You are on page 1of 16

Expert Opinion on Drug Delivery

ISSN: 1742-5247 (Print) 1744-7593 (Online) Journal homepage: https://www.tandfonline.com/loi/iedd20

Formulation strategies to modulate drug release


from poloxamer based in situ gelling systems

Hani Abdeltawab, Darren Svirskis & Manisha Sharma

To cite this article: Hani Abdeltawab, Darren Svirskis & Manisha Sharma (2020): Formulation
strategies to modulate drug release from poloxamer based in situ gelling systems, Expert Opinion
on Drug Delivery, DOI: 10.1080/17425247.2020.1731469

To link to this article: https://doi.org/10.1080/17425247.2020.1731469

Accepted author version posted online: 18


Feb 2020.
Published online: 23 Feb 2020.

Submit your article to this journal

Article views: 9

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iedd20
EXPERT OPINION ON DRUG DELIVERY
https://doi.org/10.1080/17425247.2020.1731469

REVIEW

Formulation strategies to modulate drug release from poloxamer based in situ


gelling systems
Hani Abdeltawab, Darren Svirskis and Manisha Sharma
School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland, New Zealand

ABSTRACT ARTICLE HISTORY


Introduction: Poloxamer based in situ gelling systems offer numerous advantages in drug delivery; Received 29 October 2019
however, their application as prolonged-release delivery platforms is limited mainly due to their weak Accepted 14 February 2020
mechanical properties and the interconnected aqueous network causing fast gel erosion and drug KEYWORDS
diffusion. Poloxamers; in situ gelling
Area covered: The focus of this review is to provide an insightful discussion on the formulation systems; thermo-responsive;
strategies that can be employed to sustain/prolong the drug release from poloxamer based in situ sustained drug delivery;
gelling systems. The review also outlines the formulation factors, influencing drug release from these extended release;
systems. modulating release profile;
Expert opinion: The nature, composition, and concentration of poloxamers are the most critical factors formulation strategies
in defining the rate of drug release from an in situ gelling matrix. Hydrophobic gel matrices have
compact micellar arrangements resulting in slow diffusion and erosion. Depending on the intended
clinical application, gel characteristics can be modulated, either by physical blending or by chemical
crosslinking with additive materials, to slow release and improve residence time at the administration
site. Incorporating drug-loaded particles into poloxamer gels sustains drug release by creating multiple
rate-limiting release barriers. Chemical modification of poloxamers appears to be a promising strategy
to obtain prolonged sustained release for parenteral application without compromising the rheological
properties of the formulation.

1. Introduction temperature [10]. Poloxamers are more soluble at cold tem-


peratures due to the formation of stable hydrogen bonds
Poloxamers are water-soluble tri-block copolymers with amphi-
between the PEO and PPO blocks with water molecules. At
philic and surface-active properties. Poloxamers (Figure 1) are
a critical micelle temperature, the hydrogen bonds destabilize
nonionic polymers, consisting of a central hydrophobic block of
and poloxamers aggregate and self-assemble to form spheri-
poly (propylene oxide) (PPO) and two hydrophilic terminal blocks
cal micelle with hydrophobic propylene oxide (PO) core [1].
of poly (ethylene oxide) (PEO). They have been employed as
The size of the micelles grows until the PO core becomes
dispersants for pigments/inks, detergents, foaming agents and
completely dehydrated. The outer hydrophilic chain of the
as anti-biofouling coatings [1]. Due to their biocompatibility
micelles swells and interact to form three-dimensional gel
poloxamers have been extensively used as excipients in pharma-
structures (Figure 2).
ceuticals such as, dispersing agent, emulsifiers, co-emulsifying
To date, the use of poloxamers in drug delivery is chal-
agents, solubilizing agent, tablet lubricant, and wetting agent
lenged by their limited ability to sustain drug release. Many
[2,3]. Over the last two decades, poloxamers based delivery plat-
poloxamer-based gels described, rapidly lose their integrity in
forms have grabbed the attention of formulation scientists and
an aqueous environment, causing fast release of loaded drug
have been widely employed as drug carriers in ophthalmic [4],
[11–13]. In addition, hydrophilic low molecular weight thera-
vaginal [5] and rectal drug delivery [6] due to the ease of admin-
peutics can rapidly diffuse through the interconnected porous
istration and the ability to provide controlled release of drug
network structure of the gels causing an initial burst release
following gelation [7,8].
and fast drug diffusion [14]. Furthermore, their relatively poor
Poloxamer-based delivery systems are attractive, as they
mucoadhesive properties result in short residence time and
can deliver both hydrophilic and hydrophobic drugs and exhi-
rapid clearance from the application site [6]. This raises con-
bit thermo-responsive sol-to-gel transition behavior at concen-
cerns around the safety (initial burst release) and efficacy
trations above a critical value. They can be formulated to be
(fluctuation in drug-plasma concentration) of the poloxamer
liquid at room temperature, which allows for ease of adminis-
based in situ gel delivery systems [15]. As a result, controlled
tration, with gelation occurring as temperature rises, such as
drug delivery for prolonged or extended duration is not yet
at the body temperature, forming a semisolid gel depot. The
achievable through these poloxamer-based systems alone.
polarity of PEO – PPO blocks changes with increase in the
Increasingly, research is focussing on tailoring the composition

CONTACT Manisha Sharma manisha.sharma@auckland.ac.nz School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private
Bag, Auckland 92019, New Zealand
© 2020 Informa UK Limited, trading as Taylor & Francis Group
2 H. ABDELTAWAB ET AL.

a comprehensive discussion on the release mechanism gov-


Article highlights erning each of these formulation approaches along with its
● Poloxamers are attractive thermo-responsive in situ gelling systems
limitations. The authors also suggest a potential approach for
for drug delivery; they are liquids at room temperature, facilitating future investigation, which is hypothesized to reduce the drug
their administration followed by a transformation to a gel state at initial burst release.
physiological temperature to provide sustained release.
● The use of poloxamers as sustained release drug delivery systems is
mainly limited by their rapid erosion and fast drug diffusion.
● Various approaches have been employed for sustaining drug release 2. Mechanisms of drug release
from poloxamers based in situ gelling systems such as, physical
blending, chemical crosslinking with additives, incorporation of drug- Understanding the mechanisms controlling drug release from
loaded particulates and chemical modification of poloxamers. poloxamers based in situ gel systems is an essential step
● The choice of suitable approach to achieve sustained release depends
on the desired release profile and duration of action, and route of
toward modulating their release profile. Drug release from
administration. poloxamer based in situ gels is controlled by both diffusion
and gel erosion [1] as presented in Figure 3. However, the
This box summarizes key points contained in the article.
rate-determining step for release varies according to the com-
position of the gel matrix and the physicochemical properties
of the loaded drug [14].

of the poloxamer-based formulations to modulate the release


2.1. Drug diffusion
profile of the drug-loaded moieties.
Formulation scientists have made various attempts to Drug diffusion from poloxamer-based gels plays a dominant
achieve controlled, extended release (beyond few days to role especially for small hydrophilic molecules due to the
weeks) from these systems by chemical modification or by presence of a continuous aqueous channel within the gel
developing a composite system [16,17]. It is expected that microstructure [26]. The physicochemical properties of the
developing such modified composite systems will enhance loaded drug such as its molecular weight, hydrophilicity/lipo-
patient adherence to treatment in the future. philicity balance play a significant role in controlling the diffu-
Several reviews have been published in the past, focussing sion process [26]. Gilbert et al. demonstrated that increasing
on the use of poloxamers for delivering [1,18] ocular drugs [4], the length of the alkyl chain on the benzoate ester (methyl,
oral chemotherapeutics [19], drug-resistant respiratory che- ethyl, propyl, and butyl) results in reduction in drug release
motherapeutics [20], gene delivery [21,22] and for multi-drug rate [27]. This is thought to be due to the increase in the
resistance reversal [23–25]. To the best of our knowledge, this lipophilicity of the drug, with longer alkyl chains, resulting in
is the first review, which focusses on various formulation increased affinity toward the lipophilic micellar cores and thus
strategies employed to alter the drug release from poloxamer slow diffusion from the in situ gel matrix. On the other hand,
based in situ gelling systems. This manuscript aims to provide small hydrophilic drugs tend to exist in the aqueous channels

Figure 1. Chemical structure of poloxamers showing its amphiphilic nature. x and y are the average number of repeating units of each blockchain (for poloxamer
188 x is 76 and y is 29, while for poloxamer 407 x is 100, while y is 65) [9].

Figure 2. Sol-to-gel transition of poloxamer as a function of temperature. CMC: critical micelle concentration.
EXPERT OPINION ON DRUG DELIVERY 3

Figure 3. Mechanisms of drug release from poloxamer based in situ gelling systems. Yellow dots represent drug molecules.

