Freely Available Online
JOURNAL OF WOMAN’S REPRODUCTIVE HEALTH
ISSN NO: 2381-862X
Review Article
DOI: 10.14302/issn.2381-862X.jwrh-19-3143
pH-Sensitive Nanomedicine for Treating Gynaecological Cancers
Pramod Vishwanath Prasad1*, Kakali Purkayastha2, Utkarsh Sharma3, and Mayadhar Barik4
1
Center for Biomedical Research, Population Council, The Rockefeller University, 1230 York Avenue, New York, NY
10065, USA
2
Department of Paediatrics, All India Institute of Medical Sciences, New Delhi-110 029, India
3
Patna Medical College and Hospital, Patna University Campus, Ashok Rajpath, Patna- 800001, Bihar, India
4
Department of Biochemistry, Mewar University, NH-79, Gangrar, Chittorgarh, Rajasthan-312901, India
Abstract
Emergence of various nanoscale drug carrier platforms as Drug Delivery Systems (DDS) has
revolutionized the field of medicine. Nonetheless, the side-effects due to non-specific distribution of anticancer
therapeutics in normal, healthy tissues remain to be a prime pitfall in curing cancers. Therefore, to achieve a
better therapeutic efficacy, the use of a target-specific delivery, combined with a stimuli-responsive nanocarrier
system, particularly pH-sensitive nanosystems offer an attractive strategy. Targeted drug delivery through
pH-sensitive nanosystems offer the potential to enhance the therapeutic index of anticancer agents, either by
increasing the drug concentration in tumor cells and/or by decreasing the exposure in normal host tissues.
Therefore, nanoscale-based drug delivery through pH-sensitive nanosystems seem to be a boon for treating
gynaecological cancers (as well as other cancers) without side-effects or with least harm to normal healthy
tissues.
*
Corresponding author: Pramod Vishwanath Prasad, Center for Biomedical Research, Population Council, The
Rockefeller University, 1230 York Avenue, New York, NY 10065, USA. Email:
[email protected]
Keywords: Gynaecological Cancers; pH-Sensitive nanomedicine; Liposomes; Polymeric micelles; Dendrimers;
Nanogels.
Received: Dec 24, 2019
Accepted: Jan 28, 2020
Published: Feb 11, 2020
Editor: Qiuqin Tang, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, China.
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 35
Freely Available Online
their structure and surface characteristics, causing drug
Introduction
A hope of successful treatment of cancers
without side effects has challenged oncologists and
onco-scientists since decades. Burgeoning research in
nanotechnology and the depth of understanding in
gynae-oncological pathophysiology at the cellular and
molecular levels have led to the development of
different well-tailored nanosized carriers for drug loading
and controlled delivery at the targeted site. In recent
years, nanosized carriers (nanocarriers) have gained
attention as unique drug delivery agents due to
following qualities: (i) they have abilities to incorporate
payloads with different solubilities [1], (ii) they improve
the in vivo pharmacokinetics (PK) of drugs [2], (iii) they
enhance bioavailabilities (i.e., the drug stability and
longevity in the blood circulation with or without
additional structural modifications) [3], and (iv) they
modify the carriers with targeting ligands on their
surface for tumor tissue or cell-specific delivery to
minimize side-effects on healthy cells/ tissues [4]. Some
of the nanocarriers developed till today are liposomes,
dendrimers, polymeric nanoparticles (NPs), gold or other
metallic NPs,
inorganic NPs made of iron oxide,
quantum dots etc. [5-7]. Few of them possess
unique
nature
of
stimuli-responsiveness.
Such
release
or
contrast
enhancement
at
a
particular
pathological site [11, 12]. This ease of controlled drug
delivery at the desired site, has incepted the preliminary
idea
of
developing
pH-sensitive
drug
delivery
nanosystems. Moreover, the pH-responsive NPs are one
of the most extensively studied stimuli-responsive
nanosystems. This is due to its sensitivities to the
changes in pH condition at the tumor or diseased tissue
site [13, 14].
Generally,
pH-responsive
nanoparticles
are
fabricated either using acid-sensitive linkers or ionizable
groups [15]. Varieties of pH-sensitive nanoparticles have
been designed in recent decades and have characteristic
functionalities in the molecular structure, where pKa
(negative logarithm of the acid dissociation constant)
values are close to the tumor interstitial pH. When
these
nanoparticles
reach
tumors
where
the
microenvironmental pH is slightly acidic, a pH-dependent
structural transformation occurs. The acidic environment
at the tumor site triggers the protonation of pH-sensitive
moieties, thereby disrupting the hydrophilic-hydrophobic
equilibrium within the nanoparticle, in turn causing
structural transformation and the release of therapeutic
cargo loaded inside.
Despite
stimuli-responsive nanocarriers have emerged as an
of
few
problems
with
nanoparticle-based
DDS
“intelligent” or “smart” DDS. Thus, these nanocarriers
nanomedicine,
have exhibited myriads of successful applications in
remain as a potential strategy for cancer therapy. Some
comparison with conventional DDS.
nanoparticle formulations for cancer treatment have
The main purpose of developing nanomedicine
and nanotherapies is to avoid damage to healthy organs.
So,
these
innovative
approaches
seem
to
have
tremendous potential in improving the effectiveness of
nanomedicines to treat clinical tumors with null side
effects [8]. There are several nanomaterials which have
been found to be responsive to external (viz., light,
ultrasound)
potential,
and
internal
temperature),
stimuli
and
(viz.,
have
pH,
been
redox
utilized
for cancer therapy and simultaneous diagnosis i.e.,
theranostics
[5,
9,
10].
Among
the
various
stimuli-responsive nanosystems, pH-stimuli mode is
regarded as the most general strategy because of solid
tumors acidosis. When exposed to weakly acidic tumor
microenvironment,
drug
carrying
pH-responsive
nanoplatforms can generate physicochemical changes in
pH-sensitive
associated
been already approved by regulatory agencies. These
formulations
exert
fewer
adverse
effects
than
unmodified or bare drugs [16]. Therefore, in the interest
of brevity, this review article simply retrospects and
compiles only pH-sensitive nanosystems among other
internal
stimuli-responsive
pH-sensitive
nanosystems
systems.
Some
retrospected
of
the
here
are
certainly not yet directly used for treating gynecological
tumors but paves the way for employing them for
treating gynecological cancers with some strategic
modifications depending on tissue types.
pH-Sensitive Nano-Systems
Generally, physiological pH remains 7.4 (weakly
basic) but the subcellular compartments viz., endosomes
and lysosomes exhibit remarkably lower pH of about 5-6
or 4-5, respectively. Therefore, significant lower pH in
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 36
Freely Available Online
subcellular compartments has been used as a route for
they revealed the enhanced release and accumulation of
delivering anticancer drugs by the pH-stimulated release
drugs
from endocytosed drug carriers [17]. Since the inception
comparison with liposomes without coiled coils [24]. In
of pH-sensitive NPs, myriads of innovative approaches
an attempt to develop targeted drug delivery systems
for cancer treatment have come into light. Past decades
with cancerous cell-specificity and controlled release
have witnessed synthesis and utilization of various
function inside cancer cells, Miyazaki and colleagues
pH-responsive
liposomes,
acidic
lysosomal
compartments
in
block
have designed hyaluronic acid (HA)-based pH-sensitive
polymers as multifunctional polymers. These polymers
polymer-drug conjugates, dendrimers, nanogels, and
exhibited not only pH-sensitivity but also targeting
multiple core shell complexes etc. [18]. These are
properties to cells expressing CD44 (a cancer cell
polymeric
viz.,
the
polymerosomes,
copolymers,
nanosystems
in
micelles,
briefed as follows:
surface marker). They observed that HA-derivative
I. Liposomes
modified
They are phospholipid vesicles consist of one or
more concentric lipid bilayers enclosing discrete aqueous
spaces. They can entrap both lipophilic and hydrophilic
compounds thus employed for delivering diverse range
of drugs. Moreover, its large aqueous center and
liposomes
can
be
efficiently
used
for
cell-specific intracellular drug delivery [25]. Further
research studies on the therapeutic and clinical aspects
of pH-sensitive liposomes are needed to enable their
commercial utility in gynecological cancer treatment.
II. Block Copolymers
biocompatible lipid exterior permits the delivery of
Amphiphilic block copolymers are self-assembled
different macromolecules, viz., DNA, proteins and
into polymeric micelles (10-100 nm in diameter) in
imaging agents [19]. Thus, liposomes are the most
aqueous media. These micelles possess a well-defined
common and widely sleuthed nanocarriers for targeted
hydrophobic core and a hydrophilic corona. Block
drug delivery due to their flexible physicochemical and
copolymer micelles can thus significantly improve the
biophysical
solubility of the hydrophobic drug formulated in the
properties
Doxorubucin
[19].
chemotherapeutic agent) has been observed to be
the hydrophilic end of the block copolymer, can protect
efficient
as
the micellar system from the RES elimination by
monotherapy
and
tumor-specific
liposomal
core; whereas, the densely packed corona consists of
breast
a
Pegylated
and
in
(DOX:
cancer
in
peptide
treatment
combination
both
other
reducing the interaction with serum proteins and renal
chemotherapeutics [20]. In 2016, Silva and colleagues
with
filtration [26]. The pH-sensitive block copolymers allow
have reported pH-sensitive long-circulating liposomes for
for controlled micelle dissociation and triggered drug
selective delivery of DOX into tumor [21].
release in response to the acidic pH of tumor tissue.
