ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 8, pp. 1362-1391 © The Author(s) 2024. This article is an open access publication.
1362
REVIEW
Development of Graphene-Based Materials
with the Targeted Action for Cancer Theranostics
Konstantin N. Semenov
1,2,3,a
*, Olga S. Shemchuk
1,2
, Sergei V. Ageev
1,2
,
Pavel A. Andoskin
1
, Gleb O. Iurev
1
, Igor V. Murin
2
, Pavel K. Kozhukhov
2
,
Dmitriy N. Maystrenko
3
, Oleg E. Molchanov
3
, Dilafruz K. Kholmurodova
4
,
Jasur A. Rizaev
4
, and Vladimir V. Sharoyko
1,2,3,b
*
1
Pavlov First Saint Petersburg State Medical University, 197022 Saint Petersburg, Russia
2
Saint Petersburg State University, 199034 Saint Petersburg, Russia
3
Granov Russian Research Centre for Radiology and Surgical Technologies, 197758 Saint Petersburg, Russia
4
Samarkand Medical University, 100400 Samarkand, Uzbekistan
a
e-mail: knsemenov@gmail.com 
b
e-mail: sharoyko@gmail.com
Received May 30, 2024
Revised July 11, 2024
Accepted July 13, 2024
AbstractThe review summarises the prospects in the application of graphene and graphene-based nanomate-
rials (GBNs) in nanomedicine, including drug delivery, photothermal and photodynamic therapy, and theranos-
tics in cancer treatment. The application of GBNs in various areas of science and medicine is due to the unique
properties of graphene allowing the development of novel ground-breaking biomedical applications. The review
describes current approaches to the production of new targeting graphene-based biomedical agents for the che-
motherapy, photothermal therapy, and photodynamic therapy of tumors. Analysis of publications and FDA data-
bases showed that despite numerous clinical studies of graphene-based materials conducted worldwide, there is
a lack of information on the clinical trials on the use of graphene-based conjugates for the targeted drug delivery
and diagnostics. The review will be helpful for researchers working in development of carbon nanostructures,
material science, medicinal chemistry, and nanobiomedicine.
DOI: 10.1134/S0006297924080029
Keywords: graphene, graphene-based nanomaterials, drug delivery, theranostics
Abbreviations: GBNs, graphene-based nanomaterials;
CYT, cytostatic drug cytarabine; FA, folic acid; GQDs,
graphene quantum dots; HAS, human serum albumin;
Pc,phthalocyanine; PEG,polyethylene glycol; PDT,photo-
dynamic therapy; PTT,photothermal therapy; rGO,reduced
graphene oxide; ROS,reactive oxygen species.
* To whom correspondence should be addressed.
INTRODUCTION
Graphene-based nanomaterials (GBNs), such as
graphene, graphene oxide(GO), reduced graphene ox-
ide(rGO), and graphene quantum dots(GQDs) (Fig. 1),
attract a significant attention due to their structure
and physicochemical properties. Some of the prom-
ising applications of GBNs in the field of biomedicine
include tissue engineering [1], bioimaging [2, 3] tar-
geted drug delivery [4-9], development of biosensors
[10-12] and antiviral [13-16], antibacterial [17-20],
and antifungal agents [21, 22], and delivery of biomol-
ecules, such as enzymes [23], proteins [24-26], genes
[27-29], RNA [30, 31], and DNA [32, 33] (Fig.2).
GBNs can be modified by covalent [34, 35] and
noncovalent [36, 37] functionalization to enhance their
electrical [38, 39], optical [40, 41], thermal [42, 43],
electronic [44-46], and mechanical [47, 48] proper-
ties. Monolayer graphene was first obtained in 2004
by Andre Geim and Konstantin Novoselov [49]. De-
pending on the method of synthesis, graphene can
be produced as mono- or multilayered flakes [50, 51].
It can be synthesized by chemical vapour deposition
[52-58], electrochemical exfoliation [59-62], mechano-
chemical exfoliation [63], and chemical and thermal
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1363
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Fig. 1. Classification of GBNs.
reduction of GO (synthesis of rGO) [64-70]. rGO is a
GO derivative in which almost all oxygen-containing
groups are reduced with hydrazine hydrate or biomol-
ecules [71] (see Fig. S1 in the Online Resource 1 for rGO
synthesis from GO using l-cysteine [71]).
Graphene consists of sp
2
-hybridised hexagonal
carbon atoms that form two-dimensional nanolayers,
while GO additionally has oxygen-containing groups
on the surface, e.g., carbonyl, lactol, and carboxyl
groups at the edges of GO layers and epoxy and hy-
droxyl groups on the basal plane (Fig. S2 in the Online
Resource 1) [72-74].
GBNs can be functionalized with molecules of var-
ious nature due to the presence of functional groups
on the GO surface and sp
2
-hybridised carbon atoms.
Reactions that can be carried out on the GO surface
(Fig. 3) include amidation, esterification, 1,3-dipolar
cycloaddition, and halogenation. Other types of inter-
actions are hydrogen bonding, π–π stacking, and hy-
drophobic interactions.
GQDs are graphene nanoparticles less than 100 nm
in size. Due to their exceptional properties, such as low
toxicity, stable photoluminescence, chemical stability,
and pronounced quantum confinement effect, GQDs
are considered as new promising materials for biolo-
gy, optoelectronics, energy industry, and environment
[75-78]. GQDs can be prepared using top-down or bot-
tom-up approaches (Fig.4) [79-81].
GBN CONJUGATES IN BIOMEDICINE
GBNs can be effectively used in the antitumor
therapy, e.g., for the development of platforms for the
delivery of drugs and genetic constructs, photodynam-
ic therapy (PDT), photothermal therapy (PTT), and ther-
anostics (Fig.5).
To efficacy of GBN-based antitumor nanodrugs
can be increased by using specific vectors for their de-
livery that are developed to recognise tumor-specific
receptors, such as HER2, CAIX, and receptors for Tat,
LHRH, folate, biotin, and asialoglycoprotein (Fig.6).
BIOCOMPATIBILITY
AND MECHANISMS OF ENDOCYTOSIS
Analysis of publications shows that function-
alization of graphene surface decreases hemolysis
and, therefore, increases material hemocompatibility.
Fig. 2. Publications on GBN applications.
SEMENOV et al.1364
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Fig. 3. Reactions carried out on graphene surface.
Fig. 4. Approaches for GQD synthesis: top-down degradation from various carbon sources and bottom-up synthesis from small
molecules or polymers.
Thus, noncovalent functionalization of GO with chi-
tosan produced a material with no hemolytic activity.
Pintoetal. [82] showed that the noncovalent function-
alization of graphene surface with polymers [polyvinyl
alcohol, polyethylene glycol (PEG), polyvinylpyrroli-
done (PVP), hydroxyethylcellulose, chondroitin, glucos-
amine, and hyaluronic acid (HA)] decreased hemolysis
to 1.7% for all the resulting materials at concentra-
tions below 500μg·ml
–1
. Previously, we have studied
the effect of GO enriched (about 85%) with oxygen-
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1365
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Fig. 5. Application of GBNs in cancer treatment.
containing functional groups (edge-oxidized graphene
oxide, EOGO) on the extent of spontaneous hemolysis
and found that within the studied concentration range
(C
GO
= 2.5-25mg·liter
–1
), this nanomaterial did not af-
fect the level of hemolysis after 1 and 3 h of incuba-
tion [83], while, as demonstrated in [84, 85], GO with a
lower content of oxygen-containing functional groups
(C/O ratio, 2 : 1) caused the rupture of erythrocyte
membranes with subsequent release of hemoglobin.
Our research group also showed that GO functional-
ized with L-methionine (GFM) [85], L-cysteine (GFC)
[86], glycine (GO-Gly) [87], or folic acid (GO-FA) [88]
caused no damage to the erythrocyte membrane at the
concentrations up to 25μg·ml
–1
.
In comparison with GO, GO functionalized with
amino groups caused no activation of platelet aggrega-
tion up to C= 2 µg·ml
–1
. The authors showed that GO-in-
duced aggregation was stronger than the thrombin-in-
duced aggregation [89]. Podolska etal. [90] found that
GO, rGO, and rGO-PEG (C= 50 μg mL
–1
) did not stimu-
late platelet aggregation in the presence of 2 μmol·ml
–1
adenosine diphosphate (ADP). GFC (up to 25 µg·liter
–1
)
caused no ADP-induced stimulation of platelet aggre-
gation, while GFM and EOGO demonstrated the anti-
platelet activity at the concentrations up to 25 and
100 µg·liter
–1
, respectively, in experiments on ADP- and
collagen-induced aggregation.
Ding et al. [91] showed that GO (dispersion con-
centration, C= 100 μg·ml
–1
) interacted with human
serum albumin (HSA) through various types of inter-
actions (covalent and hydrogen bonding, electrostatic
forces, hydrophobic interactions, and π–π stacking)
that resulted in the HSA dysfunction and its inability to
remove toxins due to conformational changes, which
indicated a potential toxicity of GO. Functionalization
of the GO surface with carboxyl groups (GO-COOH)
increased its biocompatibility, as GO-COOH caused no
functional changes in HSA. In contrast, Taneva et al.
[92] found that interaction of GO (8 mg·ml
–1
) with HSA
did not inactivate HSA in the blood plasma because
of the low affinity of GO for HSA. We demonstrated
that interaction of modified GO (GFM and GFC) with
HSA occurred mainly due to the formation of hydro-
gen bonds: the dissociation constants for the GFM and
GFC complexes with HSA were 185.2 [85] and 1600 [86]
μg·ml
–1
, respectively.
SEMENOV et al.1366
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Fig. 6. Tumor-specific receptors and ligands used in GBN modification.
Liu et al. [93] found that GO at the concentrations
up to 100 μg ml
–1
induced mutagenesis due to its effect
on DNA replication and gene expression. Wang etal.
[94] reported that GO (up to 100 μg·ml
−1
) displayed a
significant genotoxicity toward human lung fibroblasts
because of the DNA damage resulting from the gen-
eration of reactive oxygen species (ROS) and surface
charge of GO. The authors showed that functionaliza-
tion of the GO surface with PEG and lactobionic acid
(LA) significantly reduced the genotoxicity.
Akhavan et al. [95] showed that the genotoxicity
depends on the lateral dimensions of graphene: rGO
nanoparticles with an average lateral dimension of
11 ± 4 nm were able to penetrate into the nuclei of
human mesenchymal stem cells, leading to DNA frag-
mentation and chromosomal aberrations even at low
rGO concentrations (0.1 and 1.0 mg·ml
–1
) after 1 h of
incubation. At the same time, rGO sheets with an av-
erage lateral size of 3.8 ± 0.4 µm did not exhibit geno-
toxicity at a concentration of 100 mg·ml
–1
after 24-h
incubation. Our research group showed that GFM and
GFC did not display genotoxicity at the concentra-
tions up to 25 μg·ml
–1
, while EOGO did not exhibit the
genotoxic effect up to C= 100 μg·ml
–1
. We also studied
the mechanism of endocytosis of GO conjugates with
1,3,5-triazine-based cytostatic drugs and showed that
the transport of these conjugates could occur via two
mechanisms– pinocytosis and clathrin-dependent en-
docytosis [96].
The possibility of selective delivery of the cytostat-
ic drug cytarabine (CYT) was shown in [88]. Using a
conjugate of GO with CYT and folic acid (FA) as a vec-
tor molecule, our research group demonstrated that
the GO-FA-CYT nanoparticles localized in the vicinity of
folate receptor-expressing pancreatic carcinoma cells
(PANC-1) (Fig.7).
DRUG DELIVERY,
PHOTOTHERMAL THERAPY (PTT),
AND PHOTODYNAMIC THERAPY (PDT)
Below, we will discuss the use of GBNs in tumor
chemotherapy. GBNs can be conjugated with anti-
cancer drugs by noncovalent functionalization of the
graphene surface (see Table1).
GBNs exhibit a high photothermal conversion ef-
ficiency, i.e., they efficiently convert absorbed light
into heat. In particular, they can absorb light in the
near-infrared (NIR) region, which is a transparency
region for biological tissues (750-1700 nm), thus allow-
ing deep tissue heating [118]. Such localized heating
can selectively damage or destroy cancer cells in PTT,
representing a minimally invasive medical treatment.
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1367
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Fig. 7. Fluorescence microscopic images of folate receptor-expressing PANC-1 cells incubated with GO-FA-CYT(a) and GO(b).
Thesize of GBNs promotes their permeability, reten-
tion, and selective clustering at the tumor loci [119].
Table2 summarises information on the use of GBNs in
chemotherapy and PTT.
GO is a highly efficient nanomaterial for PDT,
since its irradiation in the NIR region results in the for-
mation of ROS insitu, leading to tumor ablation. The
presence of functional groups (epoxy, carbonyl, car-
boxyl, and hydroxyl) on the GO surface allows to load
it with drugs, including photosensitisers, which greatly
enhances the efficacy of PDT. GQDs have a significant
singlet oxygen quantum yield. Because of their prop-
erties, such as suitability for bioimaging, drug loading
capacity, and high therapeutic efficacy in PDT, they
can be used as a multifunctional nanoplatform in ther-
anostics. These properties also create the possibility of
using GBNs in the treatment of cancer. Table 3 sum-
marises the data on the use of GBNs in PDT.
DESIGN OF GBN-BASED
THERANOSTIC APPROACHES
Hatamie et al. [161] synthesized GO/cobalt nano-
composites for inducing magnetic fluid hyperthermia
(MFH) and as contrast agents in magnetic resonance
imaging (MRI) [162]. The composites were obtained
by chemical synthesis (using GO as a source material)
and assembly of 15-nm cobalt nanoparticles; the con-
centration of cobalt in the nanocomposites was80%.
The studies of hyperthermia induction showed a su-
perior conversion of electromagnetic energy into
heat at a frequency of 350 kHz for the nanocompos-
ite dispersions with the concentrations of 0.01 and
0.005 g/liter. MRI showed that negatively charged GO/
cobalt nanocomposites were suitable for T1-weighted
imaging.
Su et al. [163] engineered a noncovalent based
mitomicine C–graphene–BODIPY (4,4-difluoro-4-bora-
3a,4a-diaza-s-indacene)–mPEG (MGBP) nanoconjugate
that ensured extensive ROS production and high pho-
tothermal conversion efficiency (48%) and demonstrat-
ed an excellent therapeutic efficacy in vitro (decreased
HeLa cell viability to 17%). Apart from the synergis-
tic photo/chemo therapy, MGBP can be used in flu-
orescence and photothermal dual-mode imaging, as
BODIPY emits fluorescence when exposed to laser irra-
diation (see Fig. S3 in the Online Resource 1 for the use
of MGBP in theranostics).
Taratula et al. [164] reported a novel cancer-tar-
geting nanoplatform for imaging and treatment of
unresected ovarian cancer tumors by intraoperative
multimodal phototherapy. To develop this theranostic
system, low-oxygen-containing graphene nanosheets
were chemically modified with polypropylenimine
dendrimers loaded with phthalocyanine(Pc) as a pho-
tosensitiser. Such molecular design prevented the
quenching of Pc fluorescence by graphene nanosheets,
providing the possibility of fluorescence imaging. Fur-
thermore, the developed nanoplatform was conjugated
with PEG to improve its biocompatibility and with lu-
teinising hormone-releasing hormone (LHRH) peptide
for the tumor-targeted delivery (Fig. S4 in the Online
Resource 1). Notably, a low-power NIR irradiation at a
single wavelength was used for both heat generation by
the graphene nanosheets (PTT) and ROS production by
Pc (PDT). Such combinatorial phototherapy resulted in
an enhanced destruction of ovarian cancer cells, with
a killing efficacy of 90-95% at low doses of Pc and low-
oxygen-containing graphene, presumably, due to the
synergistic cytotoxic effect of generated ROS and mild
hyperthermia. In vivo studies confirmed that Pc loaded
into this nanoplatform can be employed as a NIR flu-
orescence agent for the imaging-guided drug delivery.
SEMENOV et al.1368
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 1. Cytotoxicity of GBN conjugates with cytostatic drugs in in vitro experiments
Type of carbon nanoconjugate Drug load
Cell lines or
type of cancer
Applied concentrations and IC
50
or cytotoxicity
(approx. %) at the highest concentration
References
GO with FA-modified β-cyclodextrin (CD)
(GO–FA–β-CD)
doxorubicin (DOX), 168% HeLa 71% inhibition [97]
Ultrasmall nano-GO (NGO) with covalent
grafting of PEG and covalently conjugated
B cell-specific antibody Rituxan (RB)
(NGO–PEG–RB)
DOX, 40% Raji 80% inhibition [98]
GO with adriamycin (ADR)
(GO–ADR)
ADR, 93.6%
MCF-7
MCF-7/ADR
IC
50
= 1.28 ± 0.26 µg/ml (MCF-7)
IC
50
= 13.95 ± 0.53 µg/ml (MCF-7/ADR)
[99]
Hybrid of graphene nanosheets (GNSs),
carbon nanotubes (CNTs), and iron oxide
nanoparticles (GNS–CNT–Fe
3
O
4
)
0.27 mg/mg at 5-fluorouracil
(5-FU) concentration
of 0.5 mg/ml
HepG2 28% viability at 80 µg/ml [100]
Polyethyleneimine (PEI)-functionalized GO
(GO–PEI, covalent)
n/a HeLa
IC
50
= 1.3 μg/ml (GO–PEI/scrambled siRNA)
IC
50
= 1.3 μg/ml (GO–PEI/Bcl-2-targeting siRNA)
[101]
NGO–PEG (noncovalent)
1 g of NGO–PEG loaded
~0.1 g of SN-38 (7-ethyl-
10- hydroxycamptothecin)
HCT-116 IC
50
= 6 nM [102]
NGO covalently modified with
diazonium salt of p-aminobenzenesulfonic
acid (NGO–SO
3
H)
DOX, more than 400% MCF-7
NGO-SO
3
H-DOX
Relative viability of 75% after 48h at 20µg/ml
(interms of DOX)
[103]
NGO covalently modified with diazonium
salt of p-aminobenzenesulfonic acid
and FA (NGO-FA)
DOX, more than 400%;
camptothecin (CPT) 4.5 %
MCF-7
NGO-FA-DOX
Relative viability of 30% after 48h at 20µg/ml
(in terms of DOX)
NGO-FA-CPT/DOX
Relative viability of 80 % after 48h at 200ng/ml
(in terms of CPT)
NGO-FA-CPT
Relative viability of 80 % after 48 h at 200 ng/ml
(in terms of CPT)
[104]
GO–chlorotoxin (CTX)
(GO–CTX, noncovalent)
570 mg DOX
per 1g of GO–CTX
C6
C = 1-5 μg/ml
% of cytotoxicity, 60%
[104]
GO–sodium alginate (SA)
(GO–SA, covalent)
1.8 mg of DOX
per 1mg of GO–SA
HeLa
C = 5-20 μg/ml
% of cytotoxicity, 69%
[105]
GO nanoplatelets (GONPs), 50 × 50 nm
2
Cisplatin (CP) loading was
not determined
A549
C = 2.5-30 μg/ml
% of cytotoxicity = 90%
[106]
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1369
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 1 (cont.)