and thus diffuse rapidly to the surrounding environment enhancement in gel strength and reduction in erosion rate [34].
[26,28]. Likewise, the gel matrix supramolecular arrangement, The gel erosion might also contribute to the release of the small
micellar volume, micellar entanglement, and the extent of hydrophilic organic drugs as demonstrated by Yang L et al. [26].
aqueous channel network affect the drug diffusion from The author reported that the release of caffeine from poloxamer-
poloxamer based in situ gelling systems [27,29–31]. based gel was governed by both diffusion and gel erosion. This
can be explained by the partitioning of the drug between the
aqueous regions and poloxamer micellar core due to its organic
2.2. Gel erosion component. On the other hand, Moore et al. concluded that the
The micellar packing arrangement of poloxamers rapidly dissoci- lipophilic/hydrophilic balance of a drug and molecular weight
ates in the presence of excess aqueous media leading to the have no significant effect on the release mechanism [37]. They
degradation of the gel matrix. The erosion process of poloxamer- studied the release of three drugs; propranolol HCl (hydrophilic),
based gels is not fully described in the literature. However, it is metronidazole (hydrophobic) and cephalexin (moderately
proposed that micelles at the surface dissolute into the release hydrophobic) from poloxamers and demonstrated that drug
media, leading to gel erosion and drug release [32,33]. Further release was a function of gel erosion irrespective of the varying
studies may be required to assess the contribution of bulk ero- physiochemical properties of the studied drugs. These contra-
sion as a result of water influx to the matrix core. Gel erosion is dictory findings might be attributed to the experimental design,
the major release mechanism under two situations. First, when as they used large dissolution medium (20 g gel in 800 ml
the loaded drug molecule has a high molecular weight and/or is deionized water, at stirring rates of 20–80 rpm). Poloxamer gels
hydrophobic, and second when the mechanical strength of the being aqueous in nature tends to degrade faster in larger
formed in situ gel is poor/weak, causing release of the loaded volumes of dissolution medium as compared to small volumes.
drug with the degradation of the gel matrix [14,34]. Indeed, these findings signify the importance of experimental
Zhang et al. demonstrated that the release of polyethylene design and its impact on the outcomes of the study.
glycol (40 kDa) from poloxamer based in situ gels following When comparing literature, it becomes obvious that there
either intramuscular or subcutaneous injection is erosion con- is considerable variations in both experimental design and
trolled [14]. Low mobility of the large molecules through the methodology used, which makes it challenging to draw any
internal aqueous channels to the external environment can conclusive outcomes from the studies. Yang et al. and Moore
explain these results. Similarly, hydrophobic drugs have higher et al. studied drug release from poloxamers using large
affinity toward the core of poloxamers micelles, and it is volumes of dissolution media and reported gel erosion as
expected that their release is mainly erosion controlled. The the dominant mechanism of release [26,37]. Whereas Inal
release of meloxicam and nonoxynol-9, two hydrophobic and Yapar studied the drug release in small volumes and
drugs, was reported to be erosion controlled, and followed zero- concluded that release is governed through diffusion [35].
order release kinetics [35,36]. Likewise, the release of acyclovir, Similarly, the type/polarity of release medium can also influ-
a practical water-insoluble antiviral drug, from poloxamer 407 ence the release mechanism. In general, release studies are
was mainly controlled by gel erosion. However, the addition of performed in aqueous medium; however, some studies have
carrageenan into poloxamers modified the release pattern to be reported the use of non-aqueous release medium such as
diffusion controlled, which is attributed to the significant isopropyl myristate [38,39]. The solubility of poloxamers in
4 H. ABDELTAWAB ET AL.

isopropyl myristate is very limited and therefore drug release hyaluronic acid (HA) into poloxamer 407 (P407) increased the
is mainly through diffusion. average micellar diameter from 15 to 400 nm, and a subse-
Another challenge in describing the drug release mechan- quent reduction in piroxicam release rate was observed [40].
ism from poloxamer-based gelling systems is the selection of Likewise, the chemical modification of poloxamers by synthe-
appropriate mathematical model. Though many studies have sizing disulfide multi-blocks increased the average micellar
reported that drug release from poloxamers follows Fickian diameter from 12 to 600 nm, with a significant extension in
diffusion and can be well described by Higuchi model [28,31], time required for complete gel erosion from 6 h to 12 days
others demonstrated that release strictly follows zero-order [41]. The increase in total poloxamer concentration also
kinetics [36,37]. Few attempts have been made to develop reduced the drug diffusion and sustained the release profile
a predictive mathematical model, describing the drug release [27,30,42].
from poloxamers based gels [26,37]. They have used large
volumes of dissolution media (300- and 800-ml deionized
3.2. Mechanical and rheological properties
water) and are therefore more appropriate to understand
drug release following oral administration of poloxamers Mechanical and rheological properties are crucial formulation
based delivery systems, where the gel encounter large attributes for sustaining the release from poloxamer based
volumes of gastric fluids. Future studies are required to take in situ gels [35]. Enhancement of these properties can lead
into consideration other clinical applications of poloxamers to a significant reduction in gel erosion and drug diffusion
based in situ gels. rates, thus sustaining the release profile [43,44]. In general,
To summarize, drug release from poloxamer based in situ poloxamer based in situ gels are highly porous structures with
gelling systems is controlled by both diffusion and gel erosion. interpenetrating aqueous channels contributing to fast ero-
The physicochemical properties of both the loaded drug and sion of the gel matrix. Creating a tightly packed micellar
the matrix, along with the experimental conditions of the structure while initiating increased intermolecular interactions,
release study determine the dominant mechanism of drug will enhance the overall strength of the gel matrix and will
release. Due to the wide applications of poloxamers, it is alter the rheological properties.
difficult to generalize a mathematical model that accurately Increasing the PPO/PEO ratio can significantly promote gel
predicts the drug release from poloxamer-based gels. Instead, strength, reduce the erosion rate, and slowdown drug release,
clinical application should be considered, when designing in- and vice versa [35,45]. Zhang et al. showed an inverse relation-
vitro release experiments to simulate the in-vivo condition as ship between the concentration of the relatively hydrophobic
closely as possible, for the reliability of the results. P407 and the gel erosion rate; P407 (25% w/v) was completely
eroded over 6 h compared to 70% and 40% erosion for 30%
w/v and 35% w/v P407 concentration, respectively [43]. This is
3. Factors influencing drug release from poloxamer explained by the increased hydrophobic interaction, causing
based in situ gelling systems reduction in the aqueous channels and enhancing the gel
mechanical properties. On the other hand, Liu et al. observed
As discussed, the release of drug from poloxamer based in situ
that increasing the ratio of the poloxamer 188 (P188) (1% to
gelling systems is through both drug diffusion and gel erosion.
6%) in the blend of P407-P188 enhanced the gel erosion rate
Thereby, it is important to understand the formulation factors
[36]. The hydrophilic PEO tends to disrupt the hydration cor-
controlling both gel erosion and drug diffusion.
ona around the hydrophobic PPO core; consequently, a higher
Comprehensive understanding of these factors will guide us
number of water molecules exist around PPO. The high ratio
toward the development of poloxamer-based systems with
of water molecules reduces the gel strength and facilitates its
the ability to provide extended or prolonged drug release.
erosion [35].
Blending/crosslinking of poloxamers with additive materials
can significantly enhance the mechanical and rheological prop-
3.1. Gel microstructures
erties of poloxamer based in situ gelling systems, thus reducing
The gel microstructure consists of a number of entangled the gel erosion and drug release rates [46–48]. This can be
micelles forming cubic/hexagonal 3D gel structure with interpe- attributed to the formation of intermolecular bonds between
netrating aqueous channels. The number, size, and arrangement the additive material and poloxamers, creating a tightly packed
of micelles critically control the porosity of the three-dimensional system. Of note, the enhancement of mechanical and rheological
gel structure, which in turn influence the diffusion of drugs to the properties is associated with the concentration and molecular
surrounding environment [27,29–31]. weight of the additive material [47,49].
An increase in the micellar diameter and/or number of Though enhancing rheological properties is usually
micelles per unit volume reduces inter-micellar distance and a successful approach to sustain the release profile from
promotes a high degree of micellar entanglement. This will poloxamer based in situ gelling systems, these properties
result in greater tortuosity in the aqueous channels and reduc- must be critically considered in the development of in situ
tion in the aqueous phase, which will ultimately slowdown the gels for parenteral and ophthalmic application as they affect
rate of drug diffusion [14,29]. Various formulation factors, such the syringe-ability, injectability, and spread-ability of these
as total poloxamer concentration, blending with additive systems. Formulations with viscosities higher than 300 mPas
material and chemical modification of poloxamers, affect the are difficult to inject intramuscularly [50]. Xuan J et al. demon-
gel microstructure and network porosity. Blending of strated that increasing the poloxamer concentration and/or
EXPERT OPINION ON DRUG DELIVERY 5

blending with additives can significantly retard the syringe- in the eye as a function of its concentration; the cumulative
ability due to the increased viscosity. Likewise, Lin et al. release over 6 h was reduced from 50% to 20% by the addition
observed that blending of alginates (≥0.7%) into P407 (14%) of 1% HA and to 10% by the incorporation of 2% HA [48]. In
renders the formulation unsuitable for ophthalmic administra- addition, an increase in mucoadhesive forces and residence time
tion due to the high viscosity and poor spread-ability [51]. Of has been reported. Similarly, the incorporation of HA to P407
note, the absence of standard methods for the determination reduced the cumulative piroxicam released over 50 h from 50%
of these properties is a challenge toward comparing the to only 20% [40]. The HA-P407 system sustained the in-vitro
results, and for setting acceptance criteria of each property. release up to 10 days and the half-life (t1/2) following intra-
articular injection increased from 24 h (poloxamer without HA)
to 86 h (poloxamer with HA). Likewise, Nascimento et al. demon-
4. Strategies for sustaining/prolonging drug release strated that the blending HA into poloxamers (P407 or P407-
from poloxamer based in situ gelling systems P108) reduced the release of lidocaine compared to HA free
Despite the advantages of poloxamer based in situ gelling formulation [29].
systems in drug delivery, their employment as sustained/pro-
longed release delivery platforms is limited by their rapid
4.1.2. Chitosan
erosion and drug diffusion as well as short residence time at
Chitosan are biocompatible and biodegradable natural polymers
the site of administration [48,51–53]. These challenges are
[66]. They form pH-responsive hydrogels; exhibit sol-to-gel tran-
explained by the poor mechanical strength, highly porous
sition as a response to pH change [66]. Chitosan blending with
gel network structure and the poor mucoadhesive properties.
poloxamer-based gel reduces the drug release rate by enhancing
Therefore, different formulation strategies have been
the gel strength via formation of inter-micellar bridges [28,67]. In
employed to overcome these limitations and prolong drug
addition, it prolongs the residence time at the administration site
release from poloxamer based in situ gelling systems as dis-
and thus is advantageous in mucosal applications [67,68]. These
cussed in the following sections.
changes are associated with chitosan concentration [69–71] and
molecular weight [72].
4.1. Physical blending with other additive(s) Sridhar et al. demonstrated that chitosan has increased the
intra-nasal residence time of a poloxamer gel by 3.2 fold [67]. In
Physical blending of poloxamers with another polymer has addition, the release profile extended from 6 to 16 h. Likewise,
been widely investigated to alter drug release from poloxamer the incorporation of chitosan into poloxamers promoted the
based in situ gel systems (Table 1). Interaction of polymer intra-nasal residence time and prolonged the in-vitro release of
additives with poloxamers alters the gel microstructure, sig- metoclopramide hydrochloride to more than 6.5 h compared to
nificantly affecting gel porosity, mechanical and rheological only 2.5 h with poloxamer formulation [73]. Qian et al. demon-
properties. These interactions occur as a result of hydrophobic strated that chitosan (0.5%) slightly reduced the in-vitro release
interactions, hydrogen bonding and/or due to supra- rate of tacrine, compared to poloxamer binary system (P407-
molecular interactions (association of polymers by intermole- P188) [74]. Chitosan blending with P407 prolonged the in-vitro
cular forces) [54]. These physicochemical changes modulate release of ketorolac tromethamine from 1 to 12 h. An in-vivo
the gel erosion and drug diffusion rates, thus altering the drug study in rabbits following intravesical administration demon-
release characteristics of the in situ gels. Various additives such strated increased residence time [68]. In ophthalmic applications,
as chitosan [55], HA [48], alginates [47], cellulose derivatives chitosan reduced ofloxacin release rate; 60% drug was released
[46], carrageenan [34,56–58], heparin [59,60], linoleic acid [61] over 3 h from the composite preparation compared to more than
and gellan gum [62] have been widely used by researchers to 90% release within 1 h from poloxamer [69]. In addition, chitosan
modulate the release profile from poloxamer based in situ increased the ocular residence time of poloxamers by 4 fold as
gelling systems. Similarly, the incorporation of salts such as demonstrated by Gratieri T et al. [55]. This significant increase in
sodium chloride into poloxamer gels has demonstrated residence time is explained by the enhanced mucoadhesive
a significant promotion in gel strength and subsequent exten- forces, which increased from 0.08 N to 0.1 N by the addition of
sion in release profile [63]. In this section, the effects of blend- chitosan.
ing additives with poloxamers are discussed. Chitosan have been used in combination with other additives
to retard the release rate from poloxamer gelling systems. Ur-
4.1.1. Hyaluronic acid Rehman et al. demonstrated that incorporation of chitosan and
Hyaluronic acid (HA) is a biocompatible and biodegradable nat- sodium tri-polyphosphate reduced the release rate of different
ural polysaccharide [64]. It has been used in several applications drugs (namely metoprolol, doxycycline, and flufenamic acid)
especially joint lubrication [65] and ocular drug delivery [64]. The from P407 as a function of chitosan concentration [70]. Likewise,
blending of HA into poloxamer solutions increases gel strength incorporation of chitosan to a blend of P407 (17% w/v)-
and reduces network porosity, due to hydrogen bonding cyclodextrin (10% w/v) reduced the fexofenadine released over
between HA and poloxamers, consequently sustaining drug 6 h from 74% to 54.5% [71]. The reduced in-vitro release rate is
release and reducing the initial burst release of loaded drug explained by the enhanced gel strength and viscosity of the
[48]. Mayol et al. reported that incorporation of HA into composite; the consistency index (shear stress/(shear rate * flow
a poloxamer gel (10% P188–15% P407) reduced acyclovir release behavioral index)) increased from 1539.4 to 1886.8, respectively.
6

