Back to Journals » International Journal of Nanomedicine » Volume 19

Graphene-Based Photodynamic Therapy and Overcoming Cancer Resistance Mechanisms: A Comprehensive Review

Authors Dilenko H , Bartoň Tománková K , Válková L , Hošíková B , Kolaříková M , Malina L , Bajgar R, Kolářová H

Received 8 February 2024

Accepted for publication 9 May 2024

Published 11 June 2024 Volume 2024:19 Pages 5637—5680

DOI https://doi.org/10.2147/IJN.S461300

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. RDK Misra



Hanna Dilenko, Kateřina Bartoň Tománková, Lucie Válková, Barbora Hošíková, Markéta Kolaříková, Lukáš Malina, Robert Bajgar, Hana Kolářová

Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic

Correspondence: Kateřina Bartoň Tománková, Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Hněvotinska 3, Olomouc, 77900, Czech Republic, Tel +420 733 690 914, Email [email protected]

Abstract: Photodynamic therapy (PDT) is a non-invasive therapy that has made significant progress in treating different diseases, including cancer, by utilizing new nanotechnology products such as graphene and its derivatives. Graphene-based materials have large surface area and photothermal effects thereby making them suitable candidates for PDT or photo-active drug carriers. The remarkable photophysical properties of graphene derivates facilitate the efficient generation of reactive oxygen species (ROS) upon light irradiation, which destroys cancer cells. Surface functionalization of graphene and its materials can also enhance their biocompatibility and anticancer activity. The paper delves into the distinct roles played by graphene-based materials in PDT such as photosensitizers (PS) and drug carriers while at the same time considers how these materials could be used to circumvent cancer resistance. This will provide readers with an extensive discussion of various pathways contributing to PDT inefficiency. Consequently, this comprehensive review underscores the vital roles that graphene and its derivatives may play in emerging PDT strategies for cancer treatment and other medical purposes. With a better comprehension of the current state of research and the existing challenges, the integration of graphene-based materials in PDT holds great promise for developing targeted, effective, and personalized cancer treatments.

Keywords: graphene quantum dots, graphene oxide, cancer research, photosensitizers, drug delivery

Introduction to Cancer Therapy

Cancer has been one of the leading causes of death in various countries, genders, and age groups in the last two decades, with an estimated total of 10 million deaths in 2020. The major types of cancer leading to fatal outcomes are lung cancer, breast cancer, and prostate cancer.1–5 Cancer is a multifactorial genetic disease, and mutations in the cellular genetic material are necessary for its development. These mutations can gradually accumulate over a lifetime, starting from a precancerous condition and developing into a malignant tumor. Malignant diseases can be divided into acquired mutations and hereditary ones, such as the familial form of retinoblastoma.3,6 External agents that trigger malignant transformation can be divided into physical carcinogens (eg, ultraviolet and ionizing radiation), chemical carcinogens (such as asbestos) and biological carcinogens (bacteria and viruses, such as human papillomavirus, hepatitis B virus), human herpesvirus-8 and H. pylori).3,6–8

The disadvantages of conventional cancer treatments, such as chemotherapy, radiation therapy, and surgery must also be taken into account when discussing cancer (Figure 1). Treatment may not only lead to many harmful side effects, but it can also cause cancer resistance. As a rule, side effects occur when healthy cells and the malignant ones are affected. Side effects usually vary according to a given type of malignancy, person and treatment used.9 These topics will be covered in more detail later in this review.

Figure 1 Advantages and disadvantages of traditional cancer treatment techniques. Green boxes represent advantages and red boxes represent disadvantages.

Chemotherapy

One of the classic types of cancer treatment is chemotherapy, the main task of which is to eliminate tumor cells without significantly damaging healthy tissues, which is obviously impossible with classic chemotherapy agents because they are not tumor cell-specific. The history of chemotherapy began in 1940 with the use of nitrogen mustard.10,11 It is usually administered through the mouth or intravenously in regular intervals called cycles so that the organism can recover after the toxic effect.11,12 Different types of drugs are administered for chemotherapy and include alkylating agents that bind to proteins and nucleic acids, antitumor antibiotics that are produced by bacteria and generate free radicals, antimetabolites (disrupt purine or pyrimidine synthesis), topoisomerase inhibitors that are responsible for disrupting the process of DNA replication and many others.11

Radiotherapy

Radiotherapy started in 1895 after the discovery of X-ray and is used in more than a half of cancer treatment regimens.13 Over the years, huge achievements have been reported in the field with the development of 3D conformal radiation methods such as stereotactic (body) radiotherapy (SBRT) and intensity-modulated radiation therapy (IMRT). In addition, accomplishments in imaging systems have minimized radiation exposure to healthy tissue.13,14 This removes some limitations imposed by the maximum tolerated dose.13 Radiation therapy is also associated with ROS production inside the cells via water radiolysis and cytosolic Rac1/NADPH oxidase system.13,15–17 The other mechanism to influence cancer via radiotherapy is through tumor hypoxia.13

Surgery

Surgery is the oldest cancer treatment and the most effective in the case of localized primary tumors.18,19 It can be used to achieve goals such as removing the entire tumor mass, debulking a tumor in case when removing the entire tumor is impossible, or easing cancer symptoms.19 Compared to both chemotherapy and radiotherapy, surgery makes it possible to eliminate all malignant cells.18

Resistance

As was written above, cancers can also outsmart therapy efforts, and the triggered therapeutic resistance will significantly contribute to cancer mortality.20 Unsuccessful treatment will result from combined factors of pharmacokinetics, TME and the resistance mentioned above.21 The vast majority of cancer therapies are chemotherapy, and, in most cases, tumor recurrence and treatment resistance are observed.20,22–25

Non-genetic/epigenetic changes that occur independently of DNA changes play an important role in cancer development.24–27 Many studies have failed to prove the genetic evolution of the disease in a large number of patients with resistance to therapy.24,28 Non-genetic resistance can occur as drug persistence, unstable non-genetic resistance and stable non-genetic resistance.24 Drug persistence for cell culture is similar to antibiotic-resistant bacteria. It occurs in the population of malignant cells with low frequency and exhibits reduced growth and altered metabolism.24 Remarkably, they are genetically identical to the entire tumor mass which proves that epigenetic mechanisms have a curtail part in them. These cells are not mitotically active but can allow other cancer cells to adapt via genetic mutations or epigenetic changes.24,29–31

Epigenetic heterogeneity refers to the variability of the epigenetic state within a cell population as a result of stimulus.24 Two different theories have been proposed for the emergence of acquired resistance to the treatment: Darwinian theory and Lamarckian theory of cancer cell evolution (Figure 2). Darwinian theory of acquired resistance says that natural selection plays the leading role in resistance and there is always a small population of tumor cells that already have therapy-resistant potential. Darwinian theory acts via heritable variability from accidental changes in the genetic material for which positive selection is necessary. It works either by gradually increasing the number of resistant cells or by gradually increasing the stability of resistance. However, in this case, the frequency of advantageous mutations in the cell population is extremely low, and adaptations are limited by these mutations.23,24,32

Figure 2 Models of cancer resistance include Darwinian cancer resistance theory and Lamarckian cancer resistance theory. The Darwinian theory of cancer resistance is based on principles of natural selection, survival advantage and genetic diversity, resulting in highly resistant population. The Lamarckian theory, proposed by Jean-Baptiste Lamarck, suggests that cancer cells can acquire traits during their lifetime and pass them on to their offspring.24–27

The second widely known theory is Lamarckian theory which postulates that the environment plays a crucial role in developing therapy resistance. During the therapy, epigenetic changes force the drug-refractory phenotype, and selection is not involved in the spread of adaptive changes.23,32–36

Nevertheless, recent research showed us that these two theories do not exclude each other and can co-exist in the same cancer cell population, what leads to the existing cancer cell plasticity theory which is very different from healthy tissue plasticity and stem cell plasticity.23

Also, the new theory “use-it or lose-it” by Catania et al combined different driving factors like environment, phenotypic plasticity, mutations, genetic drift, and others. In this theory, positive selection is not necessary and evolved adaptations stem from existing genetic features that are activated in the specific environment, while genes that are not used are being silenced or even possibly physically lost.32

Also, Charles C. Bell suggested that cancer cells adapt via “the path of most resistance”, which includes a mix of both non-genetic changes and genetic changes.24 In addition to these theories, it has been established that information between cancer cells and TME is transferred by tumor-derived exosomes which are vesicles ranging from 30 nm to 150 nm. They contain the noncoding RNA (ncRNA) which is responsible for treatment resistance and metastasis phenotypes.37,38 The cytochrome P450 enzymes could be associated with drug resistance as well, and they are usually overexpressed in some solid tumors.39 It should also be noted that during radiotherapy, immunosuppressive pathways are activated, which can lead to accumulation of tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), which are all radioresistant.40,41

Photodynamic Therapy in Cancer

On the contrary, photodynamic therapy (PDT) is one of the contemporary non-conventional methods for cancer treatment. In this field, PSs are used along with light of a specific wavelength which will activate them (Figure 3). It focuses on retention of these specific drugs at the tumor site after local or systemic administration.42,43 The antineoplastic effect of PDT originates from three different effects on the human body which area direct cytotoxic effect, damage to tumor blood vessels, and activation of both innate and adaptive immunity.44

Figure 3 PDT is a treatment method that uses a photosensitizing agent and light to destroy abnormal cells. PDT can be divided into two main types of reactions: Type I and Type II reactions which describe the mechanisms through which the ROS are generated to induce cell damage. In Type I reaction, the PS, after absorbing light, transfers an electron to a substrate molecule without participation of molecular oxygen. This creates a highly reactive radical species, often mentioned as a superoxide anion radical which leads to cell damage. It can react with cellular components, triggering oxidative stress and damage to proteins and DNA. This type of reaction is typically non-specific and can damage numerous cellular components. In Type II reaction, after light activation, the PS transfers its energy to molecular oxygen directly producing singlet oxygen, a highly reactive and cytotoxic species. Unlike Type I reactions, Type II reactions are highly specific and primarily target cells containing the PS. The balance between these two reaction types can be influenced by the type of PS used, the presence of oxygen in the target tissue, and the local environment. It is important to consider the generation of singlet as it is mainly responsible for the therapeutic effects of PDT.42

PDT is a promising cancer treatment. However, it is not without limitations as cancer tissue may develop resistance to PDT due to factors such as multiple treatment sessions, changes in protein expression, and alterations in gene expression after irradiation.45–47 PDT can also enhance many intrinsic survival pathways, such as NF-κB, autophagy, anti-apoptotic signals, p53 and many others.46 Also, tumor hypoxia contributes to resistance to oxygen-dependent treatments such as PDT.48,49

Nuclear factor-kappa B (NF-κB) is a transcription factor that plays an important role in both inflammatory and immune responses and cannot be easily considered a target to fight PDT inhibition as it can educate the immune system to fight neoplastic cells but can also help these neoplastic cells to survive the stress arisen by ROS.44,50

It is worth noting that PDT can also trigger autophagy that can add to either resistance or susceptibility to the cancer treatment. It is not a homogeneous process and consists of macroautophagy, microautophagy and chaperone-mediated autophagy.51,52 Autophagy can increase resistance to apoptosis by reusing dysfunctional organelles and cellular components damaged by PDT-induced ROS (Figure 4). This would maintain cellular homeostasis by providing enough energy for cellular vital functions and suppressing anticancer immune effector mechanisms.53

Figure 4 How PDT affects autophagy and cancer evolution. (A) Autophagy is an essential biological process that includes degrading and recycling cellular components. Through this process, cells undergo self-digestion, breaking down their organelles, proteins and other components. Throughout autophagy, a double-membrane structure (autophagosome) forms in stages such as initiation, expansion and maturation around the cellular material subjected to degradation. The autophagosome eventually fuses with lysosomes which leads to formation of autolysosomes and degradation of its contents for future recycling. There are different types of autophagy, including macroautophagy (the most common type), microautophagy and chaperone-mediated autophagy. This allows cells to remove damaged or redundant cellular materials, crucial for sustaining homeostasis. (B) As mentioned before, autophagy is responsible for degradation of dysfunctional or damaged cellular components and organelles, thereby providing cells with both energy and building blocks. Cells without proper autophagy mechanisms are vulnerable to PDT which causes apoptosis or cell death. However, in cancer, this can result in forming resistant cell population. (C) Signaling between tumor cells and their microenvironment can induce a temporary, drug-resistant state in malignant cells. Moreover, in cancer-associated fibroblasts, autophagy facilitates proliferation of adjacent cancer cells. (D) Autophagy also triggers the process of epithelial–mesenchymal transition, leading to more stem-like features in cells. In this case, anoikis, which is a form of cell death occurring after cell detachment, is less likely to happen. Additionally, by providing energy to disseminating cells, autophagy also assists with cancer cell dormancy and metastasis.51

PDT can also increase cell resistance to treatment in terms of autophagy by enhancing signaling interactions between cells and microenvironment or by protecting cells from anoikis and promoting metastasis.51 As several studies clearly show, autophagy has cytoprotective and prosurvival features, and depending on a variety of factors such as the cancer type, PS type and tumor stage, it can range from an antitumor effect to a protumor one.54

In accordance with everything written above, there are also different targets to overcome PDT resistance and enhance its therapeutic effect, mainly GRP78-targeting, survivin targeting, PS modification, two-photon absorption and use of NPs.55–57

Glucose-regulated protein 78 (GRP78) is a heat shock protein that is upregulated in tumor cells after PDT, and it was shown that GRP78 could be overexpressed in cancer cells (especially cells in malignant gliomas that are resistant to conventional chemotherapy and radiotherapy) and contribute to metastasis. Reduction of GRP78 concentration sequentially reduces metastasis development in xenograft models.55,58–60 Targeting via subtilase cytotoxin (SubAB) is the most selective targeting as it cleaves and subsequently inactivates GRP78.55 However, GRP78 suppression is under consideration as SubAB could be the reason for the hemolytic uremic syndrome as it is originally derived from Shiga toxigenic Escherichia coli (STEC) strains.55,61,62

The next target could be survivin which is an inhibitor of an apoptosis protein family, and usually the application of PDT results in an upregulation of survivin in a tumor. It plays an important role in stabilizing mitosis and cell adaptation; thus, the suppression of survival may enhance PDT treatment.57 The first antagonist of survivin is a phosphorothioate antisense oligonucleotide which provides a strong anticancer activity.57,63

It should also be noted that one of the limitations and drawbacks of PDT is a limited light penetration to the tissue, as only NIR light can penetrate deeper into the tissue and most of the PS absorbs light at a shorter wavelength than 700 nm. Therefore, one solution can be using two-photon absorption (TPA)-induced excitation as it uses fewer energy photons but a higher wavelength.56

Another drawback of the most commercially used PS is their poor solubility as most of them are hydrophobic and would aggregate in the aqueous environment (cellular cytoplasm and extracellular environment) which would limit their properties for PDT and could harm normal tissue. Nanoparticles could serve as an essential platform for enhancing PTD and drugs in general.56,64,65

Nanoparticles can be different metal NPs (GNPs), super-paramagnetic iron oxide NPs (SPIONs) or even quantum dots (QDs), and they are capable of carrying a large load of PS on their surface, changing their water solubility and its kinetics and also securing them from early degradation.56,66–69 Changes in kinetics usually occur through a specific mechanism, enhanced permeability and retention (EPR) effect.56,70 NP can be functionalized in many different ways, for instance, Master et al worked on PEG-PCL (Poly (Ethylene Glycol)-block-Poly (ε-CaproLactone) methyl ether, with Phtalo- cyanine-4 (Pc4) which further were functionalized with peptide GE11 specific from the epidermal growth factor (EGR) receptor which results in enhanced uptake in an SCC-15 head and neck cell line.56,71 Meanwhile, Gary-Bobo et al noticed the enhanced uptake of mesoporous silica NPs functionalized with galactose-carrying fluorescein by colorectal cancer cells.56,72

Introduction to Graphene

Graphene is a relatively new nanomaterial, discovered in 2004, which has attracted considerable attention in the scientific community due to its unique physical and chemical characteristics and which plays a vital role in various fields of science.73 Graphene has properties such as high specific surface area, good electrical conductivity, zero bandgap, biocompatibility, and high drug-loading efficiency.74–77 Graphene material is also used for lithium-ion batteries (LIB) due to its electrochemical properties.74,78,79

Graphene is a flat sheet one carbon atom thick (a monolayer). The atoms in its composition are sp2 hybridized and arranged in a honeycomb lattice.80–86 These atoms have four valence bonds, among which one is s and three other p orbitals.87 It can be obtained in various ways, in particular, mechanical exfoliation, chemical vapor deposition (CVD), chemical reduction of graphite oxide, epitaxial growth on SiC, liquid-phase separation, and unzipping of carbon nanotubes (Figure 5).80,86,88,89 CVD method is considered one of the most efficient methods for obtaining monolayer graphene or graphene films with only a few defects. However, this method requires a large number of high-purity gases and high energy costs.74

Figure 5 The example of graphene preparation methods.22,80,88,89,291

Graphene-based materials are often developed as smart platforms for nanocarriers and targeted drug delivery (Figure 6). Such carriers may be sensitive to the tumor microenvironment (TME), in particular, acidic pH and elevated levels of glutathione. In addition, the carriers can be activated by light, magnetic, or ultrasonic stimuli (“exogenous stimuli”). It was also shown that graphene can serve as a heat-conducting basis for increasing local temperature.87 Electricity can also be an exogenous stimulus. Servant et al developed electrosensitive scaffolds based on graphene for polymer implants for drug delivery.91

Figure 6 The versatility of graphene use. As a highly sought-after material, it is used for a wide range of applications in electronics, energy storage, water filtration, biomedicine, and composites.73,74,78,79

Graphene is also selectively absorbed by a tumor. In addition, a high yield of ROS products is characteristic.81 This material has delocalized π bonds responsible for the unique electronic properties that give graphene the ability to heat under NIR (near-infrared) irradiation photothermally. This property is used to ablate tumors.81 Furthermore, graphene is able to adsorb aromatic compounds on its surface due to π-π-electron interaction.81,87,92 Graphene nanomaterials can penetrate the skin, get into the lungs when inhaled and overcome the hemato-tissue barriers when injected, and accumulate in the tissues. Graphene itself accumulates in the kidneys, lungs, and liver when injected intravenously. Nanoparticles are also easily absorbed by mitochondria and cell nuclei. One of the most severe toxic effects of graphene is DNA fragmentation by cellular endonucleases. Moreover, elevated concentrations of heme oxygenase 1 (HO-1), heat shock protein 90 (HSP90), active caspase-3, and endonucleases such as deoxyribonuclease I and endonuclease G are observed.93,94 In addition to cytotoxicity, it is noteworthy that graphene particles can induce stem cell osteogenesis.93

Imperfections such as hydrophobicity and high cost had to be overcome, thus many graphene derivatives have been created. Such derivatives are graphene oxide (GO), reduced graphene oxide (rGO), graphene quantum dots (GQD), graphene nanoribbons (GNR), graphene nanoplates, and many more (Figure 7).80,81,95

Figure 7 The classification of carbon allotropes encompasses several distinct forms, including carbon dots, fullerenes, carbon nanotubes, graphene (a single layer of graphite), graphite (two-dimensional layers), diamonds, and other related variants.96,128

Graphene-based materials can also be used for visualization. Single-Wall Carbon Nanotubes (SWNTs), for example, have NIR photoluminescence and low autofluorescence, making SWNTs promising new NIR fluorophores.98,99 This review describes graphene derivatives such as GO, rGO, GA, and quantum dots in more detail.

Graphene Derivatives

Graphene Oxide and Reduced Graphene Oxide

Graphene Oxide

GO is a budget material with a lattice of carbon atoms bound by sp2 bonds with sp3 defects, well dispersed in water and other solvents due to functional groups on its surface. GO contains functional groups such as hydroxyl, epoxy, carboxyl, carbonyl, phenol, lactone, and quinone groups. These functional groups increase the hydrophilicity of the surface, which means that biochemical reactions and bioconjugation reactions can occur on its basal plane as well as on its edges.26,81,96,100 These functional groups create active sites for covalent or non-covalent modifications, which makes it easy to functionalize GO further using polymers, drugs, and other molecules. GO also showed a high adsorption capacity for proteins. The adsorption mechanism depends on GO morphology, oxidation state, and hydrophobicity. Polypeptides can be adsorbed on the surface of GO by the following interactions: hydrophobic–hydrophobic interaction, van der Waals interactions, electrostatic interactions, and also π–π stacking due to the large number of π-electrons on the basal plane of the GO surface.100 It also exhibits photoluminescence, the wavelength of which varies from near-UV to NIR.101

Such materials have found their way into many areas of science, including drug delivery, corrosion protection, sensors, and water treatment.82,102–104 GO also finds its application in electrochemistry and energy storage: supercapacitors, solid-state electrolytes, and GO in fuel cells.105,106 Moreover, GO can be used to fabricate composites in various forms, such as nanoparticles, hydrogels, films, and fibers.107

This material is easily obtained by oxidizing graphite with concentrated acids and strong oxidizing agents such as H2SO4, HNO3, or KMnO4.82,108,109 This method of obtaining GO was introduced in 1859 by Brodie who oxidized graphite in the presence of potassium chlorate KClO3 and fumed HNO3 + NO2 (Figure 8).109 Then, in 1937 Hofmann and Koenig made several improvements, as well as Hummers and Offeman.110 Marcano et al improved the Hummers’ method by eliminating sodium nitrate (Figure 8).110–113 This change eliminated the formation of toxic nitrous gases. Since these methods use potent oxidizing agents, the resulting GO sheets have significant defects in their crystal network.114 In addition, industrial waste can be utilized as a source of GO. It is possible to extract graphene from industrial waste, namely, the synthesis of GO from waste containing graphene precursors. Such waste may include Li batteries, biowaste (food, grass, insects), charcoal, soot, and others.115–118

Figure 8 Methods of preparation of GO.82,108,109

Mechanical characteristics of GO include internal strength, ductility, brittleness, and others. However, destroying sp2 bonds also decreases internal strength and Young’s modulus compared with graphene. To analyze the mechanical properties of GO, various methods can be used, such as tensile atomic force microscopy.81 The thermal conductivity is also relatively low, and it can be increased by imposing a polymer on the GO surface or by combining GO with metal oxide nanoparticles (for example, TiO2 or ZnO).81

Reduced Graphene Oxide

rGO is a sheet of sp2 carbon atoms with a restored π-electron graphene network and a minimum number of oxygen-containing groups.114,119 Thanks to π conjugation, improved optical absorption, and conductivity, rGO is even more suitable for PTT than GO. GO is reduced using chemical agents or physical methods, during which the carboxyl (-COOH), hydroxyl (-OH), and epoxy (-O-) groups are removed by the reducing agent, which in turn reduces the solubility in water.96 The properties of rGO are similar to those of graphene, and it is possible to change them depending on the reduction method and reduction degree. The most effective and simplest method is chemical reduction.119,120 Reductants such as hydrazine, hydrazine hydrate, dimethylhydrazine, or strong alkalis can be used, as well as green reductants such as honey, tulsi (Ocimum sanctum) leaf extract, cinnamon extract, and green tea extract.114,121,122 The following methods are available: photoreduction, solvothermal reduction, and microwave reduction (Figure 9).114,123

Figure 9 Connections between Graphite, Graphene, GO, and rGO.