Type of carbon nanoconjugate Drug load
Cell lines or
type of cancer
Applied concentrations and IC
50
or cytotoxicity
(approx. %) at the highest concentration
References
GO–PEG–FA (noncovalent) CPT, 45% MCF-7
C = 20-100 μg/ml
% of cytotoxicity, 76%
[107]
GO–Fe
3
O
4
–β-CD (covalent)
DOX, 37.4%;
methotrexate(MTX), 23.4%
K562
C = 2-16 μg/ml;
% of cytotoxicity (DOX), 65%
% of cytotoxicity (MTX), 55%
[108]
GO–PEG–FA (noncovalent)
protocatechuic
acid (PCA), 23.47%;
chlorogenic
acid (CA), 18.33%
HT29, HepG2
C = 1.56-100 μg/ml
% of cytotoxicity (HT29), 58%;
IC
50
(HT29) = 50.69 μg/ml
% of cytotoxicity (HepG2), 61%
IC
50
(HepG2) = 40.39 μg/ml
[109]
GO–FA–bovine serum albumin (BSA)
(covalent)
DOX, 437.43μg
per 1mg of GO–FA–BSA
MCF-7
(FA-receptor-
positive)
A549
(FA-receptor-
negative)
C = (0.01-20) μg/ml
IC
50
(MCF-7, 24 h) = 8.9 ± 0.7 μg/ml
IC
50
(MCF-7, 48 h) = 0.048 ± 0.010 μg/ml
IC
50
(A549, 24 h) = 5.3 ± 0.7 μg/ml
IC
50
(A549, 48 h) = 0.279 ± 0.037 μg/ml
[110]
Pegylated folate-
and peptide (Pep)-decorated GO
GO–Pep–PEG–FA (covalent)
CPT, 90% HeLa IC
50
= 3.1 μM [111]
GQD–carboxymethyl cellulose (CMC)
hydrogel
(GQD–CMC)
DOX loading depended
on the GQD dose:
GQD(10%)–CMC, ~ 4.5%%;
GQD(20%)–CMC, ~5.5%%;
GQD(30%)–CMC, ~6%
K562
C = 2-32 μg/ml
IC
50
(CMC/DOX) = 6.1 μg/ml
IC
50
(GQD 10%/CMC/DOX) = 5.7 μg/ml
IC
50
(GQD 20%/CMC/DOX) = 5.4 μg/ml
IC
50
(GQD 30%/CMC/DOX) = 5.1 μg/ml
[112]
GO–PVP and GO–β-CD
0.17 g of SN-38
per 1g of GO–PVP;
0.14g of SN-38
per 1g of GO–β-CD
MCF-7
IC
50
(GO–PVP–SN-38) = 97 μM
IC
50
(GO–β-CD–SN-38) = 170 μM
[113]
GO–DOX DOX, 87%
HEK293
A549
PA-1
IC
50
(HEK293) = 3.13 µM
IC
50
(A549) = 3.84 µM
IC
50
(PA1) = 3.35 µM
IC
50
(T98G) = 11.80 µM
IC
50
(SK-HEP-1) = 4.11 µM
[114]
SEMENOV et al.1370
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 1 (cont.)
Type of carbon nanoconjugate Drug load
Cell lines or
type of cancer
Applied concentrations and IC
50
or cytotoxicity
(approx. %) at the highest concentration
References
Multifunctional nanocomposite
of PEGylated GO with Pt(IV) complex
(NGO–PEG–Pt)
(c,c,t-[Pt(NH
3
)
2
Cl
2
(OH)
2
])
Pt(IV) content
in PEG–NGO–Pt varied
from 0.1 to 100mM,
which corresponded to the
concentration of NGO–PEG
(from 0.89 to 890mg/ml)
T98G
SK-HEP-1
HeLa
IC
50
(24 h) = 13.88 µM
IC
50
(48 h) = 6.01 µM
IC
50
(72 h) = 3.56 µM
[115]
DOX–loaded aptamer–GO aggregate (GA)
(GA–DNA–DOX)
DOX–loaded aptamer–GO complex (GC)
(GC–DNA–DOX)
DOX, ~10% (wt/wt GO) HeLa
40% inhibition for dispersion containing 4μg/ml
GA–DNA–DOX (in relation to DOX), 48h
50% inhibition for dispersion containing 4μg/ml
GC–DNA–DOX (in relation to DOX), 48h
[116]
GO covalently modified with TNF-related
apoptosis-inducing ligand (TRAIL)
conjugated with furin-cleavable peptide
via PEG linker and noncovalently
modified with DOX
(fGO–TRAIL–DOX)
DOX, up to 43.8%%
A549 IC
50
(48h) = 14ng/ml (in relation to TRAIL)
[117]
furin-
deficient
LoVo cells
IC
50
(48h) = 759ng/ml (in relation to DOX)
GO covalently modified with TRAIL
without furin-cleavable peptide
and noncovalently modified with DOX
(nGO–TRAIL–DOX)
n/a
A549 IC
50
= 33 ng/ml (in relation to TRAIL) [118]
furin-
deficient
LoVo cells
IC
50
= 884 ng/ml (in relation to DOX)
Table 2. Application of GBNs in chemotherapy and PTT
GBN Heat source; energy Cancer model References
Spark-generated carboxylic group-activated GO
(CGO)-coated hollow mesoporous silica nanoparticles
(HMSNs) loaded with topotecan (TPT)
(HMSN–NH
2
–TPT–CGO); TPT loading, 36wt.%
NIR irradiation (808nm); 2.0W/cm
2
, 5min
MDA-MB-231
(human breast cancer cell line)
[120]
results: minimal cytotoxicity was observed after treatment with blank nanoparticles; cell viability
with HMSN–NH
2
–CGO and CGO was ~91 and ~85%, respectively. After NIR irradiation, cell viability
after treatment with 200μg/ml HMSN–NH
2
–CGO decreased to ~53%. Cell viability was markedly
decreased after treatment with a low dose of HMSN–NH
2
–TPT–CGO (after exposure to NIR
irradiation), showing its advantage over treatment with free TPT
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1371
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 2 (cont.)
GBN Heat source; energy Cancer model References
GO nanoparticles modified with a conjugate
of the photothermal agent IR820 with LA
and loaded with DOX; the contents of DOX, IR820-LA,
and GO in the GO/DOX/IR820-LA nanohybrids
were 22, 42, and 36wt. %, respectively
NIR (808 nm) or visible (660nm)
irradiation; 1.0W/cm
2
, 5 min
Hep G2/Hep 1-6
(human/murine liver cells)
[121]
results: both free drug and GO/DOX/IR820-LA nanohybrids exhibited a concentration-dependent
antitumor effect. The inhibitory effect of GO/DOX/IR820-LA after laser irradiation was significantly
higher than the effects of DOX without laser irradiation and of IR820 and IR820-LA individually
after laser irradiation. To evaluate the photothermal capability of the GO/DOX/IR820-LA
nanohybrids and IR820 invivo, Hep 1-6 tumor mice were irradiated 6h after GO/DOX/IR820-LA
injection (with saline as a control). In the animals treated with the GO/DOX/IR820-LA nanohybrids,
the temperature rose to 52.9°C and the tumor growth was inhibited by 88.0%
rGO-based nanocomposites functionalized
with poly(allylamine hydro-chloride) (rGO–PAH)
and loaded with DOX; DOX loading, 36 wt. %
NIR irradiation (808 nm);
6.0W/cm
2
, 20min
MCF-7 [122]
results: the nanocomposites demonstrated a highly efficient synergistic chemo-photothermal effect
and at a concentration 5μg/ml caused the death of ~94% MCF-7 cells due to extensive ROS
generation, chromosome compression, and DNA disintegration
Nanohydrogel composed of chondroitin sulphate
multiple aldehyde (CSMA), branched polyethylenimine
(BPEI) and BPEI-conjugated graphene (BPEI–GO);
DOX loading, 60.1 wt. %
NIR irradiation (808 nm); 2.5W/cm
2
, 5min MCF-7 [123]
results: the synergetic chemo-photothermal effect promoted cell death, with 37.8, 22.8, and 9.2%
cell survival on days 1, 2, and 3 invitro. In animal treated with the DOX-loaded hydrogel,
the cancer recurred ~7 days later than in the animals treated with DOX only, indicating
that DOX loading into hydrogels provides sustainable drug release and prolonged cytotoxicity.
The combined chemo-photothermal treatment strategy yielded better results,
with only two out of six mice (33.3%) developing recurrent cancer
GO nanoparticles loaded with wedelolactone (WED)
and indocyanine green (ICG) on the surface;
WED loading, 84.91wt. %
NIR irradiation (808 nm); 2.0W/cm
2
, 1min HeLa [124]
results: cell viability in the presence of ICG–Wed–GO after laser irradiation was 12.65%.
In treated mice, the tumors reduced gradually and completely disappeared on day 10
GO nanoparticles functionalized with an amphiphilic
polymer based on poly(2-ethyl-2-oxazoline) (POx)
and co-loaded with DOX and d-α-tocopherol
succinate (TOS); DOX loading, 70 wt. %
NIR irradiation (808 nm); 1.7W/cm
2
, 5min MCF-7 [125]
results: combined application of DOX:TOS-loaded POx–GO and laser irradiation reduced
the viability of MCF-7 cells to 39%
GO nanoparticles conjugated poly(l-lysine) (GO–PLL)
deposited on cationic liposomes encapsulating DOX;
DOX encapsulation efficiency, 86.4 ± 4.7 wt. %
NIR irradiation (808 nm); 1.5 W/cm
2
, 2 min MDA-MB-231 [126]
results: MTT and live/dead cellular viability assays suggested that nanoconjugate ensured a dual
mode chemotherapy/PTT action. NIR light absorbed by GO and GO–PLL shells was converted to
heat and activated the gel leading to the liquid phase transition of the liposomal membrane and
release of encapsulated DOX. Alternatively, light absorbed by GO and GO–PLL provided the PTT
effect and killed cancer cells (cell viability was less than 20% at the DOX concentration of 5µg/ml)
SEMENOV et al.1372
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 2 (cont.)
GBN Heat source; energy Cancer model References
Nanoparticles consisting of histidine (His)-modified
amorphous zinc oxide (aZnO) shell on gold
nanoparticles (AuNPs) (AuNP–His–aZnO), integrated
onto the planar structure of PEGylated GO (PEG–GO);
DOX loading, 25 wt. %
NIR irradiation (808 nm); 1.0 W/cm
2
,
10 min or 1.5 W/cm
2
, 5min
A549 [127]
results: the synergetic effect observed at the nanoconjugate concentration of 100μg/ml upon laser
irradiation resulted in almost 100% cell death
GO nanoparticles coated with FA and AuNPs
and loaded with DOX; DOX loading, 72.4 ± 2.5 wt. %
NIR irradiation (808 nm); 0.2 W/cm
2
;
5 min or 2.0 W/cm
2
; 5 min
MCF-7/HeLa [128]
results: the viability of MCF-7 and HeLa cells at the nanoconjugate concentration of 10μg/ml
was 32.5 and 33.9%, respectively. A significant reduction in the tumor volume was observed
in mice treated with DOX–GO–AuNPs and DOX–FA–GO–AuNPs (by 27 and 35%, respectively,
on day 21). Exposure of mice treated with DOX–GO–AuNPs and DOX–FA–GO–AuNPs
to NIR irradiation caused the photothermal effect resulting in even more pronounced tumor
reduction to 32 and 43%, respectively, on the 21st day of study
Nanoplatform consisting of ruthenium nitrosyl func-
tionalized N-doped GQDs and a triphenylphosphonium
moiety and loaded with nitric oxide (NO)
NIR irradiation (808 nm); 0.6 W/cm
2
,
10 min or 1.0 W/cm
2
, 10 min
HeLa [129]
results: the nanoplatform was fluorescence-trackable and capable of targeting mitochondria
in cancer cells. Irradiation of cells led to NO release and photothermal effect, resulting
in elimination of tumor cells both invitro (reduction in cell viability to ~10%) and invivo
(pronounced inhibition of tumor growth)
Mesoporous silica (MS)-coated polydopamine-
functionalized rGO (prGO) further modified with HA
and loaded with DOX; DOX loading, 145 ± 25 wt. %
NIR irradiation (808 nm); 1.5 W/cm
2
, 5 min HeLa [130]
results: DOX-loaded prGO–MS–HA nanocomposites produced a significant synergistic
chemotherapy/PTT effect upon NIR laser irradiation (cell viability, less than 20%)
and strongly suppressed of tumor growth invivo
GO-hybridised nanogels with alginate loaded with DOX;
DOX loading, 97.2 ± 1.2 wt. %
NIR irradiation (808 nm); 4 min A549 [131]
results: the antitumor cytotoxicity was enhanced by irradiation with an 808-nm laser
(cell viability decreased from 64.0 ± 3.6 to 39.6 ± 5.7%). The temperature of the nanogel aqueous
solution increased from 25 to 50°C
Amino-modified GO (A–GO)
NIR irradiation (808 nm); 6 W/cm
2
, 6 min
HSC-3 (oral squamous cell carcinoma
cell line)
[132]
results: Invitro, A–GO (15 μg/ml) reduced the viability of HSC-3 cells to 5% upon irradiation
(temperature increased to 58.4°C). A–GO strongly reduced the tumor size to 25% of the initial size
in 1 out of 4 mice and completely ablated tumors in 3 out of 4 mice
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1373
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 2 (cont.)
GBN Heat source; energy Cancer model References
Dopamine (DOPA)-reduced GO functionalized
with sulfobetaine methacrylate (SB) brushes
and loaded with IR780 (IR780/SB/DOPA-rGO)
NIR irradiation (808 nm);
1.7W/cm
2
, 10 min
MCF-7 [133]
results: IR780/SB/DOPA–rGO elicited no cytotoxicity invitro (cell viability, >78%). In contrast,
a combination of IR780/SB/DOPA–rGO with NIR light decreased the viability of breast cancer
cells to 21%
DOPA-reduced GO covalently bound
with HA and loaded with DOX (DOX/HA–DOPA–rGO);
DOX loading, 91.2 wt. %
NIR irradiation (808 nm);
1.7W/cm
2
, 5 min
MCF-7 [134]
results: a combination of DOX/HA–DOPA–rGO with NIR light reduced cells viability to 23%
Ultrasmall GO (UGO) (average size, 30nm) loaded
with DOX; DOX loading, 52wt.%
NIR irradiation (808 nm);
1.5W/cm
2
, 300s
LO2 (human papillomavirus-related
cervical adenocarcinoma cell line)
[94]
results: a combination of enhanced chemotherapy and PTT induced cell death (cell viability, <15%)
in invitro cell tests and suppressed tumor growth in animals
MTX encapsulated into mesoporous silica
nanoparticles (MSNs) by polydopamine (PDA)
and embedded into GO nanosheets loaded
with naringin (NAR) and cystamine (CYS)
NIR irradiation, 300 min Saos-2 (osteosarcoma cells) [135]
results: in the absence of NIR irradiation, the IC
50
for NAR/CYS/MTX/MSNs@PDA@GO
and MTX/MSNs@PDA@GO were 10.3 and 27.5 mg/ml, respectively. Apparently, a significantly lower
IC
50
of NAR/CYS/MTX/MSNs@PDA@GO was due to the synergistic effect of the two drugs (MTX
and NAR). Under NIR irradiation, the IC
50
of NAR/CYS/MTX/MSNs@PDA@GO decreased further
to 3.2 mg/ml, which can be attributed to the synergistic effect of chemotherapy and PTT
Table 3. The use of GBNs in PDT
Nanomaterial Conjugated
substance
Irradiation
characteristics
Cancer model Result References
Methylene blue (MB)–GO MB red diode radiation MDA-MB-231
MB–GO (C = 20 mg/ml)
reduced cell viability by 80%
[136]
GO–MB/pluronic F127 (PF127) MB, PF127
LED irradiation
at 660 nm
SiHa (squamous
cell carcinoma)
GO–MB/PF127 (C = 10 μg/ml)
reduced cell viability by 75%
[137]
Pyropheophorbidea (PPa)–
NGO– monoclonal antibody
against αvβ3 integrin (mAb)
mAb, PPa
laser irradiation
at 30J/cm
2
for 5 min
U-87 MG (human
glioblastoma)
PPa–NGO–mAb (C = 1.5 μg/ml)
reduced cell viability by 70%
[138]
NGO– methoxy PEG (mPEG)/
zinc phthalocyanine (ZnPc)
ZnPc, mPEG
laser irradiation
at 60J/cm
2
for 5 min
MCF-7
NGO–mPEG/ZnPc (C = 60 mg/l)
reduced cell viability by 35%
[139]
SEMENOV et al.1374
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 3 (cont.)