Table 1. Physical blending and chemical crosslinking formulation strategies to sustain the drug release from poloxamer based in situ gels.
Poloxamer(s)
H. ABDELTAWAB ET AL.

composition Additives (concentration) Drug Route of Administration Modification/change in drug release profile Ref.
Physical Blending
P407 (15 wt.%)-P188 Hyaluronic acid (2 wt.%) Acyclovir Ophthalmic Reduction of in-vitro cumulative release over 6 hours from 50% to 20% [48]
(10 wt.%)
P407 (20 wt.%) Hyaluronic acid (1 wt.%) Piroxicam Intraarticular Reduction in in-vitro cumulative release over 50 hours from 50% to 20% [40]
P407 (18 wt.%) Chitosan (0.1 wt.%) Selegiline hydrochloride Intra-nasal Sustained release profile from 6 to 16 hours, with increased intranasal residence time [67]
P407 (18 wt.%) Chitosan (0.5 wt.%) Metoclopramide Intranasal Sustained release profile from 2.5 to 6.5 hours [73]
hydrochloride
P407 (20 wt.%)- P188 Chitosan (0.5 wt.%) Tacrine Intranasal Reduction in cumulative release over 90 minutes from 60% to 45% [74]
(1 wt.%)
P407 (17 wt.%) Chitosan (3 wt.%) Ketorolac Intravesical Sustained release profile from 1 to 12 hours, with increased residence time [68]
P407 (15 wt.%) Chitosan (0.33 wt.%) Ofloxacin Ophthalmic Reduction in cumulative release, over 1 hour, from 100% to 40% [69]
P407 (20 wt.%) Methylcellulose (4 wt.%) Etidronate Parenteral (intraplantar) Sustained release profile from 5 days to more than 28 days [46]
P407 (18 wt.%)- P188 Hydroxy ethyl cellulose (0.5 wt.%) Ciprofloxacin Ophthalmic Reduction in the cumulative release, over 8 hours, from 88% to 58% [81]
(13 wt.%)
P407 (20 wt.%) Alginate (0.1 wt.%) Pilocarpine Ophthalmic Sustained release profile from 4 to 6 hours, with significant increase in the AUC [51]
P407 (20 wt.%) Alginate (0.63 wt.%) Selegiline Transdermal Reduction in cumulative amount released over 48 hours, from 44 to 32 μg/cm2 [84]
P407 (18 wt.%) Alginate (0.5 wt.%) Nimesulide Rectal Only formulations containing alginate was retained inside the rabbit’s rectum without leakage, [85]
with reduced release rate.
P407 (15 wt.%)- P188 Sodium chloride (0.8 wt.%) Diclofenac sodium Rectal Reduction in the cumulative release, over 6 hours, from 50% to 25% [88]
(17 wt.%)
P407 (17, 18 and Sodium chloride (1 wt.%) Salbutamol Buccal Sustained release profile from 30 minutes to 1 hour [89]
19 wt.%)
Chemical Crosslinking
P407 (20 wt.%) Hyaluronic acid (1.18%, 6.87% and Ciprofloxacin Ophthalmic Release profile was sustained to 5, 20 and 23 hours, compared to only 3 hours for the P407 [64]
13.99% wt.%) system
P407 (25% w/v) Hyaluronic acid (1% w/v) Plasmid DNA Parenteral Release profile was sustained to more than 20 days, compared to only 3 days for poloxamers [90]
P407(18 wt.%)- P188 Chitosan (1.1 wt.%) 5-fluorouracil Intratumor Release profile was sustained from 5 to 52 hours, with significant reduction in initial burst [91]
(15 wt.%) release
P407 (5% w/v) Carboxy methylcellulose (2.5% w/v) Nepafenac Ophthalmic Release profile was sustained to 80 hours, compared to 24 hours for PBS/drug solution (no data [93]
provided for CMC free poloxamers).
P407 (20% w/v) Alginate * Selegiline Transdermal Cumulative amount released at 48 hours was reduced from 44 to only 14 µg/cm2. [84]
P407* Alginate* Lidocaine Parenteral The crosslinked hydrogel significantly increased the time of post-operative pain control in rats [96]
for 8 hours.
Concentration is not reported.
EXPERT OPINION ON DRUG DELIVERY 7

Chitosan-methyl cellulose-P407 ternary composite significantly

[102]

[108]
[105]

[109]

[104]
Ref.

[17]
[7]
sustained the in-vivo release of the soluble antigen ovalbumin
compared to non-modified poloxamer [75].

Significant reduction in release rate; cumulative release over 48 hours was reduced from 85% to 60%, with

Prolonged the onset time of chemically induced convulsion in rats by 7.5 and the death time by 14 times,
Significant reduction in the release rate over the first 24 hours; composite formulation released only 40%,
4.1.3. Cellulose derivative
Cellulose-based polymers are highly abundant and biocompa-

Release profile was sustained to 14 days, compared to only 3 hours for the microspheres alone.
tible polymers that have diverse applications in pharmaceuti-

Hypoglycemia extended for 3 days, compared to only 10 hours for the hydrogel/insulin group
cals, cosmetics, and nutritional products [76]. Blending of
cellulose derivatives with poloxamer solutions has been
applied to slow drug release by increasing gel strength and
reducing erosion rates [46]. In addition, certain cellulose deri-
vatives, such as hydroxyl propyl methylcellulose (HPMC) and
Modification/change in release profile

hydroxyl ethyl cellulose (HEC), enhance the mucoadhesive


forces, thus increasing the residence time of poloxamer-
based formulations [77]. These changes are strongly governed
Cumulative release over 5 days was reduced from 97% to 77%.

by the type, concentration, and molecular weight of the cellu-


lose derivative being used.
Rangabhatla et al. demonstrated that blending of methylcel-
lulose (MC) with P407 significantly reduced the gel erosion and
Release profile was sustained from 5 to 30 days.

etidronate release rates; release profile was extended from 5 days


increased ocular residence time by 23%.

to 4 weeks [46]. Similarly, Wang W et al. reported that MC


compared to 100% from poloxamers.

blending into P407 reduced the release rate of the Chinese


Table 2. Incorporation of drug-loaded particulates as a formulation strategy to sustain the drug release from poloxamer based in situ gels.

medicine, Cortex Moutan [78]. Dewan et al. demonstrated that


compared to the control group.

grafting of MC into poloxamers increased gel strength and visc-


osity resulting in a reduction in drug release rates as a function of
its molecular weight [49].
Carboxy methylcellulose (CMC) and HPMC blending with P407
decreased the in-vitro release constant of ceftiofur from 0.3 to 0.24
and 0.28 µg/min, respectively [43]. HPMC blending with P188
prolonged the residence time and reduced the icariin released
over 40 h from 70% to 30% [77]. Similarly, Yu et al. demonstrated
that blending sodium carboxy methylcellulose (Na CMC) and
HPMC with P188 significantly retarded the initial burst release
administration

Subcutaneous
Route of

and extended the release profile of florfenicol from 65 to 120


Intraarticular

Diclofenac sodium Intraarticular


Ophthalmic

Intratumor
Intra-nasal

Parenteral

and 128 h, respectively [79]. Hydroxypropyl cellulose (HPC) and


HPMC reduced the release rates of vancomycin from poloxamer-
based gels (61%, 72%, and 80% release within 24 h for HPC-P407
(5%-15%w/w), HPMC-P407 (10%-10% w/w), and P407 (20% w/w)
Coated crystalline

respectively) [80].
Clondronate

Clonazepam
Flurbiprofen
Drug

HEC addition to a binary system of P407 18% and P188 13% w/


Paclitaxel

insulin
Copper

w significantly reduced the ciprofloxacin HCL release over 8 h in-


vitro from 88% to 58% and promoted the mucoadhesive force by
threefold [81]. On the other hand, Marcos et al. demonstrated that
PLGA nanoparticles