Biological Properties and Bio Applications

The data on the biocompatibility of GO are rather contradictory. More and more studies show that GO has a small cytotoxicity, but its manifestation depends on the method of obtaining GO, as well as on the form of GO.81,100,124 For example, GO flakes have rough and sharp edges, which allow them to disrupt the integrity of a bilipid layer of a membrane, disrupt the membrane potential, and are widely distributed in the whole volume of tumors (in particular, tumors of the nervous system such as glioblastoma). These data demonstrate the potential of graphene as a delivery vector for both drugs and various proteins.100,119,125 This is especially important for potent aromatics that are insoluble in water. Modification of GO with polyethylene glycol (polyethylene glycol) makes it possible to create a biocompatible GO-PEG conjugate, stable in biological solutions, which can add hydrophobic aromatic molecules like SN38 (analogous to camptothecin) via π-π-stacking.126 GO-PEG can also be loaded via π-π stacking with doxorubicin (DOX), which is hydrophobic. Thus, altered GO exhibits more potent cellular toxicity.97,127 The release of imposed drugs is possible with the help of various stimuli, as discussed above. One of these incentives can be electricity. In their work, Weaver et al showed that it is possible to release the anti-inflammatory molecule, dexamethasone, in response to voltage stimulation with a linear release profile.128

Graphene itself and rGO, on the other hand, exhibit high cytotoxicity, which can also be explained by their geometry and spatial structure. rGO also has a large number of delocalized electrons due to low oxygen content, which leads to disruption of signaling pathways in the cell.81 Wang et al showed in their work that exposure to GO below 20 μg/mL on cells (Human Fibroblast Cells, HDF) exhibits low cytotoxicity with cell survival over 80%.76 At concentrations above 50 μg/mL, GO exhibits obvious cytotoxicity.76 It has been shown that GO is internalized by cells and is mainly localized within endoplasm and organelles, such as lysosomes, and mitochondria. The adhesiveness of HDF cells treated with GO was also analyzed. Western blot results showed that cells cultured with GO have markedly reduced expression levels of laminin, fibronectin, focal adhesion kinase, and the cell cycle protein cyclin D3 compared to untreated cells.76 Regarding the effects of GO on living organisms, namely mice, the injection dose of 0.1 and 0.25 mg GO per mouse did not cause death in the exposed animals and showed no clinical signs of toxicity. However, Wang et al also showed that in the group of mice treated with 0.4 mg per mouse, 4 out of 9 died, and their death was usually preceded by lethargy, inactivity, and weight loss.76

GO can also be used for work in the field of regeneration and tissue engineering, especially to restore bone tissue in severe lesions.129,130 One of the modern materials to be applied in this field is GO aerogels. These aerogels are strong, have a porous structure, and can imitate bone tissue. Another advantage is their ability to absorb growth factors on the surface.129,130

Moreover, GO and rGO show high antibacterial activity. As already mentioned, these materials mainly affect bacteria through direct contact with sharp and superoxide anion-independent oxidation.131–133 Lipid peroxidation plays an important role as well; GO nanosheets can also trap bacteria and are able to extract phospholipids from cell membranes due to dispersion interactions between GO and lipids.134,135 Also, when modifying GO and rGO with silver nanoparticles, it is possible to achieve a synergistic effect.120,134

Graphene Acid

Graphene acid (GA) is a graphene derivative that contains evenly spaced carboxylic acid groups directly bonded to the sp2 carbon backbone and has several advantages over the commonly used GO. Such advantages are aromaticity and a large number of homogeneously distributed COOH groups on the basal plane. GA luminescence has a maximum of 500nm. This product has excellent conductivity and biocompatibility and can be used as a catalyst and an electrocatalyst.136–139 By oxidation with permanganate, GA can be obtained according to Tour’s method. During the first oxidation, GO is obtained, and during the second one graphene acid. The total volume of the sample decreases by about three times, which indicates the oxidation of GO to CO2.137

Further, one of the most important applications of GA is environmental cleaning, in particular removing heavy metals, since this plays a key role in the global issue of drinking water availability. In this case, GA with 33% by weight carboxyl groups is one of the solutions to this problem, as it has proved to be able to remove highly toxic metals such as Cd2+ and Pb2+.139

It is also possible to modify GA via carboxylic acid groups. One option is covalent functionalization.136,140 For instance, Mosconi et al functionalized GA surface with ferrocene (Fc) moieties through carbodiimide chemistry.140 It allowed the introduction of up to 3.6% at. of iron as Fe2+ ions.140 The next options are non-covalent functionalization, nanoparticles, and single metal atom immobilization on GA.141,142 Bioinspired nickel bis-diphosphine HOR catalyst was grafted on GA by Reuillard et al.141 The immobilization of Sm2O3 particles by Sanad et al could be an example of functionalization with nanoparticles.143

Carbon Quantum Dots (CQDs), Graphene Quantum Dots (GQDs), and Graphene Oxide Quantum Dots (GOQDs)

Carbon Quantum Dots (CQDs)

CQDs were mentioned and obtained during the isolation and purification of single-walled carbon nanotubes for the first time by Xu et al144,145 Later, Sun et al named these fluorescent carbon nanoparticles “carbon quantum dots”.144,146 This novel material solved several problems the conventional graphene had, as CQDs have good solubility and strong luminescence, for which they are referred to as carbon nanolights.147,148

CQDs are a mixture of sp2 and sp3 carbons in a quasi-spherical crystalline structure with their properties being directly linked to the π-electron state of the sp2 carbons.149 Their size is up to 10 nm.147 The photoluminescence is size dependent, the size of the conjugated π-domains influences photoluminescence, with changes either promoting or inhibiting the direct transition of electrons from the conduction band to the valence band. This transition is responsible for generating band gap fluorescence.149–151 CQDs also show a clear dependence of photoluminescence on the excitation wavelength.147 When photoexcited, CQDs demonstrate outstanding capabilities as both electron donors and acceptors.147,152 As well, CQDs exhibit optical absorption in the UV region, with a tail extending into the visible range.147

There are various methods of CQD synthesis that are classified into top-down and bottom-up routes. Top-down methods involve the reduction and fragmentation of large sp2 carbon domains into smaller components. These techniques include arc discharge, chemical oxidation, sonication, hydrothermal methods, and others (Figure 10).144,146,153–155 Bottom-up approaches for synthesizing CQDs mean constructing the material from precursor molecules, yielding particles with consistent sizes and control over size distribution. These methods cover hydrothermal treatment, ultrasonic treatment, thermal decomposition, pyrolysis, carbonization, microwave synthesis, and the electrochemical method (Figure 10).156–158

Figure 10 Various preparation methods can be employed to obtain CQDs, utilizing different carbon sources and synthesis procedures. The most usual division of preparation methods are top/down and bottom-up methods. Commonly used carbon sources include citric acid, glucose, and carbon black, while synthesis procedures range from hydrothermal and microwave-assisted methods to electrochemical and pyrolysis techniques.156–158

Graphene Quantum Dots (GQDs)

GQDs are a new zero-dimensional material with lateral sizes up to 100 nm, most often 3–20 nm, single nanosheets of sp2 carbons with luminescence properties, exceptional optoelectronic properties, and excellent biocompatibility.159–163 Also, GQDs are low-cost, optically and chemically inert, and easy to fabricate. GQDs have applications in areas such as drug delivery, bioimaging, sensors, photovoltaic devices, and catalysis.161,164–166

GQDs have negatively charged carboxyl groups, which can provide good electrostatic properties for further functionalization. Conjugated π–π bonds also contribute to it.167 GQDs are considered non-toxic, particularly to human cell lines. However, cytotoxicity can increase due to the nonspecific adhesion of dots to the cell membrane.166,168

There are various methods for synthesizing GQDs, which are classically divided into top-down or bottom-up ones. The top-down method involves destroying the graphene sheet, CNTs, the graphite using arc discharge, chemical or laser ablation, chemical or electrochemical oxidation, and ultrasound.162,169,170 The bottom-up methods include carbonizing organic precursors such as citric acid, amino acids, carbohydrates, and some aromatic organic compounds using microwave treatment, hydrothermal treatment, solvothermal treatment, or other methods.166,169–171 GQDs were first synthesized by Pan et al in 2010.162 They had a crystalline structure of single or a few layered graphene and had an elliptical or circular shape. However, there may also be quadrate, hexagonal, as well as triangular GQDs.162,169

Graphene Oxide Quantum Dots (GOQDs)

As it was mentioned earlier, GO attracted attention among researchers due to its minimal toxicity, biocompatibility and hydrophilicity. GOQDs are nanoscale carbon-based materials that are derived from GO. These quantum dots possess unique optical and electronic properties due to their small size up to 30 nm and quantum confinement effects.160,172–174

On their basal plane and at the edges, GOQD have oxygen-rich functional groups such as epoxy, carbonyl, hydroxyl, and carboxyl groups, which facilitate further functionalization through electrostatic interaction, π–π stacking and chemical reactions.175

Preparation of GOQDs includes oxidizing, exfoliating, and cutting carbon precursors into nano-sized particles using chemical oxidation, hydrothermal, or solvothermal treatments under harsh conditions, often requiring concentrated acids like HNO3 or H2SO4 for prolonged time.172,176,177

GOQDs have applications in diverse fields, regarding their distinct characteristics. They find utility in removal water pollutants, biological imaging, optoelectronic sensors, LEDs, fluorescent agents, lithium-ion batteries, and many others.160 GOQDs hold promise for biomedical applications due to their non-toxicity, hydrophilicity, and high light-emitting efficiency, which originate from quantum confinement and edge effects associated with their oxygen-functional groups.156,177

Biological Properties and Bio Applications

Due to their properties, CQDs and GQDs can be widely used in various fields, including drug and gene delivery, biological imaging, electrochemiluminescence sensors, electrochemical sensors, and more (Figure 11). Numerous studies have confirmed the DNA fragmentation activity of therapeutic drugs when used in conjunction with GQDs.178 For example, Fang et al developed a multifunctional GQD complex of hollow carbon nanoparticles to encapsulate DOX at the average size of 120 nm. They were also able to generate heat when irradiated with an NIR laser for synergistic photothermal therapy (PTT).169,179 When functionalized with an antibody, selective destruction of cancer cells in vitro is possible.91

Figure 11 The features of CQDs.159,160,162,163,169,180

It has also been reported that GQDs are a promising potential treatment and a way to relieve the symptoms of amyloidosis, the essence of which is the aggregation and deposition of amyloid proteins in plaques around cells, which subsequently causes organ and tissue failure. Misfolded amyloid proteins are also the cause of brain tumors, Alzheimer’s disease, Parkinson’s disease, and stroke. GQDs can act as inhibitors of aggregation and, consequently, toxicity of amyloid proteins.181,182

Moreover, the fluorescence of CQDs and GQDs can be used to visualize living cells in the NIR range and to selectively recognize and bind to cancer cells, such as B-cell lymphoma.91 They are also characterized by their resistance to photobleaching due to their crystal structure. From this point of view, GQDs are superior to CQDs.183,184 GQD could be further functionalized with PEG and can selectively accumulate in the tumor after being injected as an agent for tumor fluorescence imaging.185

Shi et al developed coated GOQD magnetic nanocomplexes with high fluorescence, which can improve the diagnosis of cancer in infected blood using multiphoton luminescence.176 Pramanik et al reported short sequences of artificial RNA conjugated graphene oxide-based for improved two-photon selective imaging of breast tumor cells.186 Also, GOQD can be used as a two-photon fluorescence probe for imaging multiple drug-resistant bacteria (like Methicillin-resistant Staphylococcus aureus).187

Nanotubes and Nanohorns

Single-Walled Nanotubes and Multi-Walled Nanotubes

Carbon nanotubes can be divided into two groups: single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs). They are rather different in terms of their physical properties due to their structural differences, namely the number of carbon layers. SWCNTs, respectively, consist of a single layer of graphene with a diameter of 0.4 to 2 nm. MWCNTs, on the other hand, consist of two or more sheets of graphene with a distance between layers of 0.34 nm that form cylinders, so their diameter is from 1 to 3 nm.188 SWCNTs can be further divided into the following three groups: armchair, zigzag, and chiral (Figure 12).189 Carbon nanotubes have properties such as high rigidity (Young’s modulus 1 TPa) and strength with a tensile strength of 60 GPa for SWCNTs and 150 GPa for MWCNTs and stability at high temperatures (in vacuum and air, the limiting temperatures are 2800 °C and 750 °C, respectively). They also exhibit high electrical conductivity and high heat transfer coefficient.189–192 Their application is possible in areas such as electronics, sensors, and biomedicine, including the delivery of drugs to target organs.193–195 They have a large surface area, which enables the conjugation of various molecules on the walls in large quantities. Molecules containing aromatic groups can also be noncovalently bonded due to strong π–π interactions.195

Figure 12 Carbon nanotubes are nanostructures that take the form of cylindrical tubes by rolling up a sheet of graphene. They can be divided into the following three types: zigzag, armchair, and chiral. Zigzag nanotubes have chirality (n,0), hexagons at the tube end with edges resembling a zigzag pattern, and are typically metallic, with no band gap, affecting their electronic properties. Armchair nanotubes have chirality (n,n), and hexagons at the tube ends, with edges running parallel to the tube axis, resulting in a flat, open-ended structure. Depending on the tube’s diameter, they can either be metallic or semiconducting. The tunability of the electronic properties of chiral nanotubes makes them versatile for various applications.189,196

CNTs can typically be synthesized by the following methods: arc discharge, laser ablation, and chemical vapor deposition. The synthesis of individual SWNTs requires catalysts such as cobalt, nickel, iron, and others. In the synthesis by the arc discharge method, a high temperature of more than 3000°C is required, which mediates the evaporation of carbon atoms into the plasma to form various CNTs. Iijima used this method to synthesize MWCNTs. The next synthesis method, chemical vapor deposition, uses such precursors as methane, ethylene, and similar. The laser ablation method uses the evaporation of graphite in an electric furnace heated to 1200°C.188

Carbon nanotubes also have significant antimicrobial activity, which may be due to the synergy of physical and chemical effects. The intracellular content of bacterial cells is released through physical damage to the membrane, as described above. This process, however, depends on the size of the CNTs. Small CNTs can be internalized by bacteria and disrupt the metabolic processes in cells through oxidative stress.89

CNTs have several drawbacks, as well. Firstly, CNTs are inherently hydrophobic and insoluble in most biological media, making them difficult to use as a material for drug delivery or biomolecules. To overcome this problem, CNTs are functionalized for improved solubility and biocompatibility, allowing further drug modification with growth factors, antibodies, and so on. The methods of such functionalization can be noncovalent functionalization outside CNTs, functionalization of defects, covalent functionalization, and encapsulation of bioactive molecules inside CNTs.195,197 Also, functionalization methods can be divided into chemical and physical. The surface of CNTs can be chemically modified by oxidation, cycloaddition, and addition of functional groups such as carbenes, nitrenes, and similar. Physical modification can include methods such as the π–π stacking described above, coating with polymer chains, using surfactants, and adsorption via hydrophobic interactions.190

Secondly, CNTs may exhibit toxicity, which may be caused by their spatial conformation. Thus, carbon nanotubes and other fibrous materials will have an impact on living cells due to their jagged and flat edges.86

Nanohorns

Single-Walled Carbon Nanohorns (SWNHs) were first presented by Iijima in 1999. As a rule, SWNHs do not exist separately; they exist in a spherical aggregate of 80–100 nm in size in the amount of about a hundred.198–200 The cones are formed by cutting a graphene wedge and seamlessly joining open edges, and individual SWNHs are 1–2 nm in diameter at the ends and 4–5 nm at the base.199,201 They also have many advantages over nanotubes, they are widely used as drug delivery structures, do not require additional treatment with strong acids, and can be mass-produced with a high disassembly yield at a room temperature under Argon atmosphere at a high purity of about 95%.202–204

All production methods for SWNHs are based on disassembling and reorganizing carbon products.204 Various operating parameters optimized during synthesis, such as pressure and temperature, result in different forms of SWCNH with different purities and morphologies.202,205

The closest structural analog of SWNHs is nanotubes due to the similarity in the atom arrangement. Due to this, SWNHs acquire similar physicochemical characteristics and hold promise as catalysts, as well as fuel source and basis for electronic cells. However, there are also differences between SWNHs and carbon nanotubes when interacting with living cells, including apoptosis, protein expression, membrane disruption, and interaction with membrane proteins.130 Nanohorns are superior to na notubes due to the absence of a metal catalyst during synthesis and the possibility of their mass production at ambient temperature. Currently, SWNHs are a platform for drug delivery and can be applied to PTT.198,206,207

Fullerenes

Fullerenes, like Buckminsterfullerenes, were discovered in 1985.208,209 The most extensive group of this family consists of C60 fullerenes with 60 carbon atoms in a spherical structure, which is a truncated icosahedron containing twelve pentagonal rings separated by twenty hexagonal rings, with sp2 hybridization and resembling a soccer ball.208,210 However, sp2-hybridized carbon atoms in fullerenes have a pseudo sp3-character since the arrangement is not flat but pyramidal.199,211 Subsequently, C20, C70, C80 and even larger molecules were found.210 Upon functionalization of C60, spectrum absorption expands to the near-IR region.208

The main disadvantage of fullerenes is their low solubility in polar solvents, which complicates their use.208 In biological systems, the hydrophilicity of materials is more important than hydrophobicity, and methods such as preparation of two-phase colloidal solutions, synthesis of fullerene derivatives, encapsulation in cyclodextrins, polyvinylpyrrolidone, micelles and liposomes, and chemical modification are used to increase hydrophilicity of fullerenes and solubility in water. Chemical modification can be carried out by adding hydrophilic substances such as amino acids, carboxylic acids, and amphiphilic polymers.212 On the other hand, the apolar character of fullerenes allows their use by penetrating cell membranes and being used in lipid-like systems.212,213

Fullerenes also have extended π-conjugation of molecular orbitals, which causes the absorption of UV–visible light. Due to this property, as well as the long lifetime of the excited triplet state, fullerenes are effective PSs.208 This accounts for their significant antimicrobial activity.214 Therefore, irradiation of fullerenes and their derivatives with white light produces ROS.215 Oxidation of vital cellular structures or components without ROS formation is also possible.89,216 Several methods for synthesizing C60 are available, such as arc discharge, chemical vapor deposition, laser irradiation, combustion, and evaporation of carbon sources.217 Fields of fullerenes application include nucleic acid delivery, topical drug delivery, biosensors, and many others (Figure 13). 218–222

Figure 13 Fields of fullerenes application.218–222

Biological Properties and Bio Applications

With the help of the antioxidant activity of fullerenes described above and the ability to penetrate the skin, fullerenes can be a promising material in transdermal delivery and cosmetic applications.223 It is also possible to eliminate the side effects of chemotherapy by imposing the medicinal substance on a fullerene and its subsequent targeted delivery. For example, it is possible to eliminate the side effects of DOX associated with cardiomyopathy.223,224

The possibility of binding fullerenes and other medicinal substances was also described above. Lysozyme can serve as an example; the C60-lysozyme complex showed effective endogenous activity of ROS after the exogenous formation of H2O2 in HeLa cells.213,225 In some cases, suppression of ROS production is possible, which allows the use of fullerenes as neuroprotective agents.226

There are also reports on the ability of fullerenes to inhibit the activity of HIV-1 protease. The antiviral activity is due to the fullerenes attaching to the active center via van der Waals bonds.213,227,228

HIV-1 maturation is disrupted by altering protease activity and molecular interactions of structural proteins. New cationic derivatives of N, N-dimethyl fulleropyrrolidinium iodide inhibit more than 99% of HIV-1 infectivity at low concentrations.229

Nanoribbons

Nanoribbons were described in 1996 by Fujita et al and first mentioned in 1990 by Murayama and Maeda.230–232 GNR are characterized by remarkable electrical and mechanical properties, due to their ultra-large surface area and their structure. They can be considered unfolded CNTs with a high length-to-width ratio.230,233,234 Chemically, they are sp2-hybrid carbon networks with a crystalline structure and a honeycomb lattice shape which readily aggregate in solid or dissolved form. Their oxygenated derivatives attract attention in the field of biomedicine due to the presence of oxygen functional groups (hydroxyl, carbonyl, and carboxyl groups) at the edge and on the base plane, with the help of which subsequent functionalization is possible.233,234 Their photoluminescent nature originates from the reduction of the π-electron network.235 The three most common types of structures are “armchair” or AGNRs, “zigzag” or GNRs with zigzag edges (ZGNRs), and “cove”.230

Nanoribbons can be used in areas such as sensor fabrication (mechanical, chemical, photo, and acoustic sensors), gene and drug delivery, and tissue engineering.230,236 They are considered excellent candidates for drug and gene delivery.237,238 In addition, they have high adsorption and synergistic effects when combined materials are used.236 It is possible to create electrochemical biosensors to detect drugs such as nimesulide, dobutamine, nifedipine, and imatinib.236,239–241 Moreover, there are biosensors for detection of insecticides, pesticides, and toxins, as well as further optical biosensors, luminescent biosensors and colorimetric biosensors.236 Semiconductor nanoribbons on insulating substrates may also be useful for digital circuits.242 They also stimulate the production of ROS in the cell and inhibit proliferation, apoptosis, and DNA fragmentation. They may have toxic effects due to mechanical damage to the cell membrane, as well.230

Regarding the functionalization mentioned above, it is possible to functionalize the GNR with the help of metals. Liu et al successfully created graphene nanoribbons in conjunction with Au (111) through a stepwise reaction using two precursor molecules co-adsorbed on surfaces.243 To determine biomarkers, the GNR surface is functionalized by antigen immobilization.236,244

GNR synthesis includes the following methods: cutting graphene, electron beam lithography, plasma etching, nanoparticle etching, epitaxial growth on silicon carbide, and others.230,245–248 Graphene and its derivatives present a widely used method for preparing GNR, as cutting from graphene sheets is readily available.248 The process consisted mainly of exfoliating graphite by annealing at 1000 °C, followed by sonicating in a dichloroethane solution and centrifuging following suspension.232 Electron beam lithography consists of etching carbon atoms with the help of a scanning tunneling microscope tip.232

Chowdhury et al reported oxidized GNRs (O-GNRs) as delivery vectors for gene therapy.249 Efficient loading of double-stranded DNA was carried out on O-GNRs which were synthesized by longitudinal unzipping of multi-walled carbon nanotubes (MWCNTs). The bond between O-GNR and DNA is stable when ionic strength is reduced or treated with surfactants. O-GNR or DNA-O-GNR complexes <100 µg/mL were not cytotoxic.250

Biocompatibility and Toxicity

Derivatives such as nitrogen-doped graphene nanoribbon aerogels (N-GNRA) can be a worthy scaffold for various cell-line cultures. Superhydrophilicity and three-dimensional structure make N-GNRA a potential material in biomedicine. Liu et al reported human medulloblastoma cells (DAOY) cultured on the surface of N-GNRA for cytotoxicity analysis. Within 24 hours, most DAOY cells were found to adhere to the N-GNRA scaffold and after 3 days, DAOY cells proliferated and migrated along the skeleton and into the porous structure of N-GNRA.251

Another derivative of O-GNR, coated by amphiphilic polymer (1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N [amino (polyethylene glycol)]) (O-GNR-PEG-DSPE), also does not show a toxic effect in the case of MCF-7 and rat glial progenitor cells (CG-4) and similar. On the other hand, its complex with the antitumor drug Lucanthone (Luc-O-GNR-PEG-DSPE) was significantly toxic to malignant glioblastoma tumor cell-line U251.252,253

According to Mbeh et al, O-GNRs synthesized by oxidative decompression of MWCNT and functionalized with albumin at various concentrations may exhibit cytotoxicity.254 The loss of viability of the human epithelial cells on which the material was tested is dose-dependent. Concentrations of ≤50 μg/mL did not exhibit significant cytotoxicity on the cells, while the concentration of 100 μg/mL exhibited significant cytotoxicity, causing proliferation, inhibition, and apoptosis.254 The toxic effect of sonicating GNRs due to structural disruption is also an interesting feature.255

Use of Graphene-Based Materials to Enhance PDT

Functionalization of Graphene and Graphene Derivates

As stated before, a huge variety of NPs can be used for PDT but carbon-based and graphene-based materials stand out among them due to their properties.256,257 Such materials as graphene, its oxide, GA, carbon nanotubes, CNR, fullerenes, CQDs and many others could be used as therapeutic agents, drug carriers, photoactive sensors and diagnostic platforms in cancer and other diseases.256,258,259 Graphene nanostructures in many cases should be further functionalized for therapeutic purposes covalently or noncovalently, so their exceptional properties would be increased in biocompatibility, water solubility and many others.256,260,261

Noncovalent functionalization of graphene means functionalization via weak interactions such as π-π stacking, van der Waals forces, hydrogen bonding, electrostatic interactions, or coordination bonding, without breaking the carbon–carbon bonds of graphene. This type of functionalization offers preserving the structural integrity and its unique properties via a relatively simple process where materials and graphene interact without a chemical reaction.262 However, the noncovalent functionalization of graphene does not significantly change the electronic transport which is crucial for sensing applications.263,264 For example, molecules with π-conjugated systems can interact with the π-electron system of graphene through π-π stacking or polymers with functional groups like hydroxyl, carboxyl, or amine can interact with graphene via hydrogen or electrostatic interactions.262,265–267 Another option is the adhesion of molecules to graphene surfaces through van der Waals forces or coordination bonding.262,268

