Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Graphene-based nanozymes for revolutionizing biomedical research

Astha Tripathiab, Saumya Singh Rautelab and Pratibha Kumari*b
aDepartment of Chemistry, University of Delhi, New Delhi-110007, India
bBioorganic Material Research Laboratory, Department of Chemistry, Deshbandhu College, University of Delhi, Kalkaji, New Delhi-110019, India. E-mail: pkumari@db.du.ac.in

Received 4th July 2025 , Accepted 3rd October 2025

First published on 17th October 2025


Abstract

Nanozymes, with nanoscale dimensions and enzyme-like properties, have garnered substantial interest as they can overcome the downsides of conventional enzymes, including fragility, high cost, tedious isolation, and expensive manufacturing. Over the past decade, a diverse range of nanomaterials have demonstrated the ability to mimic enzyme-like activity by incorporating multivalent elements into nanostructures. Notably, graphene-based nanomaterials, such as graphene, graphene oxide, and reduced graphene oxide, with their precisely controlled scaffolds and electronic properties, have emerged as promising substitutes for traditional enzymes by emulating the intricately evolved catalytic centers of natural enzymes, including oxidase, peroxidase, catalase, and superoxide dismutase. The distinct electronic, mechanical, thermal, and optical properties enable graphene-based nanozymes to provide multifunctional platforms for various biomedical applications, including wound healing, tissue regeneration, cancer treatment therapies, antibacterial activity, and biosensing applications. This in-depth review examines the enzymatic features and recent advancements of graphene-based nanozymes, highlighting their significant contributions to biomedicine.


1. Introduction

The field of nanomaterials has witnessed unprecedented growth, resulting in the burgeoning of novel materials with exceptional attributes and promising applications across various scientific and technological domains.1 One such intriguing area of focus in nanomaterial research is the utilization of nanozymes. A landmark discovery in 2007 challenged the long-held belief that inorganic materials are biologically inactive. This motivated researchers to investigate the catalytic potential of nanomaterials as “nanozymes”. Nanozymes refer to a new generation of artificial enzymes that have revolutionized the landscape of biomedical research by emulating the function of natural enzymes at the nanoscale.2 These are promising areas of research at the intersection of nanotechnology and biology.3,4

Enzymes are highly efficient biocatalysts, enabling numerous chemical reactions in living organisms and playing a crucial role in various biological processes. Enzymes accelerate biochemical reactions, enabling the efficient operation of various metabolic pathways, cellular processes, and physiological functions. Enzymes exhibit high catalytic activity, high substrate specificity, and selectivity under mild conditions. However, natural enzymes have limitations, such as instability, high cost, tedious purification, and storage, which restrict their widespread technological applications.5–7 Nanozymes, on the other hand, offer several advantages over natural enzymes, such as high stability, low cost, and ease of production. Furthermore, nanozymes can be tuned to have specific catalytic properties, making them extremely versatile and suitable for numerous applications.8–10 A vast array of nanozymes is currently employed in biosensing and therapeutics. Nanozymes are usually metal and metal oxide nanoparticles,11,12 metal–organic frameworks,13 two-dimensional materials,14 inorganic nanomaterials,15 covalent-organic frameworks,16 transition metal-based nanomaterials,17 carbon-based nanomaterials,18 biomolecules,19 and supramolecules.20 These nanozymes exhibit exceptional catalytic activities and are being increasingly recognized for their potential to revolutionize the fields of biosensing and catalytic therapy, as summarized in Table 1.

Table 1 Overview of various conventional nanozymes, their enzymatic activities, and applications in biomedical fields
Nanomaterial Activity Km (H2O2) Vmax (H2O2) Biomedical applications References
HCS@Pt-Ce nanoparticles Peroxidase 0.04853 mM 0.82 × 10−8 M s−1 Photodynamic catalytic tumor therapy 21
Sm-TCPP-Pt/TPP nanosheets Catalase Photodynamic therapy 22
PVP-PtCuNCs Superoxide dismutase, catalase, and peroxidase 9.94 mM (Cat) 26.16 × 10−6 M per s (Cat) Hydroxyl radical scavenging 23
75.55 mM 35.88 × 10−8 M s−1
TiO2 nanotubes@MoS2 nanoflowers Peroxidase 0.085 mM 12.05 × 10−7 M s−1 Wound healing 24
Hemin/graphdiyne (GDY) nanocomposite Peroxidase 6.1 μM 1.14 × 10−7 M s−1 Wound healing, antibacterial activity 25
Ag NC Oxidase 119 μM 214 × 10−9 M s−1 Hg(II) sensing 26
Pt NP Oxidase 0.09 mM 7 × 10−6 M s−1 Heparin detection 27
Pd NC Oxidase Antitumor 28
Au@Pt Oxidase, and peroxidase 0.027 mM 181 × 10−9 M s−1 Immunoassays 29
NiCo2O4 Oxidase, and peroxidase 9.406 mM 25.84 × 10−8 M s−1 Glucose detection 30
CoOOH Phosphatase and oxidase Alkaline phosphatase 31
MoO3 Phosphatase and oxidase 1.6769 mM Acid phosphatase 32
V2O5 Oxidase Glutathione sensing 33
Mn3O4 Oxidase 0.025 mM 5.07 × 10−8 M s−1 Antioxidant activity of phenols 34
Co3O4 Oxidase 0.037 mM 3.20 × 10−8 M s−1 Sulfite detection 35
Ag2O Oxidase Sulfite detection 36
Tb4O7 Oxidase 0.124 mM 4.31 × 10−8 M s−1 Antibacterial 37
IrOx Oxidase and peroxidase 19.27 mM 1.79 × 10−6 M s−1 Antitumor 38
Mixed valence state Ce-MOF Oxidase 0.37 μM 5.5 × 10−6 M s−1 Biothiols sensing 39
PVP-MoS2 Peroxidase 3.66 mM 4.76 × 10−8 M s−1 H2O2 & glucose detection 40
CQDs/Fe3O4 Peroxidase 56.97 mM 5.744 × 10−8 M s−1 H2O2 & ascorbic acid detection 41
VS2NSs Peroxidase 3.49 mM 55.7 × 10−8 M s−1 Glucose detection 42
AuNR@TiO2   0.20 ± 0.02 μM Catalysis 43