14% poloxamer 123 blended with 2% HEC did not significantly


nanoparticles
Particulate

microspheres
Bioactive glass

sustain the release of griseofulvin [82]. However, the addition of


Microcrystals

10% α-cyclodextrin (αCD) to the binary system enhanced the


Liposomes

Liposomes
Liposomes
Chitosan

solubility of griseofulvin, improved rheological and mucoadhesive


properties of the gel by forming a supramolecular network struc-
ture and thus provided sustained release for up to 3 weeks. The
P407 (20 wt.%)-P188 (9 wt.%)-SA or
P407 (18 wt.%)-chitosan (0.25 wt%)

failure of the binary system to sustain the drug release was


justified by the system’s phase separation at physiological tem-
P407 (30 wt.%)-P188 (10 wt.%)

Polyether urethane (chemically


P127 (15 wt.%)-SA (0.75 wt.%)
P407 (20 wt.%)-P188 (5 wt.%)

perature, which led to a highly porous system. The elastic modulus


of the ternary system was approximately 2 and 2.5 times that of
Poloxamer composition

the poloxamer-HEC composite and poloxamers alone,


modified P407)

HPMC (2 wt.%)

respectively.
P407 (18 wt.%)

4.1.4. Alginates
Alginates are anionic mucoadhesive biocompatible polymer
with strong mucoadhesive properties [83]. The physical
8 H. ABDELTAWAB ET AL.

blending of alginate with poloxamer solutions can slow drug that sodium chloride significantly increased the gel strength of the
release from in situ gels by reducing erosion and increasing liquid suppository base (P407 15%-P188 17%) and reduced the
the residence time [51]. Lin et al. reported that the alginates cumulative release over 6 h from 50% to 25% [88]. Likewise,
reduced the release of pilocarpine from poloxamer in situ gels sodium chloride sustained salbutamol sulfate release from P407
in a concentration-dependent manner due to an increase in to 1 h, compared to only 30 min from sodium chloride-free for-
gel strength and residence time following ocular administra- mulation [89]. On the other hand, the addition of sodium chloride
tion [51]. Chen et al. reported that blending alginate into P407 to poloxamer-based gel did not significantly alter the release of
slowed the gel degradation (10% compared to 20% over meloxicam from the binary system (P407-P188) in the initial 3 h,
2 days for P407 system) and reduced the transdermal release though, faster release was observed afterward due to the ‘pore-
rate of Selegiline (cumulative amount released was 32 µg/cm2 forming effect’ discussed above [35].
compared to 44 µg/cm2 from P407) [84]. Yuan et al. demon- To conclude, physical blending of poloxamers is a successful
strated that alginates significantly reduced the in-vitro release approach for sustaining the drug release and improving the
of nimesulide from 18% w/w P407 rectal in situ forming gel mechanical properties of poloxamer based gelling systems. This
and improved its adhesion to the rectal membrane [85]. approach can significantly reduce gel erosion through increasing
the gel strength. In addition, it may hinder drug diffusion rates
because of the reduced network porosity. Furthermore, it might
4.1.5. Salts as additives increase the residence time at the application site due to the
Salts such as sodium chloride and sodium phosphate (mono enhancement of mucoadhesive properties by some additives.
or dihydrogen) crosslink with poloxamers and significantly The enhancement of mucoadhesive properties provides this
enhance the mechanical and mucoadhesive properties of approach suitable for mucosal applications such as ophthalmic,
in situ gels [35,63,86,87]. Studies have shown a 60-fold nasal, rectal, and vaginal applications. The proper choice of the
increase in gel strength and a 10-fold increase in mucoadhe- polymer type, molecular weight and concentration is critical in
sive properties following the addition of salts [63]. The achieving the set goals without affecting formulation inject-
enhancement of mechanical properties is expected to reduce ability, spread-ability, or applicability. Physical blending is
the drug release rate. However, studies have demonstrated a suitable approach to sustain drug release from several hours
that sodium salts tend to show ‘pore-forming effects’ after up to few days. Noteworthy, the duration of the release will vary
a certain time, when exposed to aqueous environment due with respect to the site of administration.
to their high-water affinity. Salts tend to diffuse out of the gel
matrix leaving pores in the matrix, allowing media to enter the
gel matrix and thus increasing the drug diffusion and gel
4.2. Crosslinking with another polymer(s)
erosion rates [35].
Yong et al. demonstrated that sodium chloride increased gel The crosslinking of poloxamers with other polymers (Figure 4)
strength of a P407-P188 liquid suppository by 67-fold. In addition, is another approach for modulating the drug release from
the mucoadhesive forces and retention inside the rabbit’s rectum poloxamer based in situ gels (Table 1). Crosslinking results in
increased proportionally with the sodium chloride concentration the preparation of compact gels with increased strength and
[86]. The authors did not report the release profile; however, the reduced erosion [90,91]. As shown in Figure 1, poloxamers
longer rectal retention time is expected to contribute into sustain- have two hydroxyl groups at chain ends. This reactive group
ing the drug release profile. Yuan Y et al. observed that sodium can be exploited for crosslinking with other polymers. The
chloride efficiently reduced nimesulide release rate from P407 reaction can be triggered by a chemical agent or by photo-
(18%) based liquid suppository; cumulative drug release over activation (photocrosslinking via UV exposure) [54]. However,
7 h was reduced from 90% to 70% [85]. Park et al. demonstrated the impact of crosslinking on the overall gel properties is

Figure 4. Schematic representation of poloxamers chemical crosslinking with additive polymers, crosslinking occurs between the hydrophilic corona of poloxamer
micelles and the additive polymer with the aid of a crosslinker, with a potential to increase the gel strength and hinder the drug diffusion.
EXPERT OPINION ON DRUG DELIVERY 9

associated with the degree of crosslinking, which should be 100% to zero [95]. In addition, the composite demonstrated better
carefully considered as per the desired application [90,91]. In flow-ability at room temperature. Noteworthy, these effects were
addition, the safety of the crosslinker is equally important, proportional to the calcium ion concentration [95]. Likewise, Chen
particularly with long-term exposure [92]. et al. demonstrated that grafting of alginate into poloxamer was
The concentration of the additive polymer significantly more efficient in sustaining selegiline release compared to either
affects the prolongation in drug release. Cho et al. demon- their physical blend or P407 single system [84]. Choi GJ et al.
strated that, at fixed crosslinker concentration, the gel showed that a lidocaine-loaded poloxamer-alginate-calcium
mechanical properties and ciprofloxacin release were asso- chloride in situ forming gel significantly sustained its effect and
ciated with the HA concentration. The release profile was provided control over the post-operative pain in mice compared
sustained for 5, 20, and 23 h by the addition of 1.18%, to calcium-free formulation [96].
6.87%, and 13.99% HA, respectively [64]. Chun et al. demon- To conclude, crosslinking of poloxamers with other poly-
strated the impact of degree of crosslinking on drug release mers sustain the drug release via reducing the gel erosion and
rate from HA-P407 composite formulations. Gel formulations drug diffusion rates. In addition, the additive polymer may
irradiated for 5 min and 20 min, demonstrated 100% and 50% enhance the mucoadhesive forces and residence time.
release, respectively, over a 20-day period [90]. Depending on the degree of crosslinking, this approach
Chung TW et al. used glutaraldehyde to crosslink chitosan with might be suitable to extend the drug release over one to
poloxamers, and demonstrated a significant reduction in the initial several days. Attention should be given to the safety of the
burst release of 5-fluorouracil with sustained release up to crosslinker and rheological properties of the final formulation.
52 h [91]. These changes can be justified by the viscosity incre-
ment of crosslinked formulation (30-fold the viscosity of poloxa-
4.3. Incorporating particulates into poloxamer based
mer solution) and the enhanced swelling ratio (13.2 times the
in situ gel systems
swelling ratio of non-cross-linked solution). Of note, the modula-
tion in rheological properties and release pattern was associated Encapsulating active drug into micro or nanoparticles and
with glutaraldehyde concentration [91]. Similarly, crosslinking of dispersing them in poloxamer solutions (Figure 5) is another
carboxyl methyl chitosan with poloxamer by glutaraldehyde suc- promising approach to sustain the drug release and combat
cessfully prolonged the release of nepafenac for up to 80 h [93]. the initial burst release from poloxamer based in situ gelling
The photocrosslinking of chitosan and poloxamer through systems [97]. Different particulates carriers such as nanoparti-
gamma radiation also enhanced the viscosity and mechanical cles [7,98–101], liposomes [102], microspheres [103] and
strength of the composite formulation [94]. Alginates can be microcrystals [104] have been dispersed in poloxamer in situ
crosslinked with poloxamers using calcium salts (biologically safe gel matrix (Table 2). It has been observed that the gel matrix
crosslinkers). Huang et al. demonstrated that the addition of prevents the wettability of the drug-loaded particulates and
calcium to the blend (poloxamer-sodium alginate-HPMC and thus consequently reduces the diffusion of drug from these
iodixanol) significantly reduced the gel erosion over 5 h from hybrid systems. Slow diffusion of drug could also be attributed

Figure 5. Schematic representation of potential mechanisms of drug release from drug-loaded particulate carriers incorporated in poloxamer-based gels; (a): Drug-
loaded particulates are shielded from the external environment by the gel matrix. Pathway 1 occurs when the particulates maintain their integrity inside the gel
matrix (b): Gel erosion and water influx into the matrix subjects particulates to the dissolution medium (c): Particulates dissolute and drug is released. Pathway 2
occurs if the drug-loaded particles dissolute at a faster rate than gel erosion (d): Particulates dissolute leading to drug release inside the aqueous channels (e): Gel
erosion causing the release of drug left in the matrix.
10 H. ABDELTAWAB ET AL.

