You are on page 1of 9

Drug Discovery Today d Volume xxx, Number xx d xxxx 2021 REVIEWS

Stimuli-responsive hydrogels for

POST-SCREEN
intratumoral drug delivery
Ana C. Marques a,⇑, Paulo J. Costa a, Sérgia Velho b,c, Maria H. Amaral a
a
UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto,
Porto, Portugal
b
i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
c
IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal

The ability of some hydrogels to exhibit a phase transition or change their structure in response to
stimuli has been extensively explored for drug depot formation and controlled drug release. Taking
advantage of the unique features of the tumor microenvironment (TME) or externally applied triggers,
several injectable stimuli-responsive hydrogels have been described as promising candidates for
intratumoral drug delivery. In this review, we provide a brief overview of the TME and highlight the
advantages of intratumoral administration, followed by a summary of the reported strategies to endow
hydrogels with responsiveness to physical (temperature and light), chemical (pH and redox potential),
or biological (enzyme) stimuli.

Keywords: Cancer; Intratumoral administration; Stimuli-responsive hydrogels

Introduction Tumor microenvironment


The standard approach to chemotherapy has been the intra- The tumor microenvironment (TME) is a complex ecosystem
venous administration of cytotoxic agents. Notwithstanding, where malignant cells coexist and interact with a heterogeneous
chemotherapeutic drugs lack specificity for cancer cells, giving group of non-malignant cells (stromal cells) and noncellular ele-
rise to suboptimal dosing and severe adverse effects. Moreover, ments comprising the extracellular matrix (ECM). Although the
this treatment modality is often restricted by dose-limiting toxi- latter provides physical support and mechanical integrity to cells,
city and drug resistance. ECM-degrading enzymes, such as matrix metalloproteinases
Despite ongoing efforts to implement targeted therapies, (MMPs), are often overexpressed in tumors. This large family of
available systemic treatment options cannot deliver drugs to endopeptidases digests ECM proteins, thereby disrupting physi-
tumors while remaining harmless to the rest of the body. Addi- cal barriers that impede cancer cells to leave the primary tumor
tionally, current locoregional therapies might help to achieve a site and invade nearby tissues.1
better sparing of healthy tissues but are associated with carcino- Solid tumors exhaust their blood supply to meet the energetic
genesis risk, cancer recurrence, or complications of invasive sur- demands of proliferating tumor cells and then experience perma-
gical techniques. From this perspective, further research and nent or transient deprivation of oxygen (hypoxia) and nutrients.
investment in developing local and minimally invasive cancer Cycles of hypoxia–reoxygenation raise intracellular levels of reac-
therapies are imperative. tive oxygen species (ROS), and mild oxidative stress stimulates

⇑ Corresponding author. Marques, A.C. (amarques@ff.up.pt)

1359-6446/Ó 2021 Elsevier Ltd. All rights reserved.


https://doi.org/10.1016/j.drudis.2021.04.012 www.drugdiscoverytoday.com 1
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
POST-SCREEN (GREY) Drug Discovery Today d Volume xx, Number xx d xxx 2021

the expression of antioxidant enzymes and reducing species, to be biocompatible and their biodegradation products to cause
such as reduced glutathione (GSH).2,3 The GSH concentration an appropriate host response without cytotoxicity, carcinogene-
in some tumor cells and tissues were found to be as high as 1– sis, and/or mutagenesis. Therefore, biocompatibility is typically
10 mM.4 assessed in vitro by cell viability assays [e.g., methylthiazolyl
In response to hypoxia and low nutrition, a metabolic switch tetrazolium (MTT) assay] and ex vivo by histological analysis of
from oxidative phosphorylation to aerobic glycolysis takes place. tumor sections and major organs after Hematoxylin and Eosin
Cancer cells increase their glucose uptake and convert it to lac- (H&E) staining. To evaluate biodegradability, hydrogels are often
tate even in the presence of oxygen (the ‘Warburg effect’). Given immersed in phosphate-buffered saline (PBS) solutions at 37 °C
that high glycolytic rates lead to excessive production of lactic and acidic pH (containing enzymes, redox species, or under light
acid, tumor cells upregulate the expression of acid-extruding irradiation if applicable) or subcutaneously injected in mice.
transporters, maintaining intracellular pH as neutral or slightly Injectable gels are either preformed gels with shear-thinning
POST-SCREEN

alkaline. As a result, the extracellular pH of solid tumors is more and self-healing properties or in situ-forming gels. The former
acidic (typically between 6.5 and 6.9) than that of normal can be extruded through a syringe needle after gelation as low
tissues.5,6 viscosity formulations by application of shear stress. When the
Besides their importance for tumor progression, one can take shear is removed, gels self-heal and quickly restore their original
advantage of these tumor traits (hypoxia, oxidative stress, and state at the site of injection.8 Conversely, in situ gelling systems
acidosis) for developing novel therapeutic strategies based on are injected as free-flowing polymer solutions that transform into
stimuli-responsive hydrogels. gels at the injection site, forming drug depots for sustained drug
release.9 The mechanism of depot formation can be: (i) in situ
crosslinking, usually using a dual-barrel syringe for co-injection
Intratumoral administration and mixing of gel precursor solutions or an external source of
Upon intravenous administration, sequential obstacles to long light to induce gelation (photo-polymerization); or (ii) in situ
circulation times affect the percentage of free and encapsulated phase transition, triggered by certain stimuli, such as changes
drugs having access to the tumor. By contrast, intratumoral (IT) in temperature or pH.
administration bypasses the bloodstream and enables local deliv-
ery of cytotoxic agents to the tumor site, achieving high concen-
trations at target cells and reducing dose requirements. Likewise, Stimuli-responsive hydrogels
it counteracts drug uptake by nontarget cells, minimizing sys- Hydrogels are 3D crosslinked networks of hydrophilic polymers
temic toxicity.7 Moreover, this administration route is indepen- that absorb and retain large amounts of water while resisting dis-
dent of the tumor vasculature and, thus, reaches poorly solution in the aqueous medium. Some polymers respond to
perfused tumor regions that systemic administration regularly external stimuli by changing their conformation, solubility, or
misses. hydrophilic/lipophilic balance.10 Such changes are reversible
Unlike other drug delivery systems, injectable biodegradable because stimuli-responsive polymers, also named ‘smart’ poly-
gels do not require surgical procedures and promote retention mers, return to their initial state once the stimulus ends. Accord-
of free drugs and drug-loaded particles at the tumor site. ing to their nature, stimuli can be classified as physical, chemical,
Biodegradable hydrogels can either be degraded after the drug or biological (Fig. 1).11
is completely released by diffusion or release the loaded drug as In the context of IT drug delivery, stimuli-responsive hydro-
they disintegrate into small fragments that are eliminated from gels can display a volume phase transition, swelling/shrinking,
the body. Besides biodegradability, injectable gels are expected or assembly/disassembly under exposure to exogenous triggers

Physical Chemical Biological

Temperature pH
Enzymes
Light
Redox
Electric field
Glucose
Magnetic field Solvent composition

Drug Discovery Today

FIGURE 1
Examples of physical, chemical, and biological stimuli responsible for changes in the volume and structure of hydrogels. Physical stimuli include temperature,
light, electric and magnetic fields. In turn, pH, redox potential, and solvent composition are considered chemical stimuli. Finally, enzymes and glucose are two
examples of biological stimuli. Temperature, light, pH, reduction, oxidation, and enzymes have been explored for the development of stimuli-responsive
hydrogels for IT drug administration.