The most promising candidates for developing nanozymes are graphene-based materials such as graphene, graphene oxide (GO), and reduced graphene oxide (rGO) (Fig. 1). Graphene and graphene-based materials are extensively studied nanomaterials with exceptional promise for a range of applications, including biomimetics and heterogeneous catalysis, due to their abundant chemical adaptability, remarkable stability, low toxicity, metal-free nature, and notable electrical and optical properties.44 The functionalization of heteroatoms and metallic or bimetallic nanoparticles enables these nanozymes to closely replicate the active sites of specific enzymes, thereby enhancing their catalytic efficiency and selectivity in biochemical reactions. Their robust structure allows them to maintain activity over extended periods, making them more reliable for long-term applications in biomedical fields, including wound healing, cancer cell detection, bioimaging, and therapeutics.45 This capability is crucial in developing biosensors and therapeutic agents that require precise activity under physiological conditions. Apart from stability, graphene-based nanozymes depict enhanced electron transfer kinetics. This property facilitates rapid reactions with reactive oxygen species (ROS), which are often implicated in inflammation and tissue damage during wound healing processes. By effectively scavenging reactive oxygen species (ROS), graphene-based nanozymes can mitigate oxidative stress and promote a more favorable microenvironment for healing.46,47 The multifunctionality of graphene-based nanozymes also sets them apart from other nanozyme materials. They can serve as scaffolds for cell proliferation and migration while providing antimicrobial properties. For example, studies have shown that graphene-reinforced scaffolds enhance fibroblast migration and effectively inhibit bacterial growth, thereby supporting wound closure and preventing infection.48 This dual functionality is less commonly observed in other nanozymes, which may focus solely on antimicrobial action or tissue regeneration but not both. The advantages and disadvantages of graphene-based nanozymes are shown in Fig. 2. By accelerating the oxidation of 3,3′,5,5′-tetramethylbenzidine (TMB) in the presence of hydrogen peroxide (H2O2), graphene-based nanozymes exhibit peroxidase-like activity and produce a blue-colored product. This catalytic process involves activation of oxygen-containing functional groups, particularly carbonyl groups, which serve as active centers for biocatalytic activity. The mechanism begins with the interaction of H2O2 with these functional groups, leading to the generation of reactive species that facilitate the oxidation of TMB.


image file: d5ra04765j-f1.tif
Fig. 1 Structures of (a) graphene, (b) graphene oxide, and (c) reduced graphene oxide.

image file: d5ra04765j-f2.tif
Fig. 2 Advantages and disadvantages of graphene-based nanozymes over conventional enzymes.

Although graphene-based nanozymes do not strictly follow the traditional ping-pong mechanism, they exhibit a similar stepwise process where the activation of the carbonyl bond is crucial for the catalytic cycle. The high surface area and electron transfer capabilities of graphene-based materials enhance their reactivity, enabling them to mimic natural peroxidases effectively.26 The graphene-based nanozymes have created highly sensitive and selective biosensors capable of detecting various biomarkers. In cancer diagnosis and therapy, peroxidase mimetic nanozymes have been extensively studied for their role in catalytic cancer therapy, complementing traditional treatments such as chemotherapy, phototherapy, and immunotherapy.49,50 The application of nanozymes in cancer treatment is multifaceted. They can kill cancer cells directly by enhancing reactive oxygen species (ROS) levels or indirectly by depleting ROS, thus exploiting the tumor microenvironment's characteristics, such as hypoxia. Nanozymes' catalytic activity is influenced by various factors, including pH, substrate concentration, and temperature, which can be optimized for specific therapeutic outcomes.

Recent advancements in graphene-based nanozymes have expanded their applications to include the detection of glucose, dopamine, cholesterol, and various disease-related biomolecules. These sensors often employ functionalization techniques to enhance their performance, using both covalent and non-covalent approaches to modify graphene for specific sensing applications.51 For instance, graphene oxide functionalization has yielded graphene-based nanozymes that closely mimic several natural enzymes.52 Graphene-based nanozymes are appealing due to their ease of synthesis and retention of catalytic activity even after prolonged storage at ambient temperature.53 The peroxidase-mimicking activities of graphene oxide and reduced graphene oxide have been widely reported, and they have been applied to the colorimetric detection of H2O2 and other substrates.54 The peroxidase-like activities of rGO have also been selectively enhanced by N-doping in carbon nanomaterials, most likely due to the adjustment of charge density and an increase in active sites. The origin of the peroxidase-mimicking activity of graphene-based nanomaterials remains unclear. Some studies indicate that the carbonyl groups are the active centers among the various oxygen moieties. Several graphene-based hybrids have been developed as nanozymes and applied to construct electrochemical biosensors involving GO-COOH and GO-COOH hybrids.54 Graphene-based nanozymes are at the forefront of innovation in biosensing and catalytic cancer therapy. Their unique properties have opened new avenues for developing sophisticated therapeutic and diagnostic tools. Wang et al. fabricated oxygen-functionalized graphene quantum dots (o-GQDs) that exhibit peroxidase enzymatic activity, surpassing that of other nanozymes.55

Interest in the enzyme-mimicking properties of graphene nanomaterials began to gain traction around 2010, with early speculation and foundational studies highlighting their potential as artificial enzymes. The first report explicitly describing graphene oxide nanosheets as peroxidase mimics appeared in 2010, where carboxyl-modified graphene oxide was shown to catalyze reactions similar to horseradish peroxidase, leading to applications such as glucose biosensing.56 The term “graphene nanozyme” was first introduced in the literature in 2011, marking a formal recognition of this new class of catalytic nanomaterials. Following these initial discoveries, research activity in the field was limited, with no publications recorded in 2012. However, after 2015, the number of studies on graphene-based nanozymes increased exponentially, reflecting the growing recognition of their versatility and catalytic efficiency. Functionalization of graphene, such as doping with nitrogen, boron, or metallic nanoparticles, has been shown to significantly enhance its enzyme-like activities, particularly for peroxidase, oxidase, and superoxide dismutase-mimetic reactions, and to improve selectivity and catalytic performance. The rapid expansion of this field is evident in bibliometric data: as of May 27, 2025, a search on CAS SciFinder reveals that there are currently 437 publications related to “graphene” and “nanozymes,” including 388 research articles, 25 reviews, and several conference and editorial pieces. Of these, 118 specifically address applications in sensing, underscoring both the breadth of research and the diverse, promising prospects of graphene-based nanozymes in biosensing, catalysis, and environmental remediation. This trajectory highlights the transition of graphene nanozymes from a speculative concept to a vibrant and multidisciplinary research area (Fig. 3).


image file: d5ra04765j-f3.tif
Fig. 3 Number of publications in journals including the keywords “graphene, nanozyme” and “graphene, nanozyme + sensing” from 2011 to 2025 (accessed on May 27, 2025 using CAS SciFinder search tool).

The objective of this review is to provide an in-depth understanding of the synthesis as well as applications of graphene-based nanozymes in wound healing, biosensing, antibacterial, and catalytic therapy. By examining the enzymatic characteristics, recent advancements, and innovative strategies employed in the development of graphene-based nanozymes, this review aims to elucidate their pivotal role in revolutionizing the field of biomedical research.