to the presence of a double barrier offered by both particulate release profile of copper to 2 weeks compared to only 3 h for
carrier and gel matrix. the microspheres [109]. On the contrary, an Eudragit RS100
Various types of nanoparticles have been incorporated into microspheres-P407 hybrid system did not significantly alter
poloxamer-based formulations for sustaining the release profile the in-vivo release of roxithromycin [103]; the microsphere
as lipid nanoparticles [98,99], poly (lacto-glycolic acid) (PLGA) released 79% compared to 75% drug release from the hybrid
nanoparticles [100] and heparin-chitosan nanoparticles [101]. system over the same time period (168 h). This may be due to
The anti–inflammatory effects of clodronate were significantly the fast gel erosion which resulted in rapid wetting of the
prolonged when drug-loaded chitosan nanoparticles were dis- microspheres. Unfortunately, the study did not report the
persed in poloxamer solution and administered intra-articularly concentration of P407 which limits the ability to draw conclu-
[7]. Küçüktürkmen et al. demonstrated that incorporating drug- sions from the study.
loaded PLGA nanoparticles into chitosan (0.25%)-P407 (18%) Microcrystal dispersion in poloxamer solutions has been
blend sustained the intra-articular release of diclofenac up to employed to prolong drug release profile and improve bioac-
30 days compared to 1 and 5 days for the nanoparticles and tivity of drug. Yang et al. compared the hypoglycemic activity
blend, respectively [17]. Unfortunately, the authors did not of parenteral insulin solution, Lantus injection in hydrogel,
report gel erosion study and therefore the exact mechanism insulin microcrystals in hydrogel and coated insulin microcrys-
behind prolonged drug release is unclear. Hence, a question tals in hydrogel [104]. They demonstrated that insulin solution
might be raised whether the drug was released while the gel resulted in rabbits’ death, which can be explained by the burst
maintained its integrity, or the gel was eroded within 5 days as release of insulin and subsequent severe hypoglycemia.
well. Similarly, paclitaxel loaded nanocrystals dispersed in P407 Coated insulin microcrystals in P407/SA or P407/HPMC
demonstrated longer release profiles and intra-tumor residence resulted in prolonged hypoglycemia for more than 3 days,
time compared to drug-loaded nanocrystals or the marketed unlike the Lantus group, which showed hypoglycemia for
product Taxol® (Taxol is a paclitaxel non-aqueous solution for only 4 h followed by normal blood glucose for 20 h. The
parenteral administration) [8]. Clonazepam loaded nanostruc- results demonstrated that both coated microcrystals and
tured lipid carriers dispersed in P407-SA composite significantly uncoated microcrystals in hydrogel system are promising stra-
prolonged the onset of pentylenetetrazole-induced convulsion tegies for controlling the release pattern.
by 1.5 times and the death time by 5 times compared to the To conclude, the incorporation of drug-loaded particulates
control/placebo group in Swiss albino mice animal models in poloxamer based in situ gelling systems significantly pro-
[105]. In addition, the loading of supramagentic iron oxide longed the drug release profile. The release pattern can be
nanoparticle into this hybrid system further prolonged the finely tuned by controlling both poloxamers concentration
onset time by 7.5 times and the death time by 14 times com- and the particulate size and composition. This technique limits
pared to the control group. Orasugh JT et al. have demon- the degradation rate of the incorporated particulates through
strated that cellulose nanocrystals and cellulose nanocrystals limiting their wettability. This approach is suitable for achiev-
grafted collagen significantly sustained the release of pilocar- ing drug release over several days to few weeks following
pine and ketorolac tromethamine, respectively, from poloxa- intramuscular or subcutaneous injections.
mer based in situ gelling systems [106,107].
Liposomes are commonly investigated nano-carriers in
drug delivery. Their incorporation into poloxamer has been
4.4. Modulating the molecular weight of poloxamers
successfully employed for sustaining the drug release. Pachis
et al. reported that a liposome-P407 (18 wt.%) hybrid system Despite the advantages of the various approaches discussed
significantly reduced the flurbiprofen release to 60% com- above to sustain drug release from poloxamer gels, they are
pared to 85% from poloxamer over 48 h [102]. In addition, limited by the significant increase in the formulation viscosity
the ocular residence time increased by 23% compared to in the sol state [110]. Consequently, they might not be the best
liposomes alone or poloxamer alone and by 73% compared choice for parenteral and ophthalmic administration. To achieve
to drug solution. A hybrid system consisting of liposomes and sustained/prolonged release by improving the gel strength with-
poloxamers has also been employed for the delivery of the out enhancing formulation viscosity, a new strategy has been
anti-cancer agent paclitaxel [108]. Incorporating paclitaxel- introduced. This involves modulating the molecular weight of
loaded liposomes into P407-P188 demonstrated slower poloxamers by chemical synthesis of multi-block copolymers
release rate; 52% and 77% over 2 and 5 days compared to [41,111]. This technique produces low viscosity poloxamers solu-
80% and 97% release from the drug-loaded liposomes over tions for easy administration, transforming into strong gels at
the same periods. Similar results were demonstrated in-vivo; body temperature, with reduced drug diffusion and gel erosion
41.61% of the drug was recovered from the tumor in the rates [110–113].
hybrid system group compared to only 3.7% from the lipo- Ahn et al. demonstrated that chemical modification of polox-
some group after 2 days. Furthermore, better efficacy, higher amers (molecular weight; 31,000 g/mol) significantly reduced the
local concentration, and enhanced safety profile were erosion rates; only 15% erosion was observed over 3 weeks
achieved with the hybrid system. compared to complete degradation in less than 3 days for non-
Similarly, microspheres have also been used in sustaining modified P407 [111]. Likewise, the chemical modification of
drug release from poloxamer matrix. Pontremoli observed that poloxamer 85 has increased the time required for complete
copper-containing bioactive glass microspheres loaded in erosion from 1 to 16 days [111]. Similarly, Cohn et al. established
polyether urethane (chemically modified P407) sustained the that by synthesizing poloxamer-based poly (ester urethane)
EXPERT OPINION ON DRUG DELIVERY 11

copolymer in varying molecular weights the erosion profile can 5. Conclusion


be tailored for up to 8 months [114].
Despite the attractive features of poloxamer based in situ
Chung HJ demonstrated that multi-block poloxamers
gelling systems for drug delivery, their rapid erosion and fast
linked by D-lactide and L-lactide oligomers reduced the ero-
drug diffusion are major challenges toward sustained drug
sion rate and sustained the release of human growth hormone
release. A variety of strategies have been reported to extend
(hGH) over 13 days compared to only 1 day for the non-
drug release; however, the desired release profile and the
modified poloxamer [115]. Likewise, Sosnik A et al. reported
route of administration for specific clinical application will
increased gel strength and slow degradation for ethoxysilane
dictate the choice of the appropriate strategy.
capped P407 thus prolonging the release profile to more than
Drug release from poloxamers is controlled by both drug
2 weeks [16].
diffusion and gel erosion, which in turn is dependent on the
Modulation of poloxamer molecular weight by chemical
gel microstructure, mechanical and rheological properties. The
synthesis is a promising strategy to achieve sustained drug
gel strength is a crucial parameter in formulation vulnerable to
release over several weeks. A superior advantage of this tech-
high mechanical strain such as intra-articular and intramuscu-
nique is that the formulations flow-ability at room tempera-
lar injections. The mucoadhesive forces are an important fac-
ture is maintained, which allows for ease of administration.
tor when considering nasal, rectal, and vaginal applications.
Further studies are required to confirm the results in in-vivo
Attention should be given to the viscosity and flow-ability of
conditions.
ophthalmic and injectable formulations.
Several strategies have been used to sustain drug release
from poloxamer-based gelling systems; physical blending and
4.5. Enhancing drug–poloxamer interaction crosslinking with polymeric additive can promote the mechan-
ical, rheological, and mucoadhesive properties. Incorporating
Enhancing the interaction between the drug moiety and the
particulates creates multiple barriers and thus slowdown the
poloxamer can be an effective approach in retarding the fast
drug release. Poloxamer multi-block synthesis could be
drug diffusion and limiting the initial burst release associated
a promising approach to achieve sustained release for
with poloxamer based in situ gel systems [116]. Gilbert et al.
extended duration. It is also hypothesized that enhancing
demonstrated that the more hydrophobic drug moieties have
drug–poloxamer interactions will significantly reduce the
lower diffusion coefficients compared to the less hydrophobic
initial burst release and provide prolonged release. This
ones [27]. The reduction in diffusion coefficient through
approach needs further investigation.
enhanced drug–polymer interaction is understood by the
energy required to break down the physical or chemical
bonds between the drug and polymer and thus slowing
6. Expert opinion
drug diffusion. This advantage can be further exploited for
sustaining drug release from poloxamer-based gels. The unique ability of poloxamers to transform from sol-to-gel
Drug–poloxamer interactions can be enhanced by the form in response to temperature makes them attractive drug
choice of the drug chemical form. The gel matrix consists of delivery platforms. However, their application as a sustained-
micelles arranged compactly with aqueous channels in release delivery system for an extended duration is limited by
between as shown in Figure 2. The drug molecules partition the poor mechanical strength and high porosity, causing fast
between these compartments based on their lipophilic/hydro- gel erosion and drug diffusion. Several formulation strategies
philic balance; water-soluble drugs (salt form) tend to stay in have been investigated to overcome these limitations.
hydrophilic corona of the micelles and diffuse readily through It has been observed that poloxamer composition plays
the aqueous channels to the external environment. On the a critical role in modulating the release profile from these
contrary, base forms of the drug with higher lipophilic proper- in situ gelling systems. The concentration and ratio of the
ties demonstrate higher affinity toward the micelle core, thus hydrophobic block (PPO) and hydrophilic block (PEO) directly
it will be shielded from the external environment and the drug influences the overall gel properties and thus dictates the
release from a gel matrix will be a function of its erosion rate. drug release rates. Therefore, it is recommended that by
Drug chemical structure and its potential to interact with increasing the concentration of hydrophobic component in
poloxamers might be considered. Drug moieties with larger poloxamers such as in poloxamer 407, can significantly
organic components/more cyclic rings tend to concentrate enhance the gel strength thereby reducing the gel erosion
into the micelles core. Previous studies done by the research and drug diffusion rates. However, careful consideration
team demonstrated covalent intermolecular interaction should be given to sol-to-gel transition temperature as this
between poloxamers and the drug lidocaine using Fourier will significantly decrease with increasing hydrophobicity of
transform infrared spectroscopy, which might be one of the the poloxamer composition. Desired gelation temperature can
contributing factors in achieving slow sustained release of the be achieved by incorporating small concentrations of polox-
drug over a period of 3 days [117]. amers with dominant PEO blocks. Attention should be given
Hence, it is hypothesized that enhancing the intermolecular to the total poloxamer concentration, as per the intended
interaction between drug and poloxamers might be a promising clinical application, because this will significantly affect the
strategy to reduce the initial burst release and retain the drug viscosity and flowability of the final formulation.
inside the gel matrix for longer periods. Future studies are Physical blending of poloxamer with other additives may
required to test this hypothesis and assess its benefits. be helpful in extending the release profile from several hours
12 H. ABDELTAWAB ET AL.