2 www.drugdiscoverytoday.com
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
Drug Discovery Today d Volume xx, Number xx d xxxx 2021 POST-SCREEN (GREY)

(light) or endogenous stimuli in the TME (temperature, pH, CS/b-GP hydrogel containing paclitaxel (PTX)-loaded poly
redox, and enzymes). The concept of stimuli responsiveness (lactide-co-glycolide) (PLGA) microparticles was at least similar
extends to hydrogels undergoing degradation in response to to that of four intraperitoneal injections of a commercial formu-
stimuli by reversible or irreversible bond cleavage in the polymer lation (PaklitaxfilÒ, Fresenius Kabi) in mammary tumor-bearing
backbone or crosslinks. mice.15 However, CS/b-GP hydrogels are prone to in vivo degrada-
Compared with conventional hydrogels, stimuli-responsive tion by erosion and the appearance of cracks that impair their
hydrogels might allow for greater control of the local and dura- mechanical properties and controlled drug release. Accordingly,
tion of the drug release. Depending on the stimulus, these hydro- significant efforts have been made to developing CS hydrogels
gels offer different advantages for IT drug administration. For based on dynamic covalent chemistry with self-repair ability
example, thermoresponsive hydrogels exhibiting sol–gel transi- after damage. To illustrate, Han et al.16 modified a CS/b-GP
tion at the tumor site are injected in the sol state with particular hydrogel with dialdehyde-functionalized poly(ethylene glycol)

POST-SCREEN
ease and form implants in situ after gelation. The in situ-forming (PEG) via chemical crosslinking to achieve autonomous self-
implants adapt their shape to the surrounding tissues and act as a healing and sustained release of doxorubicin (DOX), used as a
reservoir system (i.e., a polymer-coated drug core) that enables model drug. Alternatively, a semi-interpenetrating polymer net-
sustained drug release over longer periods compared with pre- work of N-acetylated glycol CS (or glycol chitin) and poly(acrylic
formed hydrogels. In addition to stimuli responsiveness, the acid) allows for thermal gelation at 37 °C, as well as superior gel
presence of acid-labile bonds, photocleavable moieties, or strength.17
enzyme substrates improves or imparts biodegradability to Whereas N-isopropylacrylamide is cytotoxic even at low con-
hydrogels. Moreover, the ‘on/off’ switching of a remotely applied centrations, the polymerized form is nontoxic and biocompati-
stimulus enables a pulsatile drug release (versus continuous ble with different cell types. Nevertheless, grafting poly(N-
release) with negligible release during the ‘off’ state, which can isopropylacrylamide) (PNIPAM) with natural polymers has been
be useful for certain cancer therapies. performed to impart biodegradability to the corresponding
The following sections cover stimuli-responsive hydrogels hydrogel. Fong et al.18 prepared PNIPAM end-capped with car-
developed recently for IT drug administration. boxylic acid for conjugation with CS and hyaluronic acid (HA),
obtaining a HA-CS-graft-PNIPAM copolymer with an LCST simi-
lar to that of PNIPAM (32 °C).
Thermoresponsive hydrogels Among the poloxamers, Poloxamer 407 (P407) or PluronicÒ
Hydrogels capable of responding to temperature changes are F127 (F127) solutions occupy the foreground of thermorespon-
considered thermoresponsive. For instance, the shift from room sive polymer-based vehicles for IT drug delivery. For instance,
temperature (25 °C) to body/tumor temperature (37 °C) can P407 solutions were mixed with DOX to kill MC-38 colon cancer
cause thermoresponsive polymer solutions to exhibit a sol–gel cells19 or with a topotecan solution to achieve a long-acting anti-
phase transition. The temperature at which a polymer solution tumor effect on retinoblastoma.20 Several hybrid systems inte-
undergoes a transition from one phase to two phases (an aqueous grating drug-loaded NPs and F127 hydrogels have also been
and a polymer-enriched phases) is designated the ‘critical solu- reported for this administration route. In an interesting approach
tion temperature’.12 to the treatment of melanoma, Yu et al.21 prepared an F127
Most thermoresponsive polymers have a lower critical solu- hydrogel to intratumorally deliver cisplatin (CDDP)-loaded
tion temperature (LCST) in water that is similar to the volume poly(a-L-glutamate)-graft-methoxy PEG NPs and microspheres
phase transition temperature of the corresponding hydrogel. At entrapping losartan potassium, which exerts antifibrotic effects.
temperatures below the LCST, these polymers are miscible with The combination of liposomes and a PluronicÒ F127/F68 hydro-
water but become insoluble when raising the temperature gel was proposed to stabilize the lactone form of 7-ethyl-10-
beyond the LCST, separating from the solution and yielding a hydroxycamptothecin and enhance drug retention at H22 hep-
gel. In the case of thermoresponsive polymers presenting an atoma tumors.22 By contrast, mixed micelles comprising F127
upper critical solution temperature (UCST), heating beyond the and another surfactant, such as SolutolÒ HS1523 or D-a-
UCST favors the solubilization of polymer chains. Thermore- tocopherol PEG 1000 succinate,24 have been incorporated into
sponsive polymers having the phase transition temperature near F127 hydrogels to deliver hydrophobic drugs, namely doc-
body temperature are mainly LCST-type polymers.13 etaxel23 and PTX.24 Finally, the addition of N,N,N-trimethyl
Over the past two decades, a large number of in situ-forming CS25 or the use of crosslinkers, such as genipin,26 can improve
thermoresponsive hydrogels based on natural (chitosan) or syn- the mechanical properties of P407 hydrogels.
thetic (polyacrylamides, poloxamers, and polyesters) polymers More recently, amphiphilic copolymers comprising PEG and
have been developed for IT drug administration. polyesters have been defined as first-line materials for the design
Although atypical in most polysaccharides, the cationic nat- and manufacture of thermoresponsive hydrogels for IT drug
ure of chitosan (CS) enables valuable interactions with anionic administration. The most used polyesters are polylactic acid
molecules such as b-glycerophosphate (b-GP) to produce ther- (PLA), PLGA, and PCL.
moresponsive hydrogels. For example, Bragta et al.14 selected IT Shi et al.27 used a poly(D,L-lactide)-PEG-poly(D,L-lactide)
administration as an alternative route to intravenous delivery (PDLLA-PEG-PDLLA) triblock copolymer to prepare a thermore-
of carboplatin in melanoma and formulated a CS/b-GP hydrogel sponsive hydrogel coloaded with gemcitabine and CDDP for syn-
carrying carboplatin-loaded poly(e-caprolactone) (PCL) nanopar- ergistic combination therapy in pancreatic cancer. The IT
ticles (NPs). In another study, the efficacy of one IT injection of a injection of a norcantharidin-loaded PDLLA-PEG-PDLLA hydro-

