
cancer cells. J. Cell. Biochem., 2019, 120(4), 6528-6541. 
[http://dx.doi.org/10.1002/jcb.27943] [PMID: 30362167] 
[31] Cohen, G.; Burks, S.R.; Frank, J.A. Chlorotoxin-A multimodal 
imaging platform for targeting glioma tumors. Toxins (Basel), 
2018, 10(12), E496 [http://dx.doi.org/10.3390/toxins10120496] 
[PMID: 30486274] 
[32] Wang, L.; Dong, C.; Li, X.; Han, W.; Su, X. Anticancer potential 
of bioactive peptides from animal sources (Review). Oncol. Rep., 
2017, 38(2), 637-651. 
[http://dx.doi.org/10.3892/or.2017.5778] [PMID: 28677775] 
[33] Liu, C.C.; Hao, D.J.; Zhang, Q.; An, J.; Zhao, J.J.; Chen, B.; 
Zhang, L.L.; Yang, H. Application of bee venom and its main con-
stituent melittin for cancer treatment. Cancer Chemother. Pharma-
col., 2016, 78(6), 1113-1130. 
[http://dx.doi.org/10.1007/s00280-016-3160-1] [PMID: 27677623] 
[34] Rady, I.; Siddiqui, I.A.; Rady, M.; Mukhtar, H. Melittin, a major 
peptide component of bee venom, and its conjugates in cancer ther-
apy. Cancer Lett., 2017, 402, 16-31. 
[http://dx.doi.org/10.1016/j.canlet.2017.05.010] [PMID: 28536009] 
[35] Mahadevappa, R.; Ma, R.; Kwok, H.F. Venom peptides: Improving 
specificity in cancer therapy. Trends Cancer, 2017, 3(9), 611-614. 
[http://dx.doi.org/10.1016/j.trecan.2017.07.004] 
[PMID: 28867164] 
[36] Robinson, S.D.; Safavi-Hemami, H. Venom peptides as pharmaco-
logical tools and therapeutics for diabetes. Neuropharmacology, 
2017, 127, 79-86. 
[http://dx.doi.org/10.1016/j.neuropharm.2017.07.001] 
[PMID: 28689026] 
[37] Camargo, A.C.; Ianzer, D.; Guerreiro, J.R.; Serrano, S.M. Brady-
kinin-potentiating peptides: beyond captopril. Toxicon, 2012, 59(4), 
516-523. [http://dx.doi.org/10.1016/j.toxicon.2011.07.013] 
[38] Rebello Horta, C.C.; Chatzaki, M.; Rezende, B.A.; Magalhães, 
Bde.F.; Duarte, C.G.; Felicori, L.F.; Ribeiro Oliveira-Mendes, 
B.B.; do Carmo, A.O.; Chávez-Olórtegui, C.; Kalapothakis, E. 
Cardiovascular-active venom toxins: An overview. Curr. Med. 
Chem., 2016, 23(6), 603-622. 
[http://dx.doi.org/10.2174/0929867323666160126142837] 
[PMID: 26812904] 
[39] de Queiroz, M.R.; de Sousa, B.B.; da Cunha Pereira, D.F.; 
Mamede, C.C.N.; Matias, M.S.; de Morais, N.C.G.; de Oliveira 
Costa, J.; de Oliveira, F. The role of platelets in hemostasis and the 
effects of snake venom toxins on platelet function. Toxicon, 2017, 
133, 33-47. [http://dx.doi.org/10.1016/j.toxicon.2017.04.013] 
[40] Koh,C.Y.; Kini, R.M. From snake venom toxins to therapeutics-
cardiovascular examples. Toxicon, 2012, 59(4), 497-506. 
[41] Sajevic, T.; Leonardi, A.; Krizaj, I. Haemostatically active proteins 
in snake venoms. Toxicon, 2011, 57(5), 627-645. 
[http://dx.doi.org/10.1016/j.toxicon.2011.01.006] 
[42] Sadeghi, M.; McArthur, J.R.; Finol-Urdaneta, R.K.; Adams, D.J. 
Analgesic conopeptides targeting G protein-coupled receptors re-
duce excitability of sensory neurons. Neuropharmacology, 2017, 
127, 116-123. 
[http://dx.doi.org/10.1016/j.neuropharm.2017.05.020] 
[PMID: 28533165] 
[43] Trim, S.A.; Trim, C.M. Venom: The sharp end of pain therapeutics. 