2. Synthesis of graphene and graphene-based nanozymes

Graphene and graphene-based nanozymes can be synthesized using several methods (Fig. 4), each offering distinctive advantages and applications. The different synthetic techniques can be precisely regulated to confer properties for targeted and intended applications.44 The bottom-up approach is often used for graphene synthesis, involving smaller molecules such as carbon atoms or hydrocarbons, through a series of chemical reactions. This method enables the control of graphene's morphology, crystallinity, and structure by adjusting the chemical composition and reaction conditions. The bottom-up approach is appealing for producing high-quality graphene with tailored characteristics, including excellent and consistent thickness. This method employs techniques such as chemical vapor deposition (CVD) and pyrolysis, which utilize carbon precursors in a gaseous form for deposition onto a substrate. On the contrary, the top-down approach produces graphene flakes or sheets by mechanically or chemically exfoliating graphite, a common precursor to graphene. This method is often used for bulk graphene production due to its cost-effectiveness and ease of scaling up. The top-down approach can result in a broader range of graphene morphologies and properties, including polycrystalline films and flakes of varying sizes. This method involves chemical oxidation-reduction, liquid-phase exfoliation (LPE), and electrochemical exfoliation. This method simplifies the process by directly converting graphite to graphene. Still, it can lead to challenges such as surface defects during sheet separation and the re-agglomeration of separated sheets. Each approach has its advantages and disadvantages, and the preferred method often depends on the intended application and the specific properties of the graphene.
image file: d5ra04765j-f4.tif
Fig. 4 Various synthesis methods to produce graphene-based nanozymes.

Chemical methods, such as Hummer's method, are commonly used to produce graphene oxide, providing a versatile platform for developing nanomaterials with inherent enzymatic properties. This method involves the oxidation of graphite using a mixture of concentrated sulfuric acid, sodium nitrate, and potassium permanganate, followed by sonication and filtration.57 Hummer's method was further modified to enhance the nanozymatic activity. Kim et al. synthesized Fe–N-rGO utilizing modified Hummer's method for the H2O2 detection.58 Kumari et al. also synthesized graphene oxide using a modified Hummer's method.59,60 The modified Hummers' method offers significant advantages over conventional methods, primarily by eliminating sodium nitrate. This change is crucial as it can avoid the generation of toxic gases such as nitrogen dioxide (NO2) and dinitrogen tetroxide (N2O4). Additionally, altered reactant ratios were employed to oxidize graphite to graphene oxide, such as increased amounts of KMnO4 or other oxidizing agents, like potassium persulfate, which is often used to enhance oxidation efficiency and yield. Another modification involved the use of phosphoric and sulfuric acid, which improved oxidation efficiency and safety.61 Phosphoric acid acts as an auxiliary oxidant, enhancing the oxidative chemical exfoliation of graphite when used in conjunction with sulfuric acid. This synergistic effect enables more effective intercalation and oxidation of graphite, resulting in a higher yield of graphene oxide. It also helps maintain a strong acidic environment, crucial for activating potassium permanganate, the primary oxidizing agent used in the process. This results in a more efficient oxidation reaction than methods that rely solely on sulfuric acid. This modification makes the process safer and more environmentally friendly.62 Lu et al. fabricated a surface-modified graphene-based hybrid as nanozymes, using the modified Hummer's method to synthesize graphene oxide.63 Another method commonly used is the hydrothermal approach, primarily employed for synthesizing reduced graphene oxide. The degree of reduction is controlled by temperature; higher temperatures facilitate a more rapid reduction of graphene oxide.64 With this technique, metal precursors (such as Fe3+ or Pt2+) react with graphene-based nanomaterials in an aqueous medium at high pressure and temperature to produce graphene-based nanozymes. This method often yields metal–graphene composites that exhibit excellent catalytic activity.65,66

Li et al. developed gold adsorbed on graphene sheets to exhibit peroxidase-mimic activity, which was synthesized using a pot-hydrothermal method.67 The reduction method is another top-down approach for synthesizing graphene-based nanozymes. Chemical reduction, thermal reduction, and electrochemical reduction are the approaches to synthesizing graphene-based nanozymes. The most prevalent method is the chemical reduction method, which involves reducing agents like hydrazine, sodium borohydride, ascorbic acid, and various plant extracts. This process effectively removes oxygen functional groups from the graphene oxide structures, converting them into reduced graphene oxide (rGO). The resulting rGO exhibits enhanced catalytic activity, resembling the behavior of nanozymes, which expands its potential applications across diverse fields. Liu et al. employed a green approach to synthesize a graphene-based nanozyme exhibiting peroxidase-like activity, which is also used for detecting L-cysteine.68 In their study, polysaccharides served as the reducing agent for graphene oxide, facilitating an environmentally friendly reduction process. This innovative method enhances the catalytic properties of the resulting nanozyme, underscoring the potential of using biocompatible materials to synthesize functional nanomaterials for various applications. The thermal reduction method is another approach to reducing graphene oxide, which involves heating it to high temperatures in a controlled environment. The significance of this process lies in the elimination of oxygen functional groups and the re-establishment of the sp2 carbon network, resulting in the formation of reduced graphene oxide (rGO), the active nanozyme form of graphene oxide. This is typically done inside the furnace, purged with an inert gas, and offers the most significant degree of freedom in terms of adjusting and maintaining the heating temperature and duration required to develop the desired enzyme-like properties. Careful control of such parameters is essential primarily to enhance the catalytic activity of the resulting rGO, making it beneficial for various applications, including catalysis and biosensing. Another well-known approach for synthesizing graphene-based nanozymes is leveraging graphene's unique properties for catalytic purposes. This technique is classified into solid, liquid, and electrochemical exfoliation.69 In electrochemical exfoliation, a direct voltage is applied to graphite in an electrolyte solution. Individual graphene sheets are expanded and exfoliated by gaseous species that are generated when ionic species are forced to intercalate into the graphite layers. This process has advantages in scalability and efficiency, allowing for the production of high-quality graphene with minimal defects. Furthermore, exfoliating graphene in the liquid phase is both cost-effective and environmentally friendly. Expensive and toxic solvents are avoided in this process. The resulting graphene sheets can be modified to enhance their catalytic properties, emulating those of enzymes, making them suitable for a wide range of applications.

One of the prominent methods for synthesizing graphene-based nanomaterials is solvothermal synthesis, which involves reacting precursors in a solvent at elevated temperatures and pressures. This method facilitates the formation of functionalized graphene-based nanozymes while offering scalability and versatility. However, it can lead to agglomeration effects that may affect performance. For instance, Li et al. fabricated Fe–N-GQD using a green and facile solvothermal approach.70 This process treated the precursor with DMF, and the resulting solution was transferred to a PTFE liner. It was then heated at 180 °C for 9 hours. After synthesis, the product underwent purification through multiple cycles of centrifugation and dialysis. Borthakur et al. also employed the solvothermal technique to synthesize CuS and NiS nanoparticle-decorated porous reduced graphene oxide sheets, which demonstrated peroxidase-mimicking activity. They further utilized this for the detection of Hg2+ ions.71

Chemical vapor deposition (CVD) is a widely adopted bottom-up approach for synthesizing graphene-based nanozymes, as it produces high-quality graphene with controlled properties.72 In CVD, carbon-containing gases, such as methane, are decomposed at high temperatures in the presence of a substrate, typically copper or nickel, which facilitates the growth of graphene layers. This method enables precise control over layer thickness and doping levels, making it suitable for creating functionalized graphene that can enhance enzyme-mimicking activity. One of the significant advantages of CVD is its scalability; it can produce large-area graphene films essential for various applications, including nanozymes. Furthermore, CVD can incorporate dopants, such as nitrogen, during the growth process, which can tailor the electronic properties of graphene and enhance its enzymatic performance. For instance, nitrogen-doped graphene synthesized via CVD has demonstrated enhanced charge carrier mobility and stability, making it an excellent candidate for nanozyme applications.73,74 Additionally, CVD enables the synthesis of graphene in a relatively clean environment, minimizing impurities that could affect the performance of the resulting nanozymes. However, challenges remain, such as achieving uniformity across large areas and managing production costs. CVD is a powerful technique for developing advanced graphene-based nanozymes with promising applications in biosensing and catalysis.