Nanomaterial Conjugated
substance
Irradiation
characteristics
Cancer model Result References
rGO–ZnO–HA ZnO, HA
irradiation at 365 nm,
5mW/cm
2
for 30 min
MDA-MB-231
rGO–ZnO–HA (C = 50 μg/ml)
reduced cell viability by 50%
[140]
MB/GQDs and methylene
violet (MV)/GQDs
MB or MV
irradiation at 660 nm,
210mW/cm
2
for 5 min
MCF-7
MB/GQDs and MV/GQDs reduced cell
viability by 35% and 10-20%,
respectively, at C = 50 μg/ml
[141]
GQDs GDQs
irradiation at 365 nm
up to 5 min
MCF-7 and B16F10
mouse melanoma cells
GQDs reduced viability of MCF-7
and B16F10 cells by more than 90%
depending on concentration
[142]
GQDs GDQs
irradiation at 405
and 637 nm
HeLa
GQDs (C = 1.8 µM)
reduced cell viability by 80%
[143]
GO–PEG– chlorin e6 (Ce6) PEG, Ce6
irradiation at 660 nm,
0.1 W/cm
2
for 10 min
KB (human
nasopharyngeal
epidermal carcinoma)
GO–PEG–Ce6 (C = 0.011 mg/ml)
reduced cell viability by more than 95%
[144]
FA–GO–Ce6 FA, Ce6
irradiation at 632.8 nm
with a He-Ne laser
for 10 min
MGC803 (human
gastric carcinoma)
FA–GO–Ce6 (C = 100µM)
reduced cell viability by 90%
(FA–GO to Ce6 ratio, 1:1)
[145]
Hypocrellin B (HB)–GO HB
irradiation at 470nm
with He-Ne laser
for 10min
SMMC-7721 (human he-
patocellular carcinoma),
SGC-7901 (human gastric
cancer cell), HeLa, A549
HB–GO (HB–GO ratio, 1:1; C=5μM
in terms of HB) reduced cell viability
by 80 (SMMC-7721), 90 (SGC-7901),
75 (HeLa), and 80%, (A549)
[146]
Upconversion nanoparticles
(UCNPs)–NGO/ZnPc
UCNPs,
ZnPc,
and PEG
irradiation at 630 nm,
60 mW/cm
2
for 10 min
HeLa
UCNPs–NGO/ZnPc (C=320μg/ml)
reduced cell viability by more than 90%
[147]
GO–PEG–2-(1-hexyloxyethyl)-
2-devinylpyropheophorbide-
alpha (HPPH)
PEG, HPPH
irradiation at 671 nm,
8 mW/cm
2
for 3 min
4T1 (mouse mammary
carcinoma)
GO–PEG–HPPH (C = 1µM in terms
of HPPH) reduced cell viability
by more than 80%
[148]
GO–HA–Ce6 HA, Ce6
irradiation at 670nm,
50mW/cm
2
for 3min
A549
GO–HA–Ce6 reduced cell viability
by ~80% at C = 1.8 μM (Ce6)
[149]
GO–MB MB
portable continuous
wave diode laser system
655nm, 150mW/cm
2
HeLa
GO–MB reduced cell viability
by up to 50% at C = 10 μg/ml (GO)
and C = 2 μg/ml (MB)
[150]
Hypocrellin A (HcA)/SN-38/GO HcA, SN-38
irradiation at 470 nm,
25 mW for 5 min
A549
HA/SN-38/GO reduced cell viability
by ~95% at C = 6 µM (HA)
and C = 6 µM (SN-38)
[151]
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1375
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Table 3 (cont.)
Nanomaterial Conjugated
substance
Irradiation
characteristics
Cancer model Result References
Magnetic and fluorescent
graphene (MFG)–hydropho-
bic silicon naphthalocyanine
bis(trihexylsilyl oxide) (SiNc
4
)
Fluorescent
graphene,
SiNc
4
irradiation at 775nm,
0.3 W/cm
2
for 60 min
HeLa
MFG–SiNc
4
decreased cell viability
by ~98% at C = 100 μg/ml
[152]
GO–PEG– sodium
sinoporphyrin (DVDMS)
PEG, DVDMS
irradiation at 630nm,
2J per well
PC-9 (human non-small
cell lung carcinoma)
GO–PEG–DVDMS (C = 3 μg/ml)
reduced cell viability by up to 80%
(GO–PEG: DVDMS ratio, 1:2)
[153]
p-Nanographene oxide
(pGO)–CuS/ICG
copper (II)
sulfide, ICG
irradiation at 808nm,
4 W/cm
2
for 5 min
MCF-7
pGO–CuS/ICG reduced cell viability
by ~65% at C = 50 μg/ml
[154]
GO–PEG–DVDMS PEG, DVDMS
irradiation at 630nm,
radiation dose = 50 J
U-87 MG
GO–PEG–DVDMS (C = 5 μg/ml) reduced
cell viability by more than 90%
[155]
ZnPc–PEG–Au@GO
nanocolloid (GON)
nanoparticles (NPs)
ZnPc, Au,
PEG
irradiation at 660 nm,
0.2 W/cm
2
for 10 min
HeLa
ZnPc–PEG–Au@GON NPs (C = 1.2 nM;
ZnPc content, 2.8·10
−11
M) reduced
cell viability by 90%
[156]
GO–808
PEG,
branched PEI,
heptamethine
indocyanine
dye IR-808
irradiation at 808nm,
2W/cm
2
for 5min (PTT
and PDT)
A549
GO–808 (C = 10 µM in terms of IR-808)
led to a reduced cell viability by ~90%
[157]
GO–PEG–FA PEG, FA
irradiation at 808 nm,
320 mW/cm
2
for 15 min
B16F0 (rat melanoma)
GO–PEG–FA (C = 75 μg/ml) reduced
cell viability by 60%
[158]
Hollow magnetic
nanospheres (HMNSs)
coated with the silica
shells and conjugated
with carboxylated GQDs,
loaded with DOX
and stabilized with liposomes
(HMNS/SiO
2
/GQD-DOX)
HMNSs,
liposomes,
GQDs, SiO
2
,
DOX
irradiation at 671nm
for 20min
Eca-109 (human
oesophageal carcinoma)
LP-HMNS/SiO
2
/GQD-DOX (C = 0.5 mg/ml
for HMNSs, 0.2 mg/ml for GQDs,
and 0.3 mg/ml for DOX) decreased
cell viability by ~90%
[159]
GO/gold nanostars
(AuNSs)–PEG/Ce6
PEG, AuNSs,
Ce6
irradiation at 660nm,
2 W/cm
2
for 15 min
EMT6 (mammary
carcinoma cell lines)
GO/AuNS–PEG/Ce6 (C = 3μg/ml for Ce6,
150μg/ml for GO/AuNS–PEG) decreased
cell viability by up to 80%
[160]
GO–UCNPs–Ce6 UCNPs, Ce6
irradiation at 808nm
for 10min
HeLa
GO–UCNPs–Ce6 (C = 800μg/ml)
decreased cell viability by up to 85%
[161]
SEMENOV et al.1376
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Hence, the developed Pc-graphene nanoplatform has
a significant potential as an efficient NIR theranostic
probe for imaging and combinatorial phototherapy.
Lamb et al. [165] multifunctionalized graphene
nanoflakes (GNFs) with (i) peptide-based Glu–NH–C(O)–
NH–Lys ligand capable of binding prostate-specific
membrane antigen (PSMA), (ii) potent antimitotic drug
(R)-Ispinesib, (iii) chelator desferrioxamine B (DFO),
and (iv) albumin-binding tag used to extend the half-
life of the developed agent in vivo.
68
Ga-labelled con-
jugates were used in in vitro and in vivo experiments
to evaluate the performance of GNFs as a theranostic
agent (Fig.S5 in the Online Resource1).
Using the dose-response curves and flow cytometry
analysis, it was shown that GNFs loaded with (R)-Ispine-
sib inhibited the kinesin spindle protein (KSP) and in-
duced cell cycle arrest at the G2/M checkpoint. Experi-
ments on the cellular uptake and blocking demonstrated
that GNFs functionalized with the Glu–NH–C(O)–NH–Lys
ligand showed a specificity toward PSMA-expressing
cells (LNCaP cell line). The distribution profile and the
excretion rates of
68
Ga-labelled GNFs in athymic nude
mice were evaluated using the time–activity curves de-
rived by dynamic positron-emission tomography (PET).
Imaging experiments showed that GNFs demonstrated
low accumulation and retention in background tissues
and had a rapid renal clearance.
Tomasella et al. [166] used GO and reduced thiolat-
ed GO (rGOSH) as 2D substrates to fabricate nanocom-
posites with gold nanospheres (AuNSps) or nanorods
(AuNRs) via in situ reduction of the metal salt precur-
sor and seed-mediated growth processes. The plasmon-
ic sensing capability of the gold-decorated nanosheets
was evaluated by UV-visible spectroscopy. In vitro ex-
periments on the toxicity of the obtained nanocompos-
ites in human neuroblastoma SH-SY5Y cell indicated a
high potential of these hybrids as a plasmonic thera-
nostic platform.
Usman et al. [167] synthesized a bimodal GO-based
theranostic nanodelivery system using CA as an anti-
cancer agent, while Gd and AuNPs were used as con-
trast agents for MRI. CA and Gd were simultaneously
loaded on the GO nanolayers via hydrogen bonding and
π–π noncovalent interactions to form the GOGCA nano-
composite. Subsequently, AuNPs were doped on the
GOGCA surface by means of electrostatic interactions
(Fig. S6 in the Online Resource 1). Theefficacy (cytotox-
icity) of the resulting conjugate was demonstrated in
HepG2 hepatocellular carcinoma cells (IC
50
= 25 μg/ml).
At the same time, the conjugate displayed no toxicity
toward normal 3T3 fibroblasts. TheT1-weighted im-
ages of the conjugate obtained by MRI demonstrated
contrast enhancement in comparison with the conven-
tional MRI contrast agent Gd(NO
3
)
3
.
Chawda et al. [168] engineered rGO nanoparticles
decorated with Gd
3+
ions. The resulting Gd-containing
rGO nanosheets (Gd-rGONSs) were found to enhance
the loading of 5-FU (loading capacity, 34%) (Fig. S7 in
the Online Resource 1). The drug release was sustained
and reached ~92% within 72 h. Gd–rGONSs provided
a strong contrast in comparison to the optically re-
sponsive bare GO in the swept source optical coher-
ence tomography. The longitudinal relaxivity rate (r
1
)
for Gd–rGONSs at a magnetic field strength of 1.5 T
was 16.85 mM
−1
·s
−1
, which was four times higher
than that of the commercial contrast agent Magnevist
(4mM
−1
·s
−1
).
Samadian et al. [169] developed a drug delivery
nanosystem based on AuNPs, decorated PEG, and
FA-conjugated GO. Initially, the graphite powder was
oxidised to GO and then functionalized with chloroace-
tic acid to produce carboxylated graphene oxide (GO–
COOH). The obtained GO–COOH was functionalized
with the amine end-caped PEG, FA, and 3-amino-1-pro-
panethiol to produce GO–PEG–FA–SH. AuNPs were syn-
thesized through a citrate-mediated reduction and then
decorated onto/into GO–PEG–FA–SH through the forma-
tion of the Au–S bond to produce the GO–PEG–FA/AuNP
nanosystem (Fig.S8 in the Online Resource1).
The resulting nanosystem was loaded with
DOX·HCl (76 wt. %), and its drug-loading capacity and
pH-dependent drug release were investigated. The an-
ticancer activity of the developed theranostic agent
against MCF-7 cells was evaluated using the MTT assay
(IC
50
= 20 µg/ml after 24 h). This nanomaterial can also
be used in the chemotherapy/PTT therapy of solid tu-
mors due to the presence of AuNPs.
Yang et al. [170] developed a biocompatible HA–
glutathione (GSH) conjugate (HG) with stabilised gold
nanoclusters (AuNCs) combined with GO and loaded
with 5-FU (25.3 wt. %) as a novel theranostic platform
(HG–AuNC/GO–5-FU) [170]. This multifunctional nano-
material possessed an excellent fluorescence, photo-
sensitivity, and ability to specifically target cancer cell.
Moreover, in the presence of lysosomal hyaluronidase
(HAdase) and laser illumination, the recovery of fluo-
rescence and
1
O
2
and complete release of 5-FU could
be achieved, which allows the use HG–AuNC/GO–5-FU
in imaging, tumor chemotherapy, hyperthermia treat-
ment, and PDT. This multifunctional complex holds a
great potential as a versatile theranostic platform for
application in bioimaging-assisted cancer therapy.
Guo et al. [171] double-functionalized GO with FA
and Ce6 for combined targeted PTT/PDT against MCF-7
cells and RAW264.7 macrophages (Fig. S9 in the Online
Resource 1). GO–FA/Ce6 exhibited good photothermal
properties and high ROS-generating capacity.
This nanomaterial penetrated rapidly into cancer
cells via folate receptor-mediated endocytosis, as well
as into macrophages. A combination of PTT and PDT
allowed to increase the therapeutic efficiency against
MCF-7 cancer cells (cell death, up to 65%) compared
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1377
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
toindividual treatment. GO–FA/Ce6 also efficiently elim-
inated RAW 264.7 macrophages due to the effect of
PTT/PDT (cell death, up to 94%).
Baktash et al. [172] designed and optimized a
hybrid theranostic nanosystem by combining Fe
3
O
4
magnetic nanoparticles (MNPs) for imaging and chi-
tosan-grafted GO as a pH-sensitive smart nanocarrier
(chitosans with different molecular weights and at dif-
ferent concentrations were used) and investigated the
drug (DOX) loading and release properties, biocompat-
ibility, and magnetic characteristics of the developed
Fe
3
O
4
/GO/chitosan nanosystem. It was determined that
grafting of the concentrated high-molecular-weight
chitosan on MNPs/GO provided efficient drug release
and improved DOX loading. Studying the effects of GO
and chitosan on the magnetic behavior of the Fe
3
O
4
/
GO system showed that GO decreased the contrast ef-
ficiency of the MNPs, while grafting of MNP/GO with
hydrophobic chitosan enhanced the contrast, as was
seen from a sharp decrease in the r
1
relaxivity, which
is very desirable for MRI applications (the r
2
/r
1
value
for this composite was 28.95, while the r
2
/r
1
values for
Fe
3
O
4
/GO and Fe
3
O
4
were 6.37 and 14.66, correspond-
ingly). The cytotoxicity assay using L929 cells (normal
mouse adipose fibroblasts) revealed a high biocompat-
ibility of the MNP/GO/chitosan nanosystem. Further
assays carried out using MNP/GO/chitosan loaded with
DOX demonstrated an improved performance of MNP/
GO grafted with-low-molecular weight chitosan against
MCF-7 cells (cell viability was 39% at 4 μg/ml DOX vs.
53% in the presence of DOX only).
Pan et al. synthesized a covalent conjugate based
on GO and silicon phthalocyanine (SiPc) (Fig.S10 in the
Online Resource1) [173].
In vitro studies of the GO–SiPc conjugate in cells
showed that this nanomaterial synchronously caused
the photothermal effect, intracellular fluorescence, and
ROS generation. Efficient photoablation of cancer cells
could be triggered by either 671- or 808-nm lasers due
to the synergistic PTT/PDT or NIR photothermal effect,
respectively. When systemically administered to MCF-7
xenograft mice, GO–SiPc efficiently accumulated at the
tumor loci and strongly inhibited tumor growth after
laser irradiation.
Chen et al. [174] reported a novel approach to a
one-step fabrication of magnetic graphene hybrid
nanocomposites GO–PEG–γ-Fe
2
O
3
(GPFs) using pulsed
laser ablation in liquid method [174]. Due to their good
magnetic and photothermal performance, GPFs were
employed as nanotheranostic agents for the multimod-
al imaging-guided chemo/photothermal synergistic
therapy. The results of multifunctional in vivo imaging
confirmed the GPF uptake by the tumors after intra-
venous injection. Moreover, using the GPF–DOX conju-
gate allowed to achieve a superior synergistic antitu-
mor effect via combined chemotherapy/PTT. Figure S11
inthe Online Resource 1 presents a photograph of he-
patocellular carcinoma (H22)-bearing nude mice under
different treatments (Fig.S12 in the Online Resource 1
demonstrates the difference in the relative tumor vol-
ume after the treatment).
A multifunctional theranostic nanoplatform based
on GO and MnWO
4
was developed by in situ growth of
MnWO
4
nanoparticles onto GO surfaces in a PEG-con-
taining hyperthermia polyol medium [175]. In compar-
ison with GO and MnWO
4
/PEG, the NIR absorbance of
the GO/MnWO
4
/PEG nanocomposite was significantly
improved, resulting in an enhanced photothermal con-
version capability and good photoacoustic (PA) imag-
ing performance. In addition, the longitudinal relaxivity
r
1
of GO/MnWO
4
/PEG reached 11.34 mM
−1
·s
−1
in a 0.5-T
magnetic field, which was significantly higher than
for ordinary Mn(II)-based T1 agents. In vivo MRI and
PA imaging studies demonstrated that GO/MnWO
4
/PEG
could be used as an efficient bimodal contrast agent
to guide cancer treatment. GO/MnWO
4
/PEG showed a
high loading capacity for DOX (550 mg/g); the resulting
conjugate demonstrated a pronounced cytotoxic activ-
ity towards 4T1 (human breast carcinoma) and HUVEC
(human umbilical vein endothelial cells) cell lines. For
example, cells incubated with 100 µg/ml GO/MnWO
4
/
PEG/DOX (containing 5 µg/ml DOX) and then exposed
to laser irradiation showed the highest mortality rate
(about 90%) vs. 50% in the case of DOX (C= 5 µg/ml)
orGO/MnWO
4
/PEG.
Prasad et al. [176] reported the results of invivo
photo-triggered tumor regression induced by applica-
tion of a biodegradable red emissive nanotheranostic
composite based on liposomes fortified with GO flakes
and functionalized with FA (GO–Lipo–FA) and loaded
with DOX (Fig.S13 in the Online Resource1) [176].
The synthesized nanocomposite has a good aque-
ous dispersibility, quick photothermal response (54°C in
5 min), high biocompatibility, deep intracellular local-
ization, feasibility for 4T1 visualisation, and long-term
tumor-binding ability of the injected emissive nano-
hybrid. GO enhanced the stability of the drug-loaded
liposomes in the extracellular environment, which
prevented premature release of the loaded anticancer
drug from the liposomal cavity. In addition, the authors
demonstrated the developed nanocomposite caused tu-
mor regression (~300 to 25 mm
3
) in 4T1 Balb/c mice.
Foroushani et al. [177] developed a theranostic
system based on GO integrated with PDA, BSA, dieth-
ylenetriaminepentaacetic acid (DTPA)–Mn(II) contrast
agent, FA, and 5-FU for targeting CT-26 colon cancer
cells via folate receptors overexpressed on cancer cells.
According to the results of biodistribution assessment,
the conjugate was observed mainly in the tumors
and, therefore, provided highly efficient drug deliv-
ery to CT-26 cells. In vitro and in vivo MRI and thera-
py examination confirmed the ability of the conjugate
SEMENOV et al.1378
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
toenhance the contrast in tumor imaging (diagnostics)
and to inhibit the growth of cancer cells (therapy).
Luo et al. [178] proposed an easy method for
the synthesis of a theranostic agent based on super-
paramagnetic iron oxide nanoparticles loaded onto
GO nanosheets (SPIONs@GO) and cis-aconitic anhy-
dride-DOX prodrug (CAD) attached to the carboxylic
groups of GO through the 2-poly(amidoamine) dendrim-
er (G2.NH2) linker (Fig.S14 in the Online Resource1).
The release of DOX from the conjugate was pH-sen-
sitive: 66.91 ± 3.16% at pH 5.5 and 47.51 ± 1.87% at
pH 6.5 within 12 h. The viability of 4T1 cells after treat-
ment with CAD–SPIONs@GO for 24 h decreased cell vi-
ability from 93.8% to 38.3% at the DOX concentration
of 1.3-20 µM (similar to the treatment with freeDOX).