www.drugdiscoverytoday.com 3
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
POST-SCREEN (GREY) Drug Discovery Today d Volume xx, Number xx d xxx 2021

gel prolonged drug retention and improved its curative effect on release of tamoxifen.32 Regarding commercial formulations,
hepatocellular carcinoma.28 By incorporating free bevacizumab OncoGelTM (MacroMed Inc.) is an injectable and thermorespon-
and DOX-encapsulated PDLLA-PEG-PDLLA micelles into a sive hydrogel formulated with a PLGA-PEG-PLGA triblock
PDLLA-PEG-PDLLA hydrogel, Darge et al.29 induced normaliza- copolymer (RegelTM) to deliver PTX to the tumor site in solid
tion of the tumor vasculature and potentiated the chemothera- malignancies. Despite evidence of preliminary efficacy in a Phase
peutic effects of DOX. Zhou et al.30 developed a hydrogel IIa study, IT administration of OncoGelTM failed to reveal any
composite comprising erlotinib-loaded hollow mesoporous silica impact on overall tumor response to neoadjuvant radiotherapy
NPs and a PDLLA-PEG-PDLLA hydrogel for localized treatment of and intravenous chemotherapy (CDDP and 5-fluorouracil) in a
non-small cell lung cancer (NSCLC). Phase IIb study (NCT00573131) in patients with esophageal
Shen et al.31 constructed a PEG-polyester-drug conjugate via cancer.
covalent linking of the Pt(IV) prodrug to the hydrophobic ends Amphiphilic triblock copolymers have been yielded lately by
POST-SCREEN

of two methoxy PEG (mPEG)-PLGA diblock copolymers. The bi copolymerizing e-caprolactone33 and lactide34 in the presence
(mPEG-PLGA)-Pt(IV) conjugate self-assembled into core shell- of PEG. Whereas a PEG-PCL-PEG hydrogel containing CDDP
type micelles, the hydrophobic core of which functioned as and PTX-loaded mPEG-PCL micelles was proposed for combina-
reservoir for PTX. At concentrated solutions, this thermorespon- tion chemotherapy in cervical cancer,33 a (PCL-co-lactide)-PEG-
sive polymer–Pt(IV) conjugate underwent sol–gel transition (PCL-co-lactide) hydrogel incorporating curcumin-loaded
upon heating, accomplishing an IT and synchronous co- mPEG-PLA nanopolymersomes was fabricated to treat glioma.34
delivery of Pt(IV) and PTX for treatment of ovarian cancer. In Table 1 summarizes the described thermoresponsive hydro-
addition, one IT injection of drug-loaded hydrogels based on gels regarding their phase transition temperature and their
PLGA-PEG-PLGA triblock copolymers enabled the sustained in vitro and in vivo cancer models.

TABLE 1
a
Thermoresponsive hydrogels recently developed for IT drug delivery.
Composition Drug Phase Cancer cell Cancer (in vivo) Refs
transition (in vitro)
temperatureb
CS/b-GP, PCL NPs Carboplatin 37 ± 0.8 °C B16F1 Melanoma 14
CS/b-GP, PLGA microparticles PTX – – Mammary adenocarcinoma 15
CS/DF-PEG/b-GP DOX 31.51 ± 0.31 °C – Hepatocellular carcinoma 16
Glycol chitin/PAA, PLGA microspheres PTX 17 °Cc HepG2/C3A – 17
(hepatocellular
carcinoma)
HA-CS-g-PNIPAM, folate-conjugated GO DOX 30.07 °C MCF-7 Breast cancer 18
P407 DOX – MC-38 (colon – 19
cancer)
Topotecan 37.23 ± 0.473 °C – Retinoblastoma 20
F127, PLG-g-mPEG nanoparticles and microspheres CDDP, losartan 30 °C – Melanoma 21
F127/F68, liposomes SN-38 – – Hepatocellular carcinoma 22
F127/ P188, F127 and SolutolÒ HS15 micelles DTX 37 °C – Colon cancer 23
F127/HA, F127, and TPGS micelles PTX, DOX 35 °C – – 24
F127/TMC DTX 37 °C U87MG Glioblastoma 25
PDLLA-PEG-PDLLA Gemcitabine, CDDP 35 °C Bxpc-3 Pancreatic cancer 27
Norcantharidin – – Hepatocellular carcinoma 28
PDLLA-PEG-PDLLA, PDLLA-PEG-PDLLA micelles DOX, bevacizumab 37 °C HeLa Cervical carcinoma 29
PDLLA-PEG-PDLLA, hollow mesoporous silica NPs Erlotinib 35.7°Cd36.4 °C A549 NSCLC 30
Bi(mPEG-PLGA)-Pt (IV) Pt(IV) prodrug, PTX 35 °C SKOV-3 Ovarian cancer 31
PLGA-PEG-PLGA Tamoxifen 37.14 ± 0.66 °C – Breast cancer 32
PEG-PCL-PEG, mPEG-PCL micelles CDDP, PTX 30 °C – Cervical cancer 33
PCLA-PEG-PCLA, mPEG-PLA nanopolymersomes Curcumin 32 °C – Glioma 34
a
Abbreviations: b-GP, b-glycerophosphate; Bi(mPEG-PLGA) Pt(IV), bi(poly(ethylene glycol) methyl ether-poly(lactic-co-glycolic acid))-Pt(IV) conjugate; CDDP, cisplatin; CS, chitosan; DF-PEG,
dialdehyde-functionalized poly(ethylene glycol); DOX, doxorubicin; DTX, docetaxel; F68, PluronicÒ F68; F127, PluronicÒ F127; GO, graphene oxide; HA, hyaluronic acid; HA-CS-g-PNIPAM, hyaluronic
acid-chitosan-graft-poly(N-isopropylacrylamide); mPEG-PCL, poly(ethylene glycol) methyl ether-poly(e-caprolactone); mPEG-PLA, poly(ethylene glycol) methyl ether-polylactic acid; NSCLC, non-
small cell lung cancer; P188, poloxamer 188; P407, poloxamer 407; PAA, poly(acrylic acid); PCL, poly(e-caprolactone); PCLA-PEG-PCLA, poly(e-caprolactone-co-lactide)-poly (ethylene glycol)-poly(e-
caprolactone-co-lactide): PDLLA PEG PDLLA, poly(D,L-lactide)-poly(ethylene glycol)-poly(D,L-lactide); PEG-PCL-PEG, poly(ethylene glycol)-poly(e-caprolactone)-poly(ethylene glycol); PLG-g-mPEG,
poly(a-L-glutamate)-graft-poly(ethylene glycol) methyl ether; PLGA, poly(lactic-co-glycolic acid); PLGA-PEG-PLGA, poly(lactic-co-glycolic acid)-poly(ethylene glycol)-poly(lactic-co-glycolic acid); PTX,
paclitaxel; SN-38, 7-ethyl-10-hydroxycamptothecin; TMC, N,N,N-trimethyl chitosan; TPGS, D-a-tocopherol poly(ethylene glycol) 1000 succinate.
b
The volume phase transition temperatures (sol–gel transition or gelation temperatures) were determined by the test tube inversion method, rheology, differential scanning calorimetry, or using
a thermometer.
c
This formulation comprises agent A (glycol chitin/PAA solution containing PTX-loaded PLGA microspheres, crosslinker, and initiator) and agent B (promoter). The gel forms at 37 °C upon co-
injection of agents A and B, which are liquid and stored separately in the refrigerator.
d
Incorporation of erlotinib at a higher concentration (6 mg/ml) led to a slight increase in the phase transition temperature of the hydrogel compared with that of a low concentration of erlotinib
(2 mg/ml).