The choice of synthesis method depends on the intended application of the graphene-based nanozymes. Despite its higher cost and complexity, the CVD method is superior for applications that require high-quality materials with precise control over their properties. However, hydrothermal synthesis or electrochemical reduction may be more suitable for larger-scale production where cost-effectiveness is a priority, balancing quality and scalability. Each method has its unique advantages and challenges, making it essential to tailor the synthesis approach to the specific needs of each application.

2.1 Side product formation in graphene-based nanozyme synthesis and its impact on catalytic mechanisms

The synthesis of graphene-based nanozymes inherently involves the formation of various side products and impurities, which arise from the complexity of the chemical processes and the nature of the precursors used.75,76 For example, hydrothermal synthesis employing dopants like urea and boric acid for nitrogen and boron co-doping or graphene oxide combined with zinc acetate dihydrate for ZnO/GO composites often leaves residual unreacted starting materials. These residuals can interfere with the nanozyme's structural uniformity and catalytic properties. Additionally, the formation of multi-layer graphene, such as five-layer graphene observed in nitrogen–boron doped graphene (NB-Gr), and variations in reduction or doping levels introduce structural non-uniformity that affects catalytic site distribution and electron transfer pathways.77

In chemical vapor deposition (CVD), a common side product is amorphous carbon contamination, often called carbon soot, which physically obscures the graphene surface and lacks the ordered structure necessary for catalytic activity. This contamination arises from uncontrolled carbon supply or high defect densities during growth and can be partially mitigated by hydrogen-assisted conversion to graphene. Residual oxidizing impurities like oxygen or moisture in the reactor atmosphere can etch the graphene film, creating defects and oxidized carbon clusters that degrade catalytic performance.78 Moreover, incomplete removal of reaction by-products, unreacted gases, or residual metal catalysts (e.g., nickel, cobalt, copper) used in CVD can remain on the graphene surface, further impacting catalytic efficiency.79

Graphene-based syntheses often suffer from persistent oxygen-containing functional groups (hydroxyl, carboxyl, carbonyl, epoxy) due to incomplete reduction. While some oxygen groups can enhance catalytic activity by providing active sites or improving hydrophilicity, excessive or uncontrolled oxygen functionalities can initiate photodegradation and reduce long-term stability. Reducing agents like hydrazine or sulfur-containing compounds may also leave residual impurities if purification is insufficient.80 These side products have a dual impact on catalytic mechanisms. Detrimental impurities such as amorphous carbon, excessive oxygen groups, or residual metals can block active sites, impede electron transfer, and cause structural degradation, thereby lowering catalytic efficiency and selectivity.81,82 Conversely, intentional heteroatom dopants (nitrogen, boron, sulfur) and integrated transition metals serve as engineered “impurities” that create or enhance active sites, redistribute electron density, and optimize catalytic pathways, significantly boosting nanozyme activity.83

Therefore, the challenge lies in precise impurity engineering—rigorously eliminating harmful side products while selectively introducing beneficial dopants. Advanced characterization techniques like Raman spectroscopy,84 X-ray photoelectron spectroscopy (XPS), and transmission electron microscopy (TEM) are critical for identifying impurity fingerprints, guiding synthesis optimization, and ensuring reproducible catalytic performance. Ultimately, balancing purity with controlled defect and dopant incorporation is essential for designing graphene-based nanozymes with tailored catalytic properties suitable for practical applications, especially in complex biological environments where stability and specificity are paramount85,86

3. Properties, tunability, and active sites of graphene-based nanozymes

Graphene-based nanozymes have emerged as a transformative class of nanomaterials that effectively mimic the activity of natural enzymes, offering tunable properties and diverse enzymatic functions suitable for various applications, including therapeutics and environmental remediation. Graphene-based nanozymes exhibit a range of catalytic activities, including peroxidase, oxidase, superoxide dismutase,87 and catalase activities.88 Various nanomaterials and molecular structures serve as excellent enzyme mimetics and nanozymes in various transformations.89–95 However, the tunability of graphene-based nanozymes represents a significant advancement in artificial enzymes, offering unprecedented control over catalytic properties and expanding their potential applications across various industries. Tunability in these nanozymes refers to the ability to modify and control their catalytic activity, selectivity, and stability through various structural and compositional adjustments. These activities can be modulated by altering the graphene material's surface chemistry and electronic properties, allowing it to perform complex biochemical reactions. This tunability is achieved through several methods, including surface functionalization, the choice of reducing agents during synthesis, nanoparticle size and shape control, the incorporation of metal nanoparticles, and the application of external stimuli such as ultrasound or electromagnetic fields.

Unlike the pre-formed, fixed amino acid pockets characteristic of natural enzymes, the active sites in graphene-based nanozymes are not singular, static entities. Instead, they are often dynamically engineered features on the material's surface, arising from a sophisticated interplay of its intrinsic composition, defects, and deliberately introduced structural modifications. These sites collectively form a tunable catalytic landscape. The catalytic mechanism of graphene-based nanozymes involves the adsorption, activation, and electron transfer of the substrate at carbon surfaces. Unlike natural enzymes that rely on specific binding pockets, nanozymes often utilize surface-mediated interactions, including electrostatic or hydrophobic forces, for substrate binding.

Surface functionalization, for instance, involves modifying the graphene surface with different chemical groups or molecules to enhance its interaction with specific substrates or improve its catalytic properties.96 Introducing oxygen-containing functional groups in graphene oxide (GO), a graphene derivative, enhances its dispersibility in aqueous solutions, which is beneficial for catalytic applications in biological and environmental systems. Graphene-based nanozymes host abundant active sites primarily comprising oxygen-containing functional groups, including hydroxyl (–C–OH), carbonyl (C[double bond, length as m-dash]O), and carboxyl (O[double bond, length as m-dash]C–O–) groups. These surface/defect-bound groups interact with substrates via hydrogen bonding or serve as catalytic centers, forming stable active sites. For example, carbonyl groups are key active centers for peroxidase-mimicking activities, with the activation of the C[double bond, length as m-dash]O bond being a critical step in the catalytic cycle. Similarly, carboxyl functional groups have been identified as catalytic sites for the reduction of hydrogen peroxide (H2O2) to hydroxyl radical, and also function as substrate-binding sites for H2O2 (Fig. 5a).


image file: d5ra04765j-f5.tif
Fig. 5 (a) Active sites present on graphene oxide nanozymes. (b) Cluster model depicting four distinct nitrogen configurations in nitrogen-doped reduced graphene oxide (N-rGO) that serve as active sites.