Functionalizing graphene with PEG typically also involves noncovalent functionalization. PEG is a hydrophilic polymer with terminal hydroxyl groups, which can form hydrogen bonds with the oxygen-containing functional groups on the graphene surface.269 PEGylation can increase stability of graphene materials, inhibit aggregation, reduce toxicity, and prolong blood circulation half-life improving pharmacokinetics of graphene-based materials.269,270 For PEGylation, simple mixing of both graphene material and PEG with incubation under ultrasonication for 30 min at room temperature is enough in order to initiate physical adsorption, as Mendonça et al stated.269 Graphene materials can also be functionalized with further polymers such as polystyrene sulfonate (PSS).271 Du et al incorporated GQDs into poly3,4-ethylene-dioxythiophene:polystyrenesulphonate (PEDOT:PSS) conducting polymer mixture. This incorporation was possible due to the electrostatic interaction between oxygen-containing group of GQDs and PSS and also becouse of π-π conjugation between PEDOT and GQDs.271 Another example introduced Hong et al, when PSS-rGO complex was prepared by simple mixture of GO and PSS in deionized water under 30 min sonication.272 Alternative polymer that could be coupled with graphene-based materials is dextran. Jin et al reported preparation of GO hybrids functionalized with hematin-conjugated dextran. Preparation included a mixture of dextran and GO solutions with further addition of ammonia solution and 50% hydrazine solution and stirring under nitrogen at 60 °C for 3.5 h.273 Polyvinyl alcohol (PVA) is the next important polymer that can be used for graphene functionalization.274–276 Ma et al reported a simple mixing of 10% (w/v) PVA and GO solutions at 40 °C for 3 h and further sonication treatment for 30 min to obtain a GO/PVA complex.274 A similar process was also reported by Morimune et al.275 Use of other polymers via electrostatic interactions like polyethyleneimine (PEI), polypropylene imine (PPI), and poly(amidoamine) (PAMAM) is also possible.267,277,278

It is also possible to functionalize graphene with such macromolecules as DNA. Thiolated DNA was shown to bind noncovalently to both graphene and GO surfaces.279,280 It is also possible to create a 3D hydrogel through a self-assembly process of GO/DNA material.279,281 Xu et al fabricated GO/DNA hydrogel by mixing equal volumes of the GO aqueous solution and the aqueous solution of double-stranded DNA followed by heating at 90 °C for 5 min.281 However, EDC/NHS chemistry is the most common method of attaching antibodies and ssDNA onto graphene and its derivatives. Enzymes are typically immobilized using physisorption.282 Lu et al successfully fabricated rGO sheets noncovalently functionalized with β-lactoglobulin. This hybrid material was characterized with pH-dependent water solubility and was an efficient platform for further Au nanoparticles self-assembly.279,283

Covalent bonds are much stronger than noncovalent and can be achieved through various chemical reactions, such as oxidation, reduction, diazonium chemistry, cycloaddition reactions, cross-linking, and doping. This can enhance properties like solubility, stability, and reactivity, making graphene more versatile for further applications.262 Covalent functionalization improves the water dispersibility and stability of graphene derivates.262,284,285 Carboxylic acids or hydrophilic polymers are used the most. Carboxylic treatment is simple, and attachment of carboxyl, epoxy, and hydroxyl side groups on graphene leads to its increased dispersibility in the aqueous phase.262 However, carboxylic acids can introduce other active molecules, and this may result in the creation of additional defects in graphene sheets.262 It can also be challenging to realize the covalent functionalization of pristine graphene due to its lack of conjugatable functional groups.286

Doping is a prevalent method that is used to modify the electronic characteristics of semiconductor materials. Carbon nanotubes can undergo doping with nitrogen or boron atoms to achieve n-type or p-type materials, respectively.286,287 Yalcin et al reported fabrication of GO-doped LaB6 composite for photodiode base.288 For this purpose, LaB6 was stirred into deionized water for 30 min. Gradually, GO solution was added to the mixture, and then homogenized via ultrasonication for 30 min. Subsequently, hydrothermal processing was applied on the homogenized mixture for 24 h at 120 °C with the following drying at 60 °C for 24 h in a vacuum oven.288 Zhai et al reported fabrication method for N-doped CQDs from garlic skins without any additional organic reagents that had a potential application as a fluorescent ink or smart fluorescent sensor.289 Rishabh et al reported another green synthesis of N-CQDs from Aegle Marmelos.290 Aegle Marmelos leaves were crushed with a household mixer. For nitrogen doping, a suspension solution was made with solid urea in deionized water, which was later added to the leaf paste. The mixture was further irradiated with microwave waves, filtered, and dialyzed.290

Numerous polymers are capable of serving as both covalent and noncovalent modifiers for graphene and its derivatives. Liu et al achieved successful modification of GO for delivering water-insoluble cancer drugs. They utilized PEG conjugated to the carboxylic acid groups on GO via carbodiimide-catalyzed amide formation.286,291 Kim et al reported rGO-PEI complex via covalent conjugation.277 Branched polyethyleneimine (BPEI) was linked to the carboxylic groups of graphene oxide (GO) utilizing EDC/NHS chemistry. Following this, BPEI-GO underwent reduction by hydrazine monohydrate.277

Another successful method for functionalization involves the electrophilic substitution of aryl diazonium salt onto the surface of graphene sheets.90,284,292 Aryl diazonium salt molecules have been effectively utilized to alter carbon materials, such as SWCNTs, graphite, and many others.286,293–296 This modification process can be done under mild conditions, either through simple agitation or electrochemical reduction adsorption, often accompanied by the release of N2. The use of aryl diazonium salt functionalization chemistry on nano-carbon materials was pioneered by Bahr et al.293

Covalent functionalization also represents a viable approach for the conjugation of graphene and its derivatives with biomolecules such as enzymes, antibodies, and others. Ganji Arjenaki et al fabricated an antibody-conjugated GQD complex for in vivo breast cancer imaging and biodistribution.297 For that GQDs were dissolved in a mixture of DMF and water and then, via EDC/NHS chemistry pembrolizumab, which is a humanized antibody used in cancer immunotherapy, was conjugated to GQDs.297 Kaushal et al reported the preparation of a complex system of AuNPs coated with PEGylated GO and antibody conjugated to GO for rapid visible detection of food-borne bacteria.298 Before the addition of antibody, PEG-GO-AuNPs were activated by EDC/NHS.298 As to enzyme immobilization, Royvaran et al fabricated GO nanosheets that were further decorated with superparamagnetic iron oxide nanoparticles on which xylanase was later immobilized. Both covalent and noncovalent immobilizations were utilized.299

Mechanisms of Graphene-Based Materials in PDT

There are several ways in which graphene-based materials can improve PDT efficacy. This could happen via light absorption, energy transfer, electron transfer, enhanced ROS production, synergistic effects with photosensitizers, and enhanced photothermal effect.300–304 In many ways each process cannot be separated from one another and usually, it is a combination of the abovementioned factors.

When irradiated with light, graphene can absorb photons and transfer the absorbed energy to nearby molecules, including oxygen and PSs. This energy transfer process promotes the generation of ROS, such as singlet oxygen and free radicals. However, it has been found that the light absorption of graphene is quite low, with only 2.3% of the incident light being absorbed by a single layer of graphene. This limitation affects the efficiency of graphene-based materials and calls to find ways to enhance their optical absorption, especially for the optoelectronic field.300–302,305,306 GO and rGO have been used in the synthesis of p-n heterojunction photocatalysts, which can enhance the separation of electron–hole pairs and improve photocatalytic activity. rGO has also been used as a p-type coupling semiconductor to n-type copper phosphate for high visible light photocatalytic activity.307 On the other hand, fullerenes have a capacity for visible light absorption, appealing triplet yield, and ability to ROS. With superior bioactivity and outstanding electronic properties, C60 garners the most attention as an efficient PS, boasting a near 100% quantum yield in generating singlet oxygen.303,308 The other aspect that needs to be noted is that the optical properties of graphene and its derivates rely on distance-dependent interactions and the design and fabrication of layered structures.302

Additionally, the distinctiveness of graphene as an energy acceptor strongly backs the excitation wavelength. This observation opens up new insights into the dynamics of excitation and energy transfer in systems where the characteristics of either acceptors or donors can be further controlled independently through light.302

Graphene and its derivates exhibit high electron conductivity and can act as an electron mediator in redox reactions. In the presence of light, it can transfer electrons to molecular oxygen leading to the formation of superoxide radicals.309–313 Graphene materials serve as electron sinks, boosting visible-light photodynamic capabilities, and synergistically integrating photodynamic and photothermal hyperthermia, maximizing therapeutic effect.304 For instance, GQDs function as effective Förster Resonance Energy Transfer (FRET) donors with tunable optical properties.303,314,315 In GQDs, the ROS generation mechanism is also different from the traditional one, when singlet oxygen is produced via energy transfer originating from the excited triplet state. ROS can occur from both the excited singlet (S1) and triplet (T1) states.316,317

The synergistic effect of graphene and its materials with photosensitizers is based on excellent electrical conductivity, high surface area, and efficient charge transfer.318,319 Also, novel hyaluronic acid (HA)–GO conjugate systems were reported for the switchable photoactivity of PSs.304,320 It was found that hyaluronic acid increases the stability and biocompatibility of the GO nanosheets. Cellular uptake of PS was also improved by targeting the cancer cells with overexpressed HA receptors. The loading efficiency of PS was as high as 115% via both π–π stacking and hydrophobic interactions. The photodynamic activity of Ce6, when adsorbed on HA-GO nanocarriers, was largely suppressed in aqueous solution to ensure biocompatibility. However, this suppression was recovered upon the release of Ce6 following cellular uptake. Consequently, the photodynamic therapy (PDT) efficiency of the HA-GO/Ce6 was enhanced compared to free Ce6.304,321 Other examples will be discussed further for each graphene material.

Graphene Oxide and Reduced Graphene Oxide

GO and rGO could be modified in a lot of different ways, such as through PEGylation and combination with other polymers, combination with different PS and addition of anticancer chemotherapeutic agents. Majority of the studies, however, combine several approaches. PEGylated GO co-loaded with PS to improve their hydrophilicity could be used for both PDT and PTT and it can be activated by 980 nm laser for achieving two-photon PDT, as noted by Liu et al.322 By combining these two types of therapy, it was possible to achieve improved anticancer efficacy against breast cancer while preventing damage to normal tissue.322 Similarly, Zaharie-Butucel et al successfully combined rGO, IR820 dye and DOX to prepare platforms for NIR-triggered therapy for both PTT and PDT activity under 785 nm laser irradiation.323

As mentioned before, not only PEG could be used as a sufficient modification of NP to combine it with different PS. The other widely used polymer is Pluronic (PF127) which was used by Ma et al to prepare a composite with GO and methylene blue by a thin-film hydration method.324 This platform can be used for both PDT and PTT which is beneficial as PTT is still effective when oxygen-dependent PDT is limited during hypoxia.324

As to methylene blue, several researchers show its efficiency against different cancers when combined with GO.325–327 It has pH-responsive properties and PS release could be even more efficient at acidic pH.327 In addition, a combination of PDT and PTT could provide a synergistic effect, preventing tumor regrowth and metastasis with low systemic toxicity.325,326

For both PTT and PDT, GO can also be combined with indocyanine green (ICG). In complex with sgc8 aptamer, cancer cells were efficiently killed even though GO loaded with ICG produced less singlet oxygen compared to free ICG.328,329 Also, not only PS can be loaded on the GO surface; chemotherapeutic agents and other drugs through π–π stacking interaction like wedelolactone can be as well.329

After binding rGO with hyaluronic acid, it is also possible to create an enhanced intracellular uptake and prepare a trimodal platform for cancer treatment via IR780 and DOX conjugation. IC50 of DOX was reduced by 86% in vitro as reported by Dash et al.330

Carbon Quantum Dots (CQDs), Graphene Quantum Dots (GQDs) and Graphene Oxide Quantum Dots (GOQDs)

CQDs, GQDs and GOQDs have a lot of beneficial properties according to several scientists. Remarkably, Pillar-Little et al noticed the connection between the preparation method of CQDs and its PDT-toxicity. Top-down synthesized CQDs show much stronger light-activated toxicity.331,332 Thakur et al claimed, in their research with an 808 nm laser, that GQDs are not photo-bleachable-like organic dyes and do not lose their fluorescence despite continuous irradiation.333 Also, producing such complexes of QDs as conjugation of rare-earth elements doped upconversion, nanoparticles with GQDs could produce ROS efficiently under NIR light dealing with low ROS yields with conventional PSs. As Li et al stated, such functionalized GQDs show biocompatibility and concentration-dependent PDT efficiency.334 It is also possible to use other lasers, like the one with micro-watt power (290 μW) 365 nm UV tube light, as Ahirwar et al reported.315 Ahirwar et al also showed that GQDs and GOQDs both have strong absorbance in the UV region and ROS generation. B16-F10 cells and MCF-7 cells were used in their PDT experiment, and the viability dropped dramatically after a brief 5-minute exposure.315

Fan et al also showed that doping GQDs with heteroatoms can modify their optical properties and nitrogen (N) doped GQDs (N-GQDs) for PDT are broadly researched now.335 They can be synthesized using various methods like hydrothermal method and solvothermal method.335–337 Shi et al were able to fabricate samarium doped carbon dots (Sm/CDs) via a one-step hydrothermal method. Sm/CDs exhibited noteworthy biocompatibility and remarkable absorption properties in the near-infrared spectrum. The incorporation of Sm3+ significantly enhances the production of ROS and photothermal conversion efficiencies when exposed to 808 nm and 1060 nm wavelengths.338

It is also possible to prepare PEGylated material, similar to GO and rGO particles. Zhang et al obtained GQDs-PEG material displaying a low cytotoxicity and efficient endocytosis.339 It was also demonstrated that host-immunity-related CD8+ T cells and proinflammatory cytokines significantly increased after photoactivation of GQDs-PEG material which means that this material has immunostimulatory activity. It makes them not only PDT agents but also promising immunotherapy candidates.339,340 Not only PEG but other polymers can be used. Juzenas et al also noticed that propionyl ethylenimine-co-ethylenimine coated CQDs when exposed to UV irradiation destroy malignant human prostate cancer cells, as tested on Du145 and PC3 cell lines.332,341

There is a possibility to combine GQDs with other NPs as evident from the synthesis of N-doped GQDs/titanium dioxide nanocomposites (N-GQDs/TiO2 NCs).342 Some studies also report the conjugation of GQDs and GOQDs with upconversion nanoparticles (UCNPs), as already mentioned.

Choi et al reported that UCNP-like ytterbium and erbium ions-doped sodium yttrium fluoride NPs (NaYF4:Yb3+, Er3+) were bound to the QDs by coupling chemistry with EDC and afterwards, it was possible to conjugate newly prepared material with PSs by π-π stacking interaction.343 This product was also shown to be a candidate as a photosensitizing agent and in cancer cell imaging.343

As to PSs, a lot of them can be coupled to the GQDs surface; among them are BODIPY, chlorin e6 (Ce6) and hypocrellin A (HA), curcumin, methylene blue and others.335,340,344,345 Mangalath et al described boron dipyrromethene dye derivatives in conjugation with GQDs (GQDs-BDPA) for PDT purposes.344 GQDs-BDPA exhibited a higher ROS generation yield than the free BDPA.344 Li et al reported that GQDs with disulfide-linked PEG and conjugated with Ce6 caused efficient suppression of tumor growth.340 When using methylene blue as PSs in conjugation with sulfur-doped GQDs, improved ROS generation and even antimicrobial activity were reported.346

One of the promising PS used with GQDs is curcumin, which has a low water solubility on its own, therefore further adjustments are needed.347 It is possible to dissolve curcumin in methanol or DMSO and just add curcumin solution to the solution of GQDs.348,349 Methanol or other solvents can be evaporated later via rotary evaporation.348 Another way is to add curcumin through tryptophan. Ghanbari et al reported enhanced curcumin loading capacity on the tryptophan-conjugated GQDs where π–π stacking and hydrophobic interaction played a crucial role in conjugation with an aromatic of curcumin.350 Changes in the absorbance spectra and ζ-potential were observed in the conjugated materials.349,350

Nanohorns

Moreno-Lanceta et al stated that single-walled carbon Nanohorns (SWNHs) are promising PTT agents and PDT PS carriers as they exhibit excellent photothermal features in NIR light.202 As carriers, they can be coupled with other PSs such as indocyanine green (ICG), chlorin e6, fabricated zinc phthalocyanine (ZnPc), tetrasodium salt copper phthalocyanine (TSCuPc) or IR808 covalently grafted to hyaluronic acid.207,351–357 ICG is possible to couple via the above-mentioned hydrophobic π–π stacking; Gao et al tested SWNHs coupled with ICG as a PDT agent on 4T1 triple-negative breast cancer cells and reported that ROS formation increased within activation by 808 nm wavelength and temperature increase.354 Yang et al reported the preparation of Gd3+ and chlorin e6 loaded SWNHs, which stimulated dendritic cells to secrete IL-6 and TNF-α by PTT, while PDT upregulated IFN-γ and CD80.355 As a result of this immune response, migration of Gd-Ce6@SWNHs to the tumor-draining lymph, it eliminates the distant metastases and prevents cancer reoccurrences.355 To improve its solubility in water, it was also coated first with poly(lactide-co-glycolide)-b-poly(ethylene glycol)-maleimide and maleic anhydride-alt-1-octadecene before drug loading.355

Nanotubes

CNTs have also been evaluated as candidate materials in PTT and PDT on their own or in combination with PSs.358 Single-wall CNTs in combination with polyethyleneimine and polyvinylpyrrolidone can form stable complexes, like SWNT-PEI and SWNT-PVPk30, that can act as a promising PDT agent, as Wang et al stated.359 Again, number of PSs are possible for conjugation. Among others, we can mainly note Chlorin e6, m-tetrahydroxyphenylchlorin (mTHPC), curcumin, palladium(ii)porphyrin (PdP), platinum(ii)porphyrin (PtP) and tris(triphenylamino)porphyrin palladium(ii) (Pd(TPA)3P).360–364 Chlorin e6 (Ce6) could be coupled with SWCNTs by noncovalent π–π interactions, and the resulting complex induces the significant ROS production. Optionally, this complex can also be wrapped by chitosan to improve biocompatibility and solubility in water.360,364 Li et al in their work used curcumin as the PSs of choice for PDT and it showed enhanced antitumor efficacy, cell uptake, and blood concentration compared to pure curcumin. In that case, curcumin was complexed with SWCNT in methanol.360 Arellano et al successfully linked tris(triphenylamino)porphyrin palladium(ii), Pd(TPA)3P covalently to single-walled carbon nanotubes via Sonogashira cross-coupling with the use of microwave irradiation.363

Fullerenes

Fullerenes have some unique features that make them candidates for PDT even though they have some unfavorable optical absorption properties, such as high absorption at the UVA and blue spectral region. However, poor light absorption in the range of 600–700 nm is a drawback of C60 above all, which means that we should use C70. Nevertheless, functionalization can overcome those difficulties; for instance, a light-harvesting antenna molecule could be introduced to the fullerene molecule. It is known that both pristine and functionalized fullerenes can catalyze the formation of ROS.365–369 Two widely known reactions will lead to covalent functionalization; these are the Bingel and the Prato reactions.365,370 Gündüz et al also reported the preparation of glucose-BODIPY-fullerene dyads and the same related nano micelles in the presence of Tween 80.371 These systems were tested in the K562 cell line, and sufficient ROS generation was reported, as well as absorption of the red spectrum.371 Tokuyama et al published the first report on using fullerenes in cancer cells, namely in 1993. Fullerenes functionalized with carboxylic acid and white light were used.372 Burlaka et al similarly used a mercury lamp to produce phototoxicity in cancer cells.373 It is noted that cell death was mainly caused by membrane damage.373,374

To overcome the water insolubility of fullerenes, for instance, C60 can be coupled with PEG to form conjugate C60-PEG. It is also noted that such a complex performed better than Photofrin as a PDT agent.367,375,376 It is also possible to introduce Tween 80 and PVP to enhance the solubility of the fullerenes.377 Liu et al mentioned another complex; C60-PEG was mixed with diethylenetriaminepentaacetic acid and further introduced to gadolinium which then demonstrated the significant antitumor effect after light irradiation of the spectral range from 400 to 500 nm.378

Another way to prepare an efficient complex for PDT is to complex fullerenes with porphyrins. This kind of complex can be more efficient in terms of cell penetration and ROS generation. Milanesio et al compared the PDT effect of porphyrin-C60 and its metal complex with Zn(II) to the pure porphyrin (5-(4-acetamidophenyl)-10,15,20-tris(4-methoxyphenyl)-porphyrin) in Hep-2 cell line and concluded that the porphyrin-C60 complex had the greatest impact on cells.379

Miki et al reported the preparation of amphiphilic γ-cyclodextrin-fullerene complexes and Sugikawa et al described ROS generation.375,380 With such molecules, it is also possible to prepare hybrid complexes of C60 and hydrophobic porphyrin by mixing in water in the presence of PEG. The resulting C60-porphyrin NPs are negatively charged and can be dispersed in water.375,381–383 As reported, the singlet oxygen generation ability of C60-porphyrin NPs under light radiation of 620 nm wavelength is improved compared to C60 NPs.375

Li et al also described the enhancement of Type I photoreactions using antennae-fullerene complexes and NIR irradiation.384 Type I photoreactions are considered important as they have higher oxidizing ability than Type II, and they also lead to lesser oxygen dependency. Rare-earth UCNPs can act as antennae for various PDT agents, as well as fullerenes.384

Nanoribbons

Carbon nanoribbons and their application in the biomedical field and PDT fields are still to be investigated. A small number of research papers are available, but all of them show promising properties of carbon nanoribbon materials. There is evidence of using it in the field of PTT and other cancer research areas. Nanoribbons could be modified for use in cancer research with some already mentioned chemicals, such as polyethylene glycol and DOX for synergistic effects.385,386 They were shown as an effective PTT agent under 808 nm NIR laser irradiation. This material was also tested by Lu et al in vitro on the U87 cell line and mice with no cytotoxicity.386

Recent Approaches, Future of PDT, and Conclusion

Nowadays, cancer resistance is one of the main challenges for cancer patients. Addressing this problem requires a multifaceted and complex approach, and graphene-based materials have the potential to provide a promising solution. There are several methods through which these materials can be utilized to overcome cancer resistance, like targeted drug delivery bypassing resistance mechanisms, immunotherapy; graphene-based materials can be used to modulate the immune response, gene therapy with graphene materials as gene delivery vector, hyperthermia therapy as graphene materials have outstanding photothermal and photodynamic properties.

The PDT treatment method has a distinctive approach that functions through various modalities. These modalities include targeting specific cells, causing damage to resistant cells, inducing apoptosis, modulating the immune system, preventing metastasis and reducing resistance development (Figure 14).

Figure 14 The modalities through which PDT can affect cancer resistance.

One of the most exciting future aspects of PDT is its potential for multimodal therapy. This tactic combines PDT with other therapies such as PTT, immunotherapy, chemotherapy, and targeted therapy. Multimodal therapy can strengthen different treatment modalities to achieve better therapeutic outcomes.