up to few days, while crosslinking of poloxamers with poly- poloxamers is temperature driven, the release profile of the
meric additives may offer more extended release profile. This loaded drug is likely to be greatly affected by the temperature
is explained by the presence of stronger bonds and bridging at the administration site. The physiological temperature var-
between poloxamers and the additive polymer. Some addi- ies between different body regions. For example, the normal
tives as alginate and sodium chloride significantly enhance the temperature of intra-articular joint, skin, corneal, intra-
formulation mucoadhesive properties, which is beneficial for muscular and vaginal cavity is reported as 32.6°C, 32.7°C,
mucosal applications. Modulation in physical properties and 35.7°C, and 38.2°C, respectively [118–121]. In addition, the
release profile is associated with the additive concentration temperature may increase or decrease based on the clinical
and molecular weight. Hence, the physical properties such as condition; for example, intra-articular temperature of healthy
formulation’s viscosity, flow-ability, and spread-ability should subjects is around 32.6°C, whereas the temperature in case of
be studied simultaneously along with the drug release studies. osteoarthritis and rheumatoid arthritis is recorded as 33.5°
Incorporating a drug-loaded particulate can sustain the C and 35.1°C, respectively. On the other hand, the intra-
release profile from these in situ gelling systems from few articular temperature at the time of knee surgery drops
days to weeks depending upon the particles properties and down to 29.9°C [118]. Thus, attention must be given to the
poloxamer matrix. Thereby, the size, composition, and wett- temperature at the application site as per the desired clinical
ability of the dispersed particulates are critically important in application. The volume and composition of the biological
achieving the desired release profile. In addition, the poloxa- fluids at the application site is another important factor to
mer matrix composition and strength has a significant role in be considered in the development of a sustained release
shielding the particulates from external environment. The poloxamer based in situ gelling systems, as it significantly
drug-loading efficiency and the affinity/interaction with parti- affects the gel erosion rate. Our previous findings demon-
culates should aptly considered. strated that poloxamer based gelling systems erode faster in
Chemically synthesized multi-block poloxamers tend to human synovial fluid compared to the phosphate buffer saline
provide sustained release from weeks to months, which is [122]. Future studies are required to further assess the effect of
suitable in conditions requiring drug release over extended these factors on the drug release from poloxamer based in situ
periods. This approach develops a highly strong gel matrix gelling systems.
with significantly slow erosion rate, without altering the visc- Poloxamer based in situ gelling systems have promising
osity in the sol state. These chemically modified poloxamers potential as drug delivery platforms and will continue to
therefore are more suitable for the development of sustained- gain interest in the field of modified release drug delivery. It
release injectable formulations. Nonetheless, poloxamers is postulated that an increasing number of sustained release,
obtained after chemical modification are considered new com- targeted/localized poloxamer-based delivery systems will
pounds by regulators and therefore the safety and toxicity of come into light within the coming decade.
the newly synthesized compounds need to be thoroughly
investigated.
Funding
It is hypothesized that enhancing the drug–poloxamer
interaction might significantly limit the initial burst release The authors are thankful to the New Zealand Pharmaceutical Education
and sustain the release of the loaded drug. This approach is Research Fund [NZPERF] [Reference number – 3719172] and The
simple and can be easily adopted by selecting the suitable University of Auckland for financial support.
form of the drug. More lipophilic drug form will demonstrate
higher affinity toward the micellar core, and thus rapid diffu-
sion could be avoided. On the contrary, the more hydrophilic Declaration of interest
drug forms tend to partition in the aqueous channels and The authors have no relevant affiliations or financial involvement with any
diffuse outside the gel matrix rapidly. organization or entity with a financial interest in or financial conflict with
Though several studies have demonstrated sustained the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
release of drugs from poloxamer based in situ gels, there is testimony, grants or patents received or pending, or royalties.
considerable variation and a lack of standardization in the
experimental design and methodologies used to characterize
these systems. This variation makes it difficult to directly com- Reviewer disclosures
pare the results of different studies and to fully understand
release mechanisms. In addition, the lack of information Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
regarding important parameters such as molecular weight,
concentration, and particle size of the additives, makes it
difficult to form meaningful and translatable conclusions
References
based on the data available in the existing literature.
Researchers in this field should, as far as possible, generate Papers of special note have been highlighted as either of interest (•) or of
robust data sets using established methodologies and report considerable interest (••) to readers.
all the experimental parameters available. 1. Bodratti AM, Alexandridis P. Formulation of poloxamers for drug
delivery. J Funct Biomater. 2018;9(1):11.
It is also recommended that the potential site of adminis- •• A review article providing a deep discussion on poloxamer
tration must be critically considered at the initial stage of self-assembly and various techniques to encapsulate drugs
formulation development. Since the sol-to-gel transition of into poloxamers.
EXPERT OPINION ON DRUG DELIVERY 13

2. Englert C, Brendel JC, Majdanski TC, et al. Pharmapolymers in the 24. Batrakova EV, Kabanov AV. Pluronic block copolymers: evolution of
21st century: synthetic polymers in drug delivery applications. Prog drug delivery concept from inert nanocarriers to biological
Polym Sci Pergamon. 2018;87:107–164. response modifiers. J Control Release. 2008;130(2):98–106.
3. Domb Abraham J, Kost J, Wiseman DM. Handbook of biodegrad- 25. Gong C, Qi T, Wei X, et al. Thermosensitive polymeric hydrogels as
able polymers. Boca Raton (FL): CRC/Taylor & Francis; 1997. drug delivery systems. Curr Med Chem. 2012;20(1):79–94.
4. Almeida H, Amaral MH, Lobão P, et al. Applications of poloxamers 26. Yang L, Alexandridis P. Controlled drug delivery: controlled release
in ophthalmic pharmaceutical formulations: an overview. Expert from ordered microstructures formed by poloxamer block copoly-
Opin Drug Deliv. 2013;10(9):1223–1237. mers In: Controlled Drug Delivery, Vol.752, ACS Symposium Series:
•• Excellent review providing a deep understanding of the main American Chemical Society; 2000; 364–374.
advantages of poloxamer as drug delivery systems for opthal- 27. Gilbert JC, Hadgraft J, Bye A, et al. Drug release from pluronic F-127
mic applications. gels. Int J Pharm [Internet]. 1986;32(2–3):223–228.
5. Caramella CM, Rossi S, Ferrari F, et al. Mucoadhesive and thermo- •• First article to highlight the effect of physicochemical proper-
gelling systems for vaginal drug delivery. Adv Drug Deliv Rev. ties of the loaded drug on the release mechanism and rate of
2015;92:39–52. release from poloxamer based in-situ gels.
6. Giuliano E, Paolino D, Fresta M, et al. Mucosal applications of 28. Akkari ACS, Papini JZB, Garcia GK, et al. Poloxamer 407/188 binary
poloxamer 407-based hydrogels: an overview. Pharmaceutics. thermosensitive hydrogels as delivery systems for infiltrative local
2018;10(3):1–26. anesthesia: physico-chemical characterization and pharmacological
• A review article highlighting the different mucosal applica- evaluation. Mater Sci Eng C. 2016;68:299–307.
tions of poloxamer 407. 29. Nascimento MHM, Franco MKKD, Yokaichyia F, et al. Hyaluronic
7. Russo E, Gaglianone N, Baldassari S, et al. Chitosan-clodronate acid in pluronic F-127/F-108 hydrogels for postoperative pain in
nanoparticles loaded in poloxamer gel for intra-articular arthroplasties: influence on physico-chemical properties and struc-
administration. Colloids Surf B Biointerfaces. 2016;143:88–96. tural requirements for sustained drug-release. Int J Biol Macromol.
8. Lin Z, Gao W, Hu H, et al. Novel thermo-sensitive hydrogel system 2018;111:1245–1254.
with paclitaxel nanocrystals: high drug-loading, sustained drug 30. Shin S-C, Cho C-W, Choi H-K. Permeation of piroxicam from the
release and extended local retention guaranteeing better efficacy poloxamer gels. Drug Dev Ind Pharm. 1999;25(3):273–278.
and lower toxicity. J Control Release. 2014;174(1):161–170. 31. Fathalla MAZ, Vangala A, Longman M, et al. Poloxamer-based
9. Almeida M, Magalhães M, Veiga F, et al. Poloxamers, poloxamines thermoresponsive ketorolac tromethamine in situ gel preparations:
and polymeric micelles: definition, structure and therapeutic appli- design, characterisation, toxicity and transcorneal permeation
cations in cancer. J Polym Res. 2018;25(1):31. studies. Eur J Pharm Biopharm. 2017;114:119–134.
10. Sharma M, Jingjunjiao L, Seyfoddin A, et al. Design, synthesis and 32. Hemelryck S, Van Dewulf J, Niekus H, et al. In vitro evaluation of
application in drug delivery and regenerative medicine: thermosensi- poloxamer in situ forming gels for bedaquiline fumarate salt and
tive hydrogels for drug delivery and tissue engineering. Boca Raton pharmacokinetics following intramuscular injection in rats.
(FL): CRC Press/Taylor & Francis Group; 2018. p. 184–207. [2017]. Int J Pharm X. 2019;1:100016.
11. Sosnik A, Cohn D, San Román J, et al. Crosslinkable PEO-PPO-PEO- 33. Bonacucina G, Cespi M, Mencarelli G, et al. Thermosensitive
based reverse thermo-responsive gels as potentially injectable self-assembling block copolymers as drug delivery systems.
materials. J Biomater Sci Polym Ed. 2003;14(3):227–239. Polymers. 2011;3(2):779–811.
12. Jeong B. Injectable biomaterials: science and applications: inject- 34. Liu Y, Zhu YY, Wei G, et al. Effect of carrageenan on poloxamer-based
able biodegradable materials. Cambridge (UK): Woodhead in situ gel for vaginal use: improved in vitro and in vivo
Publishing; 2011. p. 323–337. sustained-release properties. Eur J Pharm Sci. 2009;37(3–4):306–312.
13. Qiu Y, Hamilton SK, Temenoff J. Injectable biomaterials: science 35. Inal O, Yapar EA. Effect of mechanical properties on the release of
and applications: improving mechanical properties of injectable meloxicam from poloxamer gel bases. Indian J Pharm Sci. 2013;75
polymers and composites. Cambridge (UK): Woodhead Publishing; (6):700–706.
2011. p. 61–91. • Provides a deep understanding of the correlation between the
14. Zhang K, Shi X, Lin X, et al. Poloxamer-based in situ hydrogels for gel mechanical properties and the release profile from polox-
controlled delivery of hydrophilic macromolecules after intramus- amer based in-situ gels.
cular injection in rats. Drug Deliv. 2015;22(3):375–382. 36. Liu Y, Yang F, Feng L, et al. In vivo retention of poloxamer-based
15. Sathish Ummadi B, Shravani NG, Rao R, et al. Overview on con- in situ hydrogels for vaginal application in mouse and rat models.
trolled release dosage form. Int J Pharma Sci. 2013;3:258–269. Acta Pharm Sin B. 2017;7(4):502–509.
16. Sosnik A, Cohn D. Ethoxysilane-capped PEO-PPO-PEO triblocks: 37. Moore T, Croy S, Mallapragada S, et al. Experimental investigation
A new family of reverse thermo-responsive polymers. and mathematical modeling of Pluronic(®) F127 gel dissolution:
Biomaterials. 2004;25(14):2851–2858. drug release in stirred systems. J Control Release. 2000;67
17. Küçüktürkmen B, Öz UC, Bozkir A. In situ hydrogel formulation for (2–3):191–202.
intra-articular application of diclofenac sodium-loaded polymeric 38. Suh H, Jun HW. Physicochemical and release studies of naproxen in
nanoparticles. Turkish J Pharm Sci. 2017;14(1):56–64. poloxamer gels. Int J Pharm. 1996 Mar 8;129(1–2):13–20.
18. Pitto-Barry A, Barry NPE. Pluronic® block-copolymers in medicine: 39. Chi SC, Jun HW. Release rates of ketoprofen from poloxamer gels in
from chemical and biological versatility to rationalisation and clin- a membraneless diffusion cell. J Pharm Sci. 1991;80(3):280–283.
ical advances. Polym Chem. 2014;5(10):3291–3297. 40. Jung Y, Park W, Park H, et al. Thermo-sensitive injectable hydrogel
19. Bromberg L. Polymeric micelles in oral chemotherapy. J Control based on the physical mixing of hyaluronic acid and Pluronic F-127
Release. 2008;128(2):99–112. for sustained NSAID delivery. Carbohydr Polym. 2017;156:403–408.
20. Kaur P, Garg T, Rath G, et al. Surfactant-based drug delivery systems 41. Sun KH, Sohn YS, Jeong B. Thermogelling poly(ethylene
for treating drug-resistant lung cancer. Drug Deliv. 2014;23(3):1–12. oxide-b-propylene oxide-b-ethylene oxide) disulfide multiblock
21. Kabanov A, Zhu J, Alakhov V. Pluronic block copolymers for gene copolymer as a thiol-sensitive degradable polymer.
delivery. Adv Genet. 2005;53:231–261. Biomacromolecules. 2006;7(10):2871–2877.
22. Kabanov AV, Batrakova EV, Alakhov VY. Pluronic block copolymers 42. Chen-Chow P-C, Frank SG. In vitro release of lidocaine from pluro-
as novel polymer therapeutics for drug and gene delivery. J Control nic F-127 gels. Int J Pharm. 1981;8(2):89–99.
Release. 2002;82(2–3):189–212. 7. 43. Zhang L, Parsons DL, Navarre C, et al. Development and in-vitro
23. Alakhova DY, Kabanov AV. Pluronics and MDR reversal: an update. evaluation of sustained release Poloxamer 407 (P407) gel formula-
Mol Pharm. 2014;11(8):2566–2578. tions of ceftiofur. J Control Release. 2002;85(1–3):73–81.
14 H. ABDELTAWAB ET AL.