4 www.drugdiscoverytoday.com
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
Drug Discovery Today d Volume xx, Number xx d xxxx 2021 POST-SCREEN (GREY)

Photo-responsive hydrogels photothermal agents, Hou et al.44 used a derivative of an organic


These hydrogels are typically based on polymers containing a carbon source, humic acid, to endow an injectable agarose
photoreactive moiety that responds to light by way of a photo- hydrogel with photothermal conversion property. In a different
chemical reaction, such as cleavage and isomerization.35 As an work, Wang et al.45formulated an injectable self-healing hydro-
example, Zhao et al.36 prepared an injectable self-assembled gel via dynamic covalent enamine bond between polyetherimide
DOX-loaded hydrogel based on a four-arm star polymer, PEG- and a four-arm star copolymer, poly[2-(dimethylamino)ethyl
poly(c-o-nitrobenzyl-L-glutamate). The presence of o- methacrylate-co-2-hydroxyethyl methacrylate] modified with
nitrobenzyl groups in the polymer backbone rendered hydrogels tert-butyl acetoacetate. The blank hydrogel was loaded with
photodegradable by irreversible photocleavage of o-nitrobenzyl DOX and polydopamine NPs before IT injection in 4T1 breast
ester linkages after irradiation with ultraviolet (UV) light. By con- tumor-bearing mice. Under NIR irradiation (808 nm, 1.0 W/
trast, azobenzene moieties undergo reversible trans-to-cis isomer- cm2, 10 min), the temperature rose well above the LCST

POST-SCREEN
ization when irradiated with UV light, returning to trans isomers (40 °C) of the copolymer because of the photothermal effect
upon heating or higher wavelength irradiation. Given the sub- of polydopamine NPs, leading to shrinking and rapid release of
stantial limitations to the use of UV light (poor penetration DOX.
and risk of damaging tissues), approaches involving upconver-
sion of low-energy photons (e.g., near-infrared; NIR) into high- pH-responsive hydrogels
energy photons (e.g., UV) are favorable.37 Chen et al.38 devel- Referring to acidosis of tumor tissues, two strategies have been
oped an injectable ‘click’-crosslinked gelatin hydrogel loaded reported for the design of pH-responsive hydrogels for IT drug
with DOX and poly(acrylic acid)-coated upconversion NPs. The administration: (i) the selection of pH-responsive polymers with
amount of DOX released from the hydrogel was controllable numerous weak basic (e.g., amine) groups attached to the
with an ‘on/off’ switchable operation of NIR irradiation that trig- hydrophobic backbone, thereby being considered polybases (ca-
gered photo-isomerization of the azobenzene crosslinker. tionic); or (ii) the incorporation of acid-labile linkages within the
Nonetheless, NIR-I light (650–950 nm) penetrates <1 cm into polymer network or between the polymer and drug, the cleavage
the tissues, which is suitable for superficial thin tumors only. of which enables the release of entrapped or anchored drugs.46
Given that the tissue penetration depth of NIR-II light (1000– When the external pH is less than the pKa of a cationic poly-
1700 nm) is 3–5 cm, the treatment of large superficial tumors mer (e.g., acidic pH), amine pendant groups are positively
might be feasible, but its clinical application to deep tumors is charged (NH3+) by protonation. The electrostatic repulsion
still restricted.39 In general, increasing the power density (W/ between charges leads to polymer chain expansion and
cm2) and exposure time to irradiation leads to greater swelling.47 In turn, the enlarged mesh of swollen hydrogels facil-
penetration. itates drug diffusion through the network. In a different
In terms of IT drug administration, some photoresponsive approach, Raza et al.48 synthesized a pH-responsive FEFEFRFK
hydrogels are supramolecular hydrogels incorporating pho- octapeptide that self-assembled at neutral pH into an injectable
tothermal agents. These hydrogels are 3D networks of molecular hydrogel for IT administration of PTX. At pH 5.5, protonation
building blocks self-assembled via noncovalent interactions, of arginine and electrostatic repulsion between the amino acid
which exhibit a gel–sol transition under light irradiation in side chains induced a b-sheet-coil transition of peptide fibers, cul-
response to the heat generated by photothermal agents. In par- minating in hydrogel disassembly and sustained release of PTX.
ticular, Ruan et al.40 demonstrated that poly(N-phenylglycine) The second strategy involves pH-responsive bonds, which are
building blocks could act as NIR-II-absorbing mediators while cleaved in the extracellular fluid or, after endocytosis, in tumor
tethering PEG chains to form a hydrogel via host–guest interac- cell endosomes or lysosomes. Dynamic covalent bonds, namely
tion with a-cyclodextrin. Meanwhile, PEGylated poly(N- hydrazone bonds, can be used by virtue of their hydrolytic stabil-
phenylglycine) was used as photothermal polymer backbone to ity at normal physiological pH (pH 7.4) and rapid cleavage in
construct a photoresponsive hydrogel for IT delivery of DOX.41 mildly acidic pH. Before gel preparation, Sharma et al.49 conju-
Another strategy to achieve reversible photo-induced phase gated part of eight-arm PEG-hydrazine to DOX via its ketone
transition uses NIR-absorbing nanostructures embedded within moiety and covalently entrapped PEG-DOX conjugates into the
the matrix of hydrogels based on thermoresponsive polymers. polymer matrix. The resulting hydrogel provides an excellent
Specifically, a light-responsive polysaccharide-based hydrogel platform for sustained long-term delivery of DOX, with 81.33%
was obtained by dispersing small CuS NPs in a gellan aqueous of the conjugate being released after 40 days at pH 6.4. Given
solution at elevated temperature, followed by mixing with that variable pH ranges (still acidic but other than 6.5–6.9) are
DOX and cooling to room temperature. After IT injection of described for tumor tissues, one can expect this subtle change
the hydrogel in mice, CuS NPs transformed irradiated NIR light in the pH value not to impair the stimuli responsiveness of the
(808 nm, 1.0 W/cm2, 3 min) into heating beyond the UCST of hydrogel. There are also slight differences between the reported
gellan, which elicited a slow gel–sol transition and increased values of other stimuli, such as redox species or enzyme
the release of DOX at the tumor site.42 Instead, the release rate concentrations.
of DOX can be precisely modulated by light activation of black
phosphorus nanosheets integrating an agarose hydrogel that Redox-responsive hydrogels
undergoes reversible hydrolysis and softening under irradiation The basic principle of designing reduction-responsive hydrogels
with an 808-nm laser.43 Departing from the use of inorganic is the incorporation of disulfide linkages directly into polymers

www.drugdiscoverytoday.com 5
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
POST-SCREEN (GREY) Drug Discovery Today d Volume xx, Number xx d xxx 2021