Karanth and
Murthy have extensively analysed previous reports on
the cytosolic delivery of the drugs through pH-sensitive
liposomes and suggested that pH-sensitive liposomes
were more efficient in delivering anti-cancer drugs than
conventional and long-circulating liposomes due to their
fusogenic
property
[22].
Recently,
a
team
of
investigators have lucidly elaborated the developmental
and applicability status of pH-sensitive liposomes in
cancer treatment and concluded it very successful as
pharmaceutical carriers for intracytoplasmic delivery of
antineoplastic
drugs
[23].
Few
investigators
have
reported pH sensitive coiled coils and their incorporation
into the liposome as triggers for the controlled release of
encapsulated drugs. From, the drug encapsulated
liposome internalization experiments with cancer cells,
The pH-sensitive polymeric micelles assembled
from hyperbranched amphiphilic block copolymer loaded
with DOX have exhibited remarkable cytotoxicity against
HeLa cells in a dose- and time-dependent manner.
Thus, proved to be a potential carrier candidate for
pH-responsive drug delivery in treating cancer [27].
Moreover, a dual pH-sensitive micelle loaded with
Paclitaxel (PTX, a chemotherapeutic agent) has been
also proved to be a potential nanocarrier for effective
metastatic
toxicity
tumor
[28].
therapy
Poly
without
(ethylene
glycol)
significant
methyl
ether acrylate-block poly(L-lysine)-block-poly(L-histidine)
triblock
co-polypeptides
were
synthesized
for
pH-responsive drug delivery. Such nanoparticles were
found to be stable at physiological pH (7.4) but were
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 37
Freely Available Online
dramatically destabilized in acidic pH due to the
triblock
presence
urethane)-block-PEG (PEG-POEU-PEG) were found to
of
pHis
blocks
[29].
The
pH-induced
copolymer
notably
release
dose-dependent
sensitivity [35]. Last year, few investigators have
cytotoxicity in murine cancer cells. YangZhang et. al.
reported a chemo-photothermo therapy of cancer cells
(2012)
by using gold nanorods (AuNRs)-based pH-sensitive
DOX,
have
followed
reported
pH-responsive
poly
ether-b-(poly
lactic
a
by
a
series
of
DOX-loaded
(ethylene
glycol)
methyl
acid-co-poly
(β-amino
esters))
thiol-ended
at
pH 5.0
triblock
due
to
ester
destabilization of the nanoparticle enabled the controlled
of
accelerated
PEG-block-poly(ortho
copolymer
(PAA-b-PDMAEMAQ-b-PCL-SH),
in
its
pH
micelles
which
AuNRs
at
(MPEG-b-(PLA-co-PAE)) block copolymer micelles as
polymer was loaded with Methotrexate (MTX) as an
drug delivery carriers for targeted cancer therapy with
anticancer drug [36].
sustained release [30]. Investigations carried out by
III. Polymeric Micelles
Zhou et. al. (2015) have suggested that the polymeric
micelles comprising of polyethylene glycol (PEG) and a
polymethacrylamide [PEG-b-PMEA] diblock copolymer
could be useful for pH-responsive delivery of poorly
soluble
anticancer
drugs
[31].
The
pH-sensitive copolymer viz., methoxy poly(ethylene
glycol)-b-poly(hydroxypropyl
methacrylamide-g-α-
tocopheryl succinate-g-histidine) (PTH) forming micelles
in aqueous solutions were used for co-delivery of
therapeutic agents, DOX and α-TOS (α-tocopheryl
succinate) in tumor cells. In this combination therapy,
the micelles enabled the rapid release of both Dox
and α-TOS when the pH declined from 7.4 to 4.5 in
tumor tissues [32]. Mozhi and colleagues have displayed
a synergistic antitumor effect of the combination of
anticancer drug Docetaxel and the therapeutic peptide
[D(KLAKLAK)2] in an MCF-7 cell line using a pH-sensitive
copolymer
viz.,
poly(β-amino
esters)-poly(ethylene
glycol) conjugated with the dual-targeting proapoptotic
peptide CGKRKD(KLAKLAK)2. In which, CGKRK peptide
efficiently
transported
mitochondria
apoptosis
to
[33].
D(KLAKLAK)2
trigger
Few
towards
mitochondria-dependent
investigators
have
reported
synthesis of pH-sensitive copolymer through bridging
poly (2-methacryloyloxyethyl phosphorylcholine) (PMPC)
block and poly (D, L-lactide) (PLA) block by a benzoyl
imine
linkage
(Blink).
These
biomimetic
micelles
(PLA-Blink-PMPC) were prepared as carriers for PTX
delivery. Such pH-triggered drug release behaviour in
synchronization with tumoral acidic conditions was found
to be helpful for improving the utilization of drug and
facilitating antitumor efficacy [34]. Furthermore, Wang
and colleagues have exhibited antitumor efficiency of
DOX-loaded micelles. In which, ortho ester degradation
of DOX-loaded, pH-sensitive micelles consisted
of
They are self-assembling nano-constructs of
amphiphilic copolymers and are widely regarded as
efficient nano-carriers for myriads of applications,
including drug delivery, diagnostic imaging etc. These
became feasible because of their variety of favorable
properties viz., biocompatibility, bioavailability, capacity
to effectively solubilize myriads of poorly soluble drugs,
enhancing
release
profile
of
the
incorporated
pharmaceutical entities, ability to accumulate in the
targeted tissue based on the EPR effect and ability to
attach various targeting ligands to the micellar surface.
The combination of these approaches have been found
to
further
improve
specificity
and
efficacy
of
micelle-based drug delivery to promote the development
of smart multifunctional micelles [37]. Ko and colleagues
have evaluated anti-tumor activity of pH-responsive
polymeric micelles made up of methyl ether poly
(ethylene glycol) (MPEG)-poly(β-amino ester) block
copolymers, by injecting the DOX-loaded polymeric
micelles into tumor-bearing mice. These micelles notably
suppressed tumor growth and prolonged survival of the
tumor-bearing mice, compared with mice treated with
free DOX [38]. Giacomelli and coworkers have reported
pH-triggered micelles composed of a pH-responsive
PDPA
[poly(2-diisopropylamino)
ethyl
methacrylate]
inner core and a PEO [poly(ethylene oxide)] outer
shell
as
a
promising
drug
delivery
system
for
the cancer therapy. In which, pH-responsive PDPA
core was loaded with PTX [39]. In vivo evaluation of
DOX-loaded
pH-sensitive
polymeric
micelles
made up of poly(L-histidine-co-L-phenlyalanine-b-PEG
and poly(L-lactic acid)-b-PEG-folate was carried out in
multidrug-resistant (MDR) ovarian tumor-xenografted
mice. It was observed that the drug-carrying micelles
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 38
Freely Available Online
were exhibiting enhanced intracellular DOX-delivery by
pH-sensitive drug delivery nanosystems. Uthaman and
circulating for long-time (i.e., enhanced bioavailability)
his
team
(2018)
have
reported
a
variety
of
and accumulating at tumor-selective sites. Thus, they
pHis-based polymeric micelles for the delivery of
exhibited enhanced cytotoxicity to tumor cells only,
DOX [15, 29, 46-48]. In addition to these, nanocarriers
sparing the normal healthy cells [40]. Wang and
composed
colleagues
loaded
polymeric micelles were used to deliver Nonsteroidal anti
glycol)-b-poly
-inflammatory drug (NSAID) Ibuprofen in breast cancer
[PELA-PBAE]
therapy. It was observed to possess potential anti-tumor
have
shown
pH-responsive
that
PTX
Poly(ethylene
(D,L-lactide)-b-poly(β-amino
micelles
the
might
tissues [49]. The pH-responsive nanoparticles combining
the
cisplatin
Ibuprofen with chemotherapy agents have provided a
between
novel
micelles
incorporated
by
poly
with
complexation
cis-dichlorodiammineplatinum(II)
hydrophilic
utility
(CDDP)
(L-glutamic
(2-methacryloyloxyethyl
in
chitosan
metastatic breast tumor therapy [41]. In another study,
prepared
potential
biocompatible
activity, while avoiding side-effects on normal, healthy
polymeric
the
amphiphilic,
the
were
have
ester)
of
and
(PLG-b-
PMPC) diblock copolymers. Investigators observed the
sustained release of CDDP from the micelles was faster
in acidic pH (5.0 - 6.0) than the physiological pH 7.4.
Thus, CDDP-loaded polymeric micelles were developed
for targeted cancer therapy to reduce the detrimental
side
effects
of
cisplatin
CDDP
[42].
Zhou
and
coworkers have reported a pH-responsive pentablock
copolymer
made
up
of
1,2-distearoyl-sn-glycero-3-
phosphoethanolamine-N-[methoxy(polyethylene glycol)]
conjugated poly(β-amino esters) (DSPE-b-PEG-b-PAE-bPEG-b-DSPE) was able to self-assemble into polymeric
micelles. These DOX-loaded polymeric micelles displayed
pH-triggered high toxicity to tumor cells and HeLa cell
lines whereas the copolymer had negligible cytotoxicity.