For some researchers, it seems to be interesting and promising approach to combine PDT with traditional specific and non-specific immunotherapies.387,388 This would include administration of antibodies and cytokines or targeting of immune checkpoints.387,388 One of the most promising methods is a combination of PDT with programmed cell death protein 1 (PD-1) and its ligand, programmed death ligand 1 (PD-L1) immune checkpoint inhibitors. PD-1 is expressed by activated T cells, B cells, dendritic cells, and natural killer cells. On the other hand, PD-L1 is expressed in several types of tumor cells.387–389 Nagaya et al demonstrated that NIR photoimmunotherapy induced tumor cell death aiming for cell-surface CD44 and PD-1 blockade in multiple tumor models.390 This could be a target for the use of graphene materials and PDT. Wang et al reported a fabrication method of a PEGylated rGO hybridized with Fe3O4 nanoparticles for photothermal immunotherapy of cancer.391 These modifications were possible through electrostatic interactions and showed PTT effectiveness under irradiation by an 805 nm laser.391 Application of this nanocomplex resulted in PTT-induced ICD, release of danger-associated molecular patterns, and activation of dendritic cells (DCs) in lymph nodes under irradiation. Median survival time was also increased in 4T1 tumor-bearing animals after NIR irradiation. Wang et al also proposed that this novel nanosystem could be used for magnetic resonance imaging, which could potentially pave the way for MRI-guided photothermal immunotherapy for metastatic cancers.391 Wu et al synthesized hybrid nanosystems containing polydopamine stabilized GQD-photosensitizer composites (GCpD), immunostimulatory polycationic polymer nanoparticles, and Gd3+/Cy3 imaging probes for both magnetic resonance and fluorescence imaging-guided photoimmunotherapy.392 This material was denoted as PC@GCpD(Gd), was reported to act via photothermal and photodynamic effects mediated by GCpD, and was targetedly delivered to endosomal Toll-like receptor 9 (TLR9) to enhance the secretion of proinflammatory cytokines and the maturation of dendritic cells and to activate T lymphocytes. PC@GCpD(Gd) was also shown to successfully suppress the EMT6 murine mammary cancer model under laser irradiation.392 Zhao et al also stated that clinically approved carbon nanoparticle suspension injection (CNSI) inhibits the growth of primary tumors, distal tumors, and metastasis via PTT and anti-PD-1.393

Other modalities like chemotherapy and targeted therapy can also complement PDT by targeting cancer cells that may be resistant to PDT alone. Zhou et al fabricated a nanosystem that was loaded with mitoxantrone and SB-431542, which is a specific inhibitor of transforming growth factor-beta, onto reduced graphene oxide (rGO).394 The administration of this nanocomplex was followed by irradiation of a near-infrared laser and as a result, it destroyed local primary tumors and inhibited distant metastases in the 4T1 mouse tumor model, which is highly tumorigenic and invasive.394 It was also clear that mitoxantrone, SB-431542, and rGO together induced an effective tumor vaccination by the increased infiltration of tumor-specific cytotoxic CD8+ T lymphocytes and decreased infiltration of regulatory T cells in distal tumors.394 Sawy et al reported different types of carbon nanomaterials that were further employed as nanocarriers for DOX. GO with the highest surface negative charge exhibited the highest loading capacity and GQDs with the lowest surface negative charge showed the highest release effectiveness.395

The next step to further advance the use of PDT and graphene in PDT is to target cancer stem cells (CSCs). They are difficult to eradicate with conventional chemotherapy or radiation. The survival of residual CSCs can result in tumor recurrence, metastasis, and drug resistance. Fiorillo et al showed that GO hinders tumor-sphere formation in multiple cell lines such as breast cancer, ovarian cancer, and lung cancer as well as many others. GO inhibits key pathways such as Wnt signaling pathways, notch signaling and STAT signaling (Signal Transducer and Activator of Transcription) and thus inducing CSC differentiation.396 Their research was focused specifically on the influence of GO on CSCs, which can contribute to tumor recurrence, distant metastasis and drug resistance. GO was able to inhibit tumor growth of cancers such as breast, prostate, lung, ovarian, and pancreatic cancer, as well as glioblastoma (brain cancer).396 The results obtained from the Fiorillo et al study also indicate that GO specifically targets a global phenotypic property of CSCs.396 Prior research has also demonstrated the potential of GO in targeted cancer therapy, tumor growth prevention, and tumor cell migration inhibition.91,397–399 It was also shown by Burke et al that carbon nanotube-mediated thermal treatment sensitizes breast CSCs and limits their long-term proliferative capacity.400 Another research also suggested the possibility to target cancer cells with GQDs, especially GQDs that are combined with Cu2+, Zn2+, or Ni2+ ions as they are possible to target ABC transporters of multiple multidrug-resistant genes.401,402 There is also the ability to target different organelles, such as mitochondria, selectively.403

Selective targeting of malignant cells is closely related to targeting, damaging resistant cells, and modulating immune response. PDT can also work through immunogenic cell death (ICD) by inducing damage-associated molecular patterns (DAMPs), although only a few PSs can trigger that. Yu et al noted that induced immune response after PDT application can be caused by damage to cancer cells. PDT activates dendritic cells, and as a result, CD8+ infiltration will increase in the tumor site. In addition, Yu’s et al research reported that lung metastatic growth could significantly decrease after PDT treatment. It is also possible to enhance anti-metastasis response with the oxygen-boosted PDT via ICD and DAMPs release. Wang et al suggested to use nanoplatforms synthesized with polyethylene glycol altered Cu2xSe nanoparticles, β-cyclodextrin, and chlorin e6 as Fenton-like-Haber–Weiss catalyst for this purpose.404,405

It is also possible for PDT to trigger more than one cell death type, and this could be a prominent strategy to overcome cancer resistance (Figure 15).46,406–409 There are cell death types such as necroptosis, ferroptosis, pyroptosis, parthanatos, and mitotic catastrophe. Mishchenko et al suggested that there is an “ideal protocol” for PDT which should include different irradiation regimens and a combination of PSs for successful overcoming of cancer resistance.410,411

Figure 15 PDT effects on the cell, cell death types and immune boost scheme.410,412

Mitotic catastrophe (MC) is one of the onco-suppressive mechanisms that hinder the proliferation of cells with excessive DNA damage that cannot complete mitosis and generally defective mitosis. Thus, in case of triggering this cell death form, cytoskeletal components will be the most sensitive organelles and a subject for PDT treatment.410,411,413 Although some cancer cells are also reported to be able to bypass MC, some cancers will be more resistant and aggressive via aneuploidy and genomic instability as a result.409

Paraptosis is a different non-conventional cell death type that does not show chromatin condensation and cell fragmentation contrary to apoptosis.410 This cell death type should mainly occur in the cells that exhibit diminished apoptosis and are unaffected by autophagy. In case of triggering paraptosis, the endoplasmic reticulum is a key aim as paraptosis is associated with misfolded proteins. Hypericin and Verteporfin are two PSs primarily located in the ER which, after PDT, have demonstrated the induction of paraptosis.410,414–416 Decreasing the level of thiol-containing antioxidants can also facilitate the induction of paraptosis and its application in cancer therapy.417

Pyroptosis could be considered a double-edged sword as well, as on the one hand, it shows several ICD hallmarks, such as the production of DAMPs, but, on the other hand, it can provide an appropriate microenvironment for tumor development and metastasis.410,418–422 During the pyroptosis formation of pyroptotic bubbles, activation of caspase-1 and production of the cytokines IL-1β and IL-18 are typically noticed. Zhu et al reported that curcumin-loaded poly(L-lactide-co-glycolide) could be effective against liver cancer under PDT conditions, as it enhances apoptosis and pyroptosis rates.423,424 Pyruvate kinase M2 and ROS induced by chemotherapeutic drugs are also considered to be important players in PDT-induced pyroptotic cell death.425,426

There is limited evidence to support the occurrence of parthanatos during PDT, as Soriano’s research provided the only available evidence thus far.427 Parthanatos relies on the hyperactivation of the DNA damage response. It is, namely, dependent on the activation of Poly(ADP-ribose) polymerase (PARP1), which can happen after stimuli like ultraviolet irradiation and ROS generation.410,411,428,429

Necroptosis is one of the cell death modalities that resembles necrosis and includes swelling, rapid plasma membrane permeabilization and moderate chromatin condensation, although nuclear fragmentation and caspase activation are not present.410,430,431

Ferroptosis is one of the most important non-conventional cell death modalities associated with mitochondria shrinkage, mitochondrial cristae reduction or disappearance and outer mitochondrial membrane rupture. It relies on iron-dependent oxidative modification of phospholipid membranes.410,432 It is a promising perspective for apoptosis and/or necroptosis-resistant tumors.432,433 Shishido et al reported that 5-aminolevulinic acid can trigger ferroptosis through the regulation of glutathione peroxidase 4 (GPX4) and heme oxygenase 1 (HMOX1) and showed antitumor effects in esophageal squamous cell carcinoma cell lines. There are also inhibitors of ferroptosis reported: ferrostatin-1, liproxstatin-1, the iron chelator, deferoxamine, and some others.410,434 Quantum Dots are reported to be effective in ferroptosis triggering. It could be triggered by disrupting calcium homeostasis in microglia or via mitochondrial oxidative stress in microglia, as reported by Wu et al, N-GQDs are described to activate two calcium channels in the hippocampus of mice that lead to ferroptosis. Also, N-GQDs application could cause ferroptosis in the BV2 cell line via accumulation in mitochondria resulting in mitochondrial iron overload and redox imbalance.435,436

Apoptosis itself can occur through different modalities after PDT application. Mitochondrial membrane damage permeability leads to cytochrome c release into the cytoplasm with caspase activation afterward.437–440

Increasing the cellular uptake of PSs is crucial to expand the scope and efficacy of PDT. Graphene-based nanocarriers have emerged as a promising solution to enhance the absorption of PSs, as it was already mentioned before. Although further research is needed, the use of graphene materials for delivering non-water soluble photoactive agents shows great potential. This approach can overcome the challenge of limited solubility of PSs in aqueous environments, making it possible to deliver them efficiently to targeted sites. Aguilar Cosme et al reported CQDs to improve protoporphyrin IX cellular uptake and solubility.441 Klimenko et al also showed that graphene prevents the agglomeration of the aluminum phthalocyanine chloride (AlClPc).442

The application of graphene-based PDT holds promise in advancing personalized medicine methods. By using the unique properties of graphene nanomaterials, patient-specific treatment approaches can be developed to overcome specific resistance mechanisms. Functionalized graphene nanomaterials can be tailored to target genetic mutations and specific molecules associated with resistance, resulting in precise and effective delivery of treatment. This novel approach has the potential to revolutionize the field of medicine and pave the way for improved patient outcomes through individualized therapy.443 One of the techniques for achieving personalized therapy is the utilization of biosensors that are based on graphene. These biosensors can be highly sensitive and specific in detecting various biomarkers. Graphene-based biosensors have potential in early disease detection, monitoring disease progression, and assessing treatment response. Biosensors that target patient-specific biomarkers can be designed and therefore will help healthcare workers in obtaining real-time information about an individual’s health status. Graphene-based nanosystems can also be utilized for genomic analysis and genetic testing. Graphene biosensors and sequencing platforms can detect DNA mutations, gene expression patterns, and epigenetic modifications. Thus, disease risk, prognosis, and treatment response can be assessed. By integrating genomic information with clinical data, medical interventions can be customized to minimize adverse effects.444–446

Zhang et al proposed a disposable nano-biosensor for glucose monitoring using saliva. The working electrode in this biosensor was functionalized with SWNT and layers of chitosan, gold nanoparticles and glucose oxidase. It also demonstrates excellent clinical accuracy.447 Li et al also reported highly sensitive biosensor for the detection of microRNA and adenosine based on graphene oxide-gold nanoparticles (AuNPs) composites.448 GO-AuNPs composite worked as both a sensing substrate and signal amplification element. The developer surface plasmon resonance biosensor successfully detected miRNA-141 in cancer cell extractions.448 Khalil et al reported a surface-enhanced Raman scattering platform with a short DNA probe for DNA biosensing. It was based on the abovementioned GO-AuNPs.449 As to CQDs and GQDs, one of the most important biosensing mechanisms are fluorescent interactions.450 Kong et al showed that N-doped CQDs can act as α-glucosidase inhibitors based on the inner filter effect of N-doped CQDs, when CQDs absorb both the excitation light and the emitted fluorescence light, leading to a reduction in the measured fluorescence intensity.451

Graphene-based materials can also act as contrast agents to enhance the sensitivity and resolution of magnetic resonance imaging (MRI) or computed tomography (CT).443,452,453 Zhang et al fabricated PEGylated ultrasmall GO with a chelating agent DOTA and gadolinium(III) to form GO-DOTA-Gd complexes, which showed improved T1 relaxivity.453 Antoine et al reported that that GQDs are able to decrease the intensity of X-rays and form contrast images in both X-ray and computed tomography.454

Graphene-based materials also have the potential to enable real-time monitoring of treatment response and resistance development. By incorporating imaging agents or biosensors into graphene nanomaterials, it becomes possible to non-invasively observe tumor dynamics. This allows for timely adjustments to treatment strategies based on feedback, which can help improve the effectiveness of PDT or other treatment modalities. Zhang et al reviewed graphene and its derivatives as prominent materials for wearable sensors aimed at monitoring biophysical signals for healthcare applications.455 Maity et al fabricated sensors that were able to execute a real-time detection of heavy-metal ions and E. coli bacteria in flowing tap water.456 This calls for extended research and application in humans.

In conclusion, applying graphene materials in PDT holds massive potential in transforming and revolutionizing cancer treatment strategies. With their unique properties, such as high surface area, excellent conductivity and biocompatibility, graphene-based PSs have the potential to enhance PDT efficiency. Additionally, graphene-based nanomaterials present a promising potential to overcome multidrug resistance, which is a significant challenge in cancer therapy.

The synergy between graphene materials and PDT not only upgrades and optimizes the targeting and destruction of cancer cells but also improves the immune response against tumors. By triggering immunogenic cell death and changing and modulating the TME, graphene-based PDT strategies again contribute to overcoming cancer resistance mechanisms, pioneering the way for more efficient and personalized cancer treatments.

As the study of cancer biology develops and advances in huge steps, the utilization of graphene materials in PDT serves to broaden our understanding of this complex field and gives us a promising solution for patients in their fight against cancer.

Funding

Supported by Ministry of Health of the Czech Republic, grant nr. NU21-09-00357. All rights reserved.

Disclosure

The authors report no conflicts of interest in this work.

References

1. European Commission Eurostat. Causes of Death Statistics; 2024. doi:10.2908/HLTH_CD_ASDR2

2. World Health Organization. Global health estimates: leading causes of death; 2020. Available from: https://www.who.int/data/gho/data/themes/mortality-and-global-health-estimates/ghe-leading-causes-of-death. Accessed April 11, 2024.

3. World Health Organization. Cancer; 2022. Available from: https://www.who.int/news-room/fact-sheets/detail/cancer. Accessed April 11, 2024.

4. Australian Institute of Health and Welfare. Deaths in Australia; 2023. Available from: https://www.aihw.gov.au/reports/life-expectancy-deaths/deaths-in-australia/contents/leading-causes-of-death. Accessed April 11, 2024.

5. Raza A, Hayat U, Rasheed T, Bilal M, Iqbal HMN. “Smart” materials-based near-infrared light-responsive drug delivery systems for cancer treatment: a review. J Mater Res Technol. 2019;8(1):1497–1509. doi:10.1016/J.JMRT.2018.03.007

6. Chakraborty S, Rahman T. The difficulties in cancer treatment. Ecancermedicalscience. 2016:6. doi:10.3332/ecancer.2012.ed16

7. Liao JB. Viruses and Human Cancer. Eur J Cancer. 2006;79:1171.

8. Parsonnet J. Bacterial Infection as a Cause of Cancer. Environ Health Perspectives. 1995;103(suppl 8):263.

9. National Cancer Institute. Side Effects of Cancer Treatment; 2018. Available from: https://www.cancer.gov/about-cancer/treatment/side-effects. Accessed April 11, 2024.

10. Kwok KK, Vincent EC, Gibson JN. Antineoplastic Drugs. Pharmacology and Therapeutics for Dentistry. Seventh Edition. 2017;530–562. doi:10.1016/B978-0-323-39307-2.00036-9

11. Ngov L, Levin D. Chemotherapy Basics for Hospitalists. Hosp Med Clin. 2016;5(1):85–100. doi:10.1016/j.ehmc.2015.08.008

12. Yang CY, Shiranthika C, Wang CY, Chen KW, Sumathipala S. Reinforcement learning strategies in cancer chemotherapy treatments: a review. Comput Methods Programs Biomed. 2023;229. doi:10.1016/j.cmpb.2022.107280

13. Chen HHW, Kuo MT. Improving Radiotherapy in Cancer Treatment. Promises and Challenges. 2017. www.impactjournals.com/oncotarget.

14. Baumann M, Krause M, Overgaard J, et al. Radiation oncology in the era of precision medicine. Nat Rev Cancer. 2016;16(4):234–249. doi:10.1038/nrc.2016.18

15. Azzam EI, Jay-Gerin JP, Pain D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012;327(1–2):48–60. doi:10.1016/j.canlet.2011.12.012

16. Richardson RB, Harper ME Mitochondrial Stress Controls the Radiosensitivity of the Oxygen Effect: implications for Radiotherapy. Vol 7. Available from: www.impactjournals.com/oncotarget/. Accessed June 1, 2024.

17. Tateishi Y, Sasabe E, Ueta E, Yamamoto T. Ionizing irradiation induces apoptotic damage of salivary gland acinar cells via NADPH oxidase 1-dependent superoxide generation. Biochem Biophys Res Commun. 2008;366(2):301–307. doi:10.1016/j.bbrc.2007.11.039

18. Kufe M DW, Pollock MP RE, Weichselbaum M RR, et al. Holland‐Frei Cancer Medicine. Wiley; 2016. doi:10.1002/9781119000822

19. National Cancer Institute. Surgery to Treat Cancer; 2015. Available from: https://www.cancer.gov/about-cancer/treatment/types/surgery. Accessed April 11, 2024.

20. Sabnis AJ, Bivona TG. Principles of Resistance to Targeted Cancer Therapy: lessons from Basic and Translational Cancer Biology. Trends Mol Med. 2019;25(3):185–197. doi:10.1016/j.molmed.2018.12.009

21. Zargar A, Chang S, Kothari A, et al. Overcoming the challenges of cancer drug resistance through bacterial-mediated therapy. Chronic Dis Transl Med. 2019;5(4):258–266. doi:10.1016/j.cdtm.2019.11.001

22. Gu Z, Zhu S, Yan L, Zhao F, Zhao Y. Graphene-Based Smart Platforms for Combined Cancer Therapy. Adv. Mater. 2019;31(9):662. doi:10.1002/adma.201800662

23. Saha S, Pradhan N, Mahadevappa R, Minocha S, Kumar S. Cancer plasticity: investigating the causes for this agility. Semin Cancer Biol. 2023;88:138–156. doi:10.1016/j.semcancer.2022.12.005

24. Bell CC, Gilan O. Principles and mechanisms of non-genetic resistance in cancer. Br J Cancer. 2020;122(4):465–472. doi:10.1038/s41416-019-0648-6

25. Cavalli G, Heard E. Advances in epigenetics link genetics to the environment and disease. Nature. 2019;571(7766):489–499. doi:10.1038/s41586-019-1411-0

26. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–726. doi:10.1038/nrc3599

27. Huang S. Tumor progression: chance and necessity in Darwinian and Lamarckian somatic (mutationless) evolution. Prog Biophys Mol Biol. 2012;110(1):69–86. doi:10.1016/j.pbiomolbio.2012.05.001

28. Bell CC, Fennell KA, Chan YC, et al. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia. Nat Commun. 2019;10(1):652. doi:10.1038/s41467-019-10652-9

29. Brock A, Chang H, Huang S. Non-genetic heterogeneity — a mutation-independent driving force for the somatic evolution of tumours. Nat Rev Genet. 2009;10(5):336–342. doi:10.1038/nrg2556

30. Hata AN, Niederst MJ, Archibald HL, et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat Med. 2016;22(3):262–269. doi:10.1038/nm.4040

31. Bai X, Fisher DE, Flaherty KT. Cell-state dynamics and therapeutic resistance in melanoma from the perspective of MITF and IFNγ pathways. Nat Rev Clin Oncol. 2019;16(9):549–562. doi:10.1038/s41571-019-0204-6

32. Catania F, Ujvari B, Roche B, Capp JP, Thomas F. Bridging Tumorigenesis and Therapy Resistance With a Non-Darwinian and Non-Lamarckian Mechanism of Adaptive Evolution. Front Oncol. 2021;11. doi:10.3389/fonc.2021.732081

33. Foo J, Michor F. Evolution of acquired resistance to anti-cancer therapy. J Theor Biol. 2014;355:10–20. doi:10.1016/j.jtbi.2014.02.025

34. Hirata E, Sahai E. Tumor microenvironment and differential responses to therapy. Cold Spring Harb Perspect Med. 2017;7(7):1–14. doi:10.1101/cshperspect.a026781

35. Gillies RJ, Verduzco D, Gatenby RA. Evolutionary dynamics of carcinogenesis and why targeted therapy does not work. Nat Rev Cancer. 2012;12(7):487–493. doi:10.1038/nrc3298

36. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 2018;15(2):81–94. doi:10.1038/nrclinonc.2017.166

37. Guo X, Gao C, Yang DH, Li S. Exosomal circular RNAs: a chief culprit in cancer chemotherapy resistance. Drug Resist Updates. 2023;67. doi:10.1016/j.drup.2023.100937

38. Khan FH, Reza MJ, Shao YF, et al. Role of exosomes in lung cancer: a comprehensive insight from immunomodulation to theragnostic applications. Biochim Biophys Acta Rev Cancer. 2022;1877(5):776. doi:10.1016/j.bbcan.2022.188776

39. Raguz S, Yagüe E. Resistance to chemotherapy: new treatments and novel insights into an old problem. Br J Cancer. 2008;99(3):387–391. doi:10.1038/sj.bjc.6604510

40. Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15(7):409–425. doi:10.1038/nrc3958

41. Jarosz-Biej M, Smolarczyk R, Cichoń T, Kułach N. Tumor microenvironment as a “game changer” in cancer radiotherapy. Int J Mol Sci. 2019;20(13):3212. doi:10.3390/ijms20133212

42. Krishnaswami V, Natarajan B, Sethuraman V, Natesan S, RajSelvaraj B. Nano based photodynamic therapy to target tumor microenvironment. Nano Trends. 2023;1:100003. doi:10.1016/j.nwnano.2023.100003

43. Allison RR, Sibata CH. Oncologic photodynamic therapy photosensitizers: a clinical review. Photodiagnosis Photodyn Ther. 2010;7(2):61–75. doi:10.1016/j.pdpdt.2010.02.001

44. Piette J. Signalling pathway activation by photodynamic therapy: NF-κB at the crossroad between oncology and immunology. Photochem Photobiol Sci. 2015;14(8):1510–1517. doi:10.1039/c4pp00465e

45. Berg K. Resistance mechanisms in photodynamic therapy. Photochem Photobiol Sci. 2015;14(8):1376–1377. doi:10.1039/c5pp90026c

46. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi:10.1016/j.cell.2011.02.013

47. Casas A, Di Venosa G, Hasan T, Batlle A. Mechanisms of Resistance to Photodynamic Therapy. Curr Med Chem. 2011;18(16):2486.

48. Fokas E, McKenna WG, Muschel RJ. The impact of tumor microenvironment on cancer treatment and its modulation by direct and indirect antivascular strategies. Cancer Metastasis Rev. 2012;31(3–4):823–842. doi:10.1007/s10555-012-9394-4

49. Casas A, Perotti C, Di Venosa G, Batlle A. Mechanisms of Resistance to Photodynamic Therapy: an Update. J Med. 2015:29–63. doi:10.1007/978-3-319-12730-9_2

50. Giuliani C, Bucci I, Napolitano G. The role of the transcription factor Nuclear Factor-kappa B in thyroid autoimmunity and cancer. Front Endocrinol (Lausanne). 2018;9(AUG):471. doi:10.3389/fendo.2018.00471

51. Duan X, Chen B, Cui Y, et al. Ready player one? Autophagy shapes resistance to photodynamic therapy in cancers. Apoptosis. 2018;23(11–12):587–606. doi:10.1007/s10495-018-1489-0

52. Thorburn A, Debnath J. Targeting chaperone-mediated autophagy in cancer. Sci Transl Med. 2011;3(109):3390. doi:10.1126/scitranslmed.3003390

53. Garg AD, Maes H, Romano E, Agostinis P. Autophagy, a major adaptation pathway shaping cancer cell death and anticancer immunity responses following photodynamic therapy. Photochem Photobiol Sci. 2015;14(8):1410–1424. doi:10.1039/c4pp00466c

54. Garg AD, Agostinis P. Autophagy Induced by Photodynamic Therapy (PDT): shaping Resistance Against Cell Death and Anti-Tumor Immunity. Resistance Photodynamic Therapy Cancer. 2015:99–116. doi:10.1007/978-3-319-12730-9_4

55. Firczuk M, Gabrysiak M, Golab J. GRP78-targeting Sensitizes Cancer Cells to Cytotoxic Effects of Photodynamic Therapy. Resistance Photodynamic Therapy Cancer. 2015:149–161. doi:10.1007/978-3-319-12730-9_6

56. Toussaint M, Barberi-Heyob M, Pinel S, Frochot C. How Nanoparticles Can Solve Resistance and Limitation in PDT Efficiency. Resistance Photodynamic Therapy Cancer. 2015:197–211. doi:10.1007/978-3-319-12730-9_9

57. Rivarola VA, Cogno IS. Optimization of Photodynamic Therapy Response by Survivin Gene. Resistance Photodynamic Therapy Cancer. 2015:163–182. doi:10.1007/978-3-319-12730-9_7

58. Dong D, Ko B, Baumeister P, et al. Vascular Targeting and Antiangiogenesis Agents Induce Drug Resistance Effector GRP78 within the Tumor Microenvironment. Available from: http://cancerres.aacrjournals.org/. Accessed June 1, 2024.