Br. J. Pain, 2013, 7(4), 179-188. 
[http://dx.doi.org/10.1177/2049463713502005] [PMID: 26516522] 
[44] Thayer, A.M. Improving peptides (small firms develop better pep-
tide drug candidates to expand this pharmaceutical class and attract 
big pharma partners). Chem. Eng. News, 2011, 89(22), 13-20. 
[http://dx.doi.org/10.1021/cen-v089n022.p013] 
[45] Fox, J.W.; Serrano, S.M. Approaching the golden age of natural 
product pharmaceuticals from venom libraries: An overview of tox-
ins and toxin-derivatives currently involved in therapeutic or diag-
nostic applications. Curr. Pharm. Des., 2007, 13(28), 2927-2934. 
[http://dx.doi.org/10.2174/138161207782023739] 
[PMID: 17979737] 
[46] Rizvi, S.A.A.; Saleh, A.M. Applications of nanoparticle systems in 
drug delivery technology. Saudi Pharmaceut. J., 2018, 26(1), 64-
70. [http://dx.doi.org/10.1016/j.jsps.2017.10.012] 
[47] Jain, K.K. Nanobiotechnology. Reference Module in Life Sciences; 
Elsevier, 2017. 
[http://dx.doi.org/10.1016/B978-0-12-809633-8.09044-0] 
[48] Jurj, A.; Braicu, C.; Pop, L.A.; Tomuleasa, C.; Gherman, C.D.; 
Berindan-Neagoe, I. The new era of nanotechnology, an alternative 
to change cancer treatment. Drug Des. Devel. Ther., 2017, 11, 
2871-2890. [http://dx.doi.org/10.2147/DDDT.S142337] 
[PMID: 29033548] 
[49] Badr, G.; Al-Sadoon, M.K.; Rabah, D.M. Therapeutic efficacy and 
molecular mechanisms of snake (Walterinnesia aegyptia) venom-
loaded silica nanoparticles in the treatment of breast cancer- and 
prostate cancer-bearing experimental mouse models. Free Radic. 
Biol. Med., 2013, 65, 175-189. 
[http://dx.doi.org/10.1016/j.freeradbiomed.2013.06.018] 
[PMID: 23811005] 
[50] Zazo, H.; Colino, C.I.; Lanao, J.M. Current applications of nano-
particles in infectious diseases. J. Control. Release, 2016, 224, 86-
102. 
[http://dx.doi.org/10.1016/j.jconrel.2016.01.008] 
[51] Soliman, G.M. Nanoparticles as safe and effective delivery systems 
of antifungal agents: Achievements and challenges. Int. J. Pharm., 
2017, 523(1), 15-32. 
[http://dx.doi.org/10.1016/j.ijpharm.2017.03.019] 
[PMID: 28323096] 
[52] Palmer, B.C.; DeLouise, L.A. Nanoparticle-enabled transdermal 
drug delivery systems for enhanced dose control and tissue target-
ing. Molecules, 2016, 21(12), E1719. 
[http://dx.doi.org/10.3390/molecules21121719] [PMID: 27983701] 
[53] Saraiva, C.; Praca, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernar-
dino, L. Nanoparticle-mediated brain drug delivery: Overcoming 
blood-brain barrier to treat neurodegenerative diseases. J. Control. 
Release, 2016, 235, 34-47. 
[54] Chen, J.; Guo, Z.; Tian, H.; Chen, X. Production and clinical de-
velopment of nanoparticles for gene delivery. Mol. Ther. Methods 
Clin. Dev., 2016, 3, 16023. 
[http://dx.doi.org/10.1038/mtm.2016.23] [PMID: 27088105] 
[55] Shreya, A.B.; Raut, S.Y.; Managuli, R.S.; Udupa, N.; Mutalik, S. 
Active targeting of drugs and bioactive molecules via oral admin-
istration by ligand-conjugated lipidic nanocarriers: Recent advanc-
es. AAPS PharmSciTech., 2018, 20(1), 15. 
[http://dx.doi.org/10.1208/s12249-018-1262-2] [PMID: 30564942] 
[56] Yu, M.; Wu, J.; Shi, J.; Farokhzad, O.C. Nanotechnology for pro-
tein delivery: Overview and perspectives. J. Control. Release, 
2016, 240, 24-37. 