Thus, these pH-sensitive micelles have the potential to
be used for cancer chemotherapy with controlled
release [43]. Das and his team, and Zhou and
colleagues have lucidly elaborated the current advances
in the development of pH-responsive polymeric micelles/
nanoparticles, their mechanisms of action, applications
in
chemotherapy,
delivery
strategies
diagnostic
and
imaging,
provided
their
their
future
perspectives [44, 45]. This is yet to be sleuthed in
human cells and gynecological tissues.
It is well understood that, among amino acids,
Histidine (His) is an only essential amino acid having
imidazole group. The presence of lone-pair electrons on
the
unsaturated
nitrogen
of
this
group
confers
pH-sensitivity to Histidine. Therefore, poly(Histidine)
(pHis) has been extensively used for the fabrication of
system
for
both
primary
and
metastatic tumor treatment [50].
acid)-b-poly
phosphorylcholine)
nanoparticle
Targeted
efficient
delivery
and
therapeutic
efficacy of DOX have been found to be significantly
increased by using a stepwise pH-responsive nanodrug
delivery system [51]. This study has provided a
promising
strategy
for
efficient
delivery of other
antitumor agents. Similarly, some more nanocarriers like
polymeric micelles, liposomes and solid NPs have been
developed for hydrophobic as well as hydrophilic drugs
for effective therapy of cancer [52-56]. Last year, some
investigators
have
revealed
the
intracellular
pH-responsive nanoparticles of hyaluronic acid which
can provide insights into the design of potential
prodrugs for the cancer therapy [57]. Hydroxyapatite
coated iron oxide nanoparticles and pH sensitive Sodium
alginate have been developed for controlled release of
hydrophobic drugs [58]. Yandan and his colleagues have
shown
that
multifunctional
sharp
pH-responsive
nanoparticles made up of poly(2-(diisopropylamino)
ethylmethacrylate) (PDPA) polymer have great potential
to serve as a new generation nanomedicine for effective
breast cancer treatment [59].
IV. Polymersomes or Polymeric Vesicles
They are preferably prepared from amphiphilic,
biocompatible and biodegradable polymers [60]. They
have the potential to be versatile drug delivery systems
because of their tunable membrane formulations,
stabilities in vivo, various physicochemical properties,
controlled release mechanisms, targeting abilities, and
capacities to encapsulate varieties of drugs etc. [61, 62].
The pH-sensitive polymersomes have been developed to
quickly respond to small changes in the environmental
pH of tumor’s microenvironment [60, 63]. Following pH
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 39
Freely Available Online
alteration, the pendant acidic (carboxylic acids/sulfonic
multifunctional dendrimer-based theranostic nanosystem
acids) or basic groups (amine) undergo protonation or
(in which Gold nanoparticles conjugated with DOX were
deprotonation. Consequently, the structural transition
entrapped in dendrimer) to be utilized for simultaneous
induces formation/deformation of polymeric vesicles,
chemotherapy and computed tomography (CT) imaging
which confers a higher therapeutic index as a result of
of various types of cancer cells [69]. Dendritic polyester
the fast release of therapeutics at the target site.
system based on monomers 2,2-bis (hydroxymethyl)
However, the major obstacle to the application of
propanoic acid attached to DOX or hydroxyl-terminated
pH-sensitive polymersomes is the slow response to the
generation 4 PAMAM in conjugation with PTX through a
stimulus, resulting in a slow drug release, which
union with succinic acid have shown great anticancer
eventually induce drug resistance in the adjacent cells.
activity against ovarian cancer cells [65]. However, at
Therefore, polymersomes need to respond quickly as a
present the dendrimers used as drug-carriers do not
result of the decreased pH at pathological sites. Thus,
satisfactorily meet the necessary characteristic of an
pH-responsive
further
ideal dendrimer for targeted drug delivery. However, the
designed to carry, deliver, and control the release of
development and study of new dendrimers drug-carriers
therapeutic agents to the tumor tissue by relying the low
continues to be an important tool in the cancer therapy.
pH in the vicinity of tumor tissues [60, 63]. Anajafi and
(b) Acid-responsive polymers have provided enhanced
Mallik
polymersomes
(2015)
have
need
explicitely
to
be
elaborated
recent
developments of polymersomes [60]. Their utilities in
treating gynaecological cancers are yet to be sleuthed.
cis-aconityl linkers are two types of acid-sensitive linkers
(a) Dendrimers or dendritic molecules are highly
branched with a central core, nanosized, symmetric
with
well-defined,
homogenous
and
monodisperse structure with diameter 2-10 nm. They
are classified by its form as polymers, hyperbranched
polymers
or
brush-polymers
and
also
classified
by their molecular weight as low or high molecular
weight [64, 65]. Dendrimer act as a carrier for the
delivery
of
drug
to
tumor
by
encapsulation
or
conjugation. Among polymer-drug conjugates, most
widely
studied
dendrimers
non-biodegradable,
poly-amidoamine
to
cationic
(PAMAM)
date
are
amine-terminated
dendrimers
[66].
Drug
delivery to tumor site is mostly accomplished through
PAMAM, poly(propylene imine) [PPI], and poly(L-lysine)
[PLL]
dendrimers
by
either
passive
or
active
targeting [64]. Wen and colleagues have explained the
multifunctional dendrimer-modified multi-walled carbon
nanotubes for targeted and pH-responsive delivery of
DOX into various types of cancer cells [67]. In 2018,
Zhang and coworkers have reported the development of
pH-sensitive multifunctional DOX-conjugated PAMAM
dendrimers as a unique platform for targeted cancer
chemotherapy [68]. Some other investigators have
presented
the
environment, acid-sensitive linkers have provided tools
for targeted intracellular drug release. Hydrazone and
V. Polymer-Drug Conjugate
molecules
endosomal delivery of drugs. In the acidic micro
construction
of
pH-responsive
which have been commonly used for this purpose. Both
are relatively stable at physiological pH and can release
the bound drugs only under low pH conditions. The
hydrazone linker gets rapidly cleaved under low pH
conditions (which occur in endosomes, lysosomes and
tumor tissue). Through the hydrazone linker, the drug
was
found
to
be
released
in the
acidic tumor
microenvironment or in the acidic organelles after
cellular uptake by endocytosis [70-75]. Some more
sleuthed
polymer-drug
conjugates
containing
hydrazone linkages are HPMA-DOX, PEG-DOX [76, 77],
PEG-epirubicin [78] and PEG-PTXL [79]. Hydrazone
linked
acid
sensitive
PEG-based
drug
delivery
in
lysosomes was also studied by Zhu et al., 2012 [74].
They
found
that
Gemcitabine
(GemC18)
in
the
acid-sensitive micelles was more toxic toward cancer
cells than acid-insensitive micelles. There are reports of
a pH-sensitive hydrazone bridged and peptide-guided
prodrug
incorporating
DOX
for
targeted
ablation
(removal of harmful parts of the body) of cancer cells
with least cytotoxicity on normal healthy cells [80]. The
pH-sensitive N-(2-hydroxypropyl) methacrylamide-DOX
(HPMA-DOX) conjugates bearing an acid-responsive
hydrazone linker in their structure have also been widely
studied as anticancer drug delivery systems. They have
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 40
Freely Available Online
been found to significantly increase therapeutic efficacy
in different in vitro and in vivo cancer models. Liao
were constructed by encapsulating a photothermal dye
(IR 825) in the carbonized zwitterionic polymer. Before
et al. [57] have reported the synthesis of tumor
accumulating in the tumor site, these nanoparticles
targeting and pH-responsive nanoparticles for the
displayed
enhanced delivery of DOX. The nanoparticles were
hydrophobic interaction with neutral pH and π-π
prepared through the covalent bonding of DOX to
stacking. The slight change in the pH in TME enabled
hyaluronic acid (HA) backbone by hydrazone linkage. In
the charge of the nanoparticles to be altered, leading to
aqueous
acid-hydrazone
the release of IR 825 and recovered fluorescence. These
linkage-doxorubicin (HA-hyd-DOX) could self-assemble
types of nanoparticles can simultaneously be used for
into nanoparticles. Active targeting of the nanoparticles
diagnosis and photothermal therapy [90].
was achieved through receptor-mediated binding of HA
VI. Multiple Core Shell Complexes
solution,
hyaluronic
to CD 44, which are overexpressed in most cancer cells.
Studies on polymers that use cis-aconityl linker in
designing anticancer drug delivery systems included
HPMA-DOX
[81],
polyamidoamine
acid-sensitive
polyamidoamine-DOX
(PAMAM)-DOX
cis-aconityl
[83].
linked
[82]
and
Furthermore,
Polyehtylene
glycol-Chitosan (PEG-CS) micelles were found to have a
greater Docetaxel loading capacity, less cytotoxicity
toward normal cells, enhanced cellular uptake and better
accumulation
in
tumor
tissue
compared
to
acid-insensitive PCS micelles (PEG directly linked to
CS) [84]. Moreover, the pH-responsive NPs have also
been developed by conjugating nanocarriers with some
other acid-labile linkages such as orthoester [85, 86],
imine [87, 88], phosphoramidate [89], whose hydrolysis
ensured rapid release of the drug at the targeted tumor.