59. Lee E, Nichols P, Spicer D, Groshen S, Yu MC, Lee AS. GRP78 as a novel predictor of responsiveness to chemotherapy in breast cancer. Cancer Res. 2006;66(16):7849–7853. doi:10.1158/0008-5472.CAN-06-1660

60. Pyrko P, Schöntha AH, Hofman FM, Chen TC, Lee AS. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007;67(20):9809–9816. doi:10.1158/0008-5472.CAN-07-0625

61. Paton AW, Srimanote P, Talbot UM, Wang H, Paton JC. A new family of potent AB5 cytotoxins produced by Shiga toxigenic Escherichia coli. J Exp Med. 2004;200(1):35–46. doi:10.1084/jem.20040392

62. Byres E, Paton AW, Paton JC, et al. Incorporation of a non-human glycan mediates human susceptibility to a bacterial toxin. Nature. 2008;456(7222):648–652. doi:10.1038/nature07428

63. Li F, Ackermann EJ, Bennett CF, et al. Pleiotropic Cell-Division Defects and Apoptosis Induced by Interference with Survivin Function. Resistance Photodynamic Therapy Cancer. 1999;1.

64. Konan N, Gurny R, Allemann E State of the Art in the Delivery of Photosensitizers for Photodynamic Therapy. Vol 66; 2002. Available from: www.elsevier.com/locate/jphotobiol. Accessed June 1, 2024.

65. Brown SB, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 2004;5(8):497–508. doi:10.1016/S1470-2045(04)01529-3

66. Kachatkou D, Sasnouski S, Zorin V, et al. Unusual photoinduced response of mTHPC liposomal formulation (foslip). Photochem Photobiol. 2009;85(3):719–724. doi:10.1111/j.1751-1097.2008.00466.x

67. Yang Z, Ma Y, Zhao H, Yuan Y, Kim BYS. Nanotechnology platforms for cancer immunotherapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2020;12(2):e1590. doi:10.1002/wnan.1590

68. Aghebati-Maleki A, Dolati S, Ahmadi M, et al. Nanoparticles and cancer therapy: perspectives for application of nanoparticles in the treatment of cancers. J Cell Physiol. 2020;235(3):1962–1972. doi:10.1002/jcp.29126

69. Karra N, Benita S. The Ligand Nanoparticle Conjugation Approach for Targeted Cancer Therapy. Resistance Photodynamic Therapy Cancer. 2012;13.

70. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K Tumor Vascular Permeability and the EPR Effect in Macromolecular Therapeutics: a Review; 2000. Available from: www.elsevier.com/locate/jconrel. Accessed June 1, 2024.

71. Master A, Malamas A, Solanki R, Clausen DM, Eiseman JL, Sen Gupta A. A cell-targeted photodynamic nanomedicine strategy for head and neck cancers. Mol Pharm. 2013;10(5):1988–1997. doi:10.1021/mp400007k

72. Gary-Bobo M, Hocine O, Brevet D, et al. Cancer therapy improvement with mesoporous silica nanoparticles combining targeting, drug delivery and PDT. Int J Pharm. 2012;423(2):509–515. doi:10.1016/j.ijpharm.2011.11.045

73. Trivedi DN, Rachchh NV. Graphene and its application in thermoplastic polymers as nano-filler- A review. Polymer (Guildf). 2022;240. doi:10.1016/j.polymer.2021.124486

74. Foo CY, Fu RZ. Unravelling the potential of graphene in glioblastoma therapy. Mater Sci Eng C. 2021;128. doi:10.1016/j.msec.2021.112330

75. Ajala OJ, Tijani JO, Bankole MT, Abdulkareem AS. A critical review on graphene oxide nanostructured material: properties, Synthesis, characterization and application in water and wastewater treatment. Environ Nanotechnol Monit Manag. 2022;18. doi:10.1016/j.enmm.2022.100673

76. Wang K, Ruan J, Song H, et al. Biocompatibility of Graphene Oxide. Nanoscale Res Lett. 2011;6(1):1–8. doi:10.1007/s11671-010-9751-6

77. Morales-Narváez E, Sgobbi LF, Machado SAS, Merkoçi A. Graphene-encapsulated materials: synthesis, applications and trends. Prog Mater Sci. 2017;86:1–24. doi:10.1016/j.pmatsci.2017.01.001

78. Kurian M. Recent progress in the chemical reduction of graphene oxide by green reductants–A Mini review. Carbon Trends. 2021;5. doi:10.1016/j.cartre.2021.100120

79. Joshi DJ, Koduru JR, Malek NI, Hussain CM, Kailasa SK. Surface modifications and analytical applications of graphene oxide: a review. TrAC - Trends Anal Chem. 2021;144. doi:10.1016/j.trac.2021.116448

80. Kim T, Jung G, Yoo S, Suh KS, Ruoff RS. Activated graphene-based carbons as supercapacitor electrodes with macro- and mesopores. ACS Nano. 2013;7(8):6899–6905. doi:10.1021/nn402077v

81. Georgakilas V, Tiwari JN, Kemp KC, et al. Noncovalent Functionalization of Graphene and Graphene Oxide for Energy Materials, Biosensing, Catalytic, and Biomedical Applications. Chem Rev. 2016;116(9):5464–5519. doi:10.1021/acs.chemrev.5b00620

82. Dhamodharan D, Ghoderao PP, Dhinakaran V, Mubarak S, Divakaran N, Byun HS. A review on graphene oxide effect in energy storage devices. J Ind Eng Chem. 2022;106:20–36. doi:10.1016/j.jiec.2021.10.033

83. Berrio ME, Oñate A, Salas A, Fernández K, Meléndrez MF. Synthesis and applications of graphene oxide aerogels in bone tissue regeneration: a review. Mater Today Chem. 2021;20. doi:10.1016/j.mtchem.2021.100422

84. Ikram R, Jan BM, Ahmad W. An overview of industrial scalable production of graphene oxide and analytical approaches for synthesis and characterization. J Mater Res Technol. 2020;9(5):11587–11610. doi:10.1016/j.jmrt.2020.08.050

85. Qi X, Jiang F, Zhou M, Zhang W, Jiang X. Graphene oxide as a promising material in dentistry and tissue regeneration: a review. Smart Mater Med. 2021;2:280–291. doi:10.1016/j.smaim.2021.08.001

86. Fahmi T, Branch LD, Nima ZA, et al. Mechanism of graphene-induced cytotoxicity: role of endonucleases. J Appl Toxicol. 2017;37(11):1325–1332. doi:10.1002/jat.3462

87. Chaudhary K, Kumar K, Venkatesu P, Masram DT. Protein immobilization on graphene oxide or reduced graphene oxide surface and their applications: influence over activity, structural and thermal stability of protein. Adv Colloid Interface Sci. 2021;289. doi:10.1016/j.cis.2021.102367

88. Feng W, Wang Z. Biomedical applications of chitosan-graphene oxide nanocomposites. Iscience. 2022:26. doi:10.1016/j.isci

89. Liu S, Zeng TH, Hofmann M, et al. Antibacterial activity of graphite, graphite oxide, graphene oxide, and reduced graphene oxide: membrane and oxidative stress. ACS Nano. 2011;5(9):6971–6980. doi:10.1021/nn202451x

90. Lomeda JR, Doyle CD, Kosynkin DV, Hwang WF, Tour JM. Diazonium Functionalization of Surfactant-Wrapped Chemically Converted Graphene Sheets. J Am Chem Soc. 2008;130(48):16201–16206. doi:10.1021/ja806499w

91. Sun X, Liu Z, Welsher K, et al. Nano-graphene oxide for cellular imaging and drug delivery. Nano Res. 2008;1(3):203–212. doi:10.1007/s12274-008-8021-8

92. Welsher K, Liu Z, Daranciang D, Dai H. Selective probing and imaging of cells with single walled carbon nanotubes as near-infrared fluorescent molecules. Nano Lett. 2008;8(2):586–590. doi:10.1021/nl072949q

93. Zhu Y, James DK, Tour JM. New routes to graphene, graphene oxide and their related applications. Adv. Mater. 2012;24(36):4924–4955. doi:10.1002/adma.201202321

94. Chabot V, Higgins D, Yu A, Xiao X, Chen Z, Zhang J. A review of graphene and graphene oxide sponge: material synthesis and applications to energy and the environment. Energy Environ Sci. 2014;7(5):1564–1596. doi:10.1039/c3ee43385d

95. Depan D, Misra RDK. Hybrid nanoparticle architecture for cellular uptake and bioimaging: direct crystallization of a polymer immobilized with magnetic nanoparticles on carbon nanotubes. Nanoscale. 2012;4(20):6325–6335. doi:10.1039/c2nr31345f

96. Cui X, Cheng W, Xu W, Mu W, Han X. Functional Graphene Derivatives for Chemotherapy-Based Synergistic Tumor Therapy. Nano. 2019;14(11):68. doi:10.1142/S1793292019300068

97. Akhavan O, Bijanzad K, Mirsepah A. Synthesis of graphene from natural and industrial carbonaceous wastes. RSC Adv. 2014;4(39):20441–20448. doi:10.1039/c4ra01550a

98. Korkmaz S, Kariper A. Graphene and graphene oxide based aerogels: synthesis, characteristics and supercapacitor applications. J Energy Storage. 2020;27. doi:10.1016/j.est.2019.101038

99. Servant A, Leon V, Jasim D, et al. Graphene-based electroresponsive scaffolds as polymeric implants for on-demand drug delivery. Adv Healthc Mater. 2014;3(8):1334–1343. doi:10.1002/adhm.201400016

100. Colmiais I, Silva V, Borme J, Alpuim P, Mendes PM. Towards RF graphene devices: a review. FlatChem. 2022;35. doi:10.1016/j.flatc.2022.100409

101. Al NA, Lee JE, Jeong JH, Park SY. Photoresponsive fluorescent reduced graphene oxide by spiropyran conjugated hyaluronic acid for in vivo imaging and target delivery. Biomacromolecules. 2013;14(11):4082–4090. doi:10.1021/bm4012166

102. Dembereldorj U, Kim M, Kim S, Ganbold E-O, Lee SY, Joo S-W. A spatiotemporal anticancer drug release platform of PEGylated graphene oxide triggered by glutathione in vitro and in vivo. J Mater Chem. 2012;22(45):23845–23851. doi:10.1039/c2jm34853e

103. Allen MJ, Tung VC, Kaner RB. Honeycomb carbon: a review of graphene. Chem Rev. 2010;110(1):132–145. doi:10.1021/cr900070d

104. Geim AK Graphene: status and Prospects. Available from: www.sciencemag.org. Accessed June 1, 2024.

105. Talukdar Y, Rashkow JT, Lalwani G, Kanakia S, Sitharaman B. The effects of graphene nanostructures on mesenchymal stem cells. Biomaterials. 2014;35(18):4863–4877. doi:10.1016/j.biomaterials.2014.02.054

106. Chen YW, Su YL, Hu SH, Chen SY. Functionalized graphene nanocomposites for enhancing photothermal therapy in tumor treatment. Adv Drug Deliv Rev. 2016;105:190–204. doi:10.1016/j.addr.2016.05.022

107. Li R, Tian Y, Zhu B, et al. Graphene-containing metal-organic framework nanocomposites for enhanced microwave ablation of salivary adenoid cystic carcinoma. Nanoscale Adv. 2022;4(5):1308–1317. doi:10.1039/d1na00729g

108. Son IH, Park JH, Park S, et al. Graphene balls for lithium rechargeable batteries with fast charging and high volumetric energy densities. Nat Commun. 2017;8(1):1561. doi:10.1038/s41467-017-01823-7

109. Saba N, Jawaid M. Energy and environmental applications of graphene and its derivatives. In: Polymer-Based Nanocomposites for Energy and Environmental Applications: A Volume in Woodhead Publishing Series in Composites Science and Engineering. University of Ottawa Press; 2018:106–129. doi:10.1016/B978-0-08-102262-7.00004-0

110. Novoselov KS, Geim AK, Morozov SV, et al. Electric field in atomically thin carbon films. Science (1979). 2004;306(5696):666–669. doi:10.1126/science.1102896

111. Bai L, Li Z, Zhang Y, et al. Synthesis of water-dispersible graphene-modified magnetic polypyrrole nanocomposite and its ability to efficiently adsorb methylene blue from aqueous solution. Chem Eng J. 2015;279:757–766. doi:10.1016/j.cej.2015.05.068

112. Ren S, Rong P, Yu Q. Preparations, properties and applications of graphene in functional devices: a concise review. Ceram Int. 2018;44(11):11940–11955. doi:10.1016/j.ceramint.2018.04.089

113. Xiong X, Huang B, Wei X, Wang L, Zhang L. Research of graphene preparation methods. In: Lecture Notes in Electrical Engineering. Vol. 477. Springer Verlag;2018:963–971. doi:10.1007/978-981-10-7629-9_120

114. Novoselov KS, Fal’Ko VI, Colombo L, Gellert PR, Schwab MG, Kim K. A roadmap for graphene. Nature. 2012;490(7419):192–200. doi:10.1038/nature11458

115. Uhl FM, Wilkie CA. Preparation of nanocomposites from styrene and modified graphite oxides. Polym Degrad Stab. 2004;84(2):215–226. doi:10.1016/j.polymdegradstab.2003.10.014

116. Yu H, Zhang B, Bulin C, Li R, Xing R. High-efficient Synthesis of Graphene Oxide Based on Improved Hummers Method. Sci Rep. 2016;6. doi:10.1038/srep36143

117. Krishnamoorthy K, Veerapandian M, Yun K, Kim SJ. The chemical and structural analysis of graphene oxide with different degrees of oxidation. Carbon N Y. 2013;53:38–49. doi:10.1016/j.carbon.2012.10.013

118. Ding R, Li W, Wang X, et al. A brief review of corrosion protective films and coatings based on graphene and graphene oxide. J Alloys Compd. 2018;764:1039–1055. doi:10.1016/j.jallcom.2018.06.133

119. Donarelli M, Ottaviano L. 2d materials for gas sensing applications: a review on graphene oxide, mos2, ws2 and phosphorene. Sensors (Switzerland). 2018;18(11):3638. doi:10.3390/s18113638

120. Wei Y, Zhang Y, Gao X, Ma Z, Wang X, Gao C. Multilayered graphene oxide membrane for water treatment: a review. Carbon N Y. 2018;139:964–981. doi:10.1016/j.carbon.2018.07.040

121. Farjadian F, Abbaspour S, Sadatlu MAA, et al. Recent Developments in Graphene and Graphene Oxide: properties, Synthesis, and Modifications: a Review. ChemistrySelect. 2020;5(33):10200–10219. doi:10.1002/slct.202002501

122. Moosa AA, Abed MS. Graphene preparation and graphite exfoliation. Turk J Chem. 2021;45(3):493–519. doi:10.3906/kim-2101-19

123. Hummers WS, Offeman RE Preparation of Graphitic Oxide; 1958. Available from: https://pubs.acs.org/sharingguidelines. Accessed June 1, 2024.

124. Marcano DC, Kosynkin DV, Berlin JM, et al. Improved synthesis of graphene oxide. ACS Nano. 2010;4(8):4806–4814. doi:10.1021/nn1006368

125. Zhang W, Liu Z, Xia J, et al. Preparing graphene from anode graphite of spent lithium-ion batteries. Front Environ Sci Eng. 2017;11(5):993. doi:10.1007/s11783-017-0993-8

126. Ruan G, Sun Z, Peng Z, Tour JM. Growth of graphene from food, insects, and waste. ACS Nano. 2011;5(9):7601–7607. doi:10.1021/nn202625c

127. Wang R, Shou D, Lv O, Kong Y, Deng L, Shen J. pH-Controlled drug delivery with hybrid aerogel of chitosan, carboxymethyl cellulose and graphene oxide as the carrier. Int J Biol Macromol. 2017;103:248–253. doi:10.1016/j.ijbiomac.2017.05.064

128. Weaver CL, Larosa JM, Luo X, Cui XT. Electrically controlled drug delivery from graphene oxide nanocomposite films. ACS Nano. 2014;8(2):1834–1843. doi:10.1021/nn406223e

129. Karimzadeh Z, Namazi H. Nontoxic double-network polymeric hybrid aerogel functionalized with reduced graphene oxide: preparation, characterization, and evaluation as drug delivery agent. J Polym Res. 2022;29(2):2902. doi:10.1007/s10965-022-02902-0

130. Zhao H, Qu W, Hu L, et al. Coupling Facet Cu(111)/(100)-Functionalized Graphene Aerogels for a Remarkable Air Disinfection Filter: extracellular Electron Transfer and the Sharp-Edge Membrane Penetration Effect. ACS ES and T Engineering. 2022;2(12):2220–2233. doi:10.1021/acsestengg.2c00199

131. Nanda SS, Yi DK, Kim K. Study of antibacterial mechanism of graphene oxide using Raman spectroscopy. Sci Rep. 2016;6. doi:10.1038/srep28443

132. Tu Y, Lv M, Xiu P, et al. Destructive extraction of phospholipids from Escherichia coli membranes by graphene nanosheets. Nat Nanotechnol. 2013;8(8):594–601. doi:10.1038/nnano.2013.125

133. Mann R, Mitsidis D, Xie Z, et al. Antibacterial Activity of Reduced Graphene Oxide. J Nanomater. 2021;2021:577. doi:10.1155/2021/9941577

134. Prasad K, Lekshmi GS, Ostrikov K, et al. Synergic bactericidal effects of reduced graphene oxide and silver nanoparticles against Gram-positive and Gram-negative bacteria. Sci Rep. 2017;7(1):669. doi:10.1038/s41598-017-01669-5

135. Asgari S, Mohammadi Ziarani G, Badiei A, Setayeshmehr M, Kiani M, Pourjavadi A. Electrospun Ag-decorated reduced GO-graft-chitosan composite nanofibers with visible light photocatalytic activity for antibacterial performance. Chemosphere. 2022;299. doi:10.1016/j.chemosphere.2022.134436

136. Blanco M, Agnoli S, Granozzi G. Graphene Acid: a Versatile 2D Platform for Catalysis. Isr J Chem. 2022;62(3–4):118. doi:10.1002/ijch.202100118

137. Jankovský O, Nováček M, Luxa J, et al. A New Member of the Graphene Family: graphene Acid. Chem a Eur J. 2016;22(48):17416–17424. doi:10.1002/chem.201603766

138. Bakandritsos A, Pykal M, Boński P, et al. Cyanographene and Graphene Acid: emerging Derivatives Enabling High-Yield and Selective Functionalization of Graphene. ACS Nano. 2017;11(3):2982–2991. doi:10.1021/acsnano.6b08449

139. Bakandritsos A, Hobza P, Zbořil R, et al. Carboxylated graphene for radical-assisted ultra-trace-level water treatment and noble metal recovery. ACS Nano. 2021;15(2):3349–3358. doi:10.1021/acsnano.0c10093

140. Mosconi D, Blanco M, Gatti T, et al. Arene C–H insertion catalyzed by ferrocene covalently heterogenized on graphene acid. Carbon N Y. 2019;143:318–328. doi:10.1016/j.carbon.2018.11.010

141. Reuillard B, Blanco M, Calvillo L, et al. Noncovalent Integration of a Bioinspired Ni Catalyst to Graphene Acid for Reversible Electrocatalytic Hydrogen Oxidation. ACS Appl Mater Interfaces. 2020;12(5):5805–5811. doi:10.1021/acsami.9b18922

142. Zaoralová D, Mach R, Lazar P, Medveď M, Otyepka M. Anchoring of Transition Metals to Graphene Derivatives as an Efficient Approach for Designing Single-Atom Catalysts. Adv Mater Interfaces. 2021;8(8):1392. doi:10.1002/admi.202001392

143. Sanad MF, Chava VSN, Shalan AE, et al. Engineering of Electron Affinity and Interfacial Charge Transfer of Graphene for Self-Powered Nonenzymatic Biosensor Applications. ACS Appl Mater Interfaces. 2021;13(34):40731–40741. doi:10.1021/acsami.1c12423

144. Dong Z, Qi J, Yue L, et al. Biomass-based carbon quantum dots and their agricultural applications. Plant Stress. 2024:11. doi:10.1016/j.stress.2024.100411

145. Xu X, Ray R, Gu Y, et al. Electrophoretic analysis and purification of fluorescent single-walled carbon nanotube fragments. J Am Chem Soc. 2004;126(40):12736–12737. doi:10.1021/ja040082h

146. Sun YP, Zhou B, Lin Y, et al. Quantum-sized carbon dots for bright and colorful photoluminescence. J Am Chem Soc. 2006;128(24):7756–7757. doi:10.1021/ja062677d

147. Wang Y, Hu A. Carbon quantum dots: synthesis, properties and applications. J Mater Chem C Mater. 2014;2(34):6921–6939. doi:10.1039/c4tc00988f

148. Baker SN, Baker GA. Luminescent carbon nanodots: emergent nanolights. Angewandte Chemie - Int Ed. 2010;49(38):6726–6744. doi:10.1002/anie.200906623

149. Sousa HBA, Martins CSM, Prior JAV. You don’t learn that in school: an updated practical guide to carbon quantum dots. Nanomaterials. 2021;11(3):1–88. doi:10.3390/nano11030611

150. Wu YF, Wu HC, Kuan CH, et al. Multi-functionalized carbon dots as theranostic nanoagent for gene delivery in lung cancer therapy. Sci Rep. 2016:6. doi:10.1038/srep21170

151. Yan F, Sun Z, Zhang H, Sun X, Jiang Y, Bai Z. The fluorescence mechanism of carbon dots, and methods for tuning their emission color: a review. Mikrochim Acta. 2019;186(8):3688. doi:10.1007/s00604-019-3688-y

152. Xu J, Sahu S, Cao L, et al. Efficient fluorescence quenching in carbon dots by surface-doped metals - Disruption of excited state redox processes and mechanistic implications. Langmuir. 2012;28(46):16141–16147. doi:10.1021/la302506e

153. Zheng L, Chi Y, Dong Y, Lin J, Wang B. Electrochemiluminescence of water-soluble carbon nanocrystals released electrochemically from graphite. J Am Chem Soc. 2009;131(13):4564–4565. doi:10.1021/ja809073f

154. Wang X, Qu K, Xu B, Ren J, Qu X. Microwave assisted one-step green synthesis of cell-permeable multicolor photoluminescent carbon dots without surface passivation reagents. J Mater Chem. 2011;21(8):2445–2450. doi:10.1039/c0jm02963g

155. Wang F, Pang S, Wang L, Li Q, Kreiter M, Liu CY. One-step synthesis of highly luminescent carbon dots in noncoordinating solvents. Chem. Mater. 2010;22(16):4528–4530. doi:10.1021/cm101350u

156. Desmond LJ, Phan AN, Gentile P. Critical overview on the green synthesis of carbon quantum dots and their application for cancer therapy. Environ Sci Nano. 2021;8(4):848–862. doi:10.1039/d1en00017a

157. Sakdaronnarong C, Sangjan A, Boonsith S, Kim DC, Shin HS. Recent developments in synthesis and photocatalytic applications of carbon dots. Catalysts. 2020;10(3):320. doi:10.3390/catal10030320

158. Magesh V, Sundramoorthy AK, Ganapathy D. Recent Advances on Synthesis and Potential Applications of Carbon Quantum Dots. Front Mater. 2022;9. doi:10.3389/fmats.2022.906838

159. Zhao X, Gao W, Zhang H, Qiu X, Luo Y. Graphene quantum dots in biomedical applications: recent advances and future challenges. In: Handbook of Nanomaterials in Analytical Chemistry: Modern Trends in Analysis. Elsevier; 2019:493–505. doi:10.1016/B978-0-12-816699-4.00020-7

160. Kang S, Kim KM, Jung K, et al. Graphene Oxide Quantum Dots Derived from Coal for Bioimaging: facile and Green Approach. Sci Rep. 2019;9(1):3747. doi:10.1038/s41598-018-37479-6

161. Iannazzo D, Pistone A, Salamò M, et al. Graphene quantum dots for cancer targeted drug delivery. Int J Pharm. 2017;518(1–2):185–192. doi:10.1016/j.ijpharm.2016.12.060

162. Pan D, Zhang J, Li Z, Wu M. Hydrothermal route for cutting graphene sheets into blue-luminescent graphene quantum dots. Adv. Mater. 2010;22(6):734–738. doi:10.1002/adma.200902825

163. Gong P, Wang J, Hou K, et al. Small but strong: the influence of fluorine atoms on formation and performance of graphene quantum dots using a gradient F-sacrifice strategy. Carbon N Y. 2017;112:63–71. doi:10.1016/j.carbon.2016.10.091

164. Dong Y, Lin J, Chen Y, Fu F, Chi Y, Chen G. Graphene quantum dots, graphene oxide, carbon quantum dots and graphite nanocrystals in coals. Nanoscale. 2014;6(13):7410–7415. doi:10.1039/c4nr01482k

165. Yang X, Zhao Q, Chen Y, et al. Effects of graphene oxide and graphene oxide quantum dots on the osteogenic differentiation of stem cells from human exfoliated deciduous teeth. Artif Cells Nanomed Biotechnol. 2019;47(1):822–832. doi:10.1080/21691401.2019.1576706