[57] Santalices, I.; Gonella, A.; Torres, D.; José Alonso, M. Advances 
on the formulation of proteins using nanotechnologies. J. Drug De-
liv. Sci. Technol., 2017, 42, 155-180. 
[http://dx.doi.org/10.1016/j.jddst.2017.06.018] 
[58] Misra, S.K.; Schwartz-Duval, A.S.; Pan, D. Genomic DNA interac-
tions mechanize peptidotoxin-mediated anticancer nanotherapy. 
Mol. Pharm., 2017, 14(7), 2254-2261. 
[http://dx.doi.org/10.1021/acs.molpharmaceut.7b00083] 
[PMID: 28544846] 
[59] Jallouk, A.P.; Palekar, R.U.; Pan, H.; Schlesinger, P.H.; Wickline, 
S.A. Modifications of natural peptides for nanoparticle and drug 
design. Adv. Protein Chem. Struct. Biol., 2015, 98, 57-91. 
[http://dx.doi.org/10.1016/bs.apcsb.2014.12.001] 
[PMID: 25819276] 
[60] Wilczewska, A. Z.; Niemirowicz, K.; Markiewicz, K. H.; Car, H. 
Nanoparticles as drug delivery systems. Pharmacologic. Reports, 
2012, (5), 1020-1037. 
[http://dx.doi.org/10.1016/S1734-1140(12)70901-5] 
108 Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 dos Santos et al. 
[61] Lee, B.K.; Yun, Y.H.; Park, K. Smart nanoparticles for drug deliv-
ery: Boundaries and opportunities. Chem. Eng. Sci., 2015, 125, 
158-164. [http://dx.doi.org/10.1016/j.ces.2014.06.042] 
[PMID: 25684780] 
[62] Karpel, R.L.; da Silva Liberato, M.; Campeiro, J.D.; Bergeon, L.; 
Szychowski, B.; Butler, A.; Marino, G.; Cusic, J.F.; de Oliveira, 
L.C.G.; Oliveira, E.B.; de Farias, M.A.; Portugal, R.V.; Alves, 
W.A.; Daniel, M.C.; Hayashi, M.A.F. Design and characterization 
of crotamine-functionalized gold nanoparticles. Colloids Surf. B 
Biointerfaces, 2018, 163, 1-8. 
[http://dx.doi.org/10.1016/j.colsurfb.2017.12.013] 
[PMID: 29268209] 
[63] Parveen, S.; Misra, R.; Sahoo, S.K. Nanoparticles: A boon to drug 
delivery, therapeutics, diagnostics and imaging. Nanomedicine 
(Lond.), 2012, 8(2), 147-166. 
[http://dx.doi.org/10.1016/j.nano.2011.05.016] [PMID: 21703993] 
[64] Banik, B.L.; Fattahi, P.; Brown, J.L. Polymeric nanoparticles: The 
future of nanomedicine. Wiley Interdiscip. Rev. Nanomed. Nanobi-
otechnol., 2016, 8(2), 271-299. 
[http://dx.doi.org/10.1002/wnan.1364] [PMID: 26314803] 
[65] El-Say, K.M.; El-Sawy, H.S. Polymeric nanoparticles: Promising 
platform for drug delivery. Int. J. Pharm., 2017, 528(1-2), 675-691. 
[http://dx.doi.org/10.1016/j.ijpharm.2017.06.052] 
[PMID: 28629982] 
[66] Mir, M.; Ahmed, N.; Rehman, A.U. Recent applications of PLGA 
based nanostructures in drug delivery. Colloids Surf. B Biointerfac-
es, 2017, 159, 217-231. 
[http://dx.doi.org/10.1016/j.colsurfb.2017.07.038] 
[PMID: 28797972] 
[67] Mohammadpourdounighi, N.; Behfar, A.; Ezabadi, A.; Zolfaghari-
an, H.; Heydari, M. Preparation of chitosan nanoparticles contain-
ing Naja naja oxiana snake venom. Nanomedicine (Lond.), 2010, 
6(1), 137-143. [http://dx.doi.org/10.1016/j.nano.2009.06.002] 
[PMID: 19616121] 
[68] Nobre Almeida, L. Solid lipid nanoparticles: The efficiency carrier 
for topical delivery of hydrophylic drugs, World J. Pharm. Phar-
maceut. Sci., 2017, 6(4), 175-189. 