(c) Zwitterionic polymers incorporated nanoparticles
have been designed to demonstrate a pH-dependent
change in surface charge. One of the most commonly
investigated systems is based on zwitterionic polymers,
as they have cationic and anionic groups that control
surface charge in response to pH. In acidic pH, these
zwitterionic polymers have a positive charge, and in
basic pH, they have a negative charge. However, when
these zwitterionic polymers are in neutral pH, they are
overall neutral with balanced populations of positive and
negative
components
and
they
become
more
hydrophobic. However, upon entering tumor cells, the
balance between positive and negative charges alters
and thereby cause conformational changes, facilitating
drug release in tumor cells. Kang and colleagues have
reported the fabrication of tumor microenvironment
responsive
theragnostic
with
a
pH-dependent
fluorescence turn on/off property. The nanoparticles
quenching
of
fluorescence
The pH-responsive
due
to
the
drug encapsulation and
release from multiple core-shell nanoparticles become
feasible
due
to
the
presence
of
polyelectrolyte
multilayers [91]. Huang and coworkers [92] first
Gd2O3:Yb3+:Er3+,
synthesized
a
functionalized
mesoporous silica nanoparticle core, which was then
coated by multi-layers of polyelectrolytes. DOX was then
loaded onto the polyelectrolyte shell. The resulting
DOX-loaded core-shell nanoparticles exhibited more than
60% DOX release within 72 h at pH 5.2. In vitro
cytotoxicity studies on MCF-7 breast cancer cells showed
that
DOX-loaded
nanoparticles
exhibited
higher
cytotoxicity than the free DOX. Tian et al. [93]
synthesized an azide-terminated diblock copolymer from
oligo(ethylene
glycol)methyl
ether
methacrylate
(OEGMA), 2-(diisopropylamino)ethyl methacrylate (DPA),
and
glycidyl
methacrylate
(GMA).
The
resulting
copolymer was then functionalized with DOTA(Gd) and
4-(prop-2-ynyloxy)
benzaldehyde
and
the
resulting
copolymers were further co-assembled into mixed
micelles. The presence of GMA moieties inside the cores
enabled encapsulation of tetrakis[4-(2-mercaptoethoxy)
phenyl]ethylene (TPE-4SH), and thus the resulting
micelles
dual
were
capable
imaging.
surface-conjugated
of
MR
Moreover,
with
these
pH
low
and
fluorescence
micelles
insertion
were
peptide
(pHLIP), which enabled them for selective targeting
toward tumor tissues and in situ Camptothecin (a cancer
drug) release, confirmed by in vivo MR images of
tumor-bearing BALB/c nude mice. Ray et. al. (2018)[91]
and their collaborators have synthesized a unibody
core-shell (UCS) nanoparticle using a polymer platform
formed
by
resorcinol
and
1,3-phenylenediamine
monomers. In this synthesis, Gd3+was first conjugated
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 41
Freely Available Online
to the polymer backbone to form the Gd-core, and then
and the coating layer of layered double hydroxide (LDH)
DOX was encapsulated within the shell surrounding the
was used as a storehouse for MTX] as MTX delivery
Gd-core.
the
system for targeted anticancer therapy. They have
components in the core. 1,3-phenylenediamine was
Resorcinol
observed its excellent pH-sensitivity and ~85% of MTX
chosen
was released within 48h at pH 3.5 via the co-effect of
as
the
was
shell
chosen
unit
for
as
its
one
of
capability
for
pH-controllable release. In vitro and in vivo studies of
dissolution of LDH layer and ion-exchange. This study
UCS-Gd-DOX as an innovative theranostic nanoparticle
has revealed that the Fe3O4@LDH-MTX would be a
showed that the DOX in the shell is effectively and
competitive candidate for sustained, controlled release
selectively
and targeted delivery of MTX because Fe3O4@LDH-MTX
released
in
tumor
acidic
environments
(pH 5.5). In vitro pH-dependant release of DOX after 2 h
exhibited high anticancer activity with minimal toxicity to
was found to be <5%, 10%, 55%, 75%, and 80%, at
normal cells [97]. In addition to these reports, a
pH 8.0, 7.0, 6.0, 5.0, and 4.0, respectively. Enhanced
pH-responsive nanoplatform made up of a yolk-like
drug release from pH 7.0 to 6.0 verified the potential of
Fe3O4@Gd2O3 and functionalized by PEG and folic acid,
UCS-Gd-DOX for targeted therapy towards malignant
has been documented to be a potential nanotheranostic
tumor tissues. In addition, in vitro T1-weighted MR
for tumor targeted T1-T2 dual-mode Magnetic Resonance
imaging studies also reflected the pH-switchable MR
Imaging
contrast capability of UCS-Gd-DOX. The pH-responsive
HeLa cells [98]. Last year, pH-sensitive magnetic
design of the UCS nanoparticle not only improved the
composite
MRI contrast at the tumor site with respect to other
water-in-oil-in-water (W/O/W) emulsion using acetylated
tissue/organs, but also successfully suppressed growth
β-cyclodextrin as a pH-sensing material and Fe3O4 as a
and
chemotherapy
nanoparticle
using
was
Cisplatin
prepared
by
and
double
of subcutaneous human cervical cancer in mouse
component to realize magnetic response. Its in vitro
xenograft models. Therefore, theranostic nanoparticles
evaluation was performed for drug loading and release
with Gd-conjugation and DOX-doping can be synthesized
behaviour [99]. This type of study can be also extended
and further applications of UCS-Gd-DOX in the field of
for treating gynecological tumors. In a review, Lungu
cancer treatment can be anticipated [91].
and her colleagues have explicitely elucidated the utility
There are reports of pH-sensitive magnetic
of pH responsive core-shell magnetic nanoparticles in
delivery. In early years of this decade, a magnetic and
NPs were: magnetite@silicon dioxide (Fe3O4@SiO2),
nanoparticles sleuthed for targeted anticancer drug
pH dually responsive nanocarrier with a multilayer
core-shell architecture was constructed. In which, the
Fe3O4@SiO2 nanoparticles acted as a superparamagnetic
core used to target the drug loaded nanocarriers to the
pathological site. Meanwhile, the mPEG [α-methoxy poly
(ethylene glycol)] and PBLA [poly(benzyl-L-aspartate)]
segments served as a pH-sheddable hydrophilic corona
and a hydrophobic middle layer used to load the drug
DOX via hydrophobic interactions. This system appeared
to be highly promising for the targeted intracellular
delivery of hydrophobic chemotherapeutics in cancer
therapy [94, 95]. In 2017, Karimi and his colleagues
have
brought
in
light
a
pH-sensitive
magnetic
nanoparticle system for Methotrexate (MTX) targeting of
tumor [96]. In another study, Wu and his colleagues
have
performed
in
vitro
evaluation
of
magnetic
nanocomposites (Fe3O4@LDH-MTX) [in which Fe3O4
nanoparticles acted as magnetically responsive carriers
diagnosis and treatment of oncological diseases. Those
Fe3O4@titanium
dioxide
(TiO2),
beta-thiopropionate-
polyethylene glycol (PEG)-modified Fe3O4@mSiO2, Fe3O4
NPs core coated with SiO2 with an imidazole group
modified
PEG-polypeptide
polyacrylic
acid
(PAA)
the
oxide
NP
iron
(mPEG-poly-L-Asparagine),
and
core,
folic
acid
methoxy
glycol-block-polymethacrylic
coating
of
polyethylene
acid-block-polyglycerol
monomethacrylate (MPEG-b-PMAA-b-PGMA) attached by
a
PGMA
block
to
a
Fe3O4
core,
PEG-modified
polyamidoamine (PAMAM) dendrimer shell with Fe 3O4
core and mesoporous silica coated on Fe3O4, mostly
coated with an anticancer drug and used for controlled
release of cytostatic drugs into the tumor site by means
of pH change [100].
VII. Nanogels
They are three-dimensional, water soluble,
cross-linked hydrogel materials in the nanoscale size
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 42
Freely Available Online
range with a high loading capacity for guest molecules
Discussion and Conclusion
and act as drug carrier systems [101]. Nanogels are the
novel drug delivery systems for both hydrophilic and
hydrophobic drugs [102]. There are some anti-tumor
drugs
viz.,
Cisplatin,
Temozolomide
incorporation
etc.
DOX,
used
through
5-Flurouracil,
in
cancer
nanogels.
Heparin,
therapy
by
pH-
and
The
temperature-responsive nanogels made up of maleic
acid poly-(N-isopropylacrylamide) polymer loaded with
DOX have been frequently employed in the cancer
treatment, where DOX is delivered at a specific pH and
temperature. Chitin-polymerized DOX nanogels have
been also used for treatment of breast cancer [103].
Several nanogel formulations used in cancer therapy are
listed elsewhere [104]. The pH-sensitive PEGylated
nanogel loaded with anti-tumor drug has proved to be a
promising nano-sized carrier for anticancer drug delivery
systems against the human breast cancer cell line
MCF-7 [105]. Bardajee and colleagues have prepared a
thermo-/pH-sensitive
modified
graphene
nanogels
oxide
comprising
(SMGO)
with
salep
branched
N-isopropylacrylamide (NIPAM) and acrylic acid (AA).
Doxorubicin loaded SMGO/P(NIPAM-co-AA) nanogels
showed
exhibited
thermo-/pH-dependent
enhanced
toxicity
to
drug
release
HeLa
cells
and
when
compared to the equivalent dose of the free drug [101].