166. Kortel M, Mansuriya BD, Santana NV, Altintas Z. Graphene quantum dots as flourishing nanomaterials for bio-imaging, therapy development, and micro-supercapacitors. Micromachines (Basel). 2020;11(9):866. doi:10.3390/MI11090866

167. Tashkhourian J, Nami-Ana SF, Shamsipur M. Designing a modified electrode based on graphene quantum dot-chitosan application to electrochemical detection of epinephrine. J Mol Liq. 2018;266:548–556. doi:10.1016/j.molliq.2018.06.093

168. Wang T, Zhu S, Jiang X. Toxicity mechanism of graphene oxide and nitrogen-doped graphene quantum dots in RBCs revealed by surface-enhanced infrared absorption spectroscopy. Toxicol Res (Camb). 2015;4(4):885–894. doi:10.1039/c4tx00138a

169. Tajik S, Dourandish Z, Zhang K, et al. Carbon and graphene quantum dots: a review on syntheses, characterization, biological and sensing applications for neurotransmitter determination. RSC Adv. 2020;10(26):15406–15429. doi:10.1039/d0ra00799d

170. Huang H, Yang S, Li Q, et al. Electrochemical Cutting in Weak Aqueous Electrolytes: the Strategy for Efficient and Controllable Preparation of Graphene Quantum Dots. Langmuir. 2018;34(1):250–258. doi:10.1021/acs.langmuir.7b03425

171. Lu J, Yeo PSE, Gan CK, Wu P, Loh KP. Transforming C 60 molecules into graphene quantum dots. Nat Nanotechnol. 2011;6(4):247–252. doi:10.1038/nnano.2011.30

172. Lu Q, Wu C, Liu D, et al. A facile and simple method for synthesis of graphene oxide quantum dots from black carbon. Green Chem. 2017;19(4):900–904. doi:10.1039/c6gc03092k

173. Zhao Q, Zhou Y, Li Y, Gu W, Zhang Q, Liu J. Luminescent Iridium(III) Complex Labeled DNA for Graphene Oxide-Based Biosensors. Anal Chem. 2016;88(3):1892–1899. doi:10.1021/acs.analchem.5b04324

174. Loh KP, Bao Q, Eda G, Chhowalla M. Graphene oxide as a chemically tunable platform for optical applications. Nat Chem. 2010;2(12):1015–1024. doi:10.1038/nchem.907

175. Jang MH, Yang H, Chang YH, et al. Selective engineering of oxygen-containing functional groups using the alkyl ligand oleylamine for revealing the luminescence mechanism of graphene oxide quantum dots. Nanoscale. 2017;9(47):18635–18643. doi:10.1039/c7nr04150k

176. Shi Y, Pramanik A, Tchounwou C, et al. Multifunctional Biocompatible Graphene Oxide Quantum Dots Decorated Magnetic Nanoplatform for Efficient Capture and Two-Photon Imaging of Rare Tumor Cells. ACS Appl Mater Interfaces. 2015;7(20):10935–10943. doi:10.1021/acsami.5b02199

177. Tshangana CS, Muleja AA, Kuvarega AT, Malefetse TJ, Mamba BB. The applications of graphene oxide quantum dots in the removal of emerging pollutants in water: an overview. J Water Process Eng. 2021;43. doi:10.1016/j.jwpe.2021.102249

178. Wang C, Wu C, Zhou X, et al. Enhancing Cell Nucleus Accumulation and DNA Cleavage Activity of Anti-Cancer Drug via Graphene Quantum Dots. Sci Rep. 2013:3. doi:10.1038/srep02852

179. Fang J, Liu Y, Chen Y, Ouyang D, Yang G, Yu T. Graphene quantum dots-gated hollow mesoporous carbon nanoplatform for targeting drug delivery and synergistic chemo-photothermal therapy. Int J Nanomed. 2018;13:5991–6007. doi:10.2147/IJN.S175934

180. Xu Y, Wang X, Zhang WL, Lv F, Guo S. Recent progress in two-dimensional inorganic quantum dots. Chem Soc Rev. 2018;47(2):586–625. doi:10.1039/c7cs00500h

181. Ke PC, Pilkington EH, Sun Y, et al. Mitigation of Amyloidosis with Nanomaterials. Adv. Mater. 2020;32(18):1690. doi:10.1002/adma.201901690

182. Mahmoudi M, Akhavan O, Ghavami M, Rezaee F, Ghiasi SMA. Graphene oxide strongly inhibits amyloid beta fibrillation. Nanoscale. 2012;4(23):7322–7325. doi:10.1039/c2nr31657a

183. Fan Z, Nie Y, Wei Y, Zhao J, Liao X, Zhang J. Facile and large-scale synthesis of graphene quantum dots for selective targeting and imaging of cell nucleus and mitochondria. Mater Sci Eng C. 2019;103. doi:10.1016/j.msec.2019.109824

184. Liu ML, Yang L, Li RS, Chen B, Huang CZ. Large-scale simultaneous synthesis of highly photoluminescent green amorphous carbon nanodots and yellow crystalline graphene quantum dots at room temperature. Green Chem. 2017;19(15):3611–3617. doi:10.1039/c7gc01236e

185. Cao H, Qi W, Gao X, Wu Q, Tian L, Wu W. Graphene Quantum Dots prepared by Electron Beam Irradiation for Safe Fluorescence Imaging of Tumor. Nanotheranostics. 2022;6(2):205–214. doi:10.7150/ntno.67070

186. Pramanik A, Chavva SR, Fan Z, Sinha SS, Nellore BPV, Ray PC. Extremely high two-photon absorbing graphene oxide for imaging of tumor cells in the second biological window. J Phys Chem Lett. 2014;5(12):2150–2154. doi:10.1021/jz5009856

187. Pramanik A, Fan Z, Chavva SR, Sinha SS, Ray PC. Highly efficient and excitation tunable two-photon luminescence platform for targeted multi-color MDRB imaging using graphene oxide. Sci Rep. 2014;4. doi:10.1038/srep06090

188. Anzar N, Hasan R, Tyagi M, Yadav N, Narang J. Carbon nanotube - A review on Synthesis, Properties and plethora of applications in the field of biomedical science. Sensors International. 2020;1. doi:10.1016/j.sintl.2020.100003

189. Kharche G, Lokavarapu BR. Static buckling analysis of single walled carbon nanotube. Mater Today Proc. 2023. doi:10.1016/j.matpr.2023.02.047

190. Boncel S, Zajac P, Koziol KKK. Liberation of drugs from multi-wall carbon nanotube carriers. J Control Release. 2013;169(1–2):126–140. doi:10.1016/j.jconrel.2013.04.009

191. Thostenson ET, Ren Z, Chou TW Advances in the Science and Technology of Carbon Nanotubes and Their Composites: a Review. Available from: www.elsevier.com/locate/compscitech. Accessed June 1, 2024.

192. Aliev AE, Lima MH, Silverman EM, Baughman RH. Thermal conductivity of multi-walled carbon nanotube sheets: radiation losses and quenching of phonon modes. Nanotechnology. 2010;21(3):5709. doi:10.1088/0957-4484/21/3/035709

193. Ates M, Eker AA, Eker B. Carbon nanotube-based nanocomposites and their applications. J Adhes Sci Technol. 2017;31(18):1977–1997. doi:10.1080/01694243.2017.1295625

194. Soltani A, Moradi AV, Lemeski ET. The interaction of 2,6-dichlorobenzylidene-2,4-dichloroaniline (2,6-DBDA) and 2,4-dichlorobenzylidene-2,4-dichloroaniline (2,4-DBDA) with single-walled carbon nanotube: a DFT study. J Mol Struct. 2016;1105:128–134. doi:10.1016/j.molstruc.2015.10.018

195. Vashist SK, Zheng D, Pastorin G, Al-Rubeaan K, Luong JHT, Sheu FS. Delivery of drugs and biomolecules using carbon nanotubes. Carbon N Y. 2011;49(13):4077–4097. doi:10.1016/j.carbon.2011.05.049

196. Monavari SM, Marsusi F, Memarian N, Qasemnazhand M. Carbon nanotubes and nanobelts as potential materials for biosensor. Sci Rep. 2023;13(1):862. doi:10.1038/s41598-023-29862-9

197. Liu Z, Sun X, Nakayama-Ratchford N, Dai H. Supramolecular chemistry on water- Soluble carbon nanotubes for drug loading and delivery. ACS Nano. 2007;1(1):50–56. doi:10.1021/nn700040t

198. Ajima K, Yudasaka M, Murakami T, Maigné A, Shiba K, Iijima S. Carbon nanohorns as anticancer drug carriers. Mol Pharm. 2005;2(6):475–480. doi:10.1021/mp0500566

199. Georgakilas V, Perman JA, Tucek J, Zboril R. Broad Family of Carbon Nanoallotropes: classification, Chemistry, and Applications of Fullerenes, Carbon Dots, Nanotubes, Graphene, Nanodiamonds, and Combined Superstructures. Chem Rev. 2015;115(11):4744–4822. doi:10.1021/cr500304f

200. Iijima S, Yudasaka M, Yamada R, et al. Nano-Aggregates of Single-Walled Graphitic Carbon Nano-Horns. 1999. Available from: www.elsevier.nlrlocatercplett. Accessed June 1, 2024.

201. Berber S, Kwon YK, Tomá D. Electronic and Structural Properties of Carbon Nanohorns. Physical Review B. 2000;62(4):R2291.

202. Moreno-Lanceta A, Medrano-Bosch M, Melgar-Lesmes P. Single-walled carbon nanohorns as promising nanotube-derived delivery systems to treat cancer. Pharmaceutics. 2020;12(9):1–21. doi:10.3390/pharmaceutics12090850

203. Karousis N, Suarez-Martinez I, Ewels CP, Tagmatarchis N. Structure, Properties, Functionalization, and Applications of Carbon Nanohorns. Chem Rev. 2016;116(8):4850–4883. doi:10.1021/acs.chemrev.5b00611

204. Zhu S, Xu G. Single-walled carbon nanohorns and their applications. Nanoscale. 2010;2(12):2538–2549. doi:10.1039/c0nr00387e

205. Yuge R, Yudasaka M, Toyama K, Yamaguchi T, Iijima S, Manako T. Buffer gas optimization in CO 2 laser ablation for structure control of single-wall carbon nanohorn aggregates. Carbon N Y. 2012;50(5):1925–1933. doi:10.1016/j.carbon.2011.12.043

206. He B, Shi Y, Liang Y, et al. Single-walled carbon-nanohorns improve biocompatibility over nanotubes by triggering less protein-initiated pyroptosis and apoptosis in macrophages. Nat Commun. 2018;9(1):470. doi:10.1038/s41467-018-04700-z

207. Zhang M, Murakami T, Ajima K, et al. Fabrication of ZnPc/Protein Nanohorns for Double Photodynamic and Hyperthermic Cancer Phototherapy; 2008. Available from: www.pnas.orgcgidoi10.1073pnas.0801349105. Accessed June 1, 2024.

208. Heredia DA, Durantini AM, Durantini JE, Durantini EN. Fullerene C60 derivatives as antimicrobial photodynamic agents. J Photochem Photobiol C Photochem Rev. 2022;51. doi:10.1016/j.jphotochemrev.2021.100471

209. Kroto H, Heath J, O’Brien S. C60: buckminsterfullerene. Nature. 1985;318:162–163.

210. Shetti NP, Mishra A, Basu S, Aminabhavi TM. Versatile fullerenes as sensor materials. Mater Today Chem. 2021;20. doi:10.1016/j.mtchem.2021.100454

211. Haddon RC Chemistry of the Fullerenes: the Manifestation of Strain in a Class of Continuous Aromatic Molecules. Available from: www.sciencemag.org. Accessed June 1, 2024.

212. Goodarzi S, Da Ros T, Conde J, Sefat F, Mozafari M. Fullerene: biomedical engineers get to revisit an old friend. Mater Today. 2017;20(8):460–480. doi:10.1016/j.mattod.2017.03.017

213. Kazemzadeh H, Mozafari M. Fullerene-based delivery systems. Drug Discov Today. 2019;24(3):898–905. doi:10.1016/j.drudis.2019.01.013

214. Azizi-Lalabadi M, Hashemi H, Feng J, Jafari SM. Carbon nanomaterials against pathogens; the antimicrobial activity of carbon nanotubes, graphene/graphene oxide, fullerenes, and their nanocomposites. Adv Colloid Interface Sci. 2020;284. doi:10.1016/j.cis.2020.102250

215. Thomas KG, Biju V, George MV, Guldi DM, Kamat PV Excited-State Interactions in Pyrrolidinofullerenes; 1998. Available from: http://www.nd.edu/. Accessed June 1, 2024.

216. Lyon DY, Brunet L, Hinkal GW, Wiesner MR, Alvarez PJJ. Antibacterial activity of fullerene water suspensions (nC 60) is not due to ROS-mediated damage. Nano Lett. 2008;8(5):1539–1543. doi:10.1021/nl0726398

217. Mojica M, Alonso JA, Méndez F. Synthesis of fullerenes. J Phys Org Chem. 2013;26(7):526–539. doi:10.1002/poc.3121

218. Chae SR, Hotze EM, Wiesner MR. Possible Applications of Fullerene Nanomaterials in Water Treatment and Reuse. In: Nanotechnology Applications for Clean Water: Solutions for Improving Water Quality: Second Edition. Elsevier Inc.; 2014:329–338. doi:10.1016/B978-1-4557-3116-9.00021-4

219. Mao D, Wang X, Zhou G, Chen L, Chen J, Zeng S. Fullerene-intercalated graphene nanocontainers for gas storage and sustained release. J Mol Modeling. 2020:26. doi:10.1007/s00894-020-04417-1/Published

220. Agrawal PS, Belkhode PN, Brijpuriya DS, Gouda SP, Rokhum SL. Stimulation in fullerene for adsorbing pollutant gases: a review. Chemical Physics Impact. 2023;6. doi:10.1016/j.chphi.2022.100156

221. Nakamura E, Isobe H. Functionalized Fullerenes in Water. The First 10 Years of Their Chemistry, Biology, and Nanoscience. Acc Chem Res. 2003;36(11):807–815. doi:10.1021/ar030027y

222. Gopakumar DA, Abdul AK, Pai AR, Thomas S, Pasquini D, Shao-Yuan Ben L. Nanomaterials—State of Art, New Challenges, and Opportunities. In: Nanoscale Materials in Water Purification. Elsevier; 2019:1–24. doi:10.1016/B978-0-12-813926-4.00001-X

223. Montellano A, Da Ros T, Bianco A, Prato M. Fullerene C60 as a multifunctional system for drug and gene delivery. Nanoscale. 2011;3(10):4035–4041. doi:10.1039/c1nr10783f

224. Raza K, Thotakura N, Kumar P, et al. C60-fullerenes for delivery of docetaxel to breast cancer cells: a promising approach for enhanced efficacy and better pharmacokinetic profile. Int J Pharm. 2015;495(1):551–559. doi:10.1016/j.ijpharm.2015.09.016

225. Soldà A, Cantelli A, Di Giosia M, et al. C60@lysozyme: a new photosensitizing agent for photodynamic therapy. J Mater Chem B. 2017;5(32):6608–6615. doi:10.1039/c7tb00800g

226. Du Z, Gao N, Wang X, Ren J, Qu X. Near-Infrared Switchable Fullerene-Based Synergy Therapy for Alzheimer’s Disease. Small. 2018;14(33):852. doi:10.1002/smll.201801852

227. Friedman SH, Decamp DL, Sijbesma RP, Srdanov G, Wudl F, Kenyon GL Inhibition of the HIV-1 Protease by Fullerene Derivatives: model Building Studies and Experimental Verification; 1993. Available from: https://pubs.acs.org/sharingguidelines. Accessed June 1, 2024.

228. Yasuno T, Ohe T, Takahashi K, Nakamura S, Mashino T. The human immunodeficiency virus-reverse transcriptase inhibition activity of novel pyridine/pyridinium-type fullerene derivatives. Bioorg Med Chem Lett. 2015;25(16):3226–3229. doi:10.1016/j.bmcl.2015.05.086

229. Castro E, Martinez ZS, Seong CS, et al. Characterization of New Cationic N,N-Dimethyl[70]fulleropyrrolidinium Iodide Derivatives as Potent HIV-1 Maturation Inhibitors. J Med Chem. 2016;59(24):10963–10973. doi:10.1021/acs.jmedchem.6b00994

230. Zakharova OV, Mastalygina EE, Golokhvast KS, Gusev AA. Graphene nanoribbons: prospects of application in biomedicine and toxicity. Nanomaterials. 2021;11(9):2425. doi:10.3390/nano11092425

231. Nakada K, Fujita M, Dresselhaus G, Dresselhaus MS. Edge State in Graphene Ribbons: nanometer Size Effect and Edge Shape Dependence. J Mol Modeling. 1996.

232. Terrones M, Botello-Méndez AR, Campos-Delgado J, et al. Graphene and graphite nanoribbons: morphology, properties, synthesis, defects and applications. Nano Today. 2010;5(4):351–372. doi:10.1016/j.nantod.2010.06.010

233. Imani R, Mohabatpour F, Mostafavi F. Graphene-based Nano-Carrier modifications for gene delivery applications. Carbon N Y. 2018;140:569–591. doi:10.1016/j.carbon.2018.09.019

234. Johnson AP, Gangadharappa HV, Pramod K. Graphene nanoribbons: a promising nanomaterial for biomedical applications. J Control Release. 2020;325:141–162. doi:10.1016/j.jconrel.2020.06.034

235. Goenka S, Sant V, Sant S. Graphene-based nanomaterials for drug delivery and tissue engineering. J Control Release. 2014;173(1):75–88. doi:10.1016/j.jconrel.2013.10.017

236. Johnson AP, Sabu C, Swamy NK, Anto A, Gangadharappa HV, Pramod K. Graphene nanoribbon: an emerging and efficient flat molecular platform for advanced biosensing. Biosens Bioelectron. 2021;184. doi:10.1016/j.bios.2021.113245

237. Jampilek J, Kralova K. Advances in drug delivery nanosystems using graphene‐based materials and carbon nanotubes. Materials. 2021;14(5):1–39. doi:10.3390/ma14051059

238. Mousavi SM, Soroshnia S, Hashemi SA, et al. Graphene nano-ribbon based high potential and efficiency for DNA, cancer therapy and drug delivery applications. Drug Metab Rev. 2019;51(1):91–104. doi:10.1080/03602532.2019.1582661

239. Shang L, Zhao F, Zeng B. Highly dispersive hollow PdAg alloy nanoparticles modified ionic liquid functionalized graphene nanoribbons for electrochemical sensing of nifedipine. Electrochim Acta. 2015;168:330–336. doi:10.1016/j.electacta.2015.04.024

240. Govindasamy M, Mani V, Chen SM, et al. Highly sensitive determination of non-steroidal anti-inflammatory drug nimesulide using electrochemically reduced graphene oxide nanoribbons. RSC Adv. 2017;7(52):33043–33051. doi:10.1039/c7ra02844j

241. Asadian E, Shahrokhian S, Zad AI, Jokar E. In-situ electro-polymerization of graphene nanoribbon/polyaniline composite film: application to sensitive electrochemical detection of dobutamine. Sens Actuators B Chem. 2014;196:582–588. doi:10.1016/j.snb.2014.02.049

242. Wang H, Wang HS, Ma C, et al. Graphene nanoribbons for quantum electronics. Nat Rev Phys. 2021;3(12):791–802. doi:10.1038/s42254-021-00370-x

243. Liu M, Liu M, She L, et al. Graphene-like nanoribbons periodically embedded with four- and eight-membered rings. Nat Commun. 2017:8. doi:10.1038/ncomms14924

244. Zhang B, Cui T. Suspended graphene nanoribbon ion-sensitive field-effect transistors formed by shrink lithography for pH/cancer biomarker sensing. J Microelectromech Syst. 2013;22(5):1140–1146. doi:10.1109/JMEMS.2013.2254701

245. Han MY, Özyilmaz B, Zhang Y, Kim P. Energy band-gap engineering of graphene nanoribbons. Phys Rev Lett. 2007;98(20):6805. doi:10.1103/PhysRevLett.98.206805

246. Chen L, Hernandez Y, Feng X, Müllen K. From nanographene and graphene nanoribbons to graphene sheets: chemical synthesis. Angewandte Chemie - Int Ed. 2012;51(31):7640–7654. doi:10.1002/anie.201201084

247. Kosynkin DV, Lu W, Sinitskii A, Pera G, Sun Z, Tour JM. Highly conductive graphene nanoribbons by longitudinal splitting of carbon nanotubes using potassium vapor. ACS Nano. 2011;5(2):968–974. doi:10.1021/nn102326c

248. Luo H, Preparation YG. Bandgap Engineering, and Performance Control of Graphene Nanoribbons. Chem. Mater. 2022;34(8):3588–3615. doi:10.1021/acs.chemmater.1c04215

249. Mullick Chowdhury S, Zafar S, Tellez V, Sitharaman B. Graphene Nanoribbon-Based Platform for Highly Efficacious Nuclear Gene Delivery. ACS Biomater Sci Eng. 2016;2(5):798–808. doi:10.1021/acsbiomaterials.5b00562

250. Foreman HCC, Lalwani G, Kalra J, Krug LT, Sitharaman B. Gene delivery to mammalian cells using a graphene nanoribbon platform. J Mater Chem B. 2017;5(12):2347–2354. doi:10.1039/c6tb03010f

251. Liu Y, Wang X, Wan W, et al. Multifunctional nitrogen-doped graphene nanoribbon aerogels for superior lithium storage and cell culture. Nanoscale. 2016;8(4):2159–2167. doi:10.1039/c5nr05909g

252. Chowdhury SM, Surhland C, Sanchez Z, et al. Graphene nanoribbons as a drug delivery agent for lucanthone mediated therapy of glioblastoma multiforme. Nanomedicine. 2015;11(1):109–118. doi:10.1016/j.nano.2014.08.001

253. Chowdhury SM, Fang J, Sitharaman B. Interaction of graphene nanoribbons with components of the blood vascular system. Future Sci OA. 2015;1(3):17. doi:10.4155/fso.15.17

254. Mbeh DA, Akhavan O, Javanbakht T, Mahmoudi M, Yahia L. Cytotoxicity of protein Corona-graphene oxide nanoribbons on human epithelial cells. Appl Surf Sci. 2014;320:596–601. doi:10.1016/j.apsusc.2014.09.155

255. Mullick Chowdhury S, Dasgupta S, Mcelroy AE, Sitharaman B. Structural disruption increases toxicity of graphene nanoribbons. J Appl Toxicol. 2014;34(11):1235–1246. doi:10.1002/jat.3066

256. Tang L, Li J, Pan T, et al. Versatile carbon nanoplatforms for cancer treatment and diagnosis: strategies, applications and future perspectives. Theranostics. 2022;12(5):2290–2321. doi:10.7150/thno.69628

257. Augustine S, Singh J, Srivastava M, Sharma M, Das A, Malhotra BD. Recent advances in carbon based nanosystems for cancer theranostics. Biomater Sci. 2017;5(5):901–952. doi:10.1039/c7bm00008a

258. Saleem J, Wang L, Chen C. Carbon-Based Nanomaterials for Cancer Therapy via Targeting Tumor Microenvironment. Adv Healthc Mater. 2018;7(20):525. doi:10.1002/adhm.201800525

259. Su Y, Hu Y, Wang Y, et al. A precision-guided MWNT mediated reawakening the sunk synergy in RAS for anti-angiogenesis lung cancer therapy. Biomaterials. 2017;139:75–90. doi:10.1016/j.biomaterials.2017.05.046

260. Wong BS, Yoong SL, Jagusiak A, et al. Carbon nanotubes for delivery of small molecule drugs. Adv Drug Deliv Rev. 2013;65(15):1964–2015. doi:10.1016/j.addr.2013.08.005

261. Henna TK, Raphey VR, Sankar R, Ameena Shirin VK, Gangadharappa HV, Pramod K. Carbon nanostructures: the drug and the delivery system for brain disorders. Int J Pharm. 2020;587. doi:10.1016/j.ijpharm.2020.119701

262. Xiao Y, Pang YX, Yan Y, et al. Synthesis and Functionalization of Graphene Materials for Biomedical Applications: recent Advances, Challenges, and Perspectives. Adv. Sci. 2023;10(9):292. doi:10.1002/advs.202205292

263. Gosika M, Velachi V, Cordeiro MNDS, Maiti PK. Covalent Functionalization of Graphene with PAMAM Dendrimer and Its Implications on Graphene’s Dispersion and Cytotoxicity. ACS Appl Polym Mater. 2020;2(8):3587–3600. doi:10.1021/acsapm.0c00596