or via disulfide-containing crosslinkers, namely cystamine and One possible strategy to produce oxidation-responsive hydro-
its derivatives.50 Disulfide bonds can be cleaved in the presence gels involves thioketal linkages that are readily cleaved by ROS
of a reducing agent, such as GSH, which, acting as an electron into thiol and ketone groups. Xu et al.54 fabricated a L-
donor, is oxidized to glutathione disulfide. Zou et al.51 also phenylalanine-based low-molecular-weight gelator containing
described self-assembled hydrogels based on a reducible dipheny- thioketal and a control gelator without ROS-cleavable bonds.
lalanine dipeptide derivative for IT delivery of 5-aminolevulinic The corresponding hydrogels coloaded with DOX and a photo-
acid hydrochloride prodrug. Circular dichroism spectra of 10- sensitizer were intratumorally injected in 4T1 breast
mM GSH-treated hydrogels suggested a change in their sec- tumor = bearing mice to assess their in vivo antitumor efficacy.
ondary structure because of the reduction of disulfide bonds in Compared with the groups treated with drug-loaded ROS-
the dipeptide. Alternatively, diselenide bonds are expected to responsive gels, those receiving IT injections of control gels
impart more sensitivity than disulfide linkages because of their (without thioketal) showed little inhibition of tumor growth,
POST-SCREEN

lower bond energy. Generally, diselenide bonds can be cleaved owing to the absence of response to a ROS-rich
through the action of both reductants and oxidants (e.g., hydro- microenvironment.
gen peroxide, H2O2), being reduced to selenol or oxidized to sele-
ninic acid. Considering the potential of diselenide-containing Enzyme-responsive hydrogels
hydrogels for a dual redox response, Gong et al.52 produced an Enzyme responsiveness is usually introduced into hydrogels by
injectable PEG hydrogel via coupling reaction between the N- means of enzyme-mediated crosslinkages or enzyme-cleavable
hydroxysuccinimide ester ends of four-arm PEG and amine moieties.
groups of the selenocystamine crosslinker. Under the stimuli of The enzymatic degradation of hydrogels for IT drug adminis-
low concentrated GSH (0.1 mg/ml) or H2O2 (0.01%) solutions, tration requires the incorporation of an enzyme substrate or sub-
the breakage of diselenide bonds in selenocystamine led to dis- strate mimic, which can be: (i) the polymer itself in the case of
ruption of the crosslinked network and fast release of the encap- biodegradable polymers, such as polysaccharides and polyesters,
sulated drug. through the breakage of glycosidic and ester bonds, respectively;
Oxidation-responsive hydrogels depend on ROS to enable a or (ii) a peptide linker specifically recognized and cleaved by
concurrent behavior of hydrogel degradation and drug release. MMPs overexpressed in many tumor tissues. To date, 23 MMPs
These oxidative species can be generated endogenously (e.g., dur- have been identified in humans and their concentration in
ing oxidative phosphorylation in the mitochondria) or exoge- tumor tissues varies according to the type of cancer. For example,
nously (e.g., using a photosensitizer in photodynamic therapy). MMP-9 and MMP-2 are reported to be the most abundant in
In addition to oxygen free radicals, namely superoxide (O2 ) breast cancer, in a concentration of 32.27 and 17.08 ng/mg,
and hydroxyl radical (OH), ROS include nonradical species, such respectively.55 Li et al.56 prepared DOX-encapsulated PDLLA-
H2O2, usually ranging from 100 mM to 1 mM in tumor cells.53 In PEG-PDLLA micelles and selected a peptide linker containing
vitro studies are typically performed with H2O2 in PBS solutions an MMP-2-labile sequence (GPQGIWGQ) to develop an in situ
to create an oxidative environment. crosslinked MMP-responsive HA hydrogel. According to in vitro
TABLE 2
a
Dual and multiple stimuli-responsive injectable hydrogels for IT drug delivery.
Stimuli Composition Highlights Refs
Temperature; pH PDMAEMA, DOX, 131
I- PDMAEMA solutions transformed into nanogels at 37 °C 60
BSA At pH 5.8, electrostatic repulsion within polymer network led to swelling and fast in vitro
release of DOX
CS-g-(mPEG-b-PCL), Sol–gel phase transition occurred at pH 6.9 and physiological temperature 61
DOX, Curcumin CS-g-(mPEG-b-PCL) hydrogels released both hydrophilic and hydrophobic drugs in sustained
manner for up to 2 weeks
P(NIPAM-co-IA), CS/b- Incorporation of hydrophilic moiety (IA) elevated LCST of PNIPAM toward body temperature 62
GP, DOX Swollen state of cationic hydrogel quickened release of DOX
pH; enzyme; light; Gela NP, PDA NP, DOX GdL-induced acidification caused charge reversal of Gela NP to form colloidal Gela/PDA 63
temperature hydrogel
Degradation of Gela NP by proteases anticipated responsiveness to MMP-2/9 and enzymatic
promotion of DOX release
PDA NP converted NIR light into heat, eliciting gel–sol transition and increased release of DOX
pH; ROS; GSH; light; HSF, Cy7, DOX HSF molecules self-assembled into hydrogel via physical forces 64
temperature Acidity and ROS damaged b-sheet structure, whereas GSH cleaved intra- and intermolecular
disulfide bonds in HSF molecules
Hyperthermia generated by Cy7 upon NIR irradiation enhanced molecular thermal motion,
disrupting physical interactions within hydrogel
HSF hydrogels responded to multiple stimuli that together increased release rate of DOX
a
Abbreviations: CS-g-(mPEG-b-PCL), chitosan-graft-(poly(ethylene glycol) methyl ether-block-poly(e caprolactone)); CS/b-GP, chitosan/b-glycerophosphate; Cy7, cyanine-7; DOX, doxorubicin; GdL,
glucono delta-lactone; Gela, gelatin; GSH, glutathione; HSF, hydrophilic silk fibroin; 131I-BSA, iodine-131-labeled bovine serum albumin; IA, itaconic acid; LCST, lower critical solution temperature;
MMP, matrix metalloproteinase; NIR, near-infrared; NP, nanoparticles; PDA, polydopamine; PDMAEMA, poly(N,N-dimethyl aminoethyl methacrylate); PNIPAM, poly(N-isopropylacrylamide); P(NIPAM-
co-IA), poly(N-isopropylacrylamide-co-itaconic acid); ROS, reactive oxygen species.