Smaller nanoparticles with larger surface-area-to-volume ratios exhibit higher enzyme-like activity due to the presence of more unsaturated sites, although stability can be a concern. Encapsulation with a few-layer graphene can protect these particles, maintaining high catalytic activity and stability. Graphene quantum dots (GQDs) are particularly noteworthy, representing the latest addition to the nanocarbon family, characterized by their zero-dimensional structure and atomic-level thickness.97 GQDs are utilized in sensing, anti-counterfeiting, energy storage, and cancer catalytic therapy due to their unique luminescent properties and excellent water dispersibility, which prevent aggregation in aqueous solutions. Two primary approaches enhance the catalytic efficiency of GQD-based nanozymes: optimizing the number of oxygen-containing functional groups and employing heteroatom doping or combinations with metal-based materials. For example, carboxyl and carbonyl groups on GQDs can serve as substrate binding sites and catalytic centers.98 In a recent study, graphene quantum dots were synthesized using multiwalled carbon nanotubes (MWCNTs) as carbon precursors and concentrated nitric acid as an oxidizing agent through a simple oxidation reflux method. The resulting GQDs exhibited a surface enriched with carbonyl and carboxyl groups, while containing minimal hydroxyl groups, which significantly enhanced their peroxidase-like activity across a wide pH range.55 Kinetic analyses further demonstrated that the GQDs had a Michaelis constant (Km) value five times lower than other types of GQDs and an order of magnitude lower than horseradish peroxidase, indicating superior catalytic efficiency. These findings underscore the potential of GQDs in advancing nanozyme research through accessible synthesis and effective surface modification techniques.

Another approach for fine-tuning catalytic activities tailored to specific applications was the functionalization of graphene and graphene-based nanomaterials with metal oxides and heteroatom doping. The strategic incorporation of non-metallic heteroatoms, particularly nitrogen (N), but also boron (B), sulfur (S), and phosphorus (P), into the graphene lattice is a predominant and highly effective strategy for creating and significantly enhancing active sites. Nitrogen doping leads to the formation of various distinct nitrogen functionalities within the graphene structure, including pyrrolic-N, pyridinic-N, graphitic-N, and oxidized-N species (Fig. 5b). Each configuration contributes uniquely to the material's properties and catalytic activity. For example, pyridinic N is known for its higher coordination ability, enhanced transition metal loading capacity, and superior redox activity, while graphitic N offers higher electronic conductivity and structural stability. These specific nitrogen configurations are crucial as they serve as direct active catalytic sites for various reactions, including H2O2 catalysis. The incorporation of these heteroatoms into the graphene lattice induces a significant delocalization and redistribution of electronic states, which in turn modulates the material's physicochemical properties, creating highly active sites, often through bridging interactions with adjacent carbon atoms. This doping strategy dramatically increases catalytic activity; for instance, nitrogen-doped reduced graphene oxide (N-rGO) can exhibit catalytic activity several tens to hundreds of times greater than pristine rGO. Furthermore, synergistic effects from co-doping, such as with nitrogen and boron, can lead to even more remarkable enhancements, with a reported 1000-fold increase in catalytic activity for H2O2.

Integrating different metal oxides, such as Fe3O4, MnO2, and CeO2, and heteroatoms into graphene structures significantly boosts enzymatic activity, providing a means to adjust functionality for therapeutic purposes. A highly effective approach to boosting catalytic performance involves the synergistic integration of transition metals with graphene-based materials, forming sophisticated hybrid active sites. Transition metals such as iron (Fe), copper (Cu), zinc (Zn), and cobalt (Co) can be strategically integrated within nitrogen-doped graphene frameworks, leading to the formation of advanced structures like M–Nx–C, M, M′–Nx–C, or M–S, Nx–C frameworks. These hybrid structures significantly enhance the catalytic architecture by further optimizing the redistribution of electronic density and modifying electron transfer pathways. A prominent example is the Fe–N4 configuration, which remarkably mimics the active center (iron-heme group) of natural heme-containing peroxidases. This biomimetic structure exhibits profoundly enhanced peroxidase-like activity by effectively reducing the energy barrier for the generation of hydroxyl radicals. Similarly, Zn–Nx-structured GO and Cu–N4S–C/Cu–N4–C-structured GO demonstrate exceptional peroxidase-like activity. The integration of metal nanoparticles, such as Fe3O4, onto the graphene oxide surface is also highly effective, preventing particle aggregation and thereby exposing a greater number of active sites, which leads to a significant enhancement in catalytic activity.

Recent advancements include single-atom nanozymes (SANs),99 where graphene-based nanomaterials support atomically dispersed metal active sites. This integration maximizes atom utilization and enhances catalytic efficiency and stability compared to traditional enzyme mimetics. The synergy between SANs and graphene enables precise control over catalytic activity through modifications to the graphene structure and the selection of metal atoms. This tunability is essential for optimizing enzymatic functions across various applications, including sensing, environmental remediation, and biomedical uses.100,101 As research progresses, the ability to tailor the properties of SANs within graphene composites is expected to lead to breakthroughs in both fundamental research and practical applications, establishing them as a focal point in the development of next-generation nanomaterials.

The integration of machine learning techniques has further enhanced the tunability of graphene-based nanozymes by predicting and optimizing conditions for desired catalytic activities.102 This high degree of tunability has led to significant advancements in various fields, including biosensing, where these nanozymes can be used to develop sensitive and selective sensors for detecting biomolecules crucial in medical diagnostics and environmental monitoring. In clinical therapy, the enzyme-like activities of tunable graphene-based nanozymes can be harnessed for drug delivery systems, catalyzing reactions that release drugs at targeted sites within the body. Furthermore, their applications extend to environmental remediation, pollution control, and agricultural analysis, showcasing these tunable nanomaterials' versatility and potential impact across multiple industries.65,103,104 As research in this field progresses, the tunability of graphene-based nanozymes promises to revolutionize various industrial and biomedical applications by providing customizable and efficient catalytic solutions.

4. Biomedical application of graphene-based nanozymes

Graphene-based nanozymes have emerged as promising candidates in biomedicine due to their unique properties and exceptional catalytic capabilities. These nanozymes, composed of graphene,105 graphene oxide,106 reduced GO (rGO),107,108 and graphene quantum dots (GQDs)109 loaded with various metallic nanoparticles, mimic the enzymatic functions of natural enzymes. Their exceptional biocompatibility, high surface area, and tunable surface chemistry make them ideal for biomedical applications. These nanozymes exhibit strong antioxidant properties, scavenging harmful reactive oxygen species and protecting cells from oxidative stress-induced damage.110 Moreover, their peroxidase-like activity enables the efficient detection and removal of toxins, pathogens, and pollutants in biological environments.111 The biomedical application of graphene-based nanozymes holds great promise for advancing disease diagnosis, therapy, and environmental protection, making them a cutting-edge technology to pursue improved healthcare and a healthier environment.

The nanozymes play a significant role in various biomedical applications. One key area is biosensing and diagnostics.112 These nanozymes can be highly sensitive and selective platforms for detecting biomolecules and disease markers. By functionalizing graphene with specific receptors or antibodies, they can capture and detect target molecules with remarkable accuracy. This enables the early diagnosis of diseases such as cancer, infectious diseases, and cardiovascular disorders.