According to the results of biodistribution experiments,
4 h after injection, CAD–SPIONs@GO mainly localized to
the spleen and liver. The total Fe amount in all major
organs decreased greatly 12 h after injection, suggest-
ing that CAD–SPIONs@GO was cleared out of the body.
The authors proposed that the interface effect between
GO and in situ growth of SPIONs contributed to the sig-
nificant increase in the r
1
value and decrease in the
r
2
value. Invivo studies results confirmed a possibility
of conjugate application in high-resolution T1-weight-
ed MRI.
Shi et al. [179] synthesized a theranostic agent
based on rGO conjugated to the anti-CD105 antibody
(TRC105) and a complex of
64
Cu (PET label; half-life,
12.7 h) with 1,4,7-triazacyclononane-1,4,7-triacetic acid
(NOTA, chelator). Invivo experiments on the blockade of
the agent uptake by 4T1 cells with an excess of TRC105,
as well as flow cytometry and histology data, confirmed
the stability of
64
Cu–NOTA–rGO–TRC105 and its speci-
ficity for CD105 of the tumor vasculature. Noteworthy,
64
Cu–NOTA–RGO–TRC105 exhibited little extravasation
in 4T1 cells, indicating that targeting tumor vasculature
(instead of tumor cell) can be a valid and preferred
approach for the application of nanomaterials. Since
rGO can be used for PTT, the tumor-specific rGO con-
jugate may serve as a promising theranostic agent that
integrates imaging and therapeutic components.
Cheng et al. [180] developed a mild thermal an-
nealing procedure to induce blue fluorescence in GO
suspensions (Fig.S15 in the Online Resource1) [180].
The procedure preserved the oxygen functional groups,
which enabled conjugation of a cancer drug and re-
sulted in nontoxic and harmless nanomaterial. The
authors demonstrated the capability of GO to simulta-
neously act as a cellular imaging agent and a drug de-
livery agent in CT26 cancer cells without the need for
additional fluorescent protein labelling. The authors
also covalently annealed GO with CP (elemental con-
tent of Pt in the conjugate, ~3wt. %) and determined
that the annealed GO boosted the therapeutic perfor-
mance of CP in killing CT26 cancer cells.
Hu et al. [181] synthesized a new conjugate based
on rGO, PDA, and ICG for amplifying the PA imaging
and PTT effects for cancer phototheranostic (Fig. S16
in the Online Resource 1). The procedure for the
ICG–PDA–rGO preparation included the following
steps: (i)dopamine monomers were loaded on the GO
surface and spontaneously self-polymerised via the
Michael addition/Schiff reaction to form a PDA coat-
ing on the rGO surface, (ii)free ICG dye was absorbed
on the PDA–rGO surface via hydrogen bonds and π–π
stacking interactions.
ICG–PDA–rGO exhibited stronger PTT effect and
higher PA contrast than pure GO and PDA–rGO. After
PA imaging-guided PTT treatment, the tumors in 4T1
breast subcutaneous and orthotopic mice models were
suppressed completely; no treatment-induced toxicity
was observed.
Turcheniuk et al. [182] produced a theranostic
agent based on AuNRs coated with pegylated rGO
(AuNRs@rGO–PEG) and modified with sulfo-cyanine7
fluorescent dye (Cy7) and Tat protein (see Fig. S17 in
the Online Resource1).
Selective targeting of tumors was ensured by
specific interaction between the Tat protein and hu-
man glioblastoma astrocytoma cells (U87MG). Due to
the presence of NIR fluorescent dye integrated onto
the rGO shell, the conjugate acted as fluorescent cel-
lular marker. In vivo experiments in mice implanted
with U87MG cells showed that irradiation at 800nm
(0.7 W/cm
2
, 10 min) suppressed tumor growth after
5 days. Histological analysis of tumor tissues revealed
an active uptake of the nanoparticles by the tumor
stromal cells and selective damage of tumor vessels.
Wang et al. [183] synthesized a novel nanomate-
rial for the PTT/immunotherapy of cancer by the self-
assembly of oleate-capped Fe
3
O
4
nanoparticles (FNPs)
and rGO through electrostatic interaction, followed by
modification with PEG–NH
2
[182]. FNP/rGO–PEG nano-
composites can be used for the MRI-guided cancer
PTT/immunotherapy due to their excellent magnetic
properties. Under laser irradiation (805nm), FNP/rGO–
PEG improved the PTT efficacy by increasing the tem-
perature up to 60°C and killing 80% of 4T1 orthotopic
mouse breast tumor cells. In addition, FNP/rGO–PEG
nanocomposites could be used to stimulate immune
response by triggering the maturation of dendritic cells
(CD11c
+
CD86
+
) and secretion of cytokines (IL-12p70,
IL-6). Intratumoral injection of FNP/rGO–PEG nano-
composites in combination with NIR laser irradiation
significantly increased the median survival time of
tumor-bearing animals.
Bansal et al. [184] developed a theranostic agent
based on GQDs conjugated with a biosurfactant isolat-
ed from Candida parapsilosis through the amine-car-
boxyl coupling reaction and noncovalent modification
with FA. The obtained conjugate had a homogenous
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1379
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
dispersion and showed the photoluminescence proper-
ties and demonstrated enhanced uptake by cancerous
cells in comparison with non-modified GQDs. Inthe
MTT assay, the conjugate decreased the viability of
MCF-7 cells by more than 60% after 24 h of incubation
and by 75% after 48 h [184].
Ko et al. [185] synthesized GQDs for the diagnos-
tics and therapy of breast cancer via conjugation with
two precursors. DOX-disulfide-GQDs provided chemo-
therapy and PEG-disulfide-herceptin enhanced the
half-life and ensured the targeting of HER2 (Fig.S18 in
the Online Resource1) [184]. The cleavage of disulfide
links at a physiologically relevant glutathione concen-
tration in cancer cells provided controlled drug re-
lease. Theauthors demonstrated an enhanced cellular
uptake of the conjugate by SK-BR-3 cells (HER2-posi-
tive) in comparison with MDA-MB-231 cells (HER2-neg-
ative). As a result, the viability of SK-BR-3 cells was
significantly decreased (to <50%) at the conjugate con-
centration of 50mg/ml, whereas the viability of MDA-
MB-231 cells was reduced to >85%.
Iannazzo et al. [9] developed a novel conjugate
based on GQDs covalently modified with the tumor
targeting module biotin (BTN) and noncovalently mod-
ified with DOX (GQD-BTN-DOX, Fig.S19 in the Online
Resource 1), as well as the GQD-DOX conjugate [9]. The
DOX content in GQD-BTN-DOX and GQD-DOX was 16.6
and 17.8wt. %, respectively. GQD-DOX nanoparticles
were preferentially accumulated in the cytoplasm,
while DOX localized to the nuclei. At the same time,
GQD-BTN-DOX nanoparticles concentrated in the endo-
somal compartment after endocytosis-mediated inter-
nalisation. The cytotoxicity of GQD-BTN-DOX towards
A549 cells strongly depended on the uptake by the cells,
which was more pronounced and delayed for GQD-
BTN-DOX in comparison with GQD-DOX and DOX only.
Li et al. [186] synthesized a covalent GQD-FA con-
jugate and loaded it with IR780 iodide (33.19 wt. %)
via π–π stacking interactions (see Fig. S20 in the On-
line Resource 1). In vivo NIR fluorescence imaging and
biodistribution analysis demonstrated that in BALB/c
nude mice xenografted with HeLa cells, the conjugate
preferentially accumulated in the tumors. When irra-
diated with an 808-nm laser, IR780/GQDs-FA caused
hyperthermia (photothermal conversion efficiency,
87.9%) and induced apoptosis of cancer cells and tu-
mor necrosis, resulting in complete tumor disappear-
ance without relapse.
Ding et al. [187] developed a novel type of GQD-
based theranostic agent with a superior therapeutic
performance against 4T1 cancer cells both in in vitro
[IC
50
(theranostic agent) = 1.5 g/ml, IC
50
(DOX) = 4 g/ml]
and in vivo (the conjugate reduced the tumor volume
2.7 times more than DOX alone) due to the improved
tissue penetration and cellular uptake [187]. GQDs
were synthesized via facile chemical oxidation and
exfoliation technique using polyacrylonitrile carbon
fibres as a raw material. The NIR fluorescent molecule
Cy5.5 was covalently attached to GQDs via the cathep-
sin D-responsive peptide (Phe-Ala-Ala-Phe-Phe-Val-Leu-
Cys, P); functionalized GQDs were then loaded with
DOX via π–π interactions. The synthesized construct al-
lowed to track the delivery and release of the antican-
cer drug, as well as to monitor drug-induced apoptosis
of cancer cells through GQD, DOX, and Cy5.5 charac-
teristic fluorescence.
Badrigilan et al. [188] produced a theranostic agent
based on superparamagnetic iron oxide and bismuth
(III) oxide (Bi
2
O
3
) with GQDs for in vitro computed to-
mography (CT)/MR dual-mode bioimaging and PTT
(Fig.S21 in the Online Resource1).
The GQD-Fe/Bi nanocomposite had the following
advantages: (i) the photothermal conversion efficacy
was 31.8% with a high photostability upon irradiation
with a NIR 808-nm laser; (ii) photothermal ablation of
HeLa and MCF-7 cells invitro resulted in a significant
decrease in cell viability (~50% at 100µg/ml) in com-
parison with laser treatment only (3.0%); (iii) obtained
nanoparticles exhibited a superior X-ray attenuation
capability (175%) in comparison with Dotarem (mac-
rocyclic gadolinium-based contrast agent), as well as
showed a strong T2-relaxation shortening capability
(r
2
= 62.34mM
−1
·s
−1
) as a contrast agent for CT/MRI.
The same authors synthesized GQD-coated bis-
muth nanoparticles and assessed the possibility of
their application for CT imaging and PTT [189].
Lee et al. [190] developed rGQDs derived by rGO
top-down oxidation and HA-GQDs (HGQDs) that were
hydrothermally synthesized by the bottom-up method
[190]. The obtained nanomaterials possessed substan-
tial NIR absorption and fluorescence throughout the
visible and NIR regions, which is beneficial for in vivo
imaging. Aqueous dispersions of rGQDs and HGQDs
added to HeLa cells and irradiated with NIR laser
(λ = 808 nm, 0.9 W/cm
2
, 10 min) facilitated an increase in
temperature up to 54.5°C, leading to the decrease in the
HeLa cell viability from 80% for RGQDs (C = 1.5 mg/ml)
and 60% for HGQDs (C = 1.7 mg/ml) without irradiation
down to ~40% (RGQDs) and ~20% (HGQDs) after irra-
diation.
Sung et al. [191] synthesized a unique conjugate
composed of porous carbon/silica nanosponge encap-
sulated with GQDs loaded with docetaxel (DTX) via π–π
interactions; then, the particles were capped with the
red blood cell (RBC) membrane and cetuximab via fu-
sion (see Fig.S22 in the Online Resource1).
The obtained conjugate has the following advan-
tages: (i) the stability of the RBC lipids and proteins
on porous particles was higher than that of lipids of
liposomal particles due to a high adhesion energy;
(ii) the porous surface of the particles exhibited an
excellent lateral bilayer fluidity, thus improving the
SEMENOV et al.1380
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
targeting efficacy; (iii) RBC-coated nanoparticles had
a considerably longer circulation time than PEGylated
nanoparticles due to the presence of transmembrane
protein CD47 that induces signalling through the
phagocyte receptor CD172a, inhibits immune response,
and suppresses particle recognition by the immune
system (see Fig. S23 in the Online Resource 1 for the
mechanism of conjugate action).
Due to the synergistic effect of biomimetic tar-
geting and penetration of DTX/GQD nanoparticles fol-
lowed by irradiation (1.5 W/cm
2
, 10 min), it was able
to achieve a significant reduction in the size of A549
tumor during the first 10 days of treatment.
Xuan et al. [192] synthesized nanoparticles for bio-
imaging and combined chemotherapy/PTT based on
AuNSp clusters (diameter of 50nm) coated with GQDs
covalently modified by FA using carbodiimide method
and noncovalently modified with DOX (94.39 ± 0.39%)
(see Fig.S24 in the Online Resource 1 for the scheme of
conjugate synthesis).
The obtained nanoparticles formed stable aque-
ous dispersions and demonstrated an excellent PA
and CT imaging performance, low cytotoxicity, and
PTT conversion efficiency up to 51.31%. In addition,
the authors showed a significant decrease in the rel-
ative tumor volume in BALB/c nude mice (SPF males,
4-week-old) inoculated with HeLa cells (Fig. S25 in the
Online Resource1).
Wu et al. [193] developed a new type of theranos-
tic agent named PC@GCpD(Gd) [192]. First, the authors
synthesized GQDs covalently modified with the Ce6
photosensitiser (GCpD) and coated with PDA layers,
yielding water-compatible and biocompatible nanopar-
ticles with a substantial photothermal/photochemi-
cal effect. Then, the Cy3-labelled nonmethylated CpG
oligodeoxynucleotide (5′-TCC ATG ACG TTC CTG ACG
TT-3′-Cy3) was condensed with the biodegradable cat-
ionic poly(l-lysine) (PLL) polypeptide to obtain immu-
noactive nanoparticles (PCs). GCpD nanocomposites
easily self-assembled on the surface of PC nanoimmu-
nocores and then were chelated with Gd
3+
(see Fig. S26
in the Online Resource1).
The obtained photo/immunoactive hybrid PC@
GCpD(Gd) nanostructures decreased the viability of
cancer cells, released endogenous cancer cell antigens,
and contemporaneously regulated tumor microenvi-
ronment to facilitate the immunostimulatory effect.
The authors characterised the cellular uptake, MRI/flu-
orescence imaging, and phototherapeutic and immu-
nostimulatory activity towards the murine mammary
cancer EMT6 model, as well as the biosafety of PC@
GCpD(Gd) nanoparticles. It was shown that laser irra-
diation (660 nm, 1 W/cm
2
, 10 min) simulated the PTT
and PDT effects, leading to a significant decrease in the
EMT6 cell viability in mice, secretion of proinflammato-
ry cytokines, maturation of dendritic cells, and recruit-
ment of CD4
+
and CD8
+
T cells into the tumor, resulting
in a higher therapeutic efficacy. MRI/fluorescence imag-
ing traced specific accumulation and retention of PC@
GCpD(Gd) in the tumor-draining lymph nodes.
Ruiyi et al. [194] synthesized histidine (His)-
and octadecylamine (OA)-functionalized GQDs (His/
OA-GQDs). The obtained nanoparticles were used for
the fabrication of His/OA-GQD-NaYF
4
:Yb,Tm nano-
cages that exhibited a 140.2-fold enhancement of up-
conversion fluorescence, stability in aqueous solu-
tions, and high DOX-loading capacity (461.2% within
30 min) (see Fig. S27 in the Online Resource 1) [194].
The authors also developed a drug delivery system
(GYAuDOX) which included His/OA-GQD-NaYF
4
:Yb,Tm
gold nanoparticles as a core, and MGC-803 cell mem-
brane as a shell. The obtained material exhibited a
high biocompatibility, selective targeting of homotypic
tumor cells, pH- and light-stimulated DOX release, and
capacity for chemotherapy/PTT. The data on the effica-
cy of the obtained theranostic agent are presented in
Fig.S28 in the Online Resource1.
Liu et al. [195] synthesized GQDs with a strong ab-
sorption (1070 nm) in the NIR-II region (1000-1700 nm)
by a one-step solvothermal treatment using phenol
(carbon precursor) and hydrogen peroxide (oxidising
agent) in the magnetic field with an intensity of 9 T
(see Fig.S29 in the Online Resource1) [195].
The synthesized nanoparticles possessed a uni-
form size (3.6 nm), tunable fluorescence (quantum
yield, 16.67%), and high photothermal conversion ef-
ficacy (33.45%). The obtained nanomaterial ablated tu-
mor cells, inhibited tumor growth upon NIR-II irradia-
tion, and, at the same time, provided an enhanced NIR
imaging of tumors in mice.
Zhang et al. [196] developed a nanomaterial
(named R-NCNP) by coating a mesoporous carbon ni-
tride (C
3
N
4
) layer on a core–shell nitrogen-doped GQD
(N-GQD)@ HMSNs and decorated it with a P-PEG-RGD
polymer consisting of a purified hematoporphyrin
derivative photofrin (P) and the tumor-homing pep-
tide RGD (Arg-Gly-Asp) connected by PEG as a linker,
to achieve the targeted delivery (see Fig.S30 in the On-
line Resource1).
The obtained material has the following advantag-
es for biomedicine applications: (i) R-NCNPs catalyzed
water decomposition in the tumor microenvironment
with the generation of oxygen, thus decreasing local
hypoxia; (ii) the generated oxygen bubbles enhanced
generation of an echogenic signal, making them la-
ser-activatable ultrasound imaging agents; (iii) acti-
vation of the encapsulated photosensitisers and C
3
N
4
-
layered photosensitiser at λ= 630 nm stimulated ROS
formation; (iv) combination of PTT with PDT for tumor
eradication; (v) P-PEG-RGD promoted efficient accumu-
lation of particles in the tumor; (vi) R-NCNPs acted as
multimodal real-time monitoring agent.
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1381
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Fig. 8. Directions of GBN scientific applications.
Prasad et al. [197] synthesized a theranostic agent
based on GQD-embedded mesoporous silica which dis-
played a high penetration and retention ability in sol-
id tumors (see Fig. S31 in the Online Resource 1). The
obtained material had a uniform particle size distribu-
tion, improved stability, high surface area (850 m
2
/g),
DOX loading capacity of 31%, and high photothermal
response. It was shown that administration of carbano-
silica in 4T1 female Balb/c mice led to a temperature
rise (to ~55°C after 5 min of exposure to NIR light), flu-
orescence intensity of 10
8
p/s/cm
2
/sr, and as a result,
provided 68.75% tumor shrinking compared to 34.48%
without NIR irradiation.
Yang et al. [198] developed a self-assembly ap-
proach to the theranostic agent synthesis based on the
acidity-activated GQD nanotransformers (GQD NTs)
by mixing (i) GQDs (loading module) that provided
large surface area for the loading of photosensitiser
[tetrakis(4-carboxylphenyl) porphyrin, TCPP] and MRI
contrast agent (Mn-TCPP), (ii) RGD peptide as a tar-
geting module due to its affinity to α
V
β
3
integrin, and
(iii) linking module that connected the first two mod-
ules through the host–guest interactions between β-CD
and adamantine [198]. As seen from Fig.S32 in the On-
line Resource 1, the acidity of tumor microenvironment
triggered GQD NT transformation and drugs release.