[69] Beloqui, A.; Solinís, M.A.; Rodríguez-Gascón, A.; Almeida, A.J.; 
Préat, V. Nanostructured lipid carriers: Promising drug delivery 
systems for future clinics. Nanomedicine (Lond.), 2016, 12(1), 143-
161. [http://dx.doi.org/10.1016/j.nano.2015.09.004] 
[PMID: 26410277] 
[70] Patil, Y.P.; Jadhav, S. Novel methods for liposome preparation. 
Chem. Phys. Lipids, 2014, 177, 8-18. 
[http://dx.doi.org/10.1016/j.chemphyslip.2013.10.011] 
[PMID: 24220497] 
[71] Li, X.; Sui, Z.; Li, X.; Xu, W.; Guo, Q.; Sun, J.; Jing, F. Perfluo-
rooctylbromide nanoparticles for ultrasound imaging and drug de-
livery. Int. J. Nanomed., 2018, 13, 3053-3067. 
[http://dx.doi.org/10.2147/IJN.S164905] [PMID: 29872293] 
[72] Jacob, S.; Nair, A.B. Cyclodextrin complexes: Perspective from 
drug delivery and formulation. Drug Dev. Res., 2018, 79(5), 201-
217. [http://dx.doi.org/10.1002/ddr.21452] [PMID: 30188584] 
[73] Pugazhendhi, A.; Edison, T.N.J.I.; Karuppusamy, I.; Kathirvel, B. 
Inorganic nanoparticles: A potential cancer therapy for humanwel-
fare. Int. J. Pharm., 2018, 539(1-2), 104-111. 
[http://dx.doi.org/10.1016/j.ijpharm.2018.01.034] 
[PMID: 29366941] 
[74] Nafisi, S.; Samadi, N.; Houshiar, M.; Maibach, H.I. Mesoporous 
silica nanoparticles for enhanced lidocaine skin delivery. Int. J. 
Pharm., 2018, 550(1-2), 325-332. 
[http://dx.doi.org/10.1016/j.ijpharm.2018.08.004] 
[PMID: 30081227] 
[75] Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications 
and toxicities. Arab. J. Chem., 2017, 12(7), 908-931. 
[http://dx.doi.org/10.1016/j.arabjc.2017.05.011] 
[76] Kumar, B.; Jajodia, K.; Kumar, P.; Gautam, H. Recent advances in 
nanoparticle-mediated drug delivery. J. Drug Deliv. Sci. Technol., 
2017, 41. 
[77] Lin, P.C.; Lin, S.; Wang, P.C.; Sridhar, R. Techniques for physico-
chemical characterization of nanomaterials. Biotechnol. Adv., 2014, 
32(4), 711-726. 
[http://dx.doi.org/10.1016/j.biotechadv.2013.11.006] 
[PMID: 24252561] 
[78] Ding, C.; Li, Z. A review of drug release mechanisms from nanoca 
rier systems. Mater. Sci. Eng. C, 2017, 76, 1440-1453. 
[http://dx.doi.org/10.1016/j.msec.2017.03.130] [PMID: 28482511] 
[79] Villegas, M.R.; Baeza, A.; Vallet-Regí, M. Nanotechnological 
strategies for protein delivery. Molecules, 2018, 23(5), E1008. 
[http://dx.doi.org/10.3390/molecules23051008] [PMID: 29693640] 
[80] Mousavizadeh, A.; Jabbari, A.; Akrami, M.; Bardania, H. Cell 
targeting peptides as smart ligands for targeting of therapeutic or 
diagnostic agents: A systematic review. Colloids Surf. B Biointer-
faces, 2017, 158, 507-517. 
[http://dx.doi.org/10.1016/j.colsurfb.2017.07.012] 
[PMID: 28738290] 
[81] Garcia-Fuentes, M.; Gonzalez-Díaz, H.; Csaba, N. Nanocarriers & 
drug delivery: Rational design and applications. Curr. Top. Med. 
Chem., 2014, 14(9), 1095-1096. 
[http://dx.doi.org/10.2174/1568026614666140329223843] 
[PMID: 24678712] 
[82] Mishra, R.K.; Tiwari, S.K.; Mohapatra, S.; Thomas, S. Efficient 
nanocarriers for drug-delivery systems: Types and fabrication. 