A synergistic combined chemo-radioisotope therapy of
cancer using a pH-dependent hybrid nanogel (hydrogel
nanoparticle) platform based on the self-assembly of
carboxymethyl cellulose and bovine serum albumin
was
reported
[106].
The
pH
sensitive
polymeric
nano-hydrogels attached with an ionizable weak acidic
or basic moieties, cationic polymeric polyethylenimine
(PEI), polymeric nano-micelles of pH-responsive natural
polymers like albumin and gelatin have also been used
as drug delivery systems for treating
varied cancers
[107]. Peng Wei and colleagues (2018) have synthesised
a
pH-sensitive
nanogels
by
using
N-[(2,2-dimethyl-1,3-dioxolane)methyl]
a
monomer
acrylamide
(DMDOMA) bearing an acid cleavable acetal group.
These seemed to be a promising and conveniently
prepared alternative to existing carrier systems for drug
delivery
[108]. Thus, nanogels seem to be potential
candidate in the development of new nanocarriers for
anti-cancer drug delivery. So they can be further
investigated for treating gynecological cancers.
Despite
considerable
research in the
past
decades and plethora of positive results in the preclinical
studies,
the
clinical
translation
of
pH-sensitive
nanosystems assisted drug delivery platforms has not
progressed incrementally. Some of the facts which
appear to be obstacles, seem to hinder the progress are:
(i) The differences in pH between normal and tumor
tissues are not significant enough for generating the
pH-responsiveness. Moreover, pH-sensitive nanoparticles
remain
non-responsive
in
the
perivascular
region
because the acidic pH need for responsiveness is found
in region far from the blood vessels [29]. (ii) In addition,
selecting a polymer with a critical pH that matches the
desired pH range for its application is a major factor in
designing
an
ideal
pH-sensitive
system.
Thus,
understanding the chemical structure of the polymer’s
ionizable
moieties,
indispensable
appropriate
and
for
their
the
respective
design
pH-sensitive
and
DDS
pKa
synthesis
[109].
are
of
Moreover,
attempts have been made to alleviate much concerned
cytotoxicity of synthesized NPs by conjugating it
with
PEG
or
with
any
of
the
zwitterionic
polybetaines [110-114]. Further studies are still on to
nullify its cytotoxicity, if any. Considering all these, it
becomes
utmost
important
nanotechnological
to
advancement
understand
in
the
biomedical
applications to date and the challenges that still need to
be overcome. That will allow future research to improve
on existing pH-sensitive nanoplatforms and to address
the current translational and regulatory limitations.
Continued translational success will require coordinated
communication
and
collaboration
between
experts
involved in all stages of pharmaceutical development of
pH-sensitive
drug
pharmaceutical
delivery
design,
nanosystems,
including
manufacturing,
cellular
interactions and toxicology, as well as preclinical and
clinical evaluation.
In
other
elsewhere
all
of
the
pH-responsive
[50,
107,
aforementioned
nanosystems
115-117],
the
and
reported
conventional
nano-carriers have been combined with pH-responsive
systems that release drug content only under specific
acidic pH. Table 1 summarises all of the pH-sensitive
nanosystems reviewed in the present text having
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 43
Freely Available Online
Table 1. pH-Responsive Nanosystems utilized in gynaecological Cancer treatment
potential utilities in treating gynaecological Cancers.
of active compounds: concepts and applications. Adv
However, some of the systems discussed here are yet to
Drug Deliv Rev 2012; 64: 866-884.
be investigated for gynaecological cancers. But all of
them
seem
to
successfully
have
delivering
tremendous
drugs
at
potential
the
for
targeted
gynaecological tumor sites/ tissues, as the case may be.
Therefore, it will not be an exaggeration to state that
the pH sensitive nanomedicine could turn to be an
unique system for treating gynecological cancers (and
other cancers, as well), if developed and delivered with
utmost care.
Q,
Katti
nanomaterials
PS,
Gu
Z.
Enzyme-responsive
for
controlled
drug
delivery.
Nanoscale 2014; 6: 12273-12286.
2. Khadka P, Ro J, Kim H. Pharmaceutical particle
technologies:
an
approach
to
improve
drug
solubility, dissolution and bioavailability. Asian J
Pharm Sci 2014; 9: 304-316.
3. Toh
MR,
preparations
intracellular delivery. Biophys Rev. 2017 Dec; 9(6):
931-940.
6. Lehner R., Wang X., M. Wolf. Designing switchable
nanosystems for medical application. J Control
Release 2012; 161: 307-316.
7. Chen Q., Ke H., Dai Z. Nanoscale theranostics for
physical
stimulus-responsive
cancer
therapies.
Biomaterials 2015; 73: 214-230.
References
1. Hu
5. Tayo Lemmeul L. Stimuli-responsive nanocarriers for
Chiu
and
GNC.
Liposomes
limitations
of
as
sterile
sterilisation
techniques in liposomal manufacturing. Asian J
Pharm Sci 2013; 8: 88-95.
4. Fleige E, Quadir MA, Haag R. Stimuli-responsive
polymeric nanocarriers for the controlled transport
8. Camara AL, Figuero Longo JRSJP. pH-Sensitive
Nanoparticles for Cancer Therapy: Is this a Real
Innovation in Nanomedicine? Nano Res Appl 2017;
3: 1-2.
9. Frenkel V. Ultrasound mediated delivery of drugs
and genes to solid tumors. Adv Drug Deliv Rev.
2008; 60: 1193-208.
10. Liu YC, Le Ny AL, Schmidt J, Talmon Y, Chmelka BF,
Lee CT Jr. Photo-assisted gene delivery using
light-responsive catanionic vesicles. Langmuir. 2009;
25: 5713-24.
11. Sun X, Zhang G, Wu Z.
Nanostructures for
pH-sensitive Drug Delivery and Magnetic Resonance
Contrast Enhancement Systems. Curr Med Chem.
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 44
Freely Available Online
2018; 25: 3036-3057.
doxorubicin into tumors: A feasibility animal study.
12. Sun Hao, Christopher P. Kabb, Michael B. Sims,
Brent
S.
Sumerlin.
Architecture-transformable
Mol Imag Biol. 2016; 18: 898-904.
22. Karanth Hamsaraj, Murthy R S Rayasa. pH-Sensitive
polymers: Reshaping the future of stimuli-responsive
liposomes-principle
polymers. Progress in Polymer Science, 2018; 89:
therapy. J Pharm Pharmacol 2007; 59: 469-483.
61-75.
and
application
in
cancer
23. Ferreira Diego dos Santos, Sávia Caldeira de Araújo
13. GaoW, Chan JM, Farokhzad OC. pH-responsive
Lopes, Marina Santiago Franco & Mônica Cristina
nanoparticles for drug delivery. Mol Pharm. 2010; 7:
Oliveira. pH-sensitive liposomes for drug delivery in
1913-1920.
cancer treatment. Therapeutic Delivery, 2013; 4:
14. Wang
X-Q,
Zhang
Q.
pH-sensitive
polymeric
nanoparticles to improve oral bioavailability of
1099-1123.
24. Reja Rahi M.,
Mohsina Khan,
Sumeet K. Singh,
peptide/protein drugs and poorly water-soluble
Rajkumar Misra, Anjali Shiras and Hosahudya N.
drugs. Eur J Pharm Biopharm 2012; 82: 219-229.
Gopi.
15. John JV, Uthaman S, Augustine R, Chen HY, Park IK,
Kim I. pH/redox dual stimuli-responsive sheddable
nanodaisies
for
efficient
intracellular
pH sensitive coiled coils: a strategy for
enhanced liposomal drug delivery. Nanoscale 2016;
8: 5139-45.
tumour
25. Miyazaki Maiko, Eiji Yuba , Hiroshi Hayashi, Atsushi
triggered drug delivery. J Mater Chem B. 2017; 5:
Harada, and Kenji Kono. Hyaluronic Acid-Based
5027-36.
pH-Sensitive
16. Xin Yanru, Yin Mingming, Zhao L., Meng F., Luo L.
Recent progress on nanoparticle-based drug delivery
systems for cancer therapy. Cancer Biol. Med. 2017;
14: 228-242.
Polymer-Modified
Liposomes
for
Cell-Specific Intracellular Drug Delivery Systems.
Bioconjugate Chem., 2018; 29: 44-55.
26. Li Wei, Feng S.S., Guo Y. Block copolymer micelles
for nanomedicine. Nanomedicine 2012; 7: 169-172.
17. Kopansky E, Shamay Y, David A. Peptide-directed
27. Hongliang Cao, Chao Chen, Debiao Xie, Xin Chen,
HPMA copolymer-doxorubicin conjugates as targeted
Ping Wang, Yibing Wang,
therapeutics for colorectal cancer. J Drug Target
Wenxin Wang. A hyperbranched amphiphilic acetal
2011; 19: 933-42.
polymer for pH-sensitive drug delivery. Polym.
18. Bazban-Shotorbani
S,
Hasani-SadrabadiMM,
Karkhaneh A, Serpooshan V, Jacob KI, Moshaverinia
A,
Mahmoudi
Chem. 2018; 9: 169-177.