The synthesized theranostic agent provided an ef-
ficient targeting and long-term retention in the tumor
(over 96 h), possibility of MRI/fluorescence imaging,
and photothermal effect, which enhanced cell mem-
brane permeability, as well as an efficient photosen-
sitiser uptake and repeated PDT at a photosensitiser
content 10-30 times lower than in previously published
papers. As seen from Fig.S33 in the Online Resource 1
(survival and tumor growth curves of A549 tumor-
bearing mice after different treatments), the developed
nanomaterial significantly inhibited tumor growth
and increased mouse survival.
CONCLUSION
Since their discovery in 2004, graphene and its
derivatives have become some of the most promising
materials due to a broad range of potential applica-
tions in various fields of science and technology, such
as biotechnology, biomedicine, tissue engineering,
bioanalysis, etc. (Fig.8).
Graphene has a unique two-dimensional flat struc-
ture, unique physical and chemical properties, and
high biocompatibility, which promotes its application
in the creation of high-tech materials for biomedical
purposes. The use of graphene and its derivatives for
the treatment of solid tumors is one of the promising
areas of modern oncology. Along with the advantages
of GBNs, there are also some limitations that need to
be considered. One of the main problems is the lack
of information about metabolic pathways and toxi-
cokinetics of graphene materials used in biomedical
applications. This limits the ability to fully evaluate the
safety and efficacy of these materials in living organ-
isms. Another important problem is poor reproducibil-
ity of the synthesis of graphene-based materials and
common lack of comprehensive studies on their struc-
ture and composition. Both these factors lead to a poor
reproducibility of biological effects of graphene-based
materials in living systems. Also, water dispersions of
GBNs are prone to aggregation, which affects their bi-
ological activity and mechanism of biological action.
Inthis regard, it is necessary to conduct a comprehen-
sive physico-chemical investigation of their stability,
including the studies of optimal stabilizers. Let us hope
that these problems will be solved in the XXI century
the century of nanotechnology.
Supplementary information. The online version
contains supplementary material available at https://
doi.org/10.1134/S0006297924080029.
SEMENOV et al.1382
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Contributions. K.N.S., I.V.M., J.A.R., and V.V.S. creat-
ed the study concept; K.N.S., I.V.M., J.A.R., and V.V.S. de-
veloped the study methodology; K.N.S., D.N.M., D.K.K.,
J.A.R., O.E.M., and V.V.S. supervised the study; V.V.S.,
D.N.M., O.E.M., and K.N.S. acquired the funding; O.S.S.,
S.V.A., G.O.I., and P.K.K. investigated and analyzed the
data; K.N.S., P.A.A., and V.V.S. curated the data; O.S.S.
wrote the draft and prepared the figures; K.N.S., S.V.A.,
and V.V.S. reviewed and edited the manuscript.
Funding. The work was carried out with the fi-
nancial support of the Ministry of Health of the Rus-
sian Federation (state assignment on the topic "Cre-
ation of a drug based on nanoforms of innovative
synthetic antitumor antibiotics, including heterocyclic
systems with a quaternized nitrogen atom and styryl
fragments in the form of conjugates with targeted de-
livery vectors to the tumor microenvironment" EGISU:
1023022200055-4-3.2.21;3.1.3).
Ethics declarations. This work does not contain
any studies involving human and animal subjects.
Theauthors of this work declare that they have nocon-
flicts of interest.
Open access. This article is licensed under a Cre-
ative Commons Attribution 4.0 International License,
which permits use, sharing, adaptation, distribution,
and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s)
and the source, provide a link to the Creative Commons
license, and indicate if changes were made. Theimages
or other third-party material in this article are includ-
ed in the article’s Creative Commons license, unless
indicated otherwise in a credit line to the material.
Ifmaterial is not included in the article’s Creative Com-
mons license and your intended use is not permitted
by statutory regulation or exceeds the permitted use,
you will need to obtain permission directly from the
copyright holder. To view a copy of this license, visit
http://creativecommons.org/licenses/by/4.0/.
REFERENCES
1. Shin, S.R., Li, Y.C., Jang, H.L., Khoshakhlagh,P., Ak-
bari,M., Nasajpour,A., Zhang, Y.S., Tamayol,A., and
Khademhosseini,A. (2016) Graphene-based materials
for tissue engineering, Adv. Drug Deliv. Rev., 105, 255-
274, https://doi.org/10.1016/j.addr.2016.03.007.
2. Lin, J., Huang, Y., and Huang, P. (2018) Graphene-
Based Nanomaterials in Bioimaging, in Biomedical
Applications of Functionalized Nanomaterials: Con-
cepts, Development and Clinical Translation, Elsevi-
er, pp. 247-287, https://doi.org/10.1016/b978-0-323-
50878-0.00009-4.
3. Feng, L.L., Wu, Y.X., Zhang, D.L., Hu, X.X., Zhang,J.,
Wang,P., Song, Z.L., Zhang, X.B., and Tan,W. (2017)
Near infrared graphene quantum dots-based two-
photon nanoprobe for direct bioimaging of endoge-
nous ascorbic acid in living cells, Anal. Chem., 89, 4077-
4084, https://doi.org/10.1021/acs.analchem.6b04943.
4. Thapa, R.K., Kim, J.H., Jeong, J.H., Shin, B.S., Choi, H.G.,
Yong, C.S., and Kim, J.O. (2017) Silver nanoparticle-
embedded graphene oxide-methotrexate for targeted
cancer treatment, Colloids Surf. B Biointerfaces, 153,
95-103, https://doi.org/10.1016/j.colsurfb.2017.02.012.
5. Zhang,C., Liu,Z., Zheng,Y., Geng,Y., Han,C., Shi,Y.,
Sun,H., Zhang,C., Chen,Y., Zhang,L., Guo,Q., Yang,L.,
Zhou,X., and Kong,L. (2018) Glycyrrhetinic acid func-
tionalized graphene oxide for mitochondria target-
ing and cancer treatment invivo, Small, 14, 1703306,
https://doi.org/10.1002/smll.201703306.
6. Liu, J., Dong, J., Zhang, T., and Peng, Q. (2018)
Graphene-based nanomaterials and their poten-
tials in advanced drug delivery and cancer therapy,
J. Controll. Rel., 286, 64-73, https://doi.org/10.1016/
j.jconrel.2018.07.034.
7. Yang,K., Feng,L., and Liu,Z. (2016) Stimuli respon-
sive drug delivery systems based on nano-graphene
for cancer therapy, Adv. Drug Deliv. Rev., 105, 228-241,
https://doi.org/10.1016/j.addr.2016.05.015.
8. Fan,H., Yu,X., Wang,K., Yin,Y., Tang,Y., Tang,Y., and
Liang,X. (2019) Graphene quantum dots (GQDs)-based
nanomaterials for improving photodynamic therapy
in cancer treatment, Eur.J. Med. Chem., 182, 111620,
https://doi.org/10.1016/j.ejmech.2019.111620.
9. Iannazzo, D., Pistone, A., Salamò, M., Galvagno, S.,
Romeo,R., Giofré, S.V., Branca,C., Visalli,G., and Di
Pietro,A. (2017) Graphene quantum dots for cancer
targeted drug delivery, Int. J. Pharm., 518, 185-192,
https://doi.org/10.1016/j.ijpharm.2016.12.060.
10. Hai,X., Feng,J., Chen,X., and Wang,J. (2018) Tuning
the optical properties of graphene quantum dots for
biosensing and bioimaging, J.Mater. Chem.B, 6, 3219-
3234, https://doi.org/10.1039/c8tb00428e.
11. Szunerits,S., and Boukherroub,R. (2018) Graphene-
based biosensors, Interf. Focus, 8, 20160132, https://
doi.org/10.1098/rsfs.2016.0132.
12. Peña-Bahamonde, J., Nguyen, H. N., Fanourakis,
S.K., and Rodrigues, D.F. (2018) Recent advances in
graphene-based biosensor technology with applica-
tions in life sciences, J.Nanobiotechnol., 16, 75, https://
doi.org/10.1186/s12951-018-0400-z.
13. Palmieri,V., and Papi,M. (2020) Can graphene take
part in the fight against COVID-19? Nano Today, 33,
100883, https://doi.org/10.1016/j.nantod.2020.100883.
14. Yang, X.X., Li, C.M., Li, Y.F., Wang,J., and Huang, C.Z.
(2017) Synergistic antiviral effect of curcumin func-
tionalized graphene oxide against respiratory syncy-
tial virus infection, Nanoscale, 9, 16086-16092, https://
doi.org/10.1039/c7nr06520e.
15. Chen, Y.-N., Hsueh, Y.-H., Hsieh, C.-T., Tzou, D.-Y., and
Chang, P.-L. (2016) Antiviral activity of graphene-sil-
ver nanocomposites against non-enveloped and en-
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1383
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
veloped viruses, Int.J. Environ. Res. Public Health, 13,
430, https://doi.org/10.3390/ijerph13040430.
16. Du,T., Lu,J., Liu,L., Dong,N., Fang,L., Xiao,S., and
Han,H. (2018) Antiviral activity of graphene oxide-
silver nanocomposites by preventing viral entry
and activation of the antiviral innate immune re-
sponse, ACS Appl. Bio Mater., 1, 1286-1293, https://
doi.org/10.1021/acsabm.8b00154.
17. Shi,L., Chen, J., Teng, L., Wang, L., Zhu, G., Liu, S.,
Luo,Z., Shi,X., Wang,Y., and Ren,L. (2016) The an-
tibacterial applications of graphene and its deriv-
atives, Small, 12, 4165-4184, https://doi.org/10.1002/
smll.201601841.
18. Ji,H., Sun,H., and Qu,X. (2016) Antibacterial appli-
cations of graphene-based nanomaterials: recent
achievements and challenges, Adv. Drug Deliv. Rev.,
105, 176-189, https://doi.org/10.1016/j.addr.2016.04.009.
19. Xia, M. Y., Xie, Y., Yu, C. H., Chen, G. Y., Li, Y. H.,
Zhang,T., and Peng,Q. (2019) Graphene-based nanoma-
terials: the promising active agents for antibiotics-inde-
pendent antibacterial applications, J.Controll. Rel., 307,
16-31, https://doi.org/10.1016/j.jconrel.2019.06.011.
20. Kumar,P., Huo,P., Zhang,R., and Liu, B. (2019) An-
tibacterial properties of graphene-based nanomate-
rials, Nanomaterials, 9, 737, https://doi.org/10.3390/
nano9050737.
21. Wu, X., Li, H., and Xiao, N. (2018) Advancement of
Near-infrared (NIR) laser interceded surface enact-
ment of proline functionalized graphene oxide with
silver nanoparticles for proficient antibacterial, an-
tifungal and wound recuperating therapy in nurs-
ing care in hospitals, J.Photochem. Photobiol.B, 187,
89-95, https://doi.org/10.1016/j.jphotobiol.2018.07.015.
22. Kahsay, M. H., Belachew, N., Tadesse, A., and
Basavaiah,K. (2020) Magnetite nanoparticle decorat-
ed reduced graphene oxide for adsorptive removal of
crystal violet and antifungal activities, RSC Adv., 10,
34916-34927, https://doi.org/10.1039/d0ra07061k.
23. Hermanová,S., Zarevúcká,M., Bouša,D., Pumera,M.,
and Sofer, Z. (2015) Graphene oxide immobilized
enzymes show high thermal and solvent stabili-
ty, Nanoscale, 7, 5852-5858, https://doi.org/10.1039/
c5nr00438a.
24. Li, H., Fierens, K., Zhang, Z., Vanparijs, N., Schui-
js, M.J., Van Steendam,K., Feiner Gracia,N., De Ry-
cke,R., De Beer,T., De Beuckelaer,A., De Koker,S., De-
force,D., Albertazzi,L., Grooten,J., Lambrecht, B.N.,
and De Geest, B.G. (2016) Spontaneous protein ad-
sorption on graphene oxide nanosheets allowing effi-
cient intracellular vaccine protein delivery, ACS Appl.
Mater. Interfaces, 8, 1147-1155, https://doi.org/10.1021
/acsami.5b08963.
25. Kavitha, T., Kang, I. K., and Park, S. Y. (2014)
Poly(acrylic acid)-grafted graphene oxide as an intra-
cellular protein carrier, Langmuir, 30, 402-409, https://
doi.org/10.1021/la404337d.
26. Emadi, F., Amini, A., Gholami, A., and Ghasemi, Y.
(2017) Functionalized graphene oxide with chitosan
for protein nanocarriers to protect against enzymatic
cleavage and retain collagenase activity, Sci. Rep., 7,
42258, https://doi.org/10.1038/srep42258.
27. Zhao,H., Ding,R., Zhao,X., Li,Y., Qu,L., Pei,H., Yildi-
rimer,L., et al. (2017) Graphene-based nanomaterials
for drug and/or gene delivery, bioimaging, and tissue
engineering, Drug Discov. Today, 22, 1302-1317, https://
doi.org/10.1016/j.drudis.2017.04.002.
28. Paul,A., Hasan,A., Kindi, H.A., Gaharwar, A.K., Rao,
V.T.S., Nikkhah,M., Shin, S.R., Krafft,D., Dokmeci,
M.R., Shum-Tim,D., and Khademhosseini,A. (2014)
Injectable graphene oxide/hydrogel-based angiogenic
gene delivery system for vasculogenesis and cardiac
repair, ACS Nano, 8, 8050-8062, https://doi.org/10.1021/
nn5020787.
29. Chen, B., Liu, M., Zhang, L., Huang, J., Yao, J., and
Zhang, Z. (2011) Polyethylenimine-functionalized
graphene oxide as an efficient gene delivery vector,
J.Mater. Chem., 21, 7736-7741, https://doi.org/10.1039/
c1jm10341e.
30. Imani, R., Shao, W., Taherkhani, S., Emami, S. H.,
Prakash,S., and Faghihi,S. (2016) Dual-functionalized
graphene oxide for enhanced siRNA delivery to breast
cancer cells, Colloids Surf. B Biointerfaces, 147, 315-
325, https://doi.org/10.1016/j.colsurfb.2016.08.015.
31. Yue, H., Zhou, X., Cheng, M., and Xing, D. (2018)
Graphene oxide-mediated Cas9/sgRNA delivery for
efficient genome editing, Nanoscale, 10, 1063-1071,
https://doi.org/10.1039/c7nr07999k.
32. Tang,Z., Wu,H., Cort, J.R., Buchko, G.W., Zhang,Y.,
Shao,Y., Aksay, I. A., Liu,J., and Lin, Y. (2010) Con-
straint of DNA on functionalized graphene improves
its biostability and specificity, Small, 6, 1205-1209,
https://doi.org/10.1002/smll.201000024.
33. Lu, C.H., Zhu, C.L., Li,J., Liu, J.J., Chen,X., and Yang,
H. H. (2010) Using graphene to protect DNA from
cleavage during cellular delivery, Chem. Commun., 46,
3116-3118, https://doi.org/10.1039/b926893f.
34. Park,J., and Yan,M. (2013) Covalent functionalization
of graphene with reactive intermediates, Acc Chem
Res, 46, 181-189, https://doi.org/10.1021/ar300172h.
35. Criado,A., Melchionna,M., Marchesan,S., and Prato,M.
(2015) The covalent functionalization of graphene on
substrates, Angewandte Chemie Int. Edn., 54, 10734-
10750, https://doi.org/10.1002/anie.201501473.
36. Georgakilas, V., Tiwari, J. N., Kemp, K. C., Perman,
J.A., Bourlinos, A.B., et al. (2016) Noncovalent func-
tionalization of graphene and graphene oxide for en-
ergy materials, biosensing, catalytic, and biomedical
applications, Chem. Rev., 116, 5464-5519, https://doi.
org/10.1021/acs.chemrev.5b00620.
37. Georgakilas,V., Otyepka, M., Bourlinos, A. B., Chan-
dra, V., Kim, N., Kemp, K. C., Hobza, P., Zboril, R.,
and Kim, K.S. (2012) Functionalization of graphene:
SEMENOV et al.1384
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Covalent and non-covalent approaches, derivatives
and applications, Chem. Rev., 112, 6156-6214, https://
doi.org/10.1021/cr3000412.
38. Wei, D., Liu,Y., Wang,Y., Zhang, H., Huang,L., and
Yu, G. (2009) Synthesis of n-doped graphene by
chemical vapor deposition and its electrical proper-
ties, Nano Lett., 9, 1752-1758, https://doi.org/10.1021/
nl803279t.
39. Sreeprasad, T.S., and Berry,V. (2013) How do the elec-
trical properties of graphene change with its function-
alization? Small, 9, 341-350, https://doi.org/10.1002/
smll.201202196.
40. Falkovsky, L.A. (2008) Optical properties of graphene,
J.Phys. Conf. Ser., 129, 12004, https://doi.org/10.1088/
1742-6596/129/1/012004.
41. Qiu, B., Zhao, X. W., Hu, G. C., Yue, W. W., Yuan,
X.B., and Ren, J.F. (2020) Tuning optical properties
of Graphene/WSe2 heterostructure by introduc-
ing vacancy: first principles calculations, Physica E
Low Dimens Syst. Nanostruct., 116, 113729, https://
doi.org/10.1016/j.physe.2019.113729.
42. Kumar, A., Sharma, K., and Dixit, A. R. (2020) A re-
view on the mechanical and thermal properties of
graphene and graphene-based polymer nanocom-
posites: understanding of modelling and MD simula-
tion, Mol. Simul., 46, 136-154, https://doi.org/10.1080/
08927022.2019.1680844.
43. Aradhana, R., Mohanty, S., and Nayak, S. K. (2018)
Comparison of mechanical, electrical and thermal
properties in graphene oxide and reduced graphene
oxide filled epoxy nanocomposite adhesives, Poly-
mer (Guildf), 141, 109-123, https://doi.org/10.1016/
j.polymer.2018.03.005.
44. Vu, T.V., Hieu, N.V., Phuc, H.V., Hieu, N.N., Bui, H.D.,
Idrees,M., Amin,B., and Nguyen, C.V. (2020) Graphene/
WSeTe van der Waals heterostructure: Controllable
electronic properties and Schottky barrier via inter-
layer coupling and electric field, Appl. Surf. Sci., 507,
145036, https://doi.org/10.1016/j.apsusc.2019.145036.
45. Wang,Q., Li,X., Wu,L., Lu,P., and Di,Z. (2019) Elec-
tronic and interface properties in graphene oxide/hy-
drogen-passivated Ge heterostructure, Rapid Res. Lett.,
13, 1800461, https://doi.org/10.1002/pssr.201800461.