Graphene-based nanozymes also demonstrate outstanding potential in tissue engineering113,114 and regenerative medicine.115–117 Their biocompatibility, conductivity, and mechanical strength make them suitable scaffolds for promoting cell growth, proliferation, and differentiation.118 These nanozymes can be used to construct 3D tissue scaffolds113 that mimic the natural extracellular matrix, facilitating tissue regeneration and organ transplantation. Additionally, graphene-based nanozymes have been explored for their antibacterial properties.119 They can effectively inhibit bacterial growth and biofilm formation, making them valuable in combating antibiotic-resistant bacteria. Furthermore, these nanozymes can aid in wound healing by promoting tissue regeneration and preventing infections.46 Overall, the role of graphene-based nanozymes in biomedical applications is vast and diverse, as discussed in Table 2. They offer tremendous potential in biosensing, drug delivery, tissue engineering, antibacterial treatments, and other areas, revolutionizing the field of biomedicine and improving patient outcomes.

Table 2 Overview of graphene-based nanocomposites: biomimetic activities and their applications across various fieldsa
Nanocomposite Enzymatic activity Applications References
a CuS/GO NC – copper sulfide nanozymes anchored to graphene oxide nanosheets; rGO–GP – reduced graphene oxide with Ganoderma polysaccharide; CoO/N-CS–rGO – CoO nanoparticles/N-doped carbon sheets/reduced graphene oxide composites; Au–rGO – gold nanoparticle with reduced graphene oxide; Fe3O4@porous graphene – magnetite on porous graphene; Ce-GONRs – cerium nanoparticles with graphene oxide nanoribbons; rGO/MWCNT – reduced graphene oxide multiwalled carbon nanotubes; GQD–SPNs – graphene quantum dots/semiconducting polymer; NPGQDs – N and P codoped graphene quantum dots; NH2GQDs/o-phenylenediamine – amino-functionalized graphene quantum dots and o-phenylenediamine; N, B-LIGzyme – laser induced N-and B-codoped graphene nanozymes; Fe–N-rGO – iron–N embedded on reduced graphene oxide; PdNPs/N-PC-rGO – PdNPs functionalized N-doped porous C-rGO; Pt/NG – Pt nanoparticle/N-doped graphene; Cu2−xSe/rGO – Cu2−xSe nanoparticles on graphene oxide hybrids; CuONCs/rGO – CuO nanocluster supported by reduced graphene oxide; rGO–CMCS–hemin/Pt@Pd NPs – reduced graphene oxide–carboxymethyl chitosan–hemin/platinum@palladium; PRTM–RhNC@rGO – arginine-rich peptide–rhodium nanocluster@reduced graphene oxide composite; Fe3O4–NH2/GONRs – monodispersed spherical composite magnetite on graphene oxide; Ag2S@GO–silver sulfide on graphene oxide; Fe3O4@CuO–GO – Fe3O4, CuO, and cucurbit[6]uril on graphene oxide; GO–DNA–PtNPs – graphene oxide@DNA with platinum nanoparticles; GOD–GO/MnO2 – glucose oxidase-conjugated graphene oxide/MnO2 nanozymes; BN-GDY – boron and nitrogen codoped graphdiyne; Cu3/ND@G – Cu3 cluster stabilized on defect-rich nanodiamond-graphene; Cu/GO SACs – copper@graphene oxide single atom catalyst; N-GNMs – nitrogen-doped graphene nanoparticles.
CuS/GO NC Oxidase and peroxidase Antibacterial and wound healing 120
rGO–GP Peroxidase L-Cysteine detection 68
CoO/N-CS–rGO Oxidase Dopamine & uric acid detection 121
Au–rGO Laccase Phenolic substrate detection 122
Fe3O4@porous graphene Peroxidase Glucose detection 123
Ce-GONRs Peroxidase and oxidase Pesticides detection 106
rGO/MWCNT Peroxidase Glutathione detection 124
GQD–SPNs Peroxidase Photothermal cancer therapy 125
NPGQDs Peroxidase Chemodynamic cancer therapy 126
NH2GQDs/o-phenylenediamine Oxidase H2O2 & ascorbic acid 127
N, B-LIGzyme Peroxidase Anti-bacterial 128
Fe–N-rGO Peroxidase H2O2 58
PdNPs/N-PC-rGO Oxidase, peroxidase, and catalase Glutathione detection 129
Pt/NG Peroxidase Acetylcholinesterase sensing 130
Cu2−xSe/rGO Peroxidase Cancer cell detection 131
CuONCs/rGO Peroxidase H2O2 & ascorbic acid 132
rGO–CMCS–hemin/Pt@Pd NPs Peroxidase Golgi protein 133
PRTM–RhNC@rGO Uricase Uric acid & inhibition of urate crystal 134
Fe3O4–NH2/GONRs Peroxidase and catalase Sildenafil 135
Ag2S@GO Oxidase Hg(II) sensing 136
Fe3O4@CuO–GO Peroxidase Homocysteine 137
GO–DNA–PtNPs Peroxidase Hg(II) sensing 138
GOD–GO/MnO2 Peroxidase Glucose detection in blood 139
Cu-NS-rGO Peroxidase Catalytic activity 140
Cys-GO Peroxidase Hg(II) sensing 141
PtCo@graphene Oxidase Antibacterial 142
rGO/MoS2 and Fe3O4NPs Oxidase and peroxidase Cancer biomarkers 143
BN-GDY Peroxidase Cancer therapy 144
GQD/CoSnO3 Peroxidase Chemo-sono dynamic cancer therapy 145
Cu3/ND@G Oxidase Antibacterial 146
GO/CuFeSx NC Peroxidase Antibacterial 147
MoS2/rGO Oxidase, peroxidase and catalase Antibacterial 148
Cu/GO SACs Superoxide dismutase Treatment of lung injury and anti-inflammatory 149
N-GNMs Peroxidase Tumor-specific treatment 150