28. Tang S, Meng Q, Sun H, Su J, Yin Q, Zhang Z, Yu H,
Chen L, Gu W, Li Y. Dual pH-sensitive micelles with
relationship of pH-responsive polymers for drug
charge-switch for controlling cellular uptake and
delivery applications. J Control Release 2017; 253:
drug release to treat metastatic breast cancer.
46-63.
Biomaterials 2017; 114: 44-53.
Lisa,
Revisiting
and
structure-property
19. Sercombe
M.
Huajie Songa
Tejaswi
Veerati,
Fatemeh
29. Uthaman S, Kang Moo Huh, and In-Kyu Park. Tumor
Moheimani, Sherry Y. Wu, Anil K. Sood, and Susan
microenvironment-responsive
Hua. Advances and Challenges of Liposome Assisted
cancer theragnostic applications. Biomater. Res.
Drug Delivery. Front Pharmacol. 2015; 6: 286-294.
2018; 22: 22-28.
nanoparticles
for
20. Park John W. Liposome-based drug delivery in
30. YangZhang Can, You QiangYang, Tu XiongHuang,
breast cancer treatment. Breast Cancer Research
BinZhao, Xin DongGuo, Ju FangWang, Li JuanZhang.
2002; 4: 95-102.
Self-assembled pH-responsive MPEG-b-(PLA-co-PAE)
21. Silva JO, Fernandes RS, Lopes SC, Cardoso VN, Leite
EA, Cassali GD, Marzola MC, Rubello D, Oliveira MC,
de
Barros
liposomes
AL.
as
an
pH-sensitive,
alternative
long-circulating
tool
to
deliver
block
copolymer
micelles
for
anticancer
drug
delivery. Biomaterials. 2012; 33: 6273-6283.
31. Zhou X, Luo S, Tang R, Wang R, Wang J. Diblock
copolymers
of
polyethylene
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
glycol
and
Vol-2 Issue 2 Pg. no.- 45
a
Freely Available Online
polymethacrylamide with side-chains containing twin
ortho ester rings: synthesis, characterization, and
evaluation as
potential
pH-responsive
micelles.
Macromol. Biosci. 2015; 15: 385-94.
Chiang,
Lu-Yi
Yu,
Yao-An
Shen
and
Chun-Liang Lo. A pH sensitive polymeric micelle for
co-delivery of doxorubicin and α-TOS for colon
cancer therapy. J. Mater. Chem. B, 2017; 5:
5870-5880.
33. Mozhi
and
You
Han
Bae.
folate
Israr
Okeke,
receptors
and
of
targeting
endosomal
pH
in
41. Wang Jie, Gejing De, Qiaoxin Yue, Hai Ma, Jintang
Cheng, Guangwei Zhu, Maobo Du, Hong Yi, Qinghe
Ahmad,
Chan
Li,
Chukwunweike
and
Xing
JL.
Micelles Enhances Inhibitory Efficacy on Metastasis
of Murine Breast Cancer Cells. Front. Pharmacol.,
of proapoptotic peptide and anticancer drug for
fphar.2018.00543.
cancer
therapy.
Royal
Soc.
Chem.
Advances 2017; 7: 12886-12896.
pH-sensitive
polymeric
micelle
https://doi.org/10.3389/
42. Zhuang W, Ma B, Liu G, Chen X, Wang Y. A fully
absorbable biomimetic polymeric micelle loaded with
34. Ma B, Zhuang W, Liu G, Wang Y. A biomimetic and
as
carrier
for
paclitaxel delivery. Regen. Biomater. 2018; 5(1):
15-24.
cisplatin as drug carrier for cancer therapy. Regen
Biomater. 2018; 5(1): 1-8.
43. Zhou Xin Xin, Long Jin, Rui Qun Qi, and Teng Ma.
pH-responsive polymeric micelles self-assembled
35. Wang J, Lu Y, Li S, Wang X, Huang Y, Tang R.
pH-sensitive
amphiphilic
triblock
copolymers
containing ortho ester main-chains as efficient drug
delivery platforms. Mater Sci Eng C Mater Biol Appl.
2019; 94: 169-178.
Mortaza Mirzaei. A novel gold nanorods-based
pH-sensitive
thiol-ended
triblock
copolymer
for
chemo-photothermo therapy of cancer cells. Journal
of Biomaterials Sci., Polymer Edition. 2019; 30:
12-33.
37. Movassaghian
S,
Merkel
OM,
Torchilin
VP.
Applications of polymer micelles for imaging and
drug delivery. Wiley Interdiscip Rev Nanomed
Nanobiotechnol. 2015; 7(5): 691-707.
as doxorubicin delivery carriers. Royal Society Open
Sci. 2018; 5: 171654-171659.
44. Das Aparesh, N. Vishal Gupta, D. V. Gowda, Rohit R.
Sang Kim. Tumoral acidic extracellular pH targeting
of pH-responsive MPEG-poly(β-amino ester) block
copolymer micelles for cancer therapy. Journal of
Controlled Release. 2007; 123: 109-15.
Schmidt,
Eliézer
Jäger,
Alessandro Jägerb and Karel Ulbrichb. pH-triggered
on
a
pH-responsive
Journal of ChemTech Research. 2017; 10: 575-588.
45. Zhou Q, Zhang L, Yang T, Wu H. Stimuli-responsive
polymeric micelles for drug delivery and cancer
therapy. www.dovepress.com. 2018: 13; 2921-2942.
46. Johnson RP, Saji Uthaman, Rimesh Augustine, Yu
Zhang, Hua Jin, Chang In Choi, In-Kyu Park, Il Kim.
Glutathione and endosomal pH-responsive hybrid
vesicles fabricated by zwitterionic polymer block poly
platform. React Funct Polym. 2017; 119: 47-56.
47. John JV, Uthaman S, Augustine R, Lekshmi KM,
Park
IK,
Kim
I.
Biomimetic
pH/redox
dual
stimuli-responsive zwitterionic polymer block poly
((L)-histidine) micelles for intracellular delivery of
39. Giacomelli Fernando C., Petr Stepánek, Cristiano
Vanessa
Nanoparticles for Cancer Therapy. International
(L-aspartic acid) as a smart anticancer delivery
38. Ko Jinyoung, Kyeongsoon Park, Yoo-Shin Kim, Min
based
from amphiphilic copolymer modified with lipid used
Bhosale. A Review on pH -Sensitive Polymeric
36. Somayyeh Fallah iri sofla, Mojtaba Abbasian &
micelles
evaluation
micelles
(5): 1353-1362.
2018; 9: Article 543.
Giacomelli,
early
vivo
drug-resistant ovarian cancer. Mol Pharm. 2009; 6
pH-sensitive polymeric micelles for the Co-delivery
synergistic
In
polymeric
Zhao and Yanjun Chen. pH Responsive Polymer
Anbu,
Ikechukwu
40. Kim Dongin, Zhong Gao Gao, Eun Seong Lee,
doxorubicin-loaded
32. Debele Tilahun Ayane, Kuan-Yi Lee, Ning-Yu Hsu,
Yi-Ting
(poly[2-(diisopropylamino)ethyl methacrylate
PDPA
doxorubicin into tumor cells. J Polym Sci Pol Chem.
2017; 55: 2061-70.
48. Johnson RP, Uthaman S, John JV, Lee HR, Lee SJ,
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 46
Freely Available Online
Park H, Park IK, Suh H, Kim I. Poly(PEGA)-b-poly(L-
delivery with synergistic anticancer efficacy and
lysine)-b-poly(L-histidine)
reduced
Tumoral
pH-triggered
hybrid
vesicles
intracellular
for
delivery
of
doxorubicin hydrochloride. Acs Appl Mater Inter.
2015; 7: 21770-9.
toxicity.
RSC
Advances
2014;
4:
29193-29201.
57. Liao JH, Zheng H, Fei Z, Lu B, Zheng H, Li D, Xiong
X,
Yi
Y.
Tumor-targeting
and
pH-responsive
49. Marques João G, Gaspar VM, Costa EC, Paquete CM.
nanoparticles from hyaluronic acid for the enhanced
Synthesis and characterization of micelles as carriers
delivery of doxorubicin. Int J Biol Macromol. 2018;
of non-steroidal anti-inflammatory drugs (NSAID) for
113: 737-47.
application in breast cancer therapy. Colloids and
surfaces B: Biointerfaces, 2013; 113C: 375-383.
Therapeutic nanoparticles for drug delivery in
50. Zhi Zeng, Zei Liang Wei, Li-Mei Ma, Yao Xu.
pH-responsive nanoparticles based on Ibuprofen
prodrug as drug carriers for inhibition of primary
tumor growth and metastasis. J. Mater. Chem. B,
2017; 5: 6860-6868.
Ji-zhao Li, Zhi-qiang Yuan, Yang Liu, Xiao-feng
Chun
Liu,
Xue-nong
Zhang.
Stepwise
pH-responsive nanoparticles for enhanced cellular
uptake and on-demand intracellular release of
doxorubicin.
Dove
Press,
2017;
Vol.2017:
4241-4256.
52. Biswas, S., Kumari, P., Lakhani, P. M., and Ghosh, B.
Recent
advances
in
polymeric
micelles
for
anti-cancer drug delivery. Eur. J. Pharm. Sci. 2016;
83: 184-202.