Nano carriers for drug delivery; Mohapatra, S.S.; Ranjan, S.; Das-
gupta, N.; Mishra, R.K.; Thomas, S., Eds.; Elsevier, 2019, pp. 1-41. 
[http://dx.doi.org/10.1016/B978-0-12-814033-8.00001-1] 
[83] Rani, S.; Sharma, A.K.; Khan, I.; Gothwal, A.; Chaudhary, S.; 
Gupta, U. Polymeric nanoparticles in targeting and delivery of 
drugs. Nanotechnology-Based Approaches for Targeting and De-
livery of Drugs and Genes; Mishra, V.; Kesharwani, P.; Mohd 
Amin, M.C.I.; Iyer, A., Eds.; Academic Press, 2017, pp. 223-255. 
[http://dx.doi.org/10.1016/B978-0-12-809717-5.00008-7] 
[84] Wang, M.; Zhang, Y.; Feng, J.; Gu, T.; Dong, Q.; Yang, X.; Sun, 
Y.; Wu, Y.; Chen, Y.; Kong, W. Preparation, characterization, and 
in vitro and in vivo investigation of chitosan-coated poly (d,l-
lactide-co-glycolide) nanoparticles for intestinal delivery of exen-
din-4. Int. J. Nanomedicine, 2013, 8, 1141-1154. 
[PMID: 23658482] 
[85] Chen, C.; Zhu, X.; Dou, Y.; Xu, J.; Zhang, J.; Fan, T.; Du, J.; Liu, 
K.; Deng, Y.; Zhao, L.; Huang, Y. Exendin-4 loaded nanoparticles 
with a lipid shell and aqueous core containing micelles for en-
hanced intestinal absorption. J. Biomed. Nanotechnol., 2015, 11(5), 
865-876. [http://dx.doi.org/10.1166/jbn.2015.1971] 
[PMID: 26349398] 
[86] Jun, H-S.; Bae, G.; Ko, Y.T.; Oh, Y.S. Cytotoxicity and biological 
efficacy of exendin-4-encapsulated solid lipid nanoparticles in INS-
1 Cells. J. Nanomater., 2015, 2015, 6. 
[http://dx.doi.org/10.1155/2015/753569] 
[87] Zhang, X.; Liu, L.; Chai, G.; Zhang, X.; Li, F. Brain pharmacoki-
netics of neurotoxin-loaded PLA nanoparticles modified with chi-
tosan after intranasal administration in awake rats. Drug Dev. Ind. 
Pharm., 2013, 39(11), 1618-1624. 
[http://dx.doi.org/10.3109/03639045.2012.727828] 
[PMID: 24087853] 
[88] Hood, J.L.; Jallouk, A.P.; Campbell, N.; Ratner, L.; Wickline, S.A. 
Cytolytic nanoparticles attenuate HIV-1 infectivity. Antivir. Ther. 
(Lond.), 2013, 18(1), 95-103. [http://dx.doi.org/10.3851/IMP2346] 
[PMID: 22954649] 
[89] Jallouk, A.P.; Moley, K.H.; Omurtag, K.; Hu, G.; Lanza, G.M.; 
Wickline, S.A.; Hood, J.L. Nanoparticle incorporation of melittin 
reduces sperm and vaginal epithelium cytotoxicity. PLoS One, 
2014, 9(4), e95411. 
[http://dx.doi.org/10.1371/journal.pone.0095411] 
[PMID: 24748389] 
[90] Soman, N.R.; Baldwin, S.L.; Hu, G.; Marsh, J.N.; Lanza, G.M.; 
Heuser, J.E.; Arbeit, J.M.; Wickline, S.A.; Schlesinger, P.H. Mo-
lecularly targeted nanocarriers deliver the cytolytic peptide melittin 
specifically to tumor cells in mice, reducing tumor growth. J. Clin. 
Invest., 2009, 119(9), 2830-2842. 
[http://dx.doi.org/10.1172/JCI38842] [PMID: 19726870] 
[91] Mao, J.; Liu, S.; Ai, M.; Wang, Z.; Wang, D.; Li, X.; Hu, K.; Gao, 
X.; Yang, Y. A novel melittin nano-liposome exerted excellent an-
ti-hepatocellular carcinoma efficacy with better biological safety. J. 
Hematol. Oncol., 2017, 10(1), 71. 