4.1 Biosensing applications

Graphene nanozymes have revolutionized biosensing by combining the unique properties of graphene-based nanoparticles with enzymatic catalytic activity. These nanomaterials, designed to mimic natural enzymes, exhibit exceptional catalytic efficiency and specificity, making them crucial for detecting biomolecules in various applications.53,151 Graphene-based nanomaterials as colorimetric152 and electrochemical biosensors,153 provide unique properties for the in situ detection of glucose, DNA, enzymes, proteins, and microorganisms.154,155 They also show potential in cancer cell detection,156 enabling early and accurate diagnosis. This activity allows the nanozymes to catalyze the oxidation of colorimetric substrates like TMB (3,3′,5,5′-tetramethylbenzidine), and ortho-phenylenediamine in the presence of H2O2, resulting in a color change that can be detected colorimetrically (Fig. 6). Their combination with polymers and surface-decorated metal nanoparticles expands their potential in biosensing. Graphene-based nanozymes offer a versatile platform for efficient detection and advancements in biomedical research and diagnostics (Table 3).
image file: d5ra04765j-f6.tif
Fig. 6 Enzymatic activity of graphene-based nanozymes in biosensing.
Table 3 List of nanozymes with their enzyme-like activities, analyte, method, and limit of detection (LOD)a
Nanocomposite Enzyme mimic Detection Method LOD References
a NGQD@NC@Pd HNS – Pd nanoparticles decorated N-doped graphene quantum dots@N-doped carbon hollow nanospheres; NiCo2O4/3DGF – nickel–cobalt oxide decorated three-dimensional graphene; GCNT–Fe3O4 – cubic Fe3O4 nanoparticles loaded on graphene oxide-dispersed carbon nanotubes; GSF@AuNPs – gold nanoparticle-loaded mesoporous silica-coated graphene; NB-rGO – N- and B-codoped reduced graphene oxide; 3DRGO–Fe3O4–Pd – three-dimensional graphene–magnetic palladium nanohybrids; RGO–IN – reduced graphene oxide–iron nanoparticles; γ-Fe2O3/t-SG – γ-Fe2O3/thiophene-sulfur-doped graphene; b-Fe-GQDs, o-GQDs – iron and carbonyl functionalized graphene quantum dots; Pt/NG – Pt nanoparticle/N-doped graphene; AuAg-rGO – reduced graphene oxide capped AuAg bimetallic nanoparticles; Zr–GO – zirconium on graphene oxide; CoO/N-CS–rGO – CoO nanoparticles/N-doped carbon sheets/reduced graphene oxide composites; NH2-MIL-88B(Fe)@MRGO – NH2-MIL-88B(Fe)@modified reduced graphene oxide; RuO2/rGO – ruthenium oxide/reduced graphene oxide; WS2/rGO – tungsten disulfide on reduced graphene oxide; Co-His-GQD-G – cobalt histidine graphene quantum dot on graphene; FA-MnO2/GO – folic acid functionalized MnO2 on graphene oxide; g-CNQDs and GQDs – fluorescent graphitic carbon nitride and graphene oxide quantum dots; rGO/CMCNs – reduced graphene oxide Cu3(OH)2(MoO4)2 cuboidal nanostructures; CGNPs – crumpled graphene nanoparticles; Fe3+–NGQDs – nitrogen-doped graphene quantum dots and iron ion nanocomposite; GQDs and MoS2 QDs – graphene quantum dots and molybdenum disulfide quantum dots; PtCu/PSS-Gr – PtCu bimetallic nanoparticle on polystyrene sulfonate functionalized graphene.
NGQD@NC@Pd HNS Catalase H2O2, cancer cells Electrochemical 20 nM 157
NiCo2O4/3DGF Catalase Glucose and calcium ion Electrochemical 0.38 and 4.45 μM 158
GCNT–Fe3O4 Peroxidase and catalase H2O2 and glucose Colorimetric and electrochemical 0.022 mM 159
GSF@AuNPs Peroxidase Cancer cells and therapeutics Colorimetric 45
NB-rGO Peroxidase Acetylcholine and C-reactive protein Colorimetric 10 nM 160
3DRGO–Fe3O4–Pd Peroxidase Glutathione and glucose Colorimetric 13 μM 161
RGO–IN Peroxidase H2O2 and glucose Colorimetric 0.2 and 0.8 μM 151
γ-Fe2O3/t-SG Peroxidase Chlorpyrifos Colorimetric 0.17 μg mL−1 162
b-Fe-GQDs, o-GQDs Peroxidase and oxidase Alkaline phosphatase Colorimetric and fluorescence 0.21 U L−1 163
Pt/NG Peroxidase Acetylcholinesterase Colorimetric 0.0652 mU mL−1 164
AuAg-rGO Peroxidase Glutathione Colorimetric 38 nM 165
Zr–GO Peroxidase H2O2 Colorimetric 0.57 μM 166
CoO/N-CS–rGO Oxidase Dopamine and uric acid Electrochemical 1378 μA mM−1 and 1393 μA mM−1 121
NH2-MIL-88B(Fe)@MRGO Peroxidase Glucose Colorimetric 3.16 μM 154
RuO2/rGO Peroxidase Glucose Colorimetric 3.34 μM 167
WS2/rGO Peroxidase H2O2 Colorimetric 82 nM 168
Co-His-GQD-G Oxidase Chlorpyrifos Colorimetric 0.57 ng mL−1 169
FA-MnO2/GO Oxidase Cancer cells Colorimetric 20 cancer cells 170
g-CNQDs and GQDs Peroxidase Fluoride ion Colorimetric and fluorescent 4.06 μM 111
rGO/CMCNs Peroxidase Dopamine in blood serum and urine samples Colorimetric 0.17 μM 171
CGNPs Peroxidase Dopamine Colorimetric 1.15 μM 62
Fe3+–NGQDs Peroxidase Hydroquinone Colorimetric 0.2 μM 65
Fe, N-GQD Peroxidase L-Cysteine Colorimetric 140 nM 66
GQDs and MoS2 QDs Peroxidase Cholesterol Chemiluminescent 35 nM 172
PtCu/PSS-Gr Peroxidase Puerarin Colorimetric 1 × 10−5 M 173


For instance, carboxyl-modified graphene oxide (GO-COOH) possesses inherent sensing capabilities, enabling the detection of ions and small molecules.174 This functionalization enhances the surface area and facilitates binding interactions, increasing the sensitivity of biosensors. Furthermore, the integration of gold nanoparticles with graphene, creating a composite known as Au–rGO, has led to the development of advanced biosensors.122,165 This hybrid material benefits not only from the catalytic properties of graphene but also from the unique properties of gold nanoparticles, making it a versatile platform for biosensing.8,175 In point-of-care diagnosis, graphene nanozymes show great promise. One notable example is the creation of glucose-biosensing microneedle patches. These patches consist of GOx-conjugated MnO2/graphene oxide nanozymes (GOx–MnO2@GO), enabling non-invasive and efficient glucose monitoring.139 This innovation has the potential to revolutionize the management of diseases like diabetes, providing patients with convenient and accurate monitoring solutions.

The broader impact of graphene nanozymes is reflected in their diverse applications beyond biosensing. Their innate catalytic behavior, similar to biological enzymes, extends their use to other fields, such as drug delivery, environmental monitoring, and energy conversion. This versatility, coupled with their exceptional properties, positions graphene nanozymes as a cornerstone in the ongoing advancement of cutting-edge technologies for both diagnostics and therapeutics.176–178 For instance, manganese phthalocyanine-modified graphene nanosheets function as a peroxide-like nanozyme, catalyzing the oxidation of TMB by H2O2 (Fig. 7).


image file: d5ra04765j-f7.tif
Fig. 7 (a) Diagrammatic representation for the synthesis of MnPc@GNP (manganese phthalocyanine on graphene nanosheet) (b) plausible mechanism for the H2O2 mediated TMB oxidation using MnPc@GNP. Reprinted with permission from ref. 176. Copyright 2023 American Chemical Society.
4.1.1 Detection of cancer cells. Cancer is a widespread and devastating disease that claims numerous lives globally. As a result, the timely diagnosis of cancer is of utmost importance.179 Detecting cancer at its earliest stages can significantly improve patients' prognosis and increase the effectiveness of treatment.180 Given the high mortality rates associated with cancer, early detection holds the key to improving survival rates and reducing the impact of this disease on individuals and communities worldwide.181 Therefore, developing and utilizing accurate and sensitive diagnostic methods is essential in the ongoing battle against cancer. Early diagnosis not only enhances individual outcomes but also contributes to broader efforts to combat and mitigate the effects of cancer on a global scale.182 Cancer cell detection is a crucial aspect of early cancer diagnosis and treatment. Detecting cancer cells at an early stage can significantly improve the chances of successful treatment and recovery for patients. Various methods and technologies, such as imaging techniques,183 flow cytometry,184 and immunophenotyping185 are employed to identify and analyze cancer cells within the body. These detection methods aim to pinpoint abnormal cellular activity, genetic mutations, or the presence of specific biomarkers associated with cancer. Despite their established status, these approaches are expensive, time-consuming, and susceptible to operator variance. Therefore, there is a growing requirement for alternative cancer cell detection methods that are swift, sensitive, and affordable.