46. Sang,M., Shin,J., Kim,K., and Yu,K. (2019) Electron-
ic and thermal properties of graphene and recent
advances in graphene based electronics applica-
tions, Nanomaterials, 9, 374, https://doi.org/10.3390/
nano9030374.
47. Papageorgiou, D.G., Kinloch, I. A., and Young, R. J.
(2017) Mechanical properties of graphene and
graphene-based nanocomposites, Prog. Mater. Sci., 90,
75-127, https://doi.org/10.1016/j.pmatsci.2017.07.004.
48. Zhao, X., Zhang, Q., Chen, D., and Lu,P. (2010) En-
hanced mechanical properties of graphene-based
polyvinyl alcohol composites, Macromolecules, 43,
2357-2363, https://doi.org/10.1021/ma902862u.
49. Geim, A. K., and Novoselov, K.S. (2007) The rise of
graphene, Nat. Mater., 6, 183-191, https://doi.org/
10.1038/nmat1849.
50. Lee, H.C., Liu, W.-W., Chai, S.-P., Mohamed, A.R., Lai,
C.W., Khe, C.-S., Voon, C.H., Hashim,U., and Hidayah,
N.M.S. (2016) Synthesis of single-layer graphene: a
review of recent development, Procedia Chem., 19,
916-921, https://doi.org/10.1016/j.proche.2016.03.135.
51. Adetayo, A., and Runsewe, D. (2019) Synthesis and
fabrication of graphene and graphene oxide: a re-
view, Open J. Composite Mater., 09, 207-229, https://
doi.org/10.4236/ojcm.2019.92012.
52. Zhang, Y., Zhang, L., and Zhou, C. (2013) Review of
chemical vapor deposition of graphene and related
applications, Acc. Chem. Res., 46, 2329-2339, https://
doi.org/10.1021/ar300203n.
53. Muñoz,R., and Gómez-Aleixandre,C. (2013) Review of
CVD synthesis of graphene, Chem. Vapor Deposition,
19, 297-322, https://doi.org/10.1002/cvde.201300051.
54. Li,X., Colombo,L., and Ruoff, R.S. (2016) Synthesis
of graphene films on copper foils by chemical va-
por deposition, Adv. Mater., 28, 6247-6252, https://
doi.org/10.1002/adma.201504760.
55. Chen,K., Shi,L., Zhang,Y., and Liu,Z. (2018) Scalable
chemical-vapour-deposition growth of three-dimen-
sional graphene materials towards energy-related
applications, Chem. Soc. Rev., 47, 3018-3036, https://
doi.org/10.1039/c7cs00852j.
56. Yang,X., Zhang,G., Prakash,J., Chen,Z., Gauthier,M.,
and Sun, S. (2019) Chemical vapour deposition of
graphene: layer control, the transfer process, char-
acterisation, and related applications, Int. Rev. Phys.
Chem., 38, 149-199, https://doi.org/10.1080/0144235x.
2019.1634319.
57. Mattevi,C., Kim,H., and Chhowalla,M. (2011) A re-
view of chemical vapour deposition of graphene on
copper, J.Mater. Chem., 21, 3324-3334, https://doi.org/
10.1039/c0jm02126a.
58. Zhou,H., Yu, W.J., Liu,L., Cheng,R., Chen,Y., Huang,X.,
Liu,Y., Wang,Y., Huang,Y., and Duan,X. (2013) Chem-
ical vapour deposition growth of large single crystals
of monolayer and bilayer graphene, Nat. Commun.,
4, 2096, https://doi.org/10.1038/ncomms3096.
59. Yu, P., Lowe, S. E., Simon, G. P., and Zhong, Y. L.
(2015) Electrochemical exfoliation of graphite and
production of functional graphene, Curr. Opin. Col-
loid Interface Sci., 20, 329-338, https://doi.org/10.1016/
j.cocis.2015.10.007.
60. Rao, K.S., Senthilnathan,J., Liu, Y.F., and Yoshimura,M.
(2014) Role of peroxide ions in formation of graphene
nanosheets by electrochemical exfoliation of graphite,
Sci. Rep., 4, 4237, https://doi.org/10.1038/srep04237.
61. Wan,H., Wei,C., Zhu,K., Zhang,Y., Gong,C., et al. (2017)
Preparation of graphene sheets by electrochemical ex-
foliation of graphite in confined space and their appli-
cation in transparent conductive films, ACS Appl. Mater.
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1385
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Interfaces, 9, 34456-34466, https://doi.org/10.1021/
acsami.7b09891.
62. Mir,A., and Shukla,A. (2018) Bilayer-rich graphene
suspension from electrochemical exfoliation of graph-
ite, Mater Des., 156, 62-70, https://doi.org/10.1016/
j.matdes.2018.06.035.
63. Melezhik, A. V., Pershin, V. F., Memetov, N. R., and
Tkachev, A. G. (2016) Mechanochemical synthesis
of graphene nanoplatelets from expanded graphite
compound, Nanotechnol. Russ., 11, 421-429, https://
doi.org/10.1134/s1995078016040121.
64. Guex, L.G., Sacchi,B., Peuvot, K.F., Andersson, R.L.,
Pourrahimi, A.M., Ström, V., Farris, S., and Olsson,
R.T. (2017) Experimental review: chemical reduction
of graphene oxide (GO) to reduced graphene oxide
(rGO) by aqueous chemistry, Nanoscale, 9, 9562-9571,
https://doi.org/10.1039/c7nr02943h.
65. De Silva, K. K. H., Huang, H. H., Joshi, R. K., and
Yoshimura,M. (2017) Chemical reduction of graphene
oxide using green reductants, Carbon N Y, 119,
190-199, https://doi.org/10.1016/j.carbon.2017.04.025.
66. Wang,J., Salihi, E.C., and Šiller,L. (2017) Green reduc-
tion of graphene oxide using alanine, Mater. Sci. En-
gin.C, 72, 1-6, https://doi.org/10.1016/j.msec.2016.11.017.
67. Alam, S. N., Sharma, N., and Kumar, L. (2017) Syn-
thesis of graphene oxide (GO) by modified hummers
method and its thermal reduction to obtain reduced
graphene oxide (rGO)*, Graphene, 6, 1-18, https://
doi.org/10.4236/graphene.2017.61001.
68. Saleem,H., Haneef,M., and Abbasi, H.Y. (2018) Syn-
thesis route of reduced graphene oxide via thermal
reduction of chemically exfoliated graphene oxide,
Mater. Chem. Phys., 204, 1-7, https://doi.org/10.1016/
j.matchemphys.2017.10.020.
69. Oliveira, A. E. F., Braga, G. B., Tarley, C. R. T., and
Pereira, A.C. (2018) Thermally reduced graphene ox-
ide: synthesis, studies and characterization, J.Mater.
Sci., 53, 12005-12015, https://doi.org/10.1007/s10853-
018-2473-3.
70. Schedy, A., and Oetken, M. (2020) The thermal re-
duction of graphene oxide – a simple and exciting
manufacturing process of graphene, CHEMKON, 27,
244-249, https://doi.org/10.1002/ckon.201900049.
71. Abdelhalim, A.O.E., Sharoyko, V.V., Meshcheriakov,
A.A., Martynova, S. D., Ageev, S. V., Iurev, G.O., Al
Mulla,H., Petrov, A.V., Solovtsova, I.L., Vasina, L.V.,
Murin, I. V., and Semenov, K. N. (2020) Reduction
and functionalization of graphene oxide with L-cys-
teine: synthesis, characterization and biocompatibil-
ity, Nanomedicine, 29, 102284, https://doi.org/10.1016/
j.nano.2020.102284.
72. Kaplan, A., Yuan, Z., Benck, J. D., Govind Rajan,A.,
Chu, X.S., Wang, Q.H., and Strano, M.S. (2017) Cur-
rent and future directions in electron transfer chemis-
try of graphene, Chem. Soc. Rev., 46, 4530-4571, https://
doi.org/10.1039/c7cs00181a.
73. Sturala,J., Luxa,J., Pumera,M., and Sofer,Z. (2018)
Frontispiece: chemistry of graphene derivatives: syn-
thesis, applications, and perspectives, Chem. Eur.J., 24,
5992-6006, https://doi.org/10.1002/chem.201704192.
74. Gao, W. (2015) The chemistry of graphene oxide, in
Graphene Oxide: Reduction Recipes, Spectroscopy,
and Applications, Springer International Publishing,
pp.61-95, https://doi.org/10.1007/978-3-319-15500-5_3.
75. Banks, C.E., Vedyagin, A.A., Cui,Y., Liu,L., Shi,M.,
Wang, Y., Meng,X., Chen, Y., Huang, Q., and Liu,C.
(2024) A review of advances in graphene quantum
dots: from preparation and modification methods
to application, J. Carbon. Res., 10, 7, https://doi.org/
10.3390/c10010007.
76. Prakash, S.H., and Roopan, S.M. (2023) A comprehen-
sive review on recent developments in the graphene
quantum dot framework for organic transformations,
J. Organomet. Chem., 997, 122790, https://doi.org/
10.1016/j.jorganchem.2023.122790.
77. Gozali Balkanloo,P., Mohammad Sharifi,K., and Pour-
sattar Marjani, A. (2023) Graphene quantum dots:
synthesis, characterization, and application in waste-
water treatment: a review, Mater. Adv., 4, 4272-4293,
https://doi.org/10.1039/d3ma00372h.
78. Thangadurai, T. D., Manjubaashini, N., Nataraj, D.,
Gomes,V., and Lee, Y.I. (2022) A review on graphene
quantum dots, an emerging luminescent carbon
nanolights: healthcare and environmental appli-
cations, Mater. Sci. Engin. B, 278, 115633, https://
doi.org/10.1016/j.mseb.2022.115633.
79. Kadyan, P., Malik,R., Bhatia, S., Al Harrasi, A., Mo-
han,S., Yadav,M., Dalal,S., Ramniwas,S., Kumar Ka-
taria,S., and Arasu,T. (2023) Comprehensive review
on synthesis, applications, and challenges of graphene
quantum dots (GQDs), J.Nanomater., 2023, 2832964,
https://doi.org/10.1155/2023/2832964.
80. Ghaffarkhah, A., Hosseini, E., Kamkar, M., Sehat,
A.A., Dordanihaghighi,S., Allahbakhsh,A., Van Der
Kuur,C., Arjmand,M., Ghaffarkhah,A., Hosseini,E.,
Kamkar, M., Sehat, A. A., Dordanihaghighi, S., Arj-
mand,M., and Allahbakhsh,A. (2022) Synthesis, ap-
plications, and prospects of graphene quantum dots:
a comprehensive review, Small, 18, 2102683, https://
doi.org/10.1002/smll.202102683.
81. Zhao, C., Song, X., Liu, Y., Fu,Y., Ye, L., et al. (2020)
Synthesis of graphene quantum dots and their appli-
cations in drug delivery, J.Nanobiotechnol., 18, 142,
https://doi.org/10.1186/s12951-020-00698-z.
82. Pinto, A. M., Moreira, J. A., Magalhães, F. D., and
Gonçalves, I.C. (2016) Polymer surface adsorption as a
strategy to improve the biocompatibility of graphene
nanoplatelets, Colloids Surf. B Biointerfaces, 146,
818-824, https://doi.org/10.1016/j.colsurfb.2016.07.031.
83. Abdelhalim, A. O. E., Meshcheriakov, A. A., Maist-
renko, D.N., Molchanov, O.E., Ageev, S.V., Ivanova,
D. A., Iamalova, N. R., Luttsev, M. D., Vasina, L. V.,
SEMENOV et al.1386
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Sharoyko,V.V., and Semenov, K.N. (2021) Graphene
oxide enriched with oxygen-containing groups: on the
way to an increase of antioxidant activity and biocom-
patibility, Colloids Surf. B Biointerfaces, 210, 112232,
https://doi.org/10.1016/j.colsurfb.2021.112232.
84. Galebskaya, L.V., Solovtsova, I.L., Miroshnikova, E.B.,
Mikhailova, I.A., Sushkin, M.E., Razumny, A.V., Babi-
na, A.V., and Fomina, V.A. (2017) The importance of
a photosensitizer bleaching registration for the eval-
uation of mechanism of preparation action on the
photo-induced hemolysis, Biomed. Photonics, 6, 33-38,
https://doi.org/10.24931/2413-9432-2017-6-3-33-38.
85. Abdelhalim, A.O.E., Sharoyko, V.V., Meshcheriakov,
A.A., Luttsev, M.D., Potanin, A.A., Iamalova, N.R.,
Zakharov, E.E., Ageev, S.V., Petrov, A.V., Vasina, L.V.,
Solovtsova, I.L., Nashchekin, A.V., Murin, I.V., and
Semenov, K.N. (2020) Synthesis, characterisation and
biocompatibility of graphene-L-methionine nanoma-
terial, J.Mol. Liq., 314, 113605, https://doi.org/10.1016/
j.molliq.2020.113605.
86. Abdelhalim, A.O.E., Sharoyko, V.V., Ageev, S.V., Fara-
fonov, V.S., Nerukh, D.A., Postnov, V.N., Petrov, A.V.,
and Semenov, K. N. (2021) Graphene oxide of extra
high oxidation: a wafer for loading guest molecules,
J.Phys. Chem. Lett., 12, 10015-10024, https://doi.org/
10.1021/acs.jpclett.1c02766.
87. Sharoyko, V. V., Shemchuk, O. S., Meshcheriakov,
A. A., Andoskin, P. A., Petrov, A. V., Rumyantsev,
A.M., Sambuk, E.V., Maystrenko, D.N., Molchanov,
O.E., Murin, I.V., Charykov, N.A., and Semenov, K.N.
(2024) Modification of fullerene with amino acids
as a method for obtaining biocompatible materials
with a protective effect, Fullerenes Nanotubes Carbon
Nanostructures, 32, 631-639, https://doi.org/10.1080/
1536383x.2023.2291484.
88. Ageev, S.V., Semenov, K.N., Shemchuk, O.S., Iurev,
G. O., Andoskin, P. A., Rumiantsev, A. M., Sambuk,
E.V., Kozhukhov, P.K., Maistrenko, D. N., Molchan-
ov, O.E., Murin, I.V., Mazur, A.S., and Sharoyko, V.V.
(2024) Synthesis, biocompatibility and biological ac-
tivity of a graphene oxide-folic acid conjugate for cy-
tarabine delivery, Colloids Surf. A Physicochem. Eng.
Asp., 697, 134360, https://doi.org/10.1016/j.colsurfa.
2024.134360.
89. Singh, S.K., Singh, M.K., Kulkarni, P.P., Sonkar, V.K.,
Grácio, J.J.A., and Dash, D. (2012) Amine-modified
graphene: thrombo-protective safer alternative to
graphene oxide for biomedical applications, ACS
Nano, 6, 2731-2740, https://doi.org/10.1021/nn300172t.
90. Podolska, M. J., Barras, A., Alexiou, C., Frey, B.,
Gaipl, U., Boukherroub, R., Szunerits, S., Janko, C.,
and Muñoz, L.E. (2020) Graphene oxide nanosheets
for localized hyperthermia – physicochemical char-
acterization, biocompatibility, and induction of tu-
mor cell death, Cells, 9, 776, https://doi.org/10.3390/
cells9030776.
91. Ding, Z., Ma,H., and Chen, Y. (2014) Interaction of
graphene oxide with human serum albumin and its
mechanism, RSC Adv., 4, 55290-55295, https://doi.org/
10.1039/c4ra09613d.
92. Taneva, S. G., Krumova, S., Bogár, F., Kincses, A.,
Stoichev, S., Todinova,S., Danailova,A., Horváth,J.,
Násztor, Z., Kelemen,L., and Dér, A. (2021) Insights
into graphene oxide interaction with human serum
albumin in isolated state and in blood plasma, Int.J.
Biol. Macromol., 175, 19-29, https://doi.org/10.1016/
j.ijbiomac.2021.01.151.
93. Feng,L., Zhang,S., and Liu,Z. (2011) Graphene based
gene transfection, Nanoscale, 3, 1252, https://doi.org/
10.1039/c0nr00680g.
94. Li,X., Wang,Y., Liu,T., Zhang,Y., Wang,C., and Xie,B.
(2023) Ultrasmall graphene oxide for combination of
enhanced chemotherapy and photothermal therapy
of breast cancer, Colloids Surf. B Biointerfaces, 225,
113288, https://doi.org/10.1016/j.colsurfb.2023.113288.
95. Akhavan, O., Ghaderi, E., and Akhavan, A. (2012)
Size-dependent genotoxicity of graphene nanoplate-
lets in human stem cells, Biomaterials, 33, 8017-8025,
https://doi.org/10.1016/j.biomaterials.2012.07.040.
96. Sharoyko, V.V., Mikolaichuk, O.V., Shemchuk, O.S.,
Abdelhalim, A.O.E., Potanin, A.A., Luttsev, M.D., Da-
dadzhanov, D.R., Vartanyan, T.A., Petrov, A.V., Shash-
erina, A.Yu., Murin, I.V., Maistrenko, D.N., Molchan-
ov, O.E., and Semenov, K.N. (2023) Novel non-cova-
lent conjugate based on graphene oxide and alkylat-
ing agent from 1,3,5-triazine class, J.Mol. Liq., 372,
121203, https://doi.org/10.1016/j.molliq.2023.121203.
97. Yang,Q., Wang,X., Chen,J., Tian,C., Li,H., Chen,Y.,
and Lv,Q. (2012) A clinical study on regional lymphat-
ic chemotherapy using an activated carbon nanopar-
ticle-epirubicin in patients with breast cancer, Tumor
Biol., 33, 2341-2348, https://doi.org/10.1007/s13277-
012-0496-y.
98. Sun,X., Liu,Z., Welsher,K., Robinson, J.T., Goodwin,A.,
Zaric, S., and Dai, H. (2008) Nano-graphene oxide
for cellular imaging and drug delivery, Nano Res., 1,
203-212, https://doi.org/10.1007/s12274-008-8021-8.
99. Wu,J., Wang, Y.S., Yang, X.Y., Liu, Y.Y., Yang, J.R.,
Yang,R., and Zhang,N. (2012) Graphene oxide used as
a carrier for adriamycin can reverse drug resistance
in breast cancer cells, Nanotechnology, 23, 355101,
https://doi.org/10.1088/0957-4484/23/35/355101.
100. Fan, X., Jiao, G., Gao, L., Jin, P., and Li, X. (2013)
The preparation and drug delivery of a graphene-
carbon nanotube-Fe 3O4 nanoparticle hybrid, J.Ma-
ter. Chem. B, 1, 2658-2664, https://doi.org/10.1039/
c3tb00493g.