44. Ricci EJJ, Lunardi LOO, DMAMA N, et al. Sustained release of 66. Gupta H, Sharma A, Shrivastava B. Pluronic and Chitosan based
lidocaine from Poloxamer 407 gels. Int J Pharm. 2005;288 in situ gel system for periodontal application. Asian J Pharm. 2009;3
(2):235–244. (2):94.
45. Fakhari A, Corcoran M, Schwarz A. Thermogelling properties of 67. Sridhar V, Wairkar S, Gaud R, et al. Brain targeted delivery of
purified poloxamer 407. Heliyon. 2017;3(8):e00390. mucoadhesive thermosensitive nasal gel of selegiline hydrochlor-
46. Rangabhatla ASL, Tantishaiyakul V, Oungbho K, et al. Fabrication of ide for treatment of Parkinson’s disease. J Drug Target. 2018;26
pluronic and methylcellulose for etidronate delivery and their (2):150–161.
application for osteogenesis. Int J Pharm. 2016;499(1–2):110–118. 68. Sherif AY, Mahrous GM, Alanazi FK. Novel in-situ gel for intravesical
47. Kim C-K-K, Lee S-W-W, Choi H-G-G, et al. Trials of in situ-gelling and administration of ketorolac. Saudi Pharm J. 2018;26(6):845–851.
mucoadhesive acetaminophen liquid suppository in human 69. Karatas A, Boluk A, Algan A. Poloxamer/Chitosan in situ gelling
subjects. Int J Pharm. 1998;174(1–2):201–207. system for ocular delivery of ofloxacin. Curr Drug Ther. 2014;9
48. Mayol L, Quaglia F, Borzacchiello A, et al. A novel poloxamers/ (4):219–225.
hyaluronic acid in situ forming hydrogel for drug delivery: rheolo- 70. Ur-Rehman T, Tavelin S, Gröbner G. Chitosan in situ gelation for
gical, mucoadhesive and in vitro release properties. Eur J Pharm improved drug loading and retention in poloxamer 407 gels.
Biopharm. 2008;70(1):199–206. Int J Pharm. 2011;409(1–2):19–29.
49. Dewan M, Bhowmick B, Sarkar G, et al. Effect of methyl cellulose on 71. Cho HJ, Balakrishnan P, Park EK, et al. Poloxamer/cyclodextrin/
gelation behavior and drug release from poloxamer based ophthal- chitosan-based thermoreversible gel for intranasal delivery of fex-
mic formulations. Int J Biol Macromol. 2015;72:706–710. ofenadine hydrochloride. J Pharm Sci. 2011;100(2):681–691.
50. Xuan JJ, Balakrishnan P, Oh DH, et al. Rheological characterization 72. Tuğcu-Demiröz F. Development of in situ poloxamer-chitosan
and in vivo evaluation of thermosensitive poloxamer-based hydro- hydrogels for vaginal drug delivery of benzydamine hydrochloride:
gel for intramuscular injection of piroxicam. Int J Pharm. 2010;395 textural, mucoadhesive and in vitro release properties. Marmara
(1–2):317–323. Pharm J. 2017;21(4):762–770.
51. Lin H-R-R, Sung KC, Vong W-J-J. In situ gelling of alginate/pluronic 73. Zaki NM, Awad GA, Mortada ND, et al. Enhanced bioavailability of
solutions for ophthalmic delivery of pilocarpine. Biomacromolecules. metoclopramide HCl by intranasal administration of
2004;5(6):2358–2365. a mucoadhesive in situ gel with modulated rheological and muco-
52. Qi H, Chen W, Huang C, et al. Development of a poloxamer analogs/ ciliary transport properties. Eur J Pharm Sci. 2007;32(4–5):296–307.
carbopol-based in situ gelling and mucoadhesive ophthalmic delivery 74. Qian S, Wong YC, Zuo Z. Development, characterization and appli-
system for puerarin. Int J Pharm. 2007;337(1–2):178–187. cation of in situ gel systems for intranasal delivery of tacrine.
53. Aka-Any-Grah A, Bouchemal K, Koffi A, et al. Formulation of Int J Pharm. 2014;468(1–2):272–282.
mucoadhesive vaginal hydrogels insensitive to dilution with vagi- 75. Kojarunchitt T, Baldursdottir S, Da DY, et al. Modified thermore-
nal fluids. Eur J Pharm Biopharm. 2010;76(2):296–303. sponsive poloxamer 407 and chitosan sol-gels as potential
54. Parhi R. Cross-linked hydrogel for pharmaceutical applications: a sustained-release vaccine delivery systems. Eur J Pharm Biopharm.
review. Adv Pharm Bull. 2017;7(4):515–530. 2015;89:74–81.
55. Gratieri T, Gelfuso GM, Rocha EM, et al. A poloxamer/chitosan 76. Shokri J, Adibki K. Cellulose - medical, pharmaceutical and electro-
in situ forming gel with prolonged retention time for ocular nic applications: application of cellulose and cellulose derivatives in
delivery. Eur J Pharm Biopharm. 2010;75(2):186–193. pharmaceutical industries. London, UK: InTech; 2013.
56. Kianfar F, Ayensu I, Boateng JS. Development and 77. Lu C, Liu M, Fu H, et al. Novel thermosensitive in situ gel based on
physico-mechanical characterization of carrageenan and poloxamer for uterus delivery. Eur J Pharm Sci. 2015;77:24–28.
poloxamer-based lyophilized matrix as a potential buccal drug 78. Wang W, Wat E, Hui PCL, et al. Dual-functional transdermal drug
delivery system. Drug Dev Ind Pharm. 2014;40(3):361–369. delivery system with controllable drug loading based on thermo-
57. Li C, Li C, Liu Z, et al. Enhancement in bioavailability of ketorolac sensitive poloxamer hydrogel for atopic dermatitis treatment. Sci
tromethamine via intranasal in situ hydrogel based on poloxamer Rep. 2016;6(1):24112.
407 and carrageenan. Int J Pharm. 2014;474(1–2):123–133. 79. Yu Z-G, Geng Z-X, Liu T-F, et al. In vitro and in vivo evaluation of an
58. Kianfar F, Antonijevic MD, Chowdhry BZ, et al. Formulation devel- in situ forming gel system for sustained delivery of florfenicol. J Vet
opment of a carrageenan based delivery system for buccal drug Pharmacol Ther. 2015;38(3):271–277.
delivery using ibuprofen as a model drug. J Biomater 80. Talasaz AHHH, Ghahremankhani AA, Moghadam SH, et al. In situ
Nanobiotechnol. 2011;2:582–595. gel forming systems of poloxamer 407 and hydroxypropyl cellulose
59. Li R, Li Y, Wu Y, et al. Heparin-poloxamer thermosensitive hydrogel or hydroxypropyl methyl cellulose mixtures for controlled delivery
loaded with bFGF and NGF enhances peripheral nerve regenera- of vancomycin. J Appl Polym Sci. 2008;109(4):2369–2374.
tion in diabetic rats. Biomaterials. 2018;168:24–37. 81. Mansour M, Mansour S, Mortada ND, et al. Ocular poloxamer-based
60. Zhao YZ, Jiang X, Xiao J, et al. Using NGF heparin-poloxamer ciprofloxacin hydrochloride in situ forming gels. Drug Dev Ind
thermosensitive hydrogels to enhance the nerve regeneration for Pharm. 2008;34(7):744–752.
spinal cord injury. Acta Biomater. 2016;29:71–80. 82. Marcos X, Pérez-Casas S, Llovo J, et al. Poloxamer-hydroxyethyl
61. Guo DD, Xu CX, Quan JS, et al. Synergistic anti-tumor activity of cellulose-α-cyclodextrin supramolecular gels for sustained release
paclitaxel-incorporated conjugated linoleic acid-coupled poloxa- of griseofulvin. Int J Pharm. 2016;500(1–2):11–19.
mer thermosensitive hydrogel in vitro and in vivo. Biomaterials. 83. Sachan NK, Pushkar S, Jha A, et al. Sodium alginate: the wonder
2009;30(27):4777–4785. polymer for controlled drug delivery. J Pharm Res. 2009;2
62. Dewan M, Sarkar G, Bhowmik M, et al. Effect of gellan gum on the (8):1191–1199.
thermogelation property and drug release profile of poloxamer 407 84. Chen -C-C, Fang C-L, Al-Suwayeh SA, et al. Transdermal delivery of
based ophthalmic formulation. Int J Biol Macromol. selegiline from alginate–pluronic composite thermogels.
2017;102:258–265. Int J Pharm. 2011;415(1–2):119–128.
63. Choi HG, Lee MK, Kim MH, et al. Effect of additives on the physi- 85. Yuan Y, Cui Y, Zhang L, et al. Thermosensitive and mucoadhesive
cochemical properties of liquid suppository bases. Int J Pharm. in situ gel based on poloxamer as new carrier for rectal adminis-
1999;190(1):13–19. tration of nimesulide. Int J Pharm. 2012;430(1–2):114–119.
64. Cho KY, Chung TW, Kim BC, et al. Release of ciprofloxacin from 86. Yong CS, Choi JS, Quan QZ, et al. Effect of sodium chloride on the
poloxamer-graft-hyaluronic acid hydrogels in vitro. Int J Pharm. gelation temperature, gel strength and bioadhesive force of polox-
2003;260(1):83–91. amer gels containing diclofenac sodium. Int J Pharm. 2001;226
65. Strauss EJ, Hart JA, Miller MD, et al. Hyaluronic acid viscosupple- (1–2):195–205.
mentation and osteoarthritis. Am J Sports Med. 2009;37 87. Bermudez JM, Grau R. Thermosensitive poloxamer-based inject-
(8):1636–1644. ables as controlled drug release platforms for veterinary use:
EXPERT OPINION ON DRUG DELIVERY 15