53. Ahmad Z., Tang Z. H., Shah A., Lv S. X., Zhang D.,
Zhang Y., Chen X.
Cisplatin loaded methoxy poly
(ethylene glycol)-block-poly (L-glutamic acid-co-Lphenylalanine) nanoparticles against human breast
cancer cell. Macromol. Biosci. 2014; 14: 1337-1345.
54. García-Pinel Beatriz, Cristina Porras-Alcalá, Alicia
Ortega-Rodríguez, Francisco Sarabia,
Jose Prados,
Consolación Melguizo, and Juan M. López-Romero.
Lipid-Based Nanoparticles: Application and Recent
Advances in Cancer Treatment. Nanomaterials 2019;
9: 638. DOI:10.3390/nano9040638.
55. Tanbour, R., Martins, A. M., Pitt, W. G., and
Husseini, G. A. Drug delivery systems based on
polymeric micelles and ultrasound: a review. Curr.
Pharm. Des. 2016; 22: 2796-2807.
56. Jain, S.; Jain, R.; Das, M.; Agrawal, A.K.; Thanki, K.;
Kushwah,
gemcitabine
V.
Combinatorial
and
cancer. Clin Cancer Res. 2008; 14(5): 1310-1316.
59. Yandan Yao, Phei Er Saw, Yan Nie, Ping Pui Wong.
Multifunctional sharp pH-responsive nanoparticles
for targeted drug delivery and effective breast
cancer therapy. J. Materials Chem. B 2018; 7:
51. Wei-liang Chen, Fang Li, Yan Tang, Shu-di Yang,
Zhou,
58. Cho K, Wang X, Nie S, Chen ZG, Shin DM.
curcumin
bio-conjugation
enables
dual
of
drug
576-585.
60. Anajafi
Tayebeh
Polymersome-based
and
Mallik
drug-delivery
Sanku.
strategies
for
cancer therapeutics. Ther Deliv. 2015; 6: 521-534.
61. Asano, I., So, S., and Lodge, T. P. Oil-in-oil
emulsions stabilized by asymmetric polymersomes
formed by AC + BC block polymer co-assembly. J.
Am. Chem. Soc. 2016; 138: 4714-4717.
62. Zhao Yi, Xiaoming Li, X. Zhao, Y. Yang, H. Li, X.
Zhou and W. Yuan. Asymmetrical Polymer Vesicles
for Drug delivery and Other Applications. Front.
Pharmacol., 2017; 8: 374:1-9.
63. Thambi Thavasyappan, Jae Hyung Park and Doo
Sung Lee. Stimuli-Responsive Polymersomes for
Cancer Therapy. Biomaterials Science, 2015; 1-40.
64. Anitha P. , J. Bhargavi, G. Sravani, B. Aruna,
Ramkanth S. Recent Progress of Dendrimers in Drug
Delivery For Cancer Therapy. Int J App Pharm,
2018; 10(5): 34-42.
65. Castro R. I., Oscar Forero-Doria and Luis Guzman.
Perspectives of Dendrimer-based Nanoparticles in
Cancer Therapy. Anais da Academia Brasileira de
Ciências (Annals of the Brazilian Academy of
Sciences) 2018; Vol.90: 2331-2346.
66. Ekladious Iriny, Yolonda L. Colson & Mark W.
Grinstaff.
Polymer–drug
conjugate
therapeutics:
advances, insights and prospects. Nature Reviews
Drug Discovery 2019; 18: 273-294.
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 47
Freely Available Online
67. Wen S, Liu H, Cai H, Shen M, Shi X. Targeted
pH-sensitive dextran/mesoporous silica nanoparticles
and pH-responsive delivery of doxorubicin to cancer
based
cells
intracellular drug release. Int J Biol Macromol. 2017;
using
multifunctional
dendrimer-modified
multi-walled carbon nanotubes. Adv Healthc Mater.
2013; 2: 1267-76.
drug
delivery
systems
for
controlled
98: 691-700.
76. Etrych T, Subr V, Strohalm J, Sírová M, Ríhová B,
68. Zhang Mengen, Jingyi Zhu, Yun Zheng, Rui Guo,
Ulbrich K. HPMA copolymer-doxorubicin conjugates:
Shige Wang, Serge Mignani, Anne-Marie Caminade,
the effects of molecular weight and architecture on
Jean-Pierre
biodistribution and in vivo activity. J. Control.
Majoral,
and
Doxorubicin-Conjugated
pH-Responsive
Acid-Targeted
Xiangyang
PAMAM
Drug
Dendrimers
Release
Cancer
Therapy.
and
Shi.
for
Folic
Pharmaceutics.
2018; 10: 162: 1-13.
M,
Rodrigues
J,
Dendrimer-Entrapped
Shi
X.
Multifunctional
Gold
Nanoparticles
Conjugated with Doxorubicin for pH-Responsive
Drug Delivery and Targeted Computed Tomography
Imaging. Langmuir. 2018; 34: 12428-12435.
and Wenhu Zhou. A Smart pH-Sensitive Delivery
for
Enhanced
Anticancer
Efficacy
via
Paclitaxel Endosomal Escape. Front. Pharmacol.
2019; 10: 10-16.
Intracellular
Delivery
of
Anticancer
Polymer Nanomedicines. Macromol. Biosci. 2017; 17
-29.
Hydrazone-Containing
Micelles
for
Triblock
pH-Controlled
Drug
Delivery. Front Pharmacol. 2018; 9: 12-23.
78. Takahashi, A, Yamamoto Y, Yasunaga M, Koga Y,
Kuroda J, Takigahira M, Harada M, Saito H, Hayashi
T, Kato Y, Kinoshita T, Ohkohchi N, Hyodo I,
Matsumura Y. NC-6300, an epirubicin-incorporating
the cardiotoxicity of epirubicin. Cancer Sci. 2013;
104: 920-925.
79. Alani AWG, Younsoo Bae, Deepa A. Rao, and Glen S.
Kwon. Polymeric micelles for the pH-dependent
Biomaterials 2010; 31: 1765-1772.
80. Yulong Jin, Yanyan H, Hua Y, Guoquan L and Rui
Zhao. A peptide-based pH-sensitive drug delivery
system for targeted ablation of cancer cells. Chem.
72. Sirova Milada, Tomas Mrkvan, Tomas Etrych, Petr
Chytil, Rossmann P., Ibrahimova M. Preclinical
Evaluation of Linear HPMA-Doxorubicin Conjugates
with pH-Sensitive Drug Release: Efficacy, Safety,
and Immunomodulating Activity in Murine Model.
Pharmaceutical Research, 2009; 27: 200-8.
73. Talelli Marina, Maryam Iman, Wim E Hennink, Amir
K Varkouhi. Core-crosslinked polymeric micelles with
controlled
Song.
controlled, continuous low dose release of Paclitaxel.
71. Battistella Claudia, Harm‐Anton Klok. Controlling and
Monitoring
Shiyong
micelle, extends the antitumor effect and reduces
70. Yang Yihua, Zhe Wang, Ying Peng, Jinsong Ding,
System
77. Peilan Qi, Xiaohe Wu, Lei Liu, Huimin Yu, and
Copolymeric
69. Zhu J, Wang G, Alves CS, Tomás H, Xiong Z,
Shen
Release 2012; 164: 346-354.
release
of
covalently
entrapped
doxorubicin. Biomaterials 2010; 31: 7797-804·
74. Zhu S, Lansakara-P DS, Li X, Cui Z. Lysosomal
delivery of a lipophilic gemcitabine prodrug using
novel acid-sensitive micelles improved its antitumor
activity. Bioconjug Chem. 2012; 23: 966-80.
75. Zhang M, Liu J, Kuang Y, Li Q, Zheng DW, Song Q,
Chen H, Chen X, Xu Y, Li C, Jiang B. Ingenious
Communications 2015; Issue 51: 14454-14457.
81. Ulbrich K., Etrych T., Chytil P., Jelinkova M., Rihova
B. HPMA copolymers with pH-controlled release of
doxorubicin: In vitro cytotoxicity and in vivo
antitumor activity. J. Control. Release 2003; 87:
33-47.
82. Lavignac N, Johanna L Nicholls, Paolo Ferruti, Ruth
Duncan. Poly(amidoamine) Conjugates Containing
Doxorubicin Bound via an Acid-Sensitive Linker.
Macromolecular Biosci. 2009; 9: 480-7.
83. Zhu S, Hong M, Tang G, Qian L, Lin J, Jiang Y, Pei Y.
Partly PEGylated polyamidoamine dendrimer for
tumor-selective targeting of doxorubicin: the effects
of PEGylation degree and drug conjugation style.
Biomaterials 2010; 31: 1360-1371.
84. Hu Q, Rijcken CJ, Bansal R, Hennink WE, Storm G,
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 48
Freely Available Online
Prakash J. Complete regression of breast tumour
with
a
single
dose
of
docetaxel-entrapped
93. Tian
S,
Liu
G,
Wang
pH-Responsive
X,
Zhang
G,
Tumor-Targetable
Hu
J.
Theranostic
core-cross-linked polymeric micelles. Biomaterials
Nanovectors Based on Core Crosslinked (CCL)
2015; 53: 370-378.