101. Zhang, L., Lu, Z., Zhao, Q., Huang,J., Shen, H., and
Zhang,Z. (2011) Enhanced chemotherapy efficacy by
sequential delivery of siRNA and anticancer drugs
using PEI-grafted graphene oxide, Small, 7, 460-464,
https://doi.org/10.1002/smll.201001522.
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1387
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
102. Liu,Z., Robinson, J.T., Sun,X., and Dai,H. (2008) PE-
Gylated nanographene oxide for delivery of water-in-
soluble cancer drugs, J.Am. Chem. Soc., 130, 10876-
10877, https://doi.org/10.1021/ja803688x.
103. Zhang,L., Xia,J., Zhao,Q., Liu,L., and Zhang,Z. (2010)
Functional graphene oxide as a nanocarrier for con-
trolled loading and targeted delivery of mixed anti-
cancer drugs, Small, 6, 537-544, https://doi.org/10.1002/
smll.200901680.
104. Xu,Q., Wang,H., Gu, W., Xiao, N., and Ye, L. (2014)
Chlorotoxin-conjugated graphene oxide for targeted
delivery of an anticancer drug, Int.J. Nanomedicine,
9, 1433, https://doi.org/10.2147/ijn.s58783.
105. Fan,L., Ge,H., Zou,S., Xiao,Y., Wen,H., Li,Y., Feng,H.,
and Nie, M. (2016) Sodium alginate conjugated
graphene oxide as a new carrier for drug delivery sys-
tem, Int.J. Biol. Macromol., 93, 582-590, https://doi.org/
10.1016/j.ijbiomac.2016.09.026.
106. Rosli, N. F., Fojtů, M., Fisher, A. C., and Pumera, M.
(2019) Graphene oxide nanoplatelets potentiate an-
ticancer effect of cisplatin in human lung cancer
cells, Langmuir, 35, 3176-3182, https://doi.org/10.1021/
acs.langmuir.8b03086.
107. Deb, A., and Vimala, R. (2018) Camptothecin load-
ed graphene oxide nanoparticle functionalized with
polyethylene glycol and folic acid for anticancer
drug delivery, J.Drug Deliv. Sci. Technol., 43, 333-342,
https://doi.org/10.1016/j.jddst.2017.10.025.
108. Pooresmaeil,M., and Namazi,H. (2018) β-Cyclodextrin
grafted magnetic graphene oxide applicable as cancer
drug delivery agent: synthesis and characterization,
Mater. Chem. Phys., 218, 62-69, https://doi.org/10.1016/
j.matchemphys.2018.07.022.
109. Bullo, S., Buskaran, K., Baby, R., Dorniani, D., Fak-
urazi,S., and Hussein, M.Z. (2019) Dual drugs anti-
cancer nanoformulation using graphene oxide-PEG
as nanocarrier for protocatechuic acid and chloro-
genic acid, Pharm. Res., 36, 91, https://doi.org/10.1007/
s11095-019-2621-8.
110. Ma,N., Liu, J., He, W., Li, Z., Luan,Y., Song, Y., and
Garg, S. (2017) Folic acid-grafted bovine serum al-
bumin decorated graphene oxide: an efficient drug
carrier for targeted cancer therapy, J. Colloid Inter-
face Sci., 490, 598-607, https://doi.org/10.1016/j.jcis.
2016.11.097.
111. Tian, J., Luo, Y., Huang, L., Feng, Y., Ju, H., and Yu,
B. Y. (2016) Pegylated folate and peptide-decorated
graphene oxide nanovehicle for invivo targeted de-
livery of anticancer drugs and therapeutic self-mon-
itoring, Biosens. Bioelectron., 80, 519-524, https://
doi.org/10.1016/j.bios.2016.02.018.
112. Javanbakht, S., and Namazi, H. (2018) Doxorubicin
loaded carboxymethyl cellulose/graphene quantum
dot nanocomposite hydrogel films as a potential an-
ticancer drug delivery system, Mater. Sci. Engin.C, 87,
50-59, https://doi.org/10.1016/j.msec.2018.02.010.
113. Karki, N., Tiwari, H., Pal, M., Chaurasia,A., Bal, R.,
Joshi, P., and Sahoo, N. G. (2018) Functionalized
graphene oxides for drug loading, release and deliv-
ery of poorly water soluble anticancer drug: a com-
parative study, Colloids Surf. B Biointerfaces, 169,
265-272, https://doi.org/10.1016/j.colsurfb.2018.05.022.
114. Abdelhalim, A.O.E., Ageev, S.V., Petrov, A.V., Mesh-
cheriakov, A.A., Luttsev, M.D., Vasina, L.V., Nashchek-
ina, I.A., Murin, I.V., Molchanov, O.E., Maistrenko,
D.N., Potanin, A.A., Semenov, K.N., and Sharoyko, V.V.
(2022) Graphene oxide conjugated with doxorubicin:
synthesis, bioactivity, and biosafety, J.Mol. Liq., 359,
119156, https://doi.org/10.1016/j.molliq.2022.119156.
115. Li,J., Lyv,Z., Li,Y., Liu,H., Wang,J., Zhan,W., Chen,H.,
Chen,H., and Li,X. (2015) A theranostic prodrug deliv-
ery system based on Pt(IV) conjugated nano-graphene
oxide with synergistic effect to enhance the therapeu-
tic efficacy of Pt drug, Biomaterials, 51, 12-21, https://
doi.org/10.1016/j.biomaterials.2015.01.074.
116. Mo,R., Jiang,T., Sun,W., and Gu,Z. (2015) ATP-respon-
sive DNA-graphene hybrid nanoaggregates for anti-
cancer drug delivery, Biomaterials, 50, 67-74, https://
doi.org/10.1016/j.biomaterials.2015.01.053.
117. Jiang,T., Sun,W., Zhu,Q., Burns, N.A., Khan, S. A.,
Mo,R., and Gu,Z. (2015) Furin-mediated sequential
delivery of anticancer cytokine and small-molecule
drug shuttled by graphene, Adv. Mater., 27, 1021-1028,
https://doi.org/10.1002/adma.201404498.
118. Mehra, N.K., Jain, A. K., and Nahar,M. (2018) Car-
bon nanomaterials in oncology: an expanding hori-
zon, Drug Discov Today, 23, 1016-1025, https://doi.org/
10.1016/j.drudis.2017.09.013.
119. Jiang, B., Zhou, B., Lin, Z., Liang, H., and Shen, X.
(2019) Recent advances in carbon nanomaterials for
cancer phototherapy, Chem. Eur. J., 25, 3993-4004,
https://doi.org/10.1002/chem.201804383.
120. Gautam,M., Thapa, R.K., Poudel, B.K., Gupta,B., Rut-
tala, H.B., Nguyen, H.T., Soe, Z.C., Ou,W., Poudel,K.,
Choi, H.-G., Ku, S.K., Yong, C.S., and Kim, J.O. (2019)
Aerosol technique-based carbon-encapsulated hollow
mesoporous silica nanoparticles for synergistic che-
mo-photothermal therapy, Acta Biomater., 88, 448-461,
https://doi.org/10.1016/j.actbio.2019.02.029.
121. Huang, C., Hu, X., Hou, Z., Ji, J., Li, Z., and Luan, Y.
(2019) Tailored graphene oxide-doxorubicin nano-
vehicles via near-infrared dye-lactobionic acid con-
jugates for chemo-photothermal therapy, J. Colloid
Interface Sci., 545, 172-183, https://doi.org/10.1016/
j.jcis.2019.03.019.
122. Roy, S., Sarkar, A., and Jaiswal, A. (2019) Poly(al-
lylamine hydrochloride)-functionalized reduced
graphene oxide for synergistic chemo-photothermal
therapy, Nanomedicine, 14, 255-274, https://doi.org/
10.2217/nnm-2018-0320.
123. Li,Q., Wen,J., Liu,C., Jia,Y., Wu,Y., Shan,Y., Qian,Z.,
and Liao, J. (2019) Graphene-nanoparticle-based
SEMENOV et al.1388
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
self-healing hydrogel in preventing postoperative
recurrence of breast cancer, ACS Biomater. Sci. Eng.,
5, 768-779, https://doi.org/10.1021/acsbiomaterials.
8b01475.
124. Zhang, X., Luo, L., Li, L., He, Y., Cao, W., Liu, H.,
Niu,K., and Gao,D. (2019) Trimodal synergistic an-
titumor drug delivery system based on graphene ox-
ide, Nanomedicine, 15, 142-152, https://doi.org/10.1016/
j.nano.2018.09.008.
125. De Melo-Diogo,D., Costa, E.C., Alves, C.G., Lima-Sou-
sa,R., Ferreira,P., Louro, R.O., and Correia, I.J. (2018)
POxylated graphene oxide nanomaterials for com-
bination chemo-phototherapy of breast cancer cells,
Eur. J. Pharmaceut. Biopharmaceut., 131, 162-169,
https://doi.org/10.1016/j.ejpb.2018.08.008.
126. Hashemi,M., Omidi,M., Muralidharan,B., Tayebi,L.,
Herpin, M. J., Mohagheghi, M. A., Mohammadi, J.,
Smyth, H.D.C., and Milner, T.E. (2018) Layer-by-lay-
er assembly of graphene oxide on thermosensitive li-
posomes for photo-chemotherapy, Acta Biomater., 65,
376-392, https://doi.org/10.1016/j.actbio.2017.10.040.
127. Zhang,M., Wu,F., Wang,W., Shen,J., Zhou, N., and
Wu,C. (2019) Multifunctional nanocomposites for tar-
geted, photothermal, and chemotherapy, Chem. Mater.,
31, 1847-1859, https://doi.org/10.1021/acs.chemmater.
8b00934.
128. Chauhan,G., Chopra,V., Tyagi,A., Rath,G., Sharma,
R.K., and Goyal, A.K. (2017) “Gold nanoparticles com-
posite-folic acid conjugated graphene oxide nanohy-
brids” for targeted chemo-thermal cancer ablation:
invitro screening and invivo studies, Eur.J. Pharma-
ceut. Sci., 96, 351-361, https://doi.org/10.1016/j.ejps.
2016.10.011.
129. Guo, M., Xiang, H.-J., Wang, Y., Zhang, Q.-L., An, L.,
Yang, S.-P., Ma,Y., Wang,Y., and Liu, J.-G. (2017) Ru-
thenium nitrosyl functionalized graphene quantum
dots as an efficient nanoplatform for NIR-light-con-
trolled and mitochondria-targeted delivery of nitric
oxide combined with photothermal therapy, Chem.
Commun., 53, 3253-3256, https://doi.org/10.1039/
c7cc00670e.
130. Shao,L., Zhang,R., Lu,J., Zhao,C., Deng,X., and Wu,Y.
(2017) Mesoporous silica coated polydopamine func-
tionalized reduced graphene oxide for synergistic
targeted chemo-photothermal therapy, ACS Appl. Ma-
ter. Interfaces, 9, 1226-1236, https://doi.org/10.1021/
acsami.6b11209.
131. Xu,X., Wang,J., Wang,Y., Zhao,L., Li,Y., and Liu,C.
(2018) Formation of graphene oxide-hybridized nano-
gels for combinative anticancer therapy, Nanomed-
icine, 14, 2387-2395, https://doi.org/10.1016/j.nano.
2017.05.007.
132. Chen, G., Yang, Z., Yu, X., Yu, C., Sui, S., Zhang, C.,
Bao,C., Zeng,X., Chen,Q., and Peng,Q. (2023) Intra-
tumor delivery of amino-modified graphene oxide
as a multifunctional photothermal agent for efficient
antitumor phototherapy, J.Colloid Interface Sci., 652,
1108-1116, https://doi.org/10.1016/j.jcis.2023.08.126.
133. Melo, B.L., Lima-Sousa,R., Alves, C.G., Correia, I.J.,
and de Melo-Diogo, D. (2023) Sulfobetaine methac-
rylate-coated reduced graphene oxide-IR780 hybrid
nanosystems for effective cancer photothermal-pho-
todynamic therapy, Int.J. Pharm., 647, 123552, https://
doi.org/10.1016/j.ijpharm.2023.123552.
134. Lima-Sousa, R., Melo, B. L., Mendonça, A. G., Cor-
reia, I. J., and de Melo-Diogo, D. (2024) Hyaluronic
acid-functionalized graphene-based nanohybrids for
targeted breast cancer chemo-photothermal thera-
py, Int.J. Pharm., 651, 123763, https://doi.org/10.1016/
j.ijpharm.2023.123763.
135. Gao,J., Cao,C., Rui,Q., Sheng,Y., Cai,W., Li, J., and
Kong, Y. (2023) A tri-responsive dual-drug delivery
system based on mesoporous silica nanoparticles@
polydopamine@graphene oxide nanosheets for che-
mo-photothermal therapy of osteosarcoma, J. Saudi
Chem. Soc., 27, 101655, https://doi.org/10.1016/j.jscs.
2023.101655.
136. Hosseinzadeh,R., Khorsandi,K., and Hosseinzadeh,G.
(2018) Graphene oxide-methylene blue nanocompos-
ite in photodynamic therapy of human breast cancer,
J.Biomol. Struct. Dyn., 36, 2216-2223, https://doi.org/
10.1080/07391102.2017.1345698.
137. Ma,M., Cheng,L., Zhao,A., Zhang,H., and Zhang,A.
(2020) Pluronic-based graphene oxide-methylene blue
nanocomposite for photodynamic/photothermal com-
bined therapy of cancer cells, Photodiagnosis. Photo-
dyn. Ther., 29, 101640, https://doi.org/10.1016/j.pdpdt.
2019.101640.
138. Wei, Y., Zhou, F., Zhang, D., Chen, Q., and Xing, D.
(2016) A graphene oxide based smart drug deliv-
ery system for tumor mitochondria-targeting pho-
todynamic therapy, Nanoscale, 8, 3530-3538, https://
doi.org/10.1039/c5nr07785k.
139. Li, Y., Dong, H., Li, Y., and Shi, D. (2015) Graphene-
based nanovehicles for photodynamic medical
therapy, Int. J. Nanomedicine, 10, 2451-2459, https://
doi.org/10.2147/ijn.s68600.
140. Chen,Z., Li,Z., Wang,J., Ju,E., Zhou,L., Ren,J., and
Qu,X. (2014) A multi-synergistic platform for sequen-
tial irradiation-activated high-performance apoptotic
cancer therapy, Adv. Funct. Mater., 24, 522-529, https://
doi.org/10.1002/adfm.201301951.
141. Chen,J., Wu,W., Zhang,F., Zhang,J., Liu,H., Zheng,J.,
Guo,S., and Zhang,J. (2020) Graphene quantum dots
in photodynamic therapy, Nanoscale Adv., 2, 4961-
4967, https://doi.org/10.1039/d0na00631a.
142. Ahirwar, S., Mallick, S., and Bahadur,D. (2020) Pho-
todynamic therapy using graphene quantum dot de-
rivatives, J.Solid State Chem., 282, 121107, https://doi.
org/10.1016/j.jssc.2019.121107.
143. Ge,J., Lan, M., Zhou, B., Liu, W., Guo, L., Wang, H.,
Jia,Q., Niu,G., Huang,X., Zhou,H., Meng,X., Wang,P.,
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1389
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Lee, C.S., Zhang,W., and Han,X. (2014) A graphene
quantum dot photodynamic therapy agent with high
singlet oxygen generation, Nat. Commun., 5, 4596,
https://doi.org/10.1038/ncomms5596.
144. Tian, B., Wang, C., Zhang, S., Feng, L., and Liu, Z.
(2011) Photothermally enhanced photodynamic ther-
apy delivered by nano-graphene oxide, ACS Nano,
5, 7000-7009, https://doi.org/10.1021/nn201560b.
145. Huang,P., Xu,C., Lin,J., Wang,C., Wang,X., Zhang,C.,
Zhou,X., Guo,S., and Cui,D. (2011) Folic acid-conju-
gated graphene oxide loaded with photosensitizers
for targeting photodynamic therapy, Theranostics,
1, 240-250, https://doi.org/10.7150/thno/v01p0240.
146. Zhou,L., Jiang,H., Wei,S., Ge,X., Zhou,J., and Shen,J.
(2012) High-efficiency loading of hypocrellin B on
graphene oxide for photodynamic therapy, Carbon
N Y, 50, 5594-5604, https://doi.org/10.1016/j.carbon.
2012.08.013.
147. Wang, Y., Wang, H., Liu, D., Song, S., Wang, X., and
Zhang, H. (2013) Graphene oxide covalently graft-
ed upconversion nanoparticles for combined NIR
mediated imaging and photothermal/photodynamic
cancer therapy, Biomaterials, 34, 7715-7724, https://
doi.org/10.1016/j.biomaterials.2013.06.045.
148. Rong, P., Yang, K., Srivastan, A., Kiesewetter, D. O.,
Yue,X., Wang,F., Nie,L., Bhirde,A., Wang,Z., Liu,Z.,
Niu,G., Wang,W., and Chen,X. (2014) Photosensitiz-
er loaded nano-graphene for multimodality imaging
guided tumor photodynamic therapy, Theranostics,
4, 229-239, https://doi.org/10.7150/thno.8070.
149. Cho,Y., Kim,H., and Choi,Y. (2013) A graphene ox-
ide-photosensitizer complex as an enzyme-activatable
theranostic agent, Chem. Commun., 49, 1202-1204,
https://doi.org/10.1039/c2cc36297j.
150. Sahu, A., Choi, W. I., Lee, J. H., and Tae, G. (2013)
Graphene oxide mediated delivery of methylene
blue for combined photodynamic and photother-
mal therapy, Biomaterials, 34, 6239-6248, https://
doi.org/10.1016/j.biomaterials.2013.04.066.
151. Zhou, L., Zhou,L., Wei, S., Ge, X., Zhou,J., Jiang, H.,
Li,F., and Shen,J. (2014) Combination of chemothera-
py and photodynamic therapy using graphene oxide as
drug delivery system, J.Photochem. Photobiol.B, 135,
7-16, https://doi.org/10.1016/j.jphotobiol.2014.04.010.
152. Gollavelli,G., and Ling, Y.C. (2014) Magnetic and flu-
orescent graphene for dual modal imaging and single
light induced photothermal and photodynamic thera-
py of cancer cells, Biomaterials, 35, 4499-4507, https://
doi.org/10.1016/j.biomaterials.2014.02.011.