development and in-vitro evaluation. Int Res J Pharm Pharmacol. gel for magnetic brain targeting of clonazepam. J Pharm Sci.
2011;1(6):109–118. 2018;107(8):2119–2127.
88. Park YJ, Yong CS, Kim HM, et al. Effect of sodium chloride on the 106. Orasugh JT, Sarkar G, Saha NR, et al. Effect of cellulose nanocrystals
release, absorption and safety of diclofenac sodium delivered by on the performance of drug loaded in situ gelling
poloxamer gel. Int J Pharm. 2003;263(1–2):105–111. thermo-responsive ophthalmic formulations. Int J Biol Macromol.
89. Zeng N, Dumortier G, Maury M, et al. Influence of additives on 2019 Mar;1(124):235–245.
a thermosensitive hydrogel for buccal delivery of salbutamol: rela- 107. Orasugh JT, Dutta S, Das D, et al. Sustained release of ketorolac
tion between micellization, gelation, mechanic and release tromethamine from poloxamer 407/cellulose nanofibrils graft
properties. Int J Pharm. 2014;467(1–2):70–83. nanocollagen based ophthalmic formulations. Int J Biol
90. Chun KW, Lee JB, Kim SH, et al. Controlled release of plasmid DNA Macromol. 2019;140:441–453.
from photo-cross-linked pluronic hydrogels. Biomaterials. 2005;26 108. Mao Y, Li X, Chen G, et al. Thermosensitive hydrogel system with
(16):3319–3326. paclitaxel liposomes used in localized drug delivery system for
91. Chung TW, Lin SY, Liu DZ, et al. Sustained release of 5-FU from in situ treatment of tumor: better antitumor efficacy and lower
poloxamer gels interpenetrated by crosslinking chitosan network. toxicity. J Pharm Sci. 2016;105(1):194–204.
Int J Pharm. 2009;382(1–2):39–44. 109. Pontremoli C, Boffito M, Fiorilli S, et al. Hybrid injectable platforms
92. Takigawa T, Endo Y. Effects of glutaraldehyde exposure on human for the in situ delivery of therapeutic ions from mesoporous
health. J Occup Health. 2006;48(2):75–87. glasses. Chem Eng J. 2018;340:103–113.
93. Yu S, Zhang X, Tan G, et al. A novel pH-induced thermosensitive 110. Niu G, Du F, Song L, et al. Synthesis and characterization of reactive
hydrogel composed of carboxymethyl chitosan and poloxamer poloxamer 407s for biomedical applications. J Control Release.
cross-linked by glutaraldehyde for ophthalmic drug delivery. 2009;138(1):49–56.
Carbohydr Polym. 2017;155:208–217. 111. Ahn JS, Suh JM, Lee M, et al. Slow eroding biodegradable multi-
94. Leyva-Gómez G, Santillan-Reyes E, Lima E, et al. A novel hydrogel of block poloxamer copolymers. Polym Int. 2005;54(5):842–847.
poloxamer 407 and chitosan obtained by gamma irradiation exhi- 112. Niu G, Zhang H, Song L, et al. Thiol/acrylate-modified PEO-PPO-PEO
bits physicochemical properties for wound management. Mater Sci triblocks used as reactive and thermosensitive copolymers.
Eng C. 2017;74:36–46. Biomacromolecules. 2008;9(10):2621–2628.
95. Huang L, Shen M, Li R, et al. Thermo-sensitive composite hydrogels 113. Choi SG, Baek EJ, Davaa E, et al. Topical treatment of the buccal
based on poloxamer 407 and alginate and their therapeutic effect mucosa and wounded skin in rats with a triamcinolone
in embolization in rabbit VX2 liver tumors. Oncotarget. 2016;7 acetonide-loaded hydrogel prepared using an electron beam.
(45):73280–73291. Int J Pharm. 2013;447(1–2):102–108.
96. Choi GJ, Kang H, Hong ME, et al. Effects of a lidocaine-loaded polox- 114. Cohn D, Lando G, Sosnik A, et al. PEO-PPO-PEO-based poly(ether
amer/Alginate/CaCl 2 mixture on postoperative pain and adhesion in ester urethane)s as degradable reverse thermo-responsive multi-
a rat model of incisional pain. Anesth Analg. 2017;125(1):320–327. block copolymers. Biomaterials. 2006;27(9):1718–1727.
97. He Z, Wang B, Hu C, et al. An overview of hydrogel-based • A research paper with interesting findings showing how to
intra-articular drug delivery for the treatment of osteoarthritis. extend the release profile from poloxamer based in-situ gels
Vol. 154. Colloids Surf B Biointerfaces. 2017;154:33–39. up to several months.
98. Ravani L, Esposito E, Bories C, et al. Clotrimazole-loaded nanostruc- 115. Chung HJ, Lee Y, Park TG. Thermo-sensitive and biodegradable
tured lipid carrier hydrogels: thermal analysis and in vitro studies. hydrogels based on stereocomplexed pluronic multi-block copoly-
Int J Pharm. 2013;454(2):695–702. mers for controlled protein delivery. J Control Release. 2008;127
99. Hao J, Wang X, Bi Y, et al. Fabrication of a composite system (1):22–30.
combining solid lipid nanoparticles and thermosensitive hydrogel 116. Hoare TR, Kohane DS. Hydrogels in drug delivery: progress and
for challenging ophthalmic drug delivery. Colloids Surf B. challenges. Polymer. 2008;49(8):1993–2007.
2014;114:111–120. 117. Svirskis D, Chandramouli K, Bhusal P, et al. Injectable thermosensi-
100. Bansal A, Wu X, Olson V, et al. Characterization of rabies pDNA tive gelling delivery system for the sustained release of lidocaine.
nanoparticulate vaccine in poloxamer 407 gel. Int J Pharm. Ther Deliv. 2016;7(6):359–368.
2018;545(1–2):318–328. 118. Ammer K. Temperature of the human knee - A review. Vol. 22.
101. Radivojša Matanović M, Grabnar I, Gosenca M, et al. Prolonged Thermol Int. 2012;22(4):137–151.
subcutaneous delivery of low molecular weight heparin based on 119. Karlsbad A, Kopp S. Intramuscular and skin surface temperatures of
thermoresponsive hydrogels with chitosan nanocomplexes: design, the resting human superficial masseter muscle. Acta Odontol
in vitro evaluation, and cytotoxicity studies. Int J Pharm. 2015;488 Scand. 1991;49(4):225–231.
(1–2):127–135. 120. Soede NM, Hazeleger W, Broos J, et al. Vaginal temperature is not
102. Pachis K, Blazaki S, Tzatzarakis M, et al. Sustained release of intravi- related to the time of ovulation in sows. Anim Reprod Sci. 1997;47
treal flurbiprofen from a novel drug-in-liposome-in-hydrogel (3):245–252.
formulation. Eur J Pharm Sci. 2017;109:324–333. 121. Horven I. Corneal temperature in normal subjects and arterial
103. Gautam N, Singh R. Roxithromycin loaded microspheres incorpo- occlusive disease. Acta Ophthalmol. 1975;53(6):863–874.
rated in in-situ gel of poloxamer 407 for periodontitis (RLMISG). 122. Sharma M, Chandramouli K, Curley L, et al. In vitro and ex vivo
World J Pharm Pharm Sci. 2016;6(02):637–652. characterisation of an in situ gelling formulation for sustained
104. Yang G, Wang Q, Gao Y, et al. Combination of coating and inject- lidocaine release with potential use following knee arthroplasty.
able hydrogel depot to improve the sustained delivery of insulin. Drug Deliv Transl Res. 2018;8(3):820–829.
J Drug Deliv Sci Technol. 2018;45:415–421. • A research paper highlighting the effect of physiological med-
105. Abbas H, Refai H, El Sayed N. Superparamagnetic iron oxide– ium on the release profile from poloxamer based in-situ gels
loaded lipid nanocarriers incorporated in thermosensitive in situ compared to synthetic media.

You might also like