[http://dx.doi.org/10.1186/s13045-017-0442-y] [PMID: 28320480] 
[92] Li, Y.; Xu, N.; Zhu, W.; Wang, L.; Liu, B.; Zhang, J.; Xie, Z.; Liu, 
W. Nanoscale melittin@zeolitic imidazolate frameworks for en-
hanced anticancer activity and mechanism analysis. ACS Appl. Ma-
ter. Interfaces, 2018, 10(27), 22974-22984. 
[https://doi.org/10.1021/acsami.8b06125]. 
[93] Yang, L.; Cui, F.; Shi, K.; Cun, D.; Wang, R. Design of high pay-
load PLGA nanoparticles containing melittin/sodium dodecyl sul-
Nanoparticles Functionalized with Venom-Derived Peptides Current Pharmaceutical Biotechnology, 2020, Vol. 21, No. 2 109 
fate complex by the hydrophobic ion-pairing technique. Drug Dev. 
Ind. Pharm., 2009, 35(8), 959-968. 
[http://dx.doi.org/10.1080/03639040902718039] 
[PMID: 19274512] 
[94] Bhowmik, T.; Saha, P.P.; Sarkar, A.; Gomes, A. Evaluation of 
cytotoxicity of a purified venom protein from Naja kaouthia 
(NKCT1) using gold nanoparticles for targeted delivery to cancer 
cell. Chem. Biol. Interact., 2017, 261, 35-49. 
[http://dx.doi.org/10.1016/j.cbi.2016.11.007] [PMID: 27836789] 
[95] Bhowmik, T.; Saha, P.P.; Dasgupta, A.; Gomes, A. Antileukemic 
potential of PEGylated gold nanoparticle conjugated with protein 
toxin (NKCT1) isolated from Indian cobra (Naja kaouthia) venom. 
Cancer Nanotechnol., 2013, 4(1-3), 39-55. 
[http://dx.doi.org/10.1007/s12645-013-0036-5] [PMID: 26069500] 
[96] Moradhaseli, S.; Zare Mirakabadi, A.; Sarzaeem, A.; Kamalzadeh, 
M.; Haji Hosseini, R. Cytotoxicity of ICD-85 NPs on human cervi-
cal carcinoma hela cells through caspase-8 mediated pathway. Iran. 
J. Pharm. Res., 2013, 12(1), 155-163. [PMID: 24250584] 
[97] Zare Mirakabadi, A.; Moradhaseli, S. Comparative cytotoxic eval-
uation of free and sodium alginate nanoparticle-encapsulated ICD-
85 on primary lamb kidney cells. Iran. J. Cancer Prev., 2013, 6(3), 
151-159. [PMID: 25250126] 
[98] de Barros, N.B.; Macedo, S.R.A.; Ferreira, A.S.; Tagliari, M.P.; 
Zanchi, F.B.; Kayano, A.M.; Soares, A.M.; Nicolete, R. Liposomes 
containing an ASP49-phospholipase A2 from Bothrops jararacussu 
snake venom as experimental therapy against cutaneous leishmani-
asis. Int. Immunopharmacol., 2016, 36, 225-231. 
[http://dx.doi.org/10.1016/j.intimp.2016.04.025] 
[PMID: 27174621] 
[99] Bond, A. Exenatide (Byetta) as a novel treatment option for type 2 
diabetes mellitus. Proc. Bayl. Univ. Med. Cent., 2006, 19(3), 281-
284. [http://dx.doi.org/10.1080/08998280.2006.11928181] 
[PMID: 17252050] 
[100] Pencek, R.; Blickensderfer, A.; Li, Y.; Brunell, S.C.; Chen, S. 
Exenatide once weekly for the treatment of type 2 diabetes: Effec-
tiveness and tolerability in patient subpopulations. Int. J. Clin. 
Pract., 2012, 66(11), 1021-1032. [http://dx.doi.org/10.1111/j.1742-
1241.2012.03006.x] [PMID: 22925173] 
[101] Shi, Y.; Sun, X.; Zhang, L.; Sun, K.; Li, K.; Li, Y.; Zhang, Q. Fc-
modified exenatide-loaded nanoparticles for oral delivery to im-
prove hypoglycemic effects in mice. Sci. Rep., 2018, 8(1), 726. 
[http://dx.doi.org/10.1038/s41598-018-19170-y] 
[PMID: 29335533]

Continue navegando

Outros materiais