153. Yan, X., Hu, H., Lin, J., Jin, A. J., Niu, G., Zhang, S.,
Huang,P., Shen,B., and Chen,X. (2015) Optical and
photoacoustic dual-modality imaging guided synergis-
tic photodynamic/photothermal therapies, Nanoscale,
7, 2520-2526, https://doi.org/10.1039/c4nr06868h.
154. Wu,C., Zhu,A., Li,D., Wang,L., Yang,H., Zeng,H., and
Liu, Y. (2016) Photosensitizer-assembled PEGylated
graphene-copper sulfide nanohybrids as a syner-
gistic near-infrared phototherapeutic agent, Expert
Opin. Drug Deliv., 13, 155-165, https://doi.org/10.1517/
17425247.2016.1118049.
155. Yan, X., Niu, G., Lin, J., Jin, A. J., Hu, H., Tang, Y.,
Zhang,Y., Wu,A., Lu,J., Zhang,S., Huang,P., Shen,B.,
and Chen,X. (2015) Enhanced fluorescence imaging
guided photodynamic therapy of sinoporphyrin sodi-
um loaded graphene oxide, Biomaterials, 42, 94-102,
https://doi.org/10.1016/j.biomaterials.2014.11.040.
156. Kim, Y. K., Na, H. K., Kim, S., Jang, H., Chang, S.J.,
and Min, D.H. (2015) One-Pot synthesis of multifunc-
tional Au@Graphene oxide nanocolloid Core@Shell
nanoparticles for Raman bioimaging, photothermal,
and photodynamic therapy, Small, 11, 2527-2535,
https://doi.org/10.1002/smll.201402269.
157. Luo,S., Yang,Z., Tan,X., Wang,Y., Zeng,Y., Wang,Y.,
Li,C., Li,R., and Shi,C. (2016) Multifunctional photo-
sensitizer grafted on polyethylene glycol and polyeth-
ylenimine dual-functionalized nanographene oxide
for cancer-targeted near-infrared imaging and syn-
ergistic phototherapy, ACS Appl. Mater. Interfaces, 8,
17176-17186, https://doi.org/10.1021/acsami.6b05383.
158. Kalluru,P., Vankayala,R., Chiang, C.S., and Hwang,
K. C. (2016) Nano-graphene oxide-mediated in vivo
fluorescence imaging and bimodal photodynamic and
photothermal destruction of tumors, Biomaterials, 95,
1-10, https://doi.org/10.1016/j.biomaterials.2016.04.006.
159. Wo, F., Xu, R., Shao, Y., Zhang, Z., Chu, M., Shi, D.,
and Liu, S. (2016) A multimodal system with syn-
ergistic effects of magneto-mechanical, photother-
mal, photodynamic and chemo therapies of cancer
in graphene-quantum dot-coated hollow magnetic
nanospheres, Theranostics, 6, 485-500, https://doi.org/
10.7150/thno.13411.
160. Wu, C., Li, D., Wang, L., Guan, X., Tian, Y., Yang, H.,
Li,S., and Liu,Y. (2017) Single wavelength light-mediat-
ed, synergistic bimodal cancer photoablation and am-
plified photothermal performance by graphene/gold
nanostar/photosensitizer theranostics, Acta Biomater.,
53, 631-642, https://doi.org/10.1016/j.actbio.2017.01.078.
161. Gulzar,A., Xu,J., Yang,D., Xu,L., He,F., Gai,S., and
Yang,P. (2018) Nano-graphene oxide-UCNP-Ce6 cova-
lently constructed nanocomposites for NIR-mediat-
ed bioimaging and PTT/PDT combinatorial therapy,
Dalton Transactions, 47, 3931-3939, https://doi.org/
10.1039/c7dt04141a.
162. Hatamie,S., Ahadian, M.M., Ghiass, M.A., Iraji zad,A.,
Saber,R., Parseh,B., Oghabian, M.A., and Shanehsaz-
zadeh Zadeh,S. (2016) Graphene/cobalt nanocarrier
for hyperthermia therapy and MRI diagnosis, Colloids
Surf. B Biointerfaces, 146, 271-279, https://doi.org/
10.1016/j.colsurfb.2016.06.018.
163. Su,Y., Wang,N., Liu,B., Du,Y., Li,R., Meng,Y., Feng,Y.,
Shan, Z., and Meng, S. (2020) A phototheranostic
nanoparticle for cancer therapy fabricated by BODIPY
SEMENOV et al.1390
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
and graphene to realize photo-chemo synergistic
therapy and fluorescence/photothermal imaging,
Dyes Pigments, 177, 108262, https://doi.org/10.1016/
j.dyepig.2020.108262.
164. Taratula,O., Patel,M., Schumann,C., Naleway, M.A.,
Pang, A.J., He,H., and Taratula,O. (2015) Phthalocya-
nine-loaded graphene nanoplatform for imaging-guid-
ed combinatorial phototherapy, Int.J. Nanomedicine,
10, 2347, https://doi.org/10.2147/ijn.s81097.
165. Lamb,J., Fischer,E., Rosillo-Lopez,M., Salzmann, C.G.,
and Holland, J.P. (2019) Multi-functionalised graphene
nanoflakes as tumour-targeting theranostic drug-de-
livery vehicles, Chem. Sci., 10, 8880-8888, https://
doi.org/10.1039/c9sc03736e.
166. Tomasella, P., Sanfilippo, V., Bonaccorso, C., Cucci,
L.M., Consiglio,G., Nicosia,A., Mineo, P.G., Forte,G.,
and Satriano, C. (2020) Theranostic nanoplatforms
of thiolated reduced graphene oxide nanosheets and
gold nanoparticles, Appl. Sci., 10, 5529, https://doi.org/
10.3390/app10165529.
167. Usman, M.S., Hussein, M.Z., Fakurazi,S., Masarudin,
M.J., and Saad, F.F.A. (2018) A bimodal theranostic
nanodelivery system based on [graphene oxide-chlo-
rogenic acid-gadolinium/gold] nanoparticles, PLoS
One, 13, e0200760, https://doi.org/10.1371/journal.
pone.0200760.
168. Chawda,N., Basu,M., Majumdar,D., Poddar,R., Ma-
hapatra, S.K., and Banerjee,I. (2019) Engineering of
gadolinium-decorated graphene oxide nanosheets for
multimodal bioimaging and drug delivery, ACS Ome-
ga, 4, 12470-12479, https://doi.org/10.1021/acsomega.
9b00883.
169. Samadian, H., Mohammad-Rezaei, R., Jahanban-
Esfahlan,R., Massoumi,B., Abbasian,M., Jafarizad,A.,
and Jaymand,M. (2020) A de novo theranostic nano-
medicine composed of PEGylated graphene oxide and
gold nanoparticles for cancer therapy, J.Mater. Res.,
35, 430-441, https://doi.org/10.1557/jmr.2020.3.
170. Yang, Y., Wang, S., Wang, C., Tian, C., Shen, Y., and
Zhu,M. (2019) Engineered targeted hyaluronic acid-
glutathione-stabilized gold nanoclusters/graphene
oxide-5-fluorouracil as a smart theranostic plat-
form for stimulus-controlled fluorescence imag-
ing-assisted synergetic chemo/phototherapy, Chem.
As. J., 14, 1418-1423, https://doi.org/10.1002/asia.
201900153.
171. Guo,S., Song,Z., Ji, D.-K., Reina,G., Fauny, J.-D., Nishi-
na,Y., Ménard-Moyon,C., and Bianco,A. (2022) Com-
bined photothermal and photodynamic therapy for
cancer treatment using a multifunctional graphene
oxide, Pharmaceutics, 14, 1365, https://doi.org/10.3390/
pharmaceutics14071365.
172. Baktash, M. S., Zarrabi, A., Avazverdi, E., and Reis,
N. M. (2021) Development and optimization of a
new hybrid chitosan-grafted graphene oxide/mag-
netic nanoparticle system for theranostic applica-
tions, J.Mol. Liq., 322, 114515, https://doi.org/10.1016/
j.molliq.2020.114515.
173. Pan,J., Yang,Y., Fang,W., Liu,W., Le,K., Xu,D., and
Li, X. (2018) Fluorescent phthalocyanine-graphene
conjugate with enhanced nir absorbance for imaging
and multi-modality therapy, ACS Appl. Nano Mater.,
1, 2785-2795, https://doi.org/10.1021/acsanm.8b00449.
174. Chen, M.L., Gao, Z.W., Chen, X. M., Pang, S.C., and
Zhang, Y. (2018) Laser-assisted in situ synthesis of
graphene-based magnetic-responsive hybrids for
multimodal imaging-guided chemo/photothermal syn-
ergistic therapy, Talanta, 182, 433-442, https://doi.org/
10.1016/j.talanta.2018.02.030.
175. Chang, X., Zhang, Y., Xu, P., Zhang, M., Wu, H., and
Yang,S. (2018) Graphene oxide/MnWO4 nanocompos-
ite for magnetic resonance/photoacoustic dual-mod-
el imaging and tumor photothermo-chemotherapy,
Carbon N Y, 138, 397-409, https://doi.org/10.1016/
j.carbon.2018.07.058.
176. Prasad, R., Yadav, A.S., Gorain, M., Chauhan, D. S.,
Kundu, G.C., Srivastava, R., and Selvaraj,K. (2019)
Graphene oxide supported liposomes as red emis-
sive theranostics for phototriggered tissue visualiza-
tion and tumor regression, ACS Appl. Bio Mater., 2,
3312-3320, https://doi.org/10.1021/acsabm.9b00335.
177. Samiei Foroushani, M., Karimi Shervedani, R., Ke-
fayat,A., Torabi,M., Ghahremani,F., and Yaghoobi,F.
(2009) Folate-graphene chelate manganese nanoparti-
cles as a theranostic system for colon cancer MR im-
aging and drug delivery: invivo examinations, J.Drug
Deliv. Sci. Technol., 54, 101223, https://doi.org/10.1016/
j.jddst.2019.101223.
178. Luo,Y., Tang,Y., Liu,T., Chen,Q., Zhou,X., Wang,N.,
Ma, M., Cheng, Y., and Chen, H. (2019) Engineering
graphene oxide with ultrasmall SPIONs and smart
drug release for cancer theranostics, Chem. Commun.,
55, 1963-1966, https://doi.org/10.1039/c8cc09185d.
179. Shi, S., Yang, K., Hong, H., Valdovinos, H. F., Nayak,
T.R., Zhang,Y., Theuer, C.P., Barnhart, T.E., Liu,Z.,
and Cai,W. (2013) Tumor vasculature targeting and
imaging in living mice with reduced graphene ox-
ide, Biomaterials, 34, 3002, https://doi.org/10.1016/
j.biomaterials.2013.01.047.
180. Cheng, S.J., Chiu, H.Y., Kumar, P.V., Hsieh, K.Y., Yang,
J.W., Lin, Y.R., Shen, Y.C., and Chen, G.Y. (2018) Si-
multaneous drug delivery and cellular imaging us-
ing graphene oxide, Biomater Sci., 6, 813-819, https://
doi.org/10.1039/c7bm01192j.
181. Hu,D., Zhang,J., Gao,G., Sheng,Z., Cui,H., and Cai,L.
(2016) Indocyanine green-loaded polydopamine-re-
duced graphene oxide nanocomposites with amplify-
ing photoacoustic and photothermal effects for can-
cer theranostics, Theranostics, 6, 1043-1052, https://
doi.org/10.7150/thno.14566.
182. Turcheniuk, K., Dumych, T., Bilyy, R., Turcheni-
uk,V., Bouckaert,J., Vovk,V., Chopyak,V., Zaitsev,V.,
DEVELOPMENT OF GRAPHENE-BASED MATERIALS 1391
BIOCHEMISTRY (Moscow) Vol. 89 No. 8 2024
Mariot,P., Prevarskaya,N., Boukherroub,R., and Szu-
nerits,S. (2015) Plasmonic photothermal cancer ther-
apy with gold nanorods/reduced graphene oxide core/
shell nanocomposites, RSC Adv., 6, 1600-1610, https://
doi.org/10.1039/c5ra24662h.
183. Wang, L., Wang, M., Zhou, B., Zhou, F., Murray, C.,
Towner, R. A., Smith, N., Saunders, D., Xie, G., and
Chen, W. R. (2019) PEGylated reduced-graphene ox-
ide hybridized with Fe
3
O
4
nanoparticles for cancer
photothermal-immunotherapy, J. Mater. Chem. B, 7,
7406-7414, https://doi.org/10.1039/c9tb00630c.
184. Bansal,S., Singh,J., Kumari,U., Kaur, I.P., Barnwal,
R.P., Kumar,R., Singh,S., Singh,G., and Chatterjee,M.
(2019) Development of biosurfactant-based graphene
quantum dot conjugate as a novel and fluorescent
theranostic tool for cancer, Int. J. Nanomedicine, 14,
809-818, https://doi.org/10.2147/ijn.s188552.
185. Ko, N.R., Hong, S.H., Nafiujjaman,M., An, S.Y., Revu-
ri,V., Lee, S.J., Kwon, I.K., Lee, Y.ky., and Oh, S.J. (2019)
Glutathione-responsive PEGylated GQD-based nano-
materials for diagnosis and treatment of breast can-
cer, J.Industr. Engin. Chem., 71, 301-307, https://doi.org/
10.1016/j.jiec.2018.11.039.
186. Li,S., Zhou,S., Li,Y., Li,X., Zhu,J., Fan,L., and Yang,S.
(2017) Exceptionally high payload of the IR780 iodide
on folic acid-functionalized graphene quantum dots
for targeted photothermal therapy, ACS Appl. Mater.
Interfaces, 9, 22332-22341, https://doi.org/10.1021/
acsami.7b07267.
187. Ding,H., Zhang,F., Zhao,C., Lv,Y., Ma,G., Wei,W., and
Tian,Z. (2017) Beyond a carrier: graphene quantum
dots as a probe for programmatically monitoring an-
ti-cancer drug delivery, release, and response, ACS
Appl. Mater. Interfaces, 9, 27396-27401, https://doi.org/
10.1021/acsami.7b08824.
188. Badrigilan, S., Shaabani, B., Gharehaghaji, N., and
Mesbahi, A. (2019) Iron oxide/bismuth oxide nano-
composites coated by graphene quantum dots:
“Three-in-one” theranostic agents for simultaneous
CT/MR imaging-guided invitro photothermal therapy,
Photodiagnosis Photodyn. Ther., 25, 504-514, https://
doi.org/10.1016/j.pdpdt.2018.10.021.
189. Badrigilan,S., Shaabani,B., Aghaji, N.G., and Mesba-
hi,A. (2020) Graphene quantum dots-coated bismuth
nanoparticles for improved CT imaging and photo-
thermal performance, Int. J. Nanosci., 19, 1850043,
https://doi.org/10.1142/s0219581x18500436.
190. Lee, B., Stokes, G. A., Valimukhametova, A., Nguy-
en, S., Gonzalez-Rodriguez, R., Bhaloo, A., Coffer, J.,
and Naumov, A. V. (2023) Automated approach to
invitro image-guided photothermal therapy with top-
down and bottom-up-synthesized graphene quantum
dots, Nanomaterials, 13, 805, https://doi.org/10.3390/
nano13050805.
191. Sung, S.Y., Su, Y.L., Cheng,W., Hu, P.F., Chiang, C.S.,
Chen, W.T., and Hu, S.H. (2019) Graphene quantum
dots-mediated theranostic penetrative delivery of
drug and photolytics in deep tumors by targeted bio-
mimetic nanosponges, Nano Lett., 19, 69-81, https://
doi.org/10.1021/acs.nanolett.8b03249.
192. Xuan,Y., Zhang, R.Y., Zhao, D.H., Zhang, X.S., An,J.,
Cheng, K., Hou, X. L., Song, X. L., Zhao, Y. D., and
Yang, X. Q. (2019) Ultrafast synthesis of gold nano-
sphere cluster coated by graphene quantum dot for
active targeting PA/CT imaging and near-infrared
laser/pH-triggered chemo-photothermal synergistic
tumor therapy, Chem. Engin. J., 369, 87-99, https://
doi.org/10.1016/j.cej.2019.03.035.
193. Wu, C., Guan, X., Xu, J., Zhang, Y., Liu, Q., Tian, Y.,
Li,S., Qin,X., Yang,H., and Liu,Y. (2019) Highly ef-
ficient cascading synergy of cancer photo-immuno-
therapy enabled by engineered graphene quantum
dots/photosensitizer/CpG oligonucleotides hybrid
nanotheranostics, Biomaterials, 205, 106-119, https://
doi.org/10.1016/j.biomaterials.2019.03.020.
194. Ruiyi, L., Zaijun, L., Xiulan, S., Jan, J., Lin, L., Zhig-
uo, G., and Guangli, W. (2020) Graphene quantum
dot-rare earth upconversion nanocages with extreme-
ly high efficiency of upconversion luminescence, sta-
bility and drug loading towards controlled delivery
and cancer theranostics, Chem. Engin.J., 382, 122992,
https://doi.org/10.1016/j.cej.2019.122992.
195. Liu,H., Li,C., Qian,Y., Hu,L., Fang,J., Tong,W., Nie,R.,
Chen, Q., and Wang, H. (2020) Magnetic-induced
graphene quantum dots for imaging-guided photo-
thermal therapy in the second near-infrared window,
Biomaterials, 232, 119700, https://doi.org/10.1016/
j.biomaterials.2019.119700.
196. Zhang, X., Ong’achwa Machuki, J., Pan, W., Cai, W.,
Xi,Z., Shen,F., Zhang,L., Yang,Y., Gao,F., and Guan,M.
(2020) Carbon nitride hollow theranostic nanoregu-
lators executing laser-activatable water splitting for
enhanced ultrasound/fluorescence imaging and coop-
erative phototherapy, ACS Nano, 14, 4045-4060, https://
doi.org/10.1021/acsnano.9b08737.
197. Prasad,R., Jain, N. K., Yadav, A.S., Jadhav,M., Rad-
harani, N.N.V., Gorain,M., Kundu, G.C., Conde,J.,
and Srivastava,R. (2021) Ultrahigh penetration and
retention of graphene quantum dot mesoporous silica
nanohybrids for image guided tumor regression, ACS
Appl. Bio Mater., 4, 1693-1703, https://doi.org/10.1021/
acsabm.0c01478.
198. Yang,Y., Wang,B., Zhang,X., Li,H., Yue,S., Zhang,Y.,
Yang,Y., Liu,M., Ye,C., Huang,P., and Zhou,X. (2023)
Activatable graphene quantum-dot-based nanotrans-
formers for long-period tumor imaging and repeat-
ed photodynamic therapy, Adv. Mater., 35, 2211337,
https://doi.org/10.1002/adma.202211337.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.