6 www.drugdiscoverytoday.com
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
Drug Discovery Today d Volume xx, Number xx d xxxx 2021 POST-SCREEN (GREY)

release studies in the presence of MMP-2 (1.5 lg/ml), diffusion uli in human tumors might be similar, but not the same as those
and degradation by peptide cleavage were implicated in the observed in the laboratory.
release of DOX. Meanwhile, a HS-MMP-SH peptide crosslinker To understand the latter, one should consider that the in vitro
(GCREG-PQGIWGQ-ERCG) was introduced in another HA conditions sometimes fail to mimic the real environment of
hydrogel, further loaded with nano DOX and nano indocyanine tumors. In particular, if enzymes (e.g., MMPs) or biomolecules
green (photosensitizer) for chemophototherapy of head and (e.g., GSH or ROS) are used in higher concentrations than those
neck squamous cell carcinoma.57 reported for tumor tissues, a decrease in the sensitivity of the
A second prerequisite for enzymatic degradation is the acces- hydrogel is likely to be observed in vivo. Also, the contribution
sibility of enzymes to substrate within the hydrogel because it of concomitant stimuli to the hydrogel response is often
can considerably interfere with the kinetics of enzyme- neglected. Even when the in vitro studies are performed under
catalyzed reactions.58 This can be illustrated by a pentablock pathophysiological conditions, endogenous cues are somewhat

POST-SCREEN
copolymer (denoted as L-Pep-NC) synthesized from a L- different between individuals or at different stages of cancer,
peptide-PEG macroinitiator to link core-crosslinked micelles which interferes with the reproducibility of the response
and yield an injectable enzyme-responsive hydrogel. The peptide in vivo. Accordingly, additional efforts should be made to set
midblock was cleaved after 160 min of exposure to a type IV col- the range of values (e.g., pH, concentration, etc.) that trigger a
lagenase (0.5 units/ml), used as a model enzyme for MMP-2 and desirable response for each stimuli-responsive hydrogel and
MMP-9. By contrast, it took 7 days for the hydrogel crosslinked develop novel hydrogels that can change dynamically with sub-
with L-Pep-NC to be completely degraded at an enzyme concen- tle variations in the environment. Nevertheless, one can predict
tration of 30 units/ml, probably because of the limited access of the next generation of stimuli-responsive hydrogels to be multi-
collagenase to peptide block in hydrogel network.59 ple stimuli-responsive hydrogels that allow for independent tun-
ing of the individual response to each stimulus. In turn, the
Dual and multiple stimuli-responsive hydrogels combined effect of multiple stimuli would result in more precise
Dual or multiple stimuli-responsive hydrogels respond in a speci- and programmable drug delivery and, ultimately, a better thera-
fic and distinct manner to two or more stimuli available in the peutic effect.
TME and/or assisted by an external source. Compared with their Finally, most tumor models have been established by subcuta-
single stimulus-responsive counterparts, these hydrogels offer neous inoculation of human cancer cells in mice, which do not
greater control over their responsive behavior and an opportu- reflect the TME accurately. Instead, orthotopic mouse models
nity to improve overall performance and applicability to IT drug would be more clinically relevant because tumor xenografts are
delivery. placed into the tissue/organ of origin. Therefore, future preclini-
Some examples of dual and multiple stimuli-responsive inject- cal studies should be conducted in orthotopic tumor models and
able hydrogels are presented in Table 2. one can expect encouraging results to inspire further evaluation
of these formulations in clinical trials.
Concluding remarks
A better understanding of the TME, together with the advance- Declaration of Competing Interest
ments in materials chemistry, provided a solid background for The authors declare that they have no known competing
the design of stimuli-responsive hydrogels for IT drug delivery. financial interests or personal relationships that could have
Their proof-of-concept has been demonstrated by growth inhibi- appeared to influence the work reported in this paper.
tion of several tumors in different mouse models. However, sev-
eral obstacles to clinical translation remain because: (i) some
materials lack enough biocompatibility and biodegradability or Acknowledgments
need further research into these properties; (ii) the synthetic This work was supported by the Applied Molecular Bio-
routes of some polymers are too complex for industrial scale-up sciences Unit-UCIBIO, which is financed by national funds from
production; and (iii) the responsiveness of hydrogels to the stim- FCT/MCTES (UID/Multi/04378/2019).

References
1 P. Pittayapruek, J. Meephansan, O. Prapapan, M. Komine, M. Ohtsuki, Role of 6 K.M. Bailey, J.W. Wojtkowiak, H.H. Cornnell, M.C. Ribeiro, Y. Balagurunathan, A.
matrix metalloproteinases in photoaging and photocarcinogenesis, Int J Mol Sci I. Hashim, et al., Mechanisms of buffer therapy resistance, Neoplasia 16 (2014)
17 (2016) 868. 354–364.
2 W.L. Chen, C.C. Wang, Y.J. Lin, C.P. Wu, C.H. Hsieh, Cycling hypoxia induces 7 A. Fakhari, Subramony J. Anand, Engineered in-situ depot-forming hydrogels for
chemoresistance through the activation of reactive oxygen species-mediated B- intratumoral drug delivery, J Control Release 220 (2015) 465–475.
cell lymphoma extra-long pathway in glioblastoma multiforme, J Transl Med 13 8 M.H. Chen, L.L. Wang, J.J. Chung, Y.H. Kim, P. Atluri, J.A. Burdick, Methods to
(2015) 389. assess shear–thinning hydrogels for application as injectable biomaterials, ACS
3 H. Wang, H. Jiang, M. Van de Gucht, M. De Ridder, Hypoxic radioresistance: can Biomater Sci Eng 3 (2017) 3146–3160.
ROS be the key to overcome It?, Cancers 11 (2019) 112 9 T. Thambi, Y. Li, D.S. Lee, Injectable hydrogels for sustained release of therapeutic
4 G. Boysen, The glutathione conundrum: Stoichiometric disconnect between its agents, J Control Release 267 (2017) 57–66.
formation and oxidative stress, Chem Res Toxicol 30 (2017) 1113–1116. 10 W.B. Liechty, D.R. Kryscio, B.V. Slaughter, N.A. Peppas, Polymers for drug
5 A.P. Andersen, J.M. Moreira, S.F. Pedersen, Interactions of ion transporters and delivery systems, Annu Rev Chem Biomol Eng 1 (2010) 149–173.
channels with cancer cell metabolism and the tumour microenvironment, Philos 11 V.A. Ganesh, A. Baji, S. Ramakrishna, Smart functional polymers – a new route
Trans R Soc Lond B Biol Sci 369 (2014) 20130098. towards creating a sustainable environment, RSC Adv 4 (2014) 53352–53364.

www.drugdiscoverytoday.com 7
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
POST-SCREEN (GREY) Drug Discovery Today d Volume xx, Number xx d xxx 2021