264. Zhang YH, Zhou KG, Xie KF, Zeng J, Zhang HL, Peng Y. Tuning the electronic structure and transport properties of graphene by noncovalent functionalization: effects of organic donor, acceptor and metal atoms. Nanotechnology. 2010;21(6):5201. doi:10.1088/0957-4484/21/6/065201

265. Bai H, Li C, Wang X, Shi G. A pH-sensitive graphene oxide composite hydrogel. Chem. Commun. 2010;46(14):2376. doi:10.1039/c000051e

266. Chen J, Liu H, Zhao C, et al. One-step reduction and PEGylation of graphene oxide for photothermally controlled drug delivery. Biomaterials. 2014;35(18):4986–4995. doi:10.1016/j.biomaterials.2014.02.032

267. Feng L, Zhang S, Liu Z. Graphene based gene transfection. Nanoscale. 2011;3(3):1252. doi:10.1039/c0nr00680g

268. Sarkar SD, Uddin MM, Roy CK, Hossen MJ, Sujan MI, Azam MS. Mechanically tough and highly stretchable poly(acrylic acid) hydrogel cross-linked by 2D graphene oxide. RSC Adv. 2020;10(18):10949–10958. doi:10.1039/d0ra00678e

269. Mendonça MCP, Soares ES, De Jesus MB, et al. PEGylation of reduced graphene oxide induces toxicity in cells of the blood-brain barrier: an in vitro and in vivo study. Mol Pharm. 2016;13(11):3913–3924. doi:10.1021/acs.molpharmaceut.6b00696

270. Bottini M, Rosato N, Bottini N. PEG-modified carbon nanotubes in biomedicine: current status and challenges ahead. Biomacromolecules. 2011;12(10):3381–3393. doi:10.1021/bm201020h

271. Du FP, Cao NN, Zhang YF, et al. PEDOT:PSS/graphene quantum dots films with enhanced thermoelectric properties via strong interfacial interaction and phase separation. Sci Rep. 2018;8(1):258. doi:10.1038/s41598-018-24632-4

272. Hong M, Wang Y, Wang R, et al. Poly(sodium styrene sulfonate) functionalized graphene as a highly efficient adsorbent for cationic dye removal with a green regeneration strategy. J Phys Chem Solids. 2021:152. doi:10.1016/j.jpcs.2021.109973

273. Jin R, Ji X, Yang Y, Wang H, Cao A. Self-assembled graphene-dextran nanohybrid for killing drug-resistant cancer cells. ACS Appl Mater Interfaces. 2013;5(15):7181–7189. doi:10.1021/am401523y

274. Ma R, Wang Y, Qi H, et al. Nanocomposite sponges of sodium alginate/graphene oxide/polyvinyl alcohol as potential wound dressing: in vitro and in vivo evaluation. Compos B Eng. 2019;167:396–405. doi:10.1016/j.compositesb.2019.03.006

275. Morimune S, Nishino T, Goto T. Poly(vinyl alcohol)/graphene oxide nanocomposites prepared by a simple eco-process. Polym J. 2012;44(10):1056–1063. doi:10.1038/pj.2012.58

276. Mehmood A, Mubarak NM, Khalid M, Jagadish P, Walvekar R, Abdullah EC. Graphene/PVA buckypaper for strain sensing application. Sci Rep. 2020;10(1):7713. doi:10.1038/s41598-020-77139-2

277. Kim H, Kim WJ. Photothermally Controlled Gene Delivery by Reduced Graphene Oxide–Polyethylenimine Nanocomposite. Small. 2014;10(1):117–126. doi:10.1002/smll.201202636

278. Teimouri M, Nia AH, Abnous K, Eshghi H, Ramezani M. Graphene oxide-cationic polymer conjugates: synthesis and application as gene delivery vectors. Plasmid. 2016;84-85:51–60. doi:10.1016/j.plasmid.2016.03.002

279. Kemp KC, Cho Y, Chandra V, Kim KS. Noncovalent Functionalization of Graphene. In: Georgakilas V, editor. Functionalization of Graphene. First. Wiley-VCH Verlag GmbH & Co. KGaA.; 2014.

280. Liu J, Li Y, Li Y, Li J, Deng Z. Noncovalent DNA decorations of graphene oxide and reduced graphene oxide toward water-soluble metal–carbon hybrid nanostructures via self-assembly. J Mater Chem. 2010;20(5):900–906. doi:10.1039/B917752C

281. Xu Y, Wu Q, Sun Y, Bai H, Shi G. Three-dimensional self-assembly of graphene oxide and DNA into multifunctional hydrogels. ACS Nano. 2010;4(12):7358–7362. doi:10.1021/nn1027104

282. Peña-Bahamonde J, Nguyen HN, Fanourakis SK, Rodrigues DF. Recent advances in graphene-based biosensor technology with applications in life sciences. J Nanobiotechnology. 2018;16(1):400. doi:10.1186/s12951-018-0400-z

283. Lu F, Zhang S, Gao H, Jia H, Zheng L. Protein-decorated reduced oxide graphene composite and its application to SERS. ACS Appl Mater Interfaces. 2012;4(6):3278–3284. doi:10.1021/am300634n

284. Assali M, Kittana N, Badran I, Omari S. Covalent functionalization of graphene sheets for plasmid DNA delivery: experimental and theoretical study. RSC Adv. 2023;13(10):7000–7008. doi:10.1039/D3RA00727H

285. Sturala J, Luxa J, Pumera M, Sofer Z. Chemistry of Graphene Derivatives: synthesis, Applications, and Perspectives. Chem Eur J. 2018;24(23):5992–6006. doi:10.1002/chem.201704192

286. Liu J, Tang J, Gooding JJ. Strategies for chemical modification of graphene and applications of chemically modified graphene. J Mater Chem. 2012;22(25):12435. doi:10.1039/c2jm31218b

287. Czerw R, Terrones M, Charlier JC, et al. Identification of Electron Donor States in N-Doped Carbon Nanotubes. Nano Lett. 2001;1(9):457–460. doi:10.1021/nl015549q

288. Yalcin M, Al-Sehemi AG, Erol I, et al. Fabrication of photodiodes based on graphene oxide (GO) doped lanthanum hexaboride (LaB6) nanocomposites. Diam Relat Mater. 2024:141. doi:10.1016/j.diamond.2023.110585

289. Zhai Z, Xu J, Gong T, et al. Sustainable fabrication of N-doped carbon quantum dots and their applications in fluorescent inks, Fe (III) detection and fluorescent films. Inorg Chem Commun. 2022:140. doi:10.1016/j.inoche.2022.109387

290. Rishabh RM, Shanker U, Singh Kaith B, Sillanpää M. Green fabrication of fluorescent N-doped carbon quantum dots from Aegle marmelos leaves for highly selective detection of Fe3+ metal ions. Inorg Chem Commun. 2024;159. doi:10.1016/j.inoche.2023.111878

291. Liu Z, Robinson JT, Sun X, Dai H. PEGylated nanographene oxide for delivery of water-insoluble cancer drugs. J Am Chem Soc. 2008;130(33):10876–10877. doi:10.1021/ja803688x

292. Sinitskii A, Dimiev A, Corley DA, Fursina AA, Kosynkin DV, Tour JM. Kinetics of Diazonium Functionalization of Chemically Converted Graphene Nanoribbons. ACS Nano. 2010;4(4):1949–1954. doi:10.1021/nn901899j

293. Bahr JL, Yang J, Kosynkin DV, Bronikowski MJ, Smalley RE, Tour JM. Functionalization of Carbon Nanotubes by Electrochemical Reduction of Aryl Diazonium Salts: a Bucky Paper Electrode. J Am Chem Soc. 2001;123(27):6536–6542. doi:10.1021/ja010462s

294. Xu Y, Sun L, Sun Y, et al. Effect of heat treatment on sensing performance of ZIF-67@GO for the detection of copper ions. Colloids Surf a Physicochem Eng Asp. 2022:649. doi:10.1016/j.colsurfa.2022.129500

295. Adenier A, Cabet-Deliry E, Chaussé A, et al. Grafting of Nitrophenyl Groups on Carbon and Metallic Surfaces without Electrochemical Induction. Chem. Mater. 2005;17(3):491–501. doi:10.1021/cm0490625

296. Kooi SE, Schlecht U, Burghard M, Kern K. Electrochemical Modification of Single Carbon Nanotubes. Angew. Chem. Int. Ed. 2002;41(8):1353–1355. doi:10.1002/1521-3773(20020415)41:8<1353::AID-ANIE1353>3.0.CO;2-I

297. Ganji Arjenaki R, Samieepour G, Sadat Ebrahimi SE, et al. Development of novel radiolabeled antibody-conjugated graphene quantum dots for targeted in vivo breast cancer imaging and biodistribution studies. Arabian J. Chem. 2024;17(2):5518. doi:10.1016/j.arabjc.2023.105518

298. Kaushal S, Pinnaka AK, Soni S, Singhal NK. Antibody assisted graphene oxide coated gold nanoparticles for rapid bacterial detection and near infrared light enhanced antibacterial activity. Sens Actuators B Chem. 2021;329. doi:10.1016/j.snb.2020.129141

299. Royvaran M, Taheri-Kafrani A, Landarani Isfahani A, Mohammadi S. Functionalized superparamagnetic graphene oxide nanosheet in enzyme engineering: a highly dispersive, stable and robust biocatalyst. Chem Eng J. 2016;288:414–422. doi:10.1016/j.cej.2015.12.034

300. Tsakmakidis K. Coherent absorption in graphene. Nat Mater. 2013;12(8):688. doi:10.1038/nmat3732

301. Kamińska I, Bohlen J, Yaadav R, et al. Graphene Energy Transfer for Single-Molecule Biophysics, Biosensing, and Super-Resolution Microscopy. Adv. Mater. 2021;33(24):99. doi:10.1002/adma.202101099

302. Mackowski S, Kaminska I. Energy Transfer in Graphene-Based Hybrid Photosynthetic Nanostructures. Recent Advances in Graphene Research InTech. 2016. doi:10.5772/64300

303. Lin L, Song X, Dong X, Li B. Nano-photosensitizers for enhanced photodynamic therapy. Photodiagnosis Photodyn Ther. 2021;36:102597. doi:10.1016/j.pdpdt.2021.102597

304. Li Y, Dong H, Li Y, Shi D. Graphene-based nanovehicles for photodynamic medical therapy. Int J Nanomed. 2015;10:2451–2459. doi:10.2147/IJN.S68600

305. Zhu J, Li C, Ou JY, Liu QH. Perfect light absorption in graphene by two unpatterned dielectric layers and potential applications. Carbon N Y. 2019;142:430–437. doi:10.1016/j.carbon.2018.10.073

306. Ghosh A, Chizhik AI, Karedla N, Enderlein J. Graphene- and metal-induced energy transfer for single-molecule imaging and live-cell nanoscopy with (sub)-nanometer axial resolution. Nat Protoc. 2021;16(7):3695–3715. doi:10.1038/s41596-021-00558-6

307. Samal A, Das DP, Madras G. Repercussion of Solid state vs. Liquid state synthesized p-n heterojunction RGO-copper phosphate on proton reduction potential in water. Sci Rep. 2018;8(1):7. doi:10.1038/s41598-018-21239-7

308. Li Q, Hong L, Li H, Liu C. Graphene oxide-fullerene C60 (GO-C60) hybrid for photodynamic and photothermal therapy triggered by near-infrared light. Biosens Bioelectron. 2017;89:477–482. doi:10.1016/j.bios.2016.03.072

309. Pelin M, Fusco L, Martín C, et al. Graphene and graphene oxide induce ROS production in human HaCaT skin keratinocytes: the role of xanthine oxidase and NADH dehydrogenase. Nanoscale. 2018;10(25):11820–11830. doi:10.1039/C8NR02933D

310. Pan M, Zhang Y, Shan C, Zhang X, Gao G, Pan B. Flat Graphene-Enhanced Electron Transfer Involved in Redox Reactions. Environ Sci Technol. 2017;51(15):8597–8605. doi:10.1021/acs.est.7b01762

311. Liu S, Yang MQ, Xu YJ. Surface charge promotes the synthesis of large, flat structured graphene-(CdS nanowire)-TiO2 nanocomposites as versatile visible light photocatalysts. J Mater Chem a Mater. 2014;2(2):430–440. doi:10.1039/c3ta13892e

312. He Y, Del Valle A, Qian Y, Huang YF. Near infrared light-mediated enhancement of reactive oxygen species generation through electron transfer from graphene oxide to iron hydroxide/oxide. Nanoscale. 2017;9(4):1559–1566. doi:10.1039/c6nr08784a

313. Choi CH, Lim HK, Chung MW, et al. Long-range electron transfer over graphene-based catalyst for high-performing oxygen reduction reactions: importance of size, n-doping, and metallic impurities. J Am Chem Soc. 2014;136(25):9070–9077. doi:10.1021/ja5033474

314. Liu Y, Xu Y, Geng X, et al. Synergistic Targeting and Efficient Photodynamic Therapy Based on Graphene Oxide Quantum Dot-Upconversion Nanocrystal Hybrid Nanoparticles. Small. 2018;14(19):293. doi:10.1002/smll.201800293

315. Ahirwar S, Mallick S, Bahadur D. Photodynamic therapy using graphene quantum dot derivatives. J Solid State Chem. 2020;282. doi:10.1016/j.jssc.2019.121107

316. Alvarez N, Sevilla A. Current Advances in Photodynamic Therapy (PDT) and the Future Potential of PDT-Combinatorial Cancer Therapies. Int J Mol Sci. 2024;25(2):1023. doi:10.3390/ijms25021023

317. Ge J, Lan M, Zhou B, et al. A graphene quantum dot photodynamic therapy agent with high singlet oxygen generation. Nat Commun. 2014:5. doi:10.1038/ncomms5596

318. Rahimi R, Zargari S, Ghaffarinejad A, Morsali A. Investigation of the synergistic effect of porphyrin photosensitizer on graphene–TiO2nanocomposite for visible light photoactivity improvement. Environ Prog Sustain Energy. 2016;35(3):642–652. doi:10.1002/ep.12267

319. Ghulam AN, Dos Santos OAL, Hazeem L, Backx BP, Bououdina M, Bellucci S. Graphene Oxide (GO) Materials—Applications and Toxicity on Living Organisms and Environment. J Funct Biomater. 2022;13(2):77. doi:10.3390/jfb13020077

320. Li F, Park SJ, Ling D, et al. Hyaluronic acid-conjugated graphene oxide/photosensitizer nanohybrids for cancer targeted photodynamic therapy. J Mater Chem B. 2013;1(12):1678–1686. doi:10.1039/c3tb00506b

321. Cho Y, Kim H, Choi Y. A graphene oxide-photosensitizer complex as an enzyme-activatable theranostic agent. Chem. Commun. 2013;49(12):1202–1204. doi:10.1039/c2cc36297j

322. Liu J, Yuan X, Deng L, et al. Graphene oxide activated by 980 nm laser for cascading two-photon photodynamic therapy and photothermal therapy against breast cancer. Appl Mater Today. 2020:20. doi:10.1016/j.apmt.2020.100665

323. Zaharie-Butucel D, Potara M, Suarasan S, Licarete E, Astilean S. Efficient combined near-infrared-triggered therapy: phototherapy over chemotherapy in chitosan-reduced graphene oxide-IR820 dye-doxorubicin nanoplatforms. J Colloid Interface Sci. 2019;552:218–229. doi:10.1016/j.jcis.2019.05.050

324. Ma M, Cheng L, Zhao A, Zhang H, Zhang A. Pluronic-based graphene oxide-methylene blue nanocomposite for photodynamic/photothermal combined therapy of cancer cells. Photodiagnosis Photodyn Ther. 2020;29. doi:10.1016/j.pdpdt.2019.101640

325. Hosseinzadeh R, Khorsandi K, Hosseinzadeh G. Graphene oxide-methylene blue nanocomposite in photodynamic therapy of human breast cancer. J Biomol Struct Dyn. 2018;36(9):2216–2223. doi:10.1080/07391102.2017.1345698

326. Dos Santos MSC, Gouvêa AL, de Moura LD, et al. Nanographene oxide-methylene blue as phototherapies platform for breast tumor ablation and metastasis prevention in a syngeneic orthotopic murine model. J Nanobiotechnology. 2018;16(1):333. doi:10.1186/s12951-018-0333-6

327. Sahu A, Choi W, Lee JH, Tae G. Graphene oxide mediated delivery of methylene blue for combined photodynamic and photothermal therapy. Biomaterials. 2013;34(26):6239–6248. doi:10.1016/j.biomaterials.2013.04.066

328. Ocsoy I, Isiklan N, Cansiz S, Ozdemir N, Tan W. ICG-Conjugated magnetic graphene oxide for dual photothermal and photodynamic therapy. RSC Adv. 2016;6(36):30285–30292. doi:10.1039/C6RA06798K

329. Zhang X, Luo L, Li L, et al. Trimodal synergistic antitumor drug delivery system based on graphene oxide. Nanomedicine. 2019;15(1):142–152. doi:10.1016/j.nano.2018.09.008

330. Dash BS, Lu YJ, Pejrprim P, Lan YH, Chen JP. Hyaluronic acid-modified, IR780-conjugated and doxorubicin-loaded reduced graphene oxide for targeted cancer chemo/photothermal/photodynamic therapy. Biomaterials Advances. 2022;136. doi:10.1016/j.bioadv.2022.212764

331. Pillar-Little TJ, Wanninayake N, Nease L, Heidary DK, Glazer EC, Kim DY. Superior photodynamic effect of carbon quantum dots through both type I and type II pathways: detailed comparison study of top-down-synthesized and bottom-up-synthesized carbon quantum dots. Carbon N Y. 2018;140:616–623. doi:10.1016/j.carbon.2018.09.004

332. Soumya K, More N, Choppadandi M, Aishwarya DA, Singh G, Kapusetti G. A comprehensive review on carbon quantum dots as an effective photosensitizer and drug delivery system for cancer treatment. Biomedical Technology. 2023;4:11–20. doi:10.1016/j.bmt.2023.01.005

333. Thakur M, Kumawat MK, Srivastava R. Multifunctional graphene quantum dots for combined photothermal and photodynamic therapy coupled with cancer cell tracking applications. RSC Adv. 2017;7(9):5251–5261. doi:10.1039/c6ra25976f

334. Li Y, Wang Y, Shang H, Wu J. Graphene Quantum Dots Modified Upconversion Nanoparticles for Photodynamic Therapy. Int J Mol Sci. 2022;23(20):12558. doi:10.3390/ijms232012558

335. Yang FH, Hua YX, Wang K, et al. Graphene quantum dots (GQDs)-based nanomaterials for improving photodynamic therapy in cancer treatment. Eur J Med Chem. 2019:182. doi:10.1016/j.ejmech.2019.111620

336. Sun J, Xin Q, Yang Y, et al. Nitrogen-doped graphene quantum dots coupled with photosensitizers for one-/two-photon activated photodynamic therapy based on a FRET mechanism. Chem. Commun. 2018;54(7):715–718. doi:10.1039/C7CC08820E

337. Ju J, Regmi S, Fu A, Lim S, Liu Q. Graphene quantum dot based charge‐reversal nanomaterial for nucleus‐targeted drug delivery and efficiency controllable photodynamic therapy. J Biophotonics. 2019;12(6):367. doi:10.1002/jbio.201800367

338. Shi H, Yin Y, Xu H, Qu X, Wang H, An Z. Samarium doped carbon dots for near-infrared photo-therapy. Chem Eng J. 2024;488. doi:10.1016/j.cej.2024.150661

339. Zhang X, Li H, Yi C, et al. Host immune response triggered by graphene quantum-dot-mediated photodynamic therapy for oral squamous cell carcinoma. Int J Nanomed. 2020;15:9627–9638. doi:10.2147/IJN.S276153

340. Li Y, Wu Z, Du D, Dong H, Shi D, Li Y. A graphene quantum dot (GQD) nanosystem with redox-triggered cleavable PEG shell facilitating selective activation of the photosensitiser for photodynamic therapy. RSC Adv. 2016;6(8):6516–6522. doi:10.1039/c5ra23622c

341. Juzenas P, Kleinauskas A, George Luo P, Sun YP. Photoactivatable carbon nanodots for cancer therapy. Appl Phys Lett. 2013;103(6):7787. doi:10.1063/1.4817787

342. Ramachandran P, Khor BK, Lee CY, et al. N-Doped Graphene Quantum Dots/Titanium Dioxide Nanocomposites: a Study of ROS-Forming Mechanisms, Cytotoxicity and Photodynamic Therapy. Biomedicines. 2022;10(2):421. doi:10.3390/biomedicines10020421

343. Choi SY, Baek SH, Chang SJ, et al. Synthesis of upconversion nanoparticles conjugated with graphene oxide quantum dots and their use against cancer cell imaging and photodynamic therapy. Biosens Bioelectron. 2017;93:267–273. doi:10.1016/j.bios.2016.08.094

344. Mangalath S, Saneesh Babu PS, Nair RR, et al. Graphene Quantum Dots Decorated with Boron Dipyrromethene Dye Derivatives for Photodynamic Therapy. ACS Appl Nano Mater. 2021;4(4):4162–4171. doi:10.1021/acsanm.1c00486

345. Du D, Wang K, Wen Y, Li Y, Li YY. Photodynamic Graphene Quantum Dot: reduction Condition Regulated Photoactivity and Size Dependent Efficacy. ACS Appl Mater Interfaces. 2016;8(5):3287–3294. doi:10.1021/acsami.5b11154

346. Kholikov K, Ilhom S, Sajjad M, et al. Improved singlet oxygen generation and antimicrobial activity of sulphur-doped graphene quantum dots coupled with methylene blue for photodynamic therapy applications. Photodiagnosis Photodyn Ther. 2018;24:7–14. doi:10.1016/j.pdpdt.2018.08.011

347. Kazantzis KT, Koutsonikoli K, Mavroidi B, et al. Curcumin derivatives as photosensitizers in photodynamic therapy: photophysical properties and: in vitro studies with prostate cancer cells. Photochem Photobiol Sci. 2020;19(2):193–206. doi:10.1039/c9pp00375d

348. Arvapalli DM, Sheardy AT, Allado K, Chevva H, Yin Z, Wei J. Design of Curcumin Loaded Carbon Nanodots Delivery System: enhanced Bioavailability, Release Kinetics, and Anticancer Activity. ACS Appl Bio Mater. 2020;3(12):8776–8785. doi:10.1021/acsabm.0c01144

349. De D, Das CK, Mandal D, et al. Curcumin Complexed with Graphene Derivative for Breast Cancer Therapy. ACS Appl Bio Mater. 2020;3(9):6284–6296. doi:10.1021/acsabm.0c00771

350. Ghanbari N, Salehi Z, Khodadadi AA, Shokrgozar MA, Saboury AA, Farzaneh F. Tryptophan-functionalized graphene quantum dots with enhanced curcumin loading capacity and pH-sensitive release. J Drug Deliv Sci Technol. 2021;61. doi:10.1016/j.jddst.2020.102137

351. Gazzi A, Fusco L, Khan A, et al. Photodynamic therapy based on graphene and MXene in cancer theranostics. Front Bioeng Biotechnol. 2019;7(OCT):295. doi:10.3389/fbioe.2019.00295

352. Agostinis P, Berg K, Cengel KA, et al. Photodynamic therapy of cancer: an update. CA Cancer J Clin. 2011;61(4):250–281. doi:10.3322/caac.20114

353. Jiang BP, Hu LF, Shen XC, et al. One-step preparation of a water-soluble carbon nanohorn/phthalocyanine hybrid for dual-modality photothermal and photodynamic therapy. ACS Appl Mater Interfaces. 2014;6(20):18008–18017. doi:10.1021/am504860c

354. Gao C, Dong P, Lin Z, et al. Near-Infrared Light Responsive Imaging-Guided Photothermal and Photodynamic Synergistic Therapy Nanoplatform Based on Carbon Nanohorns for Efficient Cancer Treatment. Chem a Eur J. 2018;24(49):12827–12837. doi:10.1002/chem.201802611

355. Yang J, Hou M, Sun W, et al. Sequential PDT and PTT Using Dual-Modal Single-Walled Carbon Nanohorns Synergistically Promote Systemic Immune Responses against Tumor Metastasis and Relapse. Adv. Sci. 2020;7(16):88. doi:10.1002/advs.202001088

356. Curcio M, Cirillo G, Saletta F, et al. Carbon Nanohorns as Effective Nanotherapeutics in Cancer Therapy. C (Basel). 2020;7(1):3. doi:10.3390/c7010003

357. Gao C, Jian J, Luo L, et al. Single-walled carbon nanohorns-based smart nanotheranostic: from phototherapy to enzyme-activated fluorescence imaging-guided photodynamic therapy. J Colloid Interface Sci. 2022;628:273–286. doi:10.1016/j.jcis.2022.07.168

358. Son KH, Hong JH, Lee JW. Carbon nanotubes as cancer therapeutic carriers and mediators. Int J Nanomed. 2016;11:5163–5185. doi:10.2147/IJN.S112660

359. Wang L, Shi J, Liu R, et al. Photodynamic effect of functionalized single-walled carbon nanotubes: a potential sensitizer for photodynamic therapy. Nanoscale. 2014;6(9):4642–4651. doi:10.1039/c3nr06835h

360. Xiao H, Zhu B, Wang D, et al. Photodynamic effects of chlorin e6 attached to single wall carbon nanotubes through noncovalent interactions. Carbon N Y. 2012;50(4):1681–1689. doi:10.1016/j.carbon.2011.12.013

361. Marangon I, Ménard-Moyon C, Silva AKA, Bianco A, Luciani N, Gazeau F. Synergic mechanisms of photothermal and photodynamic therapies mediated by photosensitizer/carbon nanotube complexes. Carbon N Y. 2016;97:110–123. doi:10.1016/j.carbon.2015.08.023

362. Li H, Zhang N, Hao Y, Wang Y, Jia S, Zhang H. Enhancement of curcumin antitumor efficacy and further photothermal ablation of tumor growth by single-walled carbon nanotubes delivery system in vivo. Drug Deliv. 2019;26(1):1017–1026. doi:10.1080/10717544.2019.1672829

363. Arellano LM, Gobeze HB, Gómez-Escalonilla MJ, Fierro JLG, D’Souza F, Langa F. Triplet photosensitizer-nanotube conjugates: synthesis, characterization and photochemistry of charge stabilizing, palladium porphyrin/carbon nanotube conjugates. Nanoscale. 2020;12(17):9890–9898. doi:10.1039/d0nr02136a

364. Zhu Z, Tang Z, Phillips JA, Yang R, Wang H, Tan W. Regulation of singlet oxygen generation using single-walled carbon nanotubes. J Am Chem Soc. 2008;130(33):10856–10857. doi:10.1021/ja802913f

365. Hamblin MR. Fullerenes as photosensitizers in photodynamic therapy: pros and cons. Photochem Photobiol Sci. 2018;17(11):1515–1533. doi:10.1039/c8pp00195b

366. Mroz P, Tegos GP, Gali H, Wharton T, Sarna T, Hamblin MR. Photodynamic therapy with fullerenes. Photochem Photobiol Sci. 2007;6(11):1139–1149. doi:10.1039/b711141j

367. Huang YY, Sharma SK, Yin R, Agrawal T, Chiang LY, Hamblin MR. Functionalized fullerenes in photodynamic therapy. J Biomed Nanotechnol. 2014;10(9):1918–1936. doi:10.1166/jbn.2014.1963

368. Yano S, Hirohara S, Obata M, et al. Current states and future views in photodynamic therapy. J Photochem Photobiol C Photochem Rev. 2011;12(1):46–67. doi:10.1016/j.jphotochemrev.2011.06.001

369. Hamano T, Okuda K, Mashino T, et al. Singlet Oxygen Production from Fullerene Derivatives: effect of Sequential Functionalization of the Fullerene Core. J Mol Modeling. 1997;12.