Micelles with Fluorescence and Magnetic Resonance
85. Thambi T, Deepagan VG, Yoo CK, Park JH. Synthesis
and physicochemical characterization of amphiphilic
block copolymers bearing acid-sensitive orthoester
(MR) Dual Imaging Modalities and Drug Delivery
Performance. Polymers (Basel). 2016; 8: E226.
94. Yu S, Wu G, Gu X, Wang J, Wang Y, Gao H, Ma J.
linkage as the drug carrier. Polymer 2011; 52:
Magnetic
4753-9.
antitumor drug delivery. Colloids and Surfaces B:
86. Zha Q, Wang X, Cheng X, Fu SX, Yang GQ, Yao WJ,
and
pH-sensitive
nanoparticles
for
Biointerfaces, 2013; 103: 15-22.
Tang RP. Acid degradable carboxymethyl chitosan
95. Wang Jingjing, Chu Gong, Yinong Wang and Guolin
nanogels via an ortho ester linkage mediated
Wu. Magnetic and pH sensitive drug delivery system
improved penetration and growth inhibition of 3-D
through NCA chemistry for tumor targeting. Royal
tumor spheroids in vitro. Mat Sci Eng C-Mater. 2017;
Society
78: 246-57.
15856-15862.
87. Belali S, Karimi AR, Hadizadeh M. Cell-specific and
pH-sensitive
nanostructure
hydrogel
based
on
of
Chemistry
Advances,
2014;
4:
96. Karimi Z, Abbasi S, Shokrollahi H, Yousefi G,
Fahham M, Karimi L, Firuzi O. Pegylated and
chitosan as a photosensitizer carrier for selective
amphiphilic Chitosan coated
photodynamic therapy. Int J Biol Macromol. 2018;
nanoparticles
110: 437-48.
methotrexate: Synthesis and characterization. Mater
88. Tao YC, Liu SW, Zhang Y, Chi ZG, Xu JR. A
for
manganese
pH-sensitive
ferrite
delivery
of
Sci Eng. 2017; 71: 504-511.
pH-responsive polymer based on dynamic imine
97. Wu Juan, Aipeng Deng, Wei Jiang, Renbing Tian,
bonds as a drug delivery material with pseudo target
Shen Y. Synthesis and in vitro evaluation of
release behavior. Polym Chem-Uk. 2018; 9: 878-84.
pH-sensitive
89. Popat A, Liu J, Lu GQ, Qiao SZ. A pH-responsive
drug delivery system based on chitosan coated
mesoporous silica nanoparticles. J Mater Chem.
90. Kang EB, Lee JE, Mazrad ZAI, In I, Jeong JH, Park
SY. pH-responsible fluorescent carbon nanoparticles
for tumor selective theranostics via pH turn on/off
fluorescence and photothermal effect in vivo and in
vitro. Nanoscale. 2018; 10: 2512-23.
91. Ray Sayoni, Zhao Li, Chao-Hsiung Hsu, Lian-Pin
Hwang, Ying-Chih Lin, Pi-Tai Chou, Yung-Ya Lin.
Dendrimer- and copolymer-based nanoparticles for
resonance
cancer
theranostics.
Theranostics, 2018; 8: 6322-6349.
Deng,
as
methotrexate delivery system for targeted cancer
therapy. Materials Science and Engineering: C.
2016; 71: 132-140.
Ping'an
Ma
Tian X, Zhou J, He J, Wang Y, Wu Y, Zhong K, Cai
D, Zou D, Wu Z. A pH-Responsive Yolk-Like
Nanoplatform
for
Tumor
Targeted
Dual-Mode
Magnetic Resonance Imaging and Chemotherapy.
ACS Nano. 2017; 11: 7049-7059.
99. Wang Xin, Ziyu Gao, Long Zhang, Huiming Wang,
and Xiaohong Hu. A Magnetic and pH-Sensitive
Composite
Nanoparticle
for
Drug
Delivery.
J.
Nanomater. 2018; Article ID 1506342, pp.7.
100.Lungu II, Rădulescu M, Mogoşanu GD, Grumezescu
92. Huang S, Ziyong Cheng, Yinyin Chen, Bei Liu,
Xiaoran
nanocomposites
98. Sun X, Du R, Zhang L, Zhang G, Zheng X, Qian J,
2012; 22: 11173-8.
magnetic
magnetic
and
Jun
Lin.
Multifunctional polyelectrolyte multilayers coated
AM. pH sensitive core-shell magnetic nanoparticles
for targeted drug delivery in cancer therapy. Rom J
Morphol Embryol. 2016; 57: 23-32.
onto Gd2O3:Yb3+,Er3+@MSNs can be used as drug
101.Bardajee GR, Hooshyar Z, Farsi M, Mobini A, Sang
carriers and imaging agents. RSC Advances 2015; 5:
G. Synthesis of a novel thermo/pH sensitive nanogel
41985-41993.
based on salep modified graphene oxide for drug
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 49
Freely Available Online
release.
Materials
Science
&
Engineering.
C,
110.Lee ES, Gao Z, Kim D, Park K, Kwon IC, Bae YH.
Materials for Biological Applications. 2016; 72:
Super pH sensitive multifunctional polymeric micelle
558-565.
for tumor pH(e) specific TATexposure and multidrug
102.Yadav HKS, Al Halabi NA, Alsalloum GA. Nanogels as
Novel Drug Delivery Systems - A Review. J Pharm
Pharm Res. 2017; 1: 5: 1-8.
resistance. J Control Release 2008; 129: 228-236.
111.Rao NV, Mane S, Kishore A, Das Sarma J,
Shunmugam R. Norbornene derived doxorubicin
103.Garg T, Arora S, Murthy R, Goyal AK. Development,
optimization & evaluation of porous chitosan scaffold
formulation of gliclazide for the treatment of type-2
diabetes mellitus. Drug Delive Lett. 2012; 2:
251-261.
copolymers as drug carriers with pH responsive
hydrazone linker. Biomacromolecules 2011; 13:
221-230.
112.Wei
H,
Zhuo
R-X,
Zhang
X-Z.
Design
and
development of polymeric micelles with cleavable
104.Sharma A, Tarun Garg, Amrinder Aman, Kushan
Panchal, Rajiv Sharma, Sahil Kumar. Nanogel-an
advanced drug delivery tool: Current and future.
links for intracellular drug delivery. Prog Polym Sci.
2013; 38: 503-535.
113.Liu
M,
Du
H,
Zhang
W,
Zhai
G.
Internal
Artificial Cells, Nanomedicine, and Biotechnology: An
stimuli-responsive nanocarriers for drug delivery:
International Journal 2016; 44: 165-77.
design strategies and applications. Mater Sci Eng C
105.Oishi M, Hisato Hayashi, Michihiro Iijima and Yukio
Nagasaki.
Endosomal
release
and
2017; 71: 1267-1280.
intracellular
114.Shih Y, Venault A, Tayo LL, Chen SH, Higuchi A,
delivery of anticancer drugs using pH-sensitive
Deratani A, Chinnathambi A, Alharbi SA, Quemener
PEGylated nanogels. J. Materials Chem. 2007; 17:
D, Chang Y. A Zwitterionic-shielded carrier with
3720-3725.
pH-modulated reversible self assembly for gene
106.Liu K., Dan Zheng, Jingyang Zhao, Yinghua Tao,
transfection. Langmuir 2017; 33: 1914-1926.
Yingsa Wang, Jing He, Jiandu Lei and Xingjun
115.Shen Y, Huadong Tang, Maciej Radosz, Edward Van
Xi. pH-Sensitive nanogels based on the electrostatic
Kirk. pH-Responsive Nanoparticles for Cancer Drug
self-assembly of radionuclide 131I labeled albumin
Delivery. Methods in Molecular Biology 2008; 437:
and
183-216.
carboxymethyl
cellulose
for
synergistic
combined chemo-radioisotope therapy of cancer. J.
Materials Chem. B 2018; 6: 4738-4746.
Responsive nanoparticles for drug delivery. Mol.
107.Taghizadeh Bita, Shahrouz Taranejoo, Seyed Ali
Monemian, Zoha Salehi Moghaddam, Karim Daliri,
Hossein
Derakhshankhah,
Zaynab
Classification of stimuli-responsive
116.Weiwei Gao, Juliana M Chan, Omid C Farokhzad. pH
Derakhshani.
polymers
as
anticancer drug delivery systems. Drug Delivery
2015; 22: 145-155.
Pharmaceutics 2010; 7: 1913-1920.
117.Liu J, Huang Y, Kumar A, Tan A, Jin S, Mozhi A,
Liang
XJ.
pH-sensitive
nano-systems
for
drug
delivery in cancer therapy. Biotechnol adv. 2014; 32:
693-710.
108.Peng Wei, Gauri Gangapurwala, David Pretzel,
Meike Nicole Leiske. Smart pH-sensitive Nanogels for
Controlled
Release
in
Acidic
Environment.
Biomacromolecules 2018; 20: 130-140.
109.Urban-Klein B, Werth S, Abuharbeid S, Czubayko F,
Aigner A. RNAi-mediated gene-targeting through
systemic
application
of
polyethylenimine
(PEI)-complexed siRNA in vivo. Gene Ther. 2005;
12: 461-6.
www.openaccesspub.org | JWRH CC-license DOI : 10.14302/issn.2381-862X.jwrh-19-3143
Vol-2 Issue 2 Pg. no.- 50