12 D.D. Chakraborty, L.K. Nath, P. Chakraborty, Recent progress in smart polymers: 33 S. Xu, X. Du, G. Feng, Y. Zhang, J. Li, B. Lin, et al., Efficient inhibition of cervical
behavior, mechanistic understanding and application, Polym Plast Technol Eng cancer by dual drugs loaded in biodegradable thermosensitive hydrogel
57 (2018) 945–957. composites, Oncotarget 9 (2018) 282–292.
13 L. Klouda, Thermoresponsive hydrogels in biomedical applications: a seven-year 34 M. Babaei, J. Davoodi, R. Dehghan, M. Zahiri, K. Abnous, S.M. Taghdisi, et al.,
update, Eur J Pharm Biopharm 97 (2015) 338–349. Thermosensitive composite hydrogel incorporated with curcumin-loaded
14 P. Bragta, R.K. Sidhu, K. Jyoti, A. Baldi, U.K. Jain, R. Chandra, J. Mada, et al., nanopolymersomes for prolonged and localized treatment of glioma, J Drug
Intratumoral administration of carboplatin bearing poly (e-caprolactone) Deliv Sci Technol 59 (2020) 101885.
nanoparticles amalgamated with in situ gel tendered augmented drug delivery, 35 I. Tomatsu, K. Peng, A. Kros, Photoresponsive hydrogels for biomedical
cytotoxicity, and apoptosis in melanoma tumor, Colloids Surf B 166 (2018) 339– applications, Adv Drug Deliv Rev 63 (2011) 1257–1266.
348. 36 D. Zhao, Q. Tang, Q. Zhou, K. Peng, H. Yang, X. Zhang, A photo-degradable
15 J.I. Pesoa, M.J. Rico, V.R. Rozados, O.G. Scharovsky, J.A. Luna, L.N. Mengatto, injectable self-healing hydrogel based on star poly(ethylene glycol)-b-polypeptide
Paclitaxel delivery system based on poly(lactide-co-glycolide) microparticles and as a potential pharmaceuticals delivery carrier, Soft Matter 14 (2018) 7420–7428.
chitosan thermo–sensitive gel for mammary adenocarcinoma treatment, J Pharm 37 A. Barhoumi, Q. Liu, D.S. Kohane, Ultraviolet light-mediated drug delivery:
Pharmacol 70 (2018) 1494–1502. principles, applications, and challenges, J Control Release 219 (2015) 31–42.
POST-SCREEN

16 X. Han, X. Meng, Z. Wu, Z. Wu, X. Qi, Dynamic imine bond cross-linked self- 38 Y. Chen, Y. Hao, Y. Huang, W. Wu, X. Liu, Y. Li, et al., An injectable, near-
healing thermosensitive hydrogels for sustained anticancer therapy via infrared light-responsive click cross-linked azobenzene hydrogel for breast cancer
intratumoral injection, Mater Sci Eng C Mater Biol Appl 93 (2018) 1064–1072. chemotherapy, J Biomed Nanotechnol 15 (2019) 1923–1936.
17 H.R. Lin, Y.T. Chen, Y.C. Wu, Y.J. Lin, Glycol chitin/PAA hydrogel composite 39 X. Wang, Z. Xuan, X. Zhu, H. Sun, J. Li, Z. Xie, Near-infrared photoresponsive
incorporated bio-functionalized PLGA microspheres intended for sustained drug delivery nanosystems for cancer photo-chemotherapy, J
release of anticancer drug through intratumoral injection, J Biomater Sci Polym Nanobiotechnology 18 (2020) 108.
Ed 29 (2018) 1839–1858. 40 C. Ruan, C. Liu, H. Hu, X.L. Guo, B.P. Jiang, H. Liang, et al., NIR-II light-
18 Y.T. Fong, C.H. Chen, J.P. Chen, Intratumoral delivery of doxorubicin on folate– modulated thermosensitive hydrogel for light–triggered cisplatin release and
conjugated graphene oxide by in-situ forming thermo-sensitive hydrogel for repeatable chemo-photothermal therapy, Chem Sci 10 (2019) 4699–4706.
breast cancer therapy, Nanomaterials 7 (2017) 388. 41 C. Liu, X. Guo, C. Ruan, H. Hu, B.P. Jiang, H. Liang, et al., An injectable
19 C.K. Chung, J. García-Couce, Y. Campos, D. Kralisch, K. Bierau, A. Chan, et al., thermosensitive photothermal-network hydrogel for near-infrared-triggered drug
Doxorubicin loaded poloxamer thermosensitive hydrogels: chemical, delivery and synergistic photothermal–chemotherapy, Acta Biomater 96 (2019)
pharmacological and biological evaluation, Molecules 25 (2020) 2219. 281–294.
20 Y. Huo, Q. Wang, Y. Liu, J. Wang, Q. Li, Z. Li, et al., A temperature-sensitive 42 Y. Zheng, Y. Liang, D. Zhang, Z. Zhou, J. Li, X. Sun, et al., Fabrication of injectable
phase-change hydrogel of topotecan achieves a long-term sustained antitumor CuS nanocomposite hydrogels based on UCST-type polysaccharides for NIR-
effect on retinoblastoma cells, OncoTargets Ther 12 (2019) 6069–6082. triggered chemo-photothermal therapy, Chem Commun 54 (2018) 13805–
21 M. Yu, C. Zhang, Z. Tang, X. Tang, H. Xu, Intratumoral injection of gels 13808.
containing losartan microspheres and (PLG-g-mPEG)-cisplatin nanoparticles 43 M. Qiu, D. Wang, W. Liang, L. Liu, Y. Zhang, X. Chen, et al., Novel concept of the
improves drug penetration, retention and anti-tumor activity, Cancer Lett 442 smart NIR-light-controlled drug release of black phosphorus nanostructure for
(2019) 396–408. cancer therapy, Proc Natl Acad Sci U S A 115 (2018) 501–506.
22 R. Bai, X. Deng, Q. Wu, X. Cao, T. Ye, S. Wang, Liposome-loaded thermo- 44 M. Hou, R. Yang, L. Zhang, L. Zhang, G. Liu, Z. Xu, et al., Injectable and natural
sensitive hydrogel for stabilization of SN-38 via intratumoral injection: humic acid/agarose hybrid hydrogel for localized light-driven photothermal
Optimization, characterization, and antitumor activity, Pharm Dev Technol 23 ablation and chemotherapy of cancer, ACS Biomater Sci Eng 4 (2018) 4266–4277.
(2018) 106–115. 45 C. Wang, N. Zhao, W. Yuan, NIR/thermoresponsive injectable self-healing
23 M. Xu, Y. Mou, M. Hu, W. Dong, X. Su, R. Wu, et al., Evaluation of micelles hydrogels containing polydopamine nanoparticles for efficient synergistic
incorporated into thermosensitive hydrogels for intratumoral delivery and cancer thermochemotherapy, ACS Appl Mater Interfaces 12 (2020) 9118–9131.
controlled release of docetaxel: a dual approach for in situ treatment of tumors, 46 M. Norouzi, B. Nazari, D.W. Miller, Injectable hydrogel-based drug delivery
Asian J Pharm Sci 13 (2018) 373–382. systems for local cancer therapy, Drug Discov Today 21 (2016) 1835–1849.
24 M. Rezazadeh, V. Akbari, E. Amuaghae, J. Emami, Preparation and 47 M. Rizwan, R. Yahya, A. Hassan, M. Yar, A.D. Azzahari, V. Selvanathan, et al., pH
characterization of an injectable thermosensitive hydrogel for simultaneous Sensitive hydrogels in drug delivery: brief history, properties, swelling, and
delivery of paclitaxel and doxorubicin, Res Pharm Sci 13 (2018) 181–191. release mechanism, material selection and applications, Polymers 9 (2017) 137.
25 M.H. Turabee, T.H. Jeong, P. Ramalingam, J.H. Kang, Y.T. Ko, N, N, N-trimethyl 48 F. Raza, Y. Zhu, L. Chen, X. You, J. Zhang, A. Khan, et al., Paclitaxel-loaded pH
chitosan embedded in situ Pluronic F127 hydrogel for the treatment of brain responsive hydrogel based on self-assembled peptides for tumor targeting,
tumor, Carbohydr Polym 203 (2019) 302–309. Biomater Sci 7 (2019) 2023–2036.
26 Kelly H, Duffy G, Rossi S, Hastings C. A thermo-responsive hydrogel for 49 P.K. Sharma, Y. Singh, Glyoxylic hydrazone linkage-based PEG hydrogels for
intratumoral administration as a treatment in solid tumor cancers. Royal covalent entrapment and controlled delivery of doxorubicin, Biomacromolecules
College of Surgeons in Ireland. EP3706718 20 (2019) 2174–2184.
27 K. Shi, B. Xue, Y. Jia, L. Yuan, R. Han, F. Yang, et al., Sustained co-delivery of 50 J. Zhang, X. Jiang, X. Wen, Q. Xu, H. Zeng, Y. Zhao, et al., Bio-responsive smart
gemcitabine and cis-platinum via biodegradable thermo-sensitive hydrogel for polymers and biomedical applications, J Phys Mater 2 (2019) 032004.
synergistic combination therapy of pancreatic cancer, Nano Res 12 (2019) 1389– 51 Q. Zou, R. Chang, R. Xing, C. Yuan, X. Yan, Injectable self-assembled bola-
1399. dipeptide hydrogels for sustained photodynamic prodrug delivery and enhanced
28 B. Xue, M. Lei, K. Shi, M. Wang, Y. Hao, Y. Xiao, et al., Intratumoral injection of tumor therapy, J Control Release 319 (2020) 344–351.
norcantharidin-loaded poly(D, L–lactide)-b-poly(ethylene glycol)-b-poly(D, L- 52 C. Gong, M. Shan, B. Li, G. Wu, Injectable dual redox responsive diselenide-
lactide) thermosensitive hydrogel for the treatment of primary hepatocellular containing poly(ethylene glycol) hydrogel, J Biomed Mater Res A 105 (2017)
carcinoma, J Biomed Nanotechnol 15 (2019) 2025–2044. 2451–2460.
29 H.F. Darge, A.T. Andrgie, E.Y. Hanurry, Y.S. Birhan, T.W. Mekonnen, H.Y. Chou, 53 D. Wang, R. Shi, J. Zhou, S. Shi, H. Wu, P. Xu, et al., Photo-enhanced singlet
et al., Localized controlled release of bevacizumab and doxorubicin by thermo- oxygen generation of Prussian blue-based nanocatalyst for augmented
sensitive hydrogel for normalization of tumor vasculature and to enhance the photodynamic therapy, iScience 9 (2018) 14–26.
efficacy of chemotherapy, Int J Pharm 572 (2019) 118799. 54 L. Xu, M. Zhao, Y. Yang, Y. Liang, C. Sun, W. Gao, et al., A reactive oxygen species
30 X. Zhou, X. He, K. Shi, L. Yuan, Y. Yang, Q. Liu, et al., Injectable thermosensitive (ROS)-responsive low molecular weight gel co-loaded with doxorubicin and Zn(ii)
hydrogel containing erlotinib–loaded hollow mesoporous silica nanoparticles as phthalocyanine tetrasulfonic acid for combined chemo-photodynamic therapy, J
a localized drug delivery system for NSCLC therapy, Adv Sci 7 (2020) 2001442. Mater Chem B 5 (2017) 9157–9164.
31 W. Shen, X. Chen, J. Luan, D. Wang, L. Yu, J. Ding, Sustained codelivery of 55 K.J. Isaacson, M. Martin Jensen, N.B. Subrahmanyam, H. Ghandehari, Matrix–
cisplatin and paclitaxel via an injectable prodrug hydrogel for ovarian cancer metalloproteinases as targets for controlled delivery in cancer: An analysis of
treatment, ACS Appl Mater Interfaces 9 (2017) 40031–40046. upregulation and expression, J Control Release 259 (2017) 62–75.
32 D. Meng, H. Lei, X. Zheng, Y. Han, R. Sun, D. Zhao, et al., A temperature-sensitive 56 W. Li, C. Tao, J. Wang, Y. Le, J. Zhang, MMP-responsive in situ forming hydrogel
phase-change hydrogel of tamoxifen achieves the long-acting antitumor loaded with doxorubicin-encapsulated biodegradable micelles for local
activation on breast cancer cells, OncoTargets Ther 12 (2019) 3919–3931. chemotherapy of oral squamous cell carcinoma, RSC Adv 9 (2019) 31264–31273.