370. Castro E, Cerón MR, Garcia AH, et al. A new family of fullerene derivatives: fullerene-curcumin conjugates for biological and photovoltaic applications. RSC Adv. 2018;8(73):41692–41698. doi:10.1039/c8ra08334g

371. Gündüz EÖ, Gedik ME, Günaydın G, Okutan E. Amphiphilic Fullerene-BODIPY Photosensitizers for Targeted Photodynamic Therapy. ChemMedChem. 2022;17(6):693. doi:10.1002/cmdc.202100693

372. Tokuyama H, Yamago S, Nakamura E, Shiraki T, Sugiura Y. Photoinduced biochemical activity of fullerene carboxylic acid. J Am Chem Soc. 1993;115(17):7918–7919. doi:10.1021/ja00070a064

373. Burlaka AP, Sidorik YP, Prylutska SV, et al. Catalytic system of the reactive oxygen species on the C60 fullerene basis. Exp Oncol. 2004;26(4):326–327.

374. Rancan F, Rosan S, Boehm F, et al. Cytotoxicity and photocytotoxicity of a dendritic C(60) mono-adduct and a malonic acid C(60) tris-adduct on Jurkat cells. J Photochem Photobiol B. 2002;67(3):157–162. doi:10.1016/s1011-1344(02)00320-2

375. Sugikawa K, Masuda K, Kozawa K, Kawasaki R, Ikeda A. Fullerene–porphyrin hybrid nanoparticles that generate activated oxygen by photoirradiation. RSC Adv. 2021;11(3):1564–1568. doi:10.1039/D0RA09387D

376. Tabata Y, Murakami Y, Ikada Y. Photodynamic effect of polyethylene glycol-modified fullerene on tumor. Jpn J Cancer Res. 1997;88(11):1108–1116. doi:10.1111/j.1349-7006.1997.tb00336.x

377. Kop TJ, Bjelaković MS, Živković L, Žekić A, Milić DR. Stable colloidal dispersions of fullerene C60, curcumin and C60-curcumin in water as potential antioxidants. Colloids Surf a Physicochem Eng Asp. 2022;648. doi:10.1016/j.colsurfa.2022.129379

378. Liu J, Ohta S, Sonoda A, et al. Preparation of PEG-conjugated fullerene containing Gd3+ ions for photodynamic therapy. J Control Release. 2007;117(1):104–110. doi:10.1016/j.jconrel.2006.10.008

379. Milanesio ME, Alvarez MG, Rivarola V, Silber JJ, Durantini EN. Porphyrin‐fullerene C60 Dyads with High Ability to Form Photoinduced Charge‐separated State as Novel Sensitizers for Photodynamic Therapy. Photochem Photobiol. 2005;81(4):891–897. doi:10.1111/j.1751-1097.2005.tb01459.x

380. Miki K, Dan Zhang Z, Kaneko K, et al. Amphiphilic γ-cyclodextrin-fullerene complexes with photodynamic activity. Mater Adv. 2022;3(1):312–317. doi:10.1039/d1ma00743b

381. Sugikawa K, Kozawa K, Ueda M, Ikeda A. Size controlled fullerene nanoparticles prepared by guest exchange of γ-cyclodextrin complexes in water. RSC Adv. 2016;6(78):74696–74699. doi:10.1039/c6ra16513c

382. Sugikawa K, Kozawa K, Ueda M, Ikeda A. Stepwise Growth of Fullerene Nanoparticles through Guest Exchange of γ-Cyclodextrin Complexes in Water. Chem a Eur J. 2017;23(55):13704–13710. doi:10.1002/chem.201701717

383. Sugikawa K, Inoue Y, Kozawa K, Ikeda A. Introduction of Fullerenes into Hydrogels via Formation of Fullerene Nanoparticles. ChemNanoMat. 2018;4(7):682–687. doi:10.1002/cnma.201800143

384. Li Q, Huang C, Liu L, Hu R, Qu J. Enhancing Type I Photochemistry in Photodynamic Therapy Under Near Infrared Light by Using Antennae–Fullerene Complexes. Cytometry Part A. 2018;93(10):997–1003. doi:10.1002/cyto.a.23596

385. Lu YJ, Lin CW, Yang HW, et al. Biodistribution of PEGylated graphene oxide nanoribbons and their application in cancer chemo-photothermal therapy. Carbon N Y. 2014;74:83–95. doi:10.1016/j.carbon.2014.03.007

386. Sun CL, Chang CT, Lee HH, et al. Microwave-assisted synthesis of a core-shell MWCNT/GONR heterostructure for the electrochemical detection of ascorbic acid, dopamine, and uric acid. ACS Nano. 2011;5(10):7788–7795. doi:10.1021/nn2015908

387. Warszyńska M, Repetowski P, Dąbrowski JM. Photodynamic therapy combined with immunotherapy: recent advances and future research directions. Coord Chem Rev. 2023;495. doi:10.1016/j.ccr.2023.215350

388. Shen L, Zhou T, Fan Y, et al. Recent progress in tumor photodynamic immunotherapy. Chin. Chem. Lett. 2020;31(7):1709–1716. doi:10.1016/j.cclet.2020.02.007

389. Dermani FK, Samadi P, Rahmani G, Kohlan AK, Najafi R. PD‐1/PD‐L1 immune checkpoint: potential target for cancer therapy. J Cell Physiol. 2019;234(2):1313–1325. doi:10.1002/jcp.27172

390. Nagaya T, Friedman J, Maruoka Y, et al. Host immunity following near-infrared photoimmunotherapy is enhanced with PD-1 checkpoint blockade to eradicate established antigenic tumors. Cancer Immunol Res. 2019;7(3):401–413. doi:10.1158/2326-6066.CIR-18-0546

391. Wang L, Wang M, Zhou B, et al. PEGylated reduced-graphene oxide hybridized with Fe3O4 nanoparticles for cancer photothermal-immunotherapy. J Mater Chem B. 2019;7(46):7406–7414. doi:10.1039/C9TB00630C

392. Wu C, Guan X, Xu J, et al. Highly efficient cascading synergy of cancer photo-immunotherapy enabled by engineered graphene quantum dots/photosensitizer/CpG oligonucleotides hybrid nanotheranostics. Biomaterials. 2019;205:106–119. doi:10.1016/j.biomaterials.2019.03.020

393. Zhao M, Li Z, Yu C, Sun Q, Wang K, Xie Z. Clinically approved carbon nanoparticles for enhanced photothermal-immunotherapy toward cancer metastasis. Chem Eng J. 2024;482. doi:10.1016/j.cej.2024.149039

394. Zhou F, Wang M, Luo T, Qu J, Chen WR. Photo-activated chemo-immunotherapy for metastatic cancer using a synergistic graphene nanosystem. Biomaterials. 2021;265. doi:10.1016/j.biomaterials.2020.120421

395. Sawy AM, Barhoum A, Abdel Gaber SA, et al. Insights of doxorubicin loaded graphene quantum dots: synthesis, DFT drug interactions, and cytotoxicity. Mater Sci Eng C. 2021:122. doi:10.1016/j.msec.2021.111921

396. Fiorillo M, Verre AF, Iliut M, et al. Graphene Oxide Selectively Targets Cancer Stem Cells, across Multiple Tumor Types: implications for Non-Toxic Cancer Treatment, via “Differentiation-Based Nano-Therapy”. Available from: www.impactjournals.com/oncotarget/. Accessed June 1, 2024.

397. Qin W, Huang G, Chen Z, Zhang Y. Nanomaterials in targeting cancer stem cells for cancer therapy. Front Pharmacol. 2017;8(JAN):1. doi:10.3389/fphar.2017.00001

398. Tian B, Wang C, Zhang S, Feng L, Liu Z. Photothermally Enhanced Photodynamic Therapy Delivered by Nano-Graphene Oxide. ACS Nano. 2011;5(9):7000–7009. doi:10.1021/nn201560b

399. Gurunathan S, Han JW, Park JH, et al. Reduced graphene oxide-silver nanoparticle nanocomposite: a potential anticancer nanotherapy. Int J Nanomed. 2015;10:6257–6276. doi:10.2147/IJN.S92449

400. Burke AR, Singh RN, Carroll DL, et al. The resistance of breast cancer stem cells to conventional hyperthermia and their sensitivity to nanoparticle-mediated photothermal therapy. Biomaterials. 2012;33(10):2961–2970. doi:10.1016/j.biomaterials.2011.12.052

401. Qi L, Pan T, Ou L, et al. Biocompatible nucleus-targeted graphene quantum dots for selective killing of cancer cells via DNA damage. Commun Biol. 2021;4(1):1713. doi:10.1038/s42003-021-01713-1

402. Luo C, Li Y, Guo L, et al. Graphene Quantum Dots Downregulate Multiple Multidrug-Resistant Genes via Interacting with Their C-Rich Promoters. Adv Healthc Mater. 2017;6(21):328. doi:10.1002/adhm.201700328

403. Tabish TA, Narayan RJ. Mitochondria-targeted graphene for advanced cancer therapeutics. Acta Biomater. 2021;129:43–56. doi:10.1016/j.actbio.2021.04.054

404. Wang T, Zhang H, Han Y, et al. Light-Enhanced O 2 -Evolving Nanoparticles Boost Photodynamic Therapy to Elicit Antitumor Immunity. ACS Appl Mater Interfaces. 2019;11(18):16367–16379. doi:10.1021/acsami.9b03541

405. Liang R, Liu L, He H, et al. Oxygen-boosted immunogenic photodynamic therapy with gold nanocages@manganese dioxide to inhibit tumor growth and metastases. Biomaterials. 2018;177:149–160. doi:10.1016/j.biomaterials.2018.05.051

406. Turubanova VD, Mishchenko TA, Balalaeva IV, et al. Novel porphyrazine-based photodynamic anti-cancer therapy induces immunogenic cell death. Sci Rep. 2021;11(1):354. doi:10.1038/s41598-021-86354-4

407. Krysko O, Aaes TL, Kagan VE, et al. Necroptotic cell death in anti-cancer therapy. Immunol Rev. 2017;280(1):207–219. doi:10.1111/imr.12583

408. Wang Y, Hao F, Nan Y, et al. PKM2 inhibitor shikonin overcomes the cisplatin resistance in bladder cancer by inducing necroptosis. Int J Biol Sci. 2018;14(13):1883–1891. doi:10.7150/ijbs.27854

409. Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23(16 REV. ISS. 2):2825–2837. doi:10.1038/sj.onc.1207528

410. Mishchenko T, Balalaeva I, Gorokhova A, Vedunova M, Krysko DV. Which cell death modality wins the contest for photodynamic therapy of cancer? Cell Death Dis. 2022;13(5):455. doi:10.1038/s41419-022-04851-4

411. Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486–541. doi:10.1038/s41418-017-0012-4

412. Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour immunity. Nat Rev Cancer. 2006;6(7):535–545. doi:10.1038/nrc1894

413. Alzeibak R, Mishchenko TA, Shilyagina NY, Balalaeva IV, Vedunova MV, Krysko DV. Targeting immunogenic cancer cell death by photodynamic therapy: past, present and future. J Immunother Cancer. 2021;9(1):1926. doi:10.1136/jitc-2020-001926

414. Kessel D, Reiners JJ. Effects of Combined Lysosomal and Mitochondrial Photodamage in a Non-small-Cell Lung Cancer Cell Line: the Role of Paraptosis. Photochem Photobiol. 2017;93(6):1502–1508. doi:10.1111/php.12805

415. Kessel D. Exploring Modes of Photokilling by Hypericin. Photochem Photobiol. 2020;96(5):1101–1104. doi:10.1111/php.13275

416. Kessel D. Apoptosis, Paraptosis and Autophagy: death and Survival Pathways Associated with Photodynamic Therapy. Photochem Photobiol. 2019;95(1):119–125. doi:10.1111/php.12952

417. Seo MJ, Lee DM, Kim IY, et al. Gambogic acid triggers vacuolization-associated cell death in cancer cells via disruption of thiol proteostasis. Cell Death Dis. 2019;10(3):1360. doi:10.1038/s41419-019-1360-4

418. Hu B, Elinav E, Huber S, et al. Inflammation-induced tumorigenesis in the colon is regulated by caspase-1 and NLRC4. Proc Natl Acad Sci. 2010;107(50):21635–21640. doi:10.1073/pnas.1016814108

419. cheng ZC, Guang LC, feng WY, et al. Chemotherapeutic paclitaxel and cisplatin differentially induce pyroptosis in A549 lung cancer cells via caspase-3/GSDME activation. Apoptosis. 2019;24(3–4):312–325. doi:10.1007/s10495-019-01515-1

420. Wang Y, Gao W, Shi X, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547(7661):99–103. doi:10.1038/nature22393

421. Erkes DA, Cai W, Sanchez IM, et al. Mutant BRAF and MEK inhibitors regulate the tumor immune microenvironment via pyroptosis. Cancer Discov. 2020;10(2):255–269. doi:10.1158/2159-8290.CD-19-0672

422. Yang D, He Y, Muñoz-Planillo R, Liu Q, Núñez G. Caspase-11 Requires the Pannexin-1 Channel and the Purinergic P2X7 Pore to Mediate Pyroptosis and Endotoxic Shock. Immunity. 2015;43(5):923–932. doi:10.1016/j.immuni.2015.10.009

423. Wu M, Liu X, Chen H, et al. Activation of Pyroptosis by Membrane-Anchoring AIE Photosensitizer Design: new Prospect for Photodynamic Cancer Cell Ablation. Angewandte Chemie - Int Ed. 2021;60(16):9093–9098. doi:10.1002/anie.202016399

424. Zhu JX, Zhu WT, Hu JH, et al. Curcumin-Loaded Poly(L-lactide-co-glycolide) Microbubble-Mediated Sono-photodynamic Therapy in Liver Cancer Cells. Ultrasound Med Biol. 2020;46(8):2030–2043. doi:10.1016/j.ultrasmedbio.2020.03.030

425. Li L, Song D, Qi L, et al. Photodynamic therapy induces human esophageal carcinoma cell pyroptosis by targeting the PKM2/caspase-8/caspase-3/GSDME axis. Cancer Lett. 2021;520:143–159. doi:10.1016/j.canlet.2021.07.014

426. Zhou B, yuan ZJ, Shuo LX, et al. Tom20 senses iron-activated ROS signaling to promote melanoma cell pyroptosis. Cell Res. 2018;28(12):1171–1185. doi:10.1038/s41422-018-0090-y

427. Soriano J, Mora-Espí I, Alea-Reyes ME, et al. Cell death mechanisms in Tumoral and Non-Tumoral human cell lines triggered by photodynamic treatments: apoptosis, necrosis and parthanatos. Sci Rep. 2017;7. doi:10.1038/srep41340

428. Nayak KB, Sajitha IS, Kumar TRS, Chakraborty S. Ecotropic viral integration site 1 promotes metastasis independent of epithelial mesenchymal transition in colon cancer cells article. Cell Death Dis. 2018;9(2):36. doi:10.1038/s41419-017-0036-1

429. Chiu LY, Ho FM, Shiah SG, Chang Y, Lin WW. Oxidative stress initiates DNA damager MNNG-induced poly(ADP-ribose) polymerase-1-dependent parthanatos cell death. Biochem Pharmacol. 2011;81(3):459–470. doi:10.1016/j.bcp.2010.10.016

430. Van der Meeren L, Verduijn J, Krysko DV, Skirtach AG. AFM Analysis Enables Differentiation between Apoptosis, Necroptosis, and Ferroptosis in Murine Cancer Cells. iScience. 2020;23(12):1816. doi:10.1016/j.isci.2020.101816

431. Moreno-Gonzalez G, Vandenabeele P, Krysko DV. Necroptosis: a novel cell death modality and its potential relevance for critical care medicine. Am J Respir Crit Care Med. 2016;194(4):415–428. doi:10.1164/rccm.201510-2106CI

432. Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19(7):405–414. doi:10.1038/s41568-019-0149-1

433. Efimova I, Catanzaro E, Van Der Meeren L, et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J Immunother Cancer. 2020;8(2):1369. doi:10.1136/jitc-2020-001369

434. Tang D, Kang R, Berghe T, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29(5):347–364. doi:10.1038/s41422-019-0164-5

435. Wu T, Wang X, Cheng J, et al. Nitrogen-doped graphene quantum dots induce ferroptosis through disrupting calcium homeostasis in microglia. Part Fibre Toxicol. 2022;19(1):464. doi:10.1186/s12989-022-00464-z

436. Wu T, Liang X, Liu X, et al. Induction of ferroptosis in response to graphene quantum dots through mitochondrial oxidative stress in microglia. Part Fibre Toxicol. 2020;17(1):363. doi:10.1186/s12989-020-00363-1

437. Oleinick NL, Morris RL, Belichenko I. The role of apoptosis in response to photodynamic therapy: what, where, why, and how. Photochem Photobiol Sci. 2002;1(1):1–21. doi:10.1039/b108586g

438. Tan L, Shen X, He Z, Lu Y. The Role of Photodynamic Therapy in Triggering Cell Death and Facilitating Antitumor Immunology. Front Oncol. 2022;12. doi:10.3389/fonc.2022.863107

439. Igney FH, Krammer PH. Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer. 2002;2(4):277–288. doi:10.1038/nrc776

440. Wu S, Xing D. Mechanism of mitochondrial membrane permeabilization during apoptosis under Photofrin-mediated photodynamic therapy. J Xray Sci Technol. 2012;20:363–372. doi:10.3233/XST-2012-344

441. Aguilar Cosme JR, Bryant HE, Claeyssens F. Carbon dot-protoporphyrin IX conjugates for improved drug delivery and bioimaging. PLoS One. 2019;14(7):210. doi:10.1371/journal.pone.0220210

442. Klimenko IV, Trusova EA, Shchegolikhin AN, Lobanov AV, Jurina LV. Surface modification of graphene sheets with aluminum phthalocyanine complex. Fuller Nanotub Car Nanostruct. 2022;30(1):133–139. doi:10.1080/1536383X.2021.1976754

443. Wang Y, Staudinger JN, Mindt TL, Gasser G. Theranostics with photodynamic therapy for personalized medicine: to see and to treat. Theranostics. 2023;13(15):5501–5544. doi:10.7150/thno.87363

444. Suvarnaphaet P, Pechprasarn S. Graphene-based materials for biosensors: a review. Sensors (Switzerland). 2017;17(10):2161. doi:10.3390/s17102161

445. Xue M, Mackin C, Weng WH, et al. Integrated biosensor platform based on graphene transistor arrays for real-time high-accuracy ion sensing. Nat Commun. 2022;13(1):749. doi:10.1038/s41467-022-32749-4

446. Yildiz G, Bolton-Warberg M, Awaja F. Graphene and graphene oxide for bio-sensing: general properties and the effects of graphene ripples. Acta Biomater. 2021;131:62–79. doi:10.1016/j.actbio.2021.06.047

447. Zhang W, Du Y, Wang ML. Noninvasive glucose monitoring using saliva nano-biosensor. Sens Biosensing Res. 2015;4:23–29. doi:10.1016/j.sbsr.2015.02.002

448. Li Q, Wang Q, Yang X, Wang K, Zhang H, Nie W. High sensitivity surface plasmon resonance biosensor for detection of microRNA and small molecule based on graphene oxide-gold nanoparticles composites. Talanta. 2017;174:521–526. doi:10.1016/j.talanta.2017.06.048

449. Khalil I, Yehye WA, Julkapli NM, et al. Graphene oxide and gold nanoparticle based dual platform with short DNA probe for the PCR free DNA biosensing using surface-enhanced Raman scattering. Biosens Bioelectron. 2019;131:214–223. doi:10.1016/j.bios.2019.02.028

450. Walther BK, Dinu CZ, Guldi DM, et al. Nanobiosensing with graphene and carbon quantum dots: recent advances. Mater Today. 2020;39:23–46. doi:10.1016/j.mattod.2020.04.008

451. Kong W, Wu D, Xia L, et al. Carbon dots for fluorescent detection of α-glucosidase activity using enzyme activated inner filter effect and its application to anti-diabetic drug discovery. Anal Chim Acta. 2017;973:91–99. doi:10.1016/j.aca.2017.03.050

452. Gulati S, Mansi N, Vijayan S, et al. Magnetic nanocarriers adorned on graphene: promising contrast-enhancing agents with state-of-the-art performance in magnetic resonance imaging (MRI) and theranostics. Mater Adv. 2022;3(7):2971–2989. doi:10.1039/d1ma01071a

453. Zhang M, Liu X, Huang J, et al. Ultrasmall graphene oxide based T1 MRI contrast agent for in vitro and in vivo labeling of human mesenchymal stem cells. Nanomedicine. 2018;14(7):2475–2483. doi:10.1016/j.nano.2017.03.019

454. Antoine C, Sahylí Ortega Pijeira M, Ricci-Junior E, Magalhães Rebelo Alencar L, Santos-Oliveira R. Graphene quantum dots as bimodal imaging agent for X-ray and Computed Tomography. Eur. J. Pharm. Biopharm. 2022;179:74–78. doi:10.1016/j.ejpb.2022.08.020

455. Zhang H, He R, Niu Y, et al. Graphene-enabled wearable sensors for healthcare monitoring. Biosens Bioelectron. 2022:197. doi:10.1016/j.bios.2021.113777

456. Maity A, Pu H, Sui X, et al. Scalable graphene sensor array for real-time toxins monitoring in flowing water. Nat Commun. 2023;14(1):701. doi:10.1038/s41467-023-39701-0

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.