8 www.drugdiscoverytoday.com
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012
Drug Discovery Today d Volume xx, Number xx d xxxx 2021 POST-SCREEN (GREY)

57 H.H. Wang, Z.G. Fu, W. Li, Y.X. Li, L.S. Zhao, L. Wen, et al., The synthesis and temperature suitable for tumor acidic environment, Carbohydr Polym 198 (2018)
application of nano doxorubicin–indocyanine green matrix metalloproteinase- 486–494.
responsive hydrogel in chemophototherapy for head and neck squamous cell 62 M. Fathi, M. Alami-Milani, M.H. Geranmayeh, J. Barar, H. Erfan-Niya, Y. Omidi,
carcinoma, Int J Nanomed 14 (2019) 623–638. Dual thermo-and pH-sensitive injectable hydrogels of chitosan/(poly(N-
58 R. Chandrawati, Enzyme-responsive polymer hydrogels for therapeutic delivery, isopropylacrylamide-co-itaconic acid)) for doxorubicin delivery in breast cancer,
Exp Biol Med 241 (2016) 972–979. Int J Biol Macromol 128 (2019) 957–964.
59 M. Najafi, H. Asadi, J. van den Dikkenberg, M.J. van Steenbergen, M.H.A.M. Fens, 63 G. He, S. Chen, Y. Xu, Z. Miao, Y. Ma, H. Qian, et al., Charge reversal induced
W.E. Hennink, et al., Conversion of an injectable MMP–degradable hydrogel into colloidal hydrogel acts as a multi–stimuli responsive drug delivery platform for
core-cross-linked micelles, Biomacromolecules 21 (2020) 1739–1751. synergistic cancer therapy, Mater Horizons 6 (2019) 711–716.
60 D. Maiti, Y. Chao, Z. Dong, X. Yi, J. He, Z. Liu, et al., Development of a 64 S. Gou, D. Xie, Y. Ma, Y. Huang, F. Dai, C. Wang, et al., Injectable, thixotropic,
thermosensitive protein conjugated nanogel for enhanced radio-chemotherapy and multiresponsive silk fibroin hydrogel for localized and synergistic tumor
of cancer, Nanoscale 10 (2018) 13976–13985. therapy, ACS Biomater Sci Eng 6 (2020) 1052–1063.
61 N. Jommanee, C. Chanthad, K. Manokruang, Preparation of injectable hydrogels
from temperature and pH responsive grafted chitosan with tuned gelation

POST-SCREEN

www.drugdiscoverytoday.com 9
Please cite this article in press as: A.C. Marques et al., Drug Discovery Today (2021), https://doi.org/10.1016/j.drudis.2021.04.012

You might also like