Graphene-based nanozymes are good alternatives for the early detection of cancer cells. Liu et al. fabricated a MnO2 nanosheet/graphene oxide hybrid functionalized with folic acid that served as an oxidase mimic.186 According to these findings, FA-MnO2/GO might preferentially bind to cancer cells by interacting with the folate receptors that are overexpressed in many tumor and cancer cell lines. Additionally, it catalyzed the oxidation of TMB without the use of H2O2. The overexpression of the folate receptor on hybrid and cancer cells was made possible by the functionalization of folate acid (FA), and the binding events were then transformed into quantifiable colorimetric signals by the oxidation of the chromogenic substrate TMB. In another study, Tao et al. discussed a coordinated strategy for cancerous cell detection, reporting the incorporation of AuNCs in graphene oxide-based colorimetric biosensors with amplified signals for cancer cell detection due to the favorable optical properties, biocompatibility, and low toxicity of AuNCs.187 Good catalytic oxidation of TMB occurred even at neutral pH by incorporating AuNC in graphene oxide, enabling selective, sensitive cancer cells detection by leveraging the specific binding between folic acid conjugated to the graphene oxide and folate receptors on cell membranes, achieving a detection limit of 1000 MCF-7 cells. In another study, Kim et al. significantly improved the activity of a nanohybrid by modifying the surface of graphene oxide with platinum nanoparticles (PtNPs) and Fe3O4 magnetic nanoparticles.188 This nanohybrid exhibited a 30 times higher maximal reaction velocity (Vmax) compared to bare graphene oxide and blue color signals obtained in just 5 minutes, proportional to the amount of target cancer cells.

Guo et al. utilized a nanocomposite of Cu2−xSe/reduced graphene oxide, aided by folic acid, to visualize cancerous cells. To functionalize Cu2−xSe/rGO, folic acid was chosen as the target chemical since it can efficiently target various tumor cells with overexpressed folate receptors on their cell membranes.131 FA–Cu2−xSe/rGO was demonstrated to possess the capacity to distinguish between target cells and control cells. Furthermore, Zhang et al. designed the colorimetric test to quickly and accurately identify cancer cells using PtNPs/GO as a signal transducer. Folic acid was used as the recognition component to functionalize PtNPs/GO.189 When the folate receptor on a tumor cell's membrane was overexpressed, folic acid efficiently targeted those cells. PtNPs/GO on tumor cells transformed the identification process into a quantifiable colorimetric signal, providing an accurate and convenient method for identifying malignant cells. Folic acid was conjugated with hybrid nanomaterials, resulting in the FA–GO–AuNCs composite (GFA), which served as an effective probe for the rapid, selective, and quantitative colorimetric detection of cancer cells. As reported by Yu Tao et al., this technology could substantially improve diagnostic accuracy and prognostic monitoring in oncology.187 To create a quick, low-cost, highly accurate, and sensitive colorimetric test to identify cancer cells, they used the folic acid-attached GO–AuNCs hybrid.187

Electrochemical biosensors are being extensively used in fields such as clinical diagnostics, environmental monitoring, and food safety analysis, as they are user-friendly, cost-effective, highly stable, and offer excellent sensitivity.155 Alizadeh et al. created a 3-electrode electrochemical cell, a cost-effective, rapid, and highly sensitive technique for cancer cell identification.156 They discovered that the electrochemical detection of cancer cells required electrodes and a sensing platform. The CuO/WO3 nanoparticles, which exhibit peroxidase-like activity, may be incorporated into this sensing platform. CuO/WO3 nanoparticles catalytically converted H2O2 generated by cancer cells into ROS upon contact. This enzymatic activity enhanced the electrochemical signal, making the presence of cancer cells more straightforward to detect. Liu et al. revealed that a graphene hemin composite coated with Au nanoflowers can exhibit enhanced peroxidase-like characteristics, making it a promising electrochemical aptamer biosensor for detecting K562 leukemia cells.190 They also employed a three-electrode arrangement, with the gold electrode as the working electrode. Cancerous cells can be detected for several reasons. For example, the biosensor contains aptamers that are precisely chosen to recognize and bind to the target molecules on the surface of K562 leukemia cancer cells. When malignant cells are present in the sample, the aptamers bind specifically to them, enabling their recognition. The signal amplification process plays a significant role in their experiment. AuNFs inserted within the biosensor provide additional catalytic sites, enabling signal amplification. The catalytic capabilities of AuNFs increase the electrochemical process, resulting in an amplified signal that correlates to the presence or amount of K562 leukemia cancer cells. This amplification step improves detection sensitivity. Nanozymes capture electroactive molecules due to their large surface area and numerous binding sites, which enable more effective electrochemical reactions.

In a recent study, Song et al. introduced an innovative composite called folic acid-linked graphene–hemin (GFH).191 This compound demonstrated the ability to rapidly, selectively, and precisely target cancer cells via a colorimetric signal enhanced by the peroxidase-mimicking activity of GFH. This method can detect cancer cells at a threshold of 1000 cells or less. Compared to other methods that rely on antibodies or natural enzymes, this composite offers superior stability and resistance to denaturation or degradation. Additionally, graphene in this composite offers several advantages over other materials, such as carbon nanotubes or mesoporous silica. The significant size of the GFH complex allows for its visualization under an optical microscope, facilitating its entry into cells. The system is capable of detecting cancer cells within cellular environments by focusing on surface receptors or specific biomarkers. The strong π–π stacking interactions between graphene and hemin facilitate the attachment of multiple hemin molecules to both sides of a single graphene layer. This arrangement ensures that each hemin active site is easily accessible for interaction with substrates, significantly enhancing catalytic efficiency. Graphene oxide (GO) also possesses numerous functional groups, including epoxy, hydroxyl, and carboxyl, allowing biomolecules to attach flexibly. As a result, our system shows significant promise for a wide range of applications. The findings from this research could enhance the application of functionalized graphene derivatives in various fields, including accurate and rapid recognition of specific cells with outstanding sensitivity, DNA and protein analysis, and the development of new imaging methods.191 The GO possesses numerous functional groups, allowing biomolecules to attach readily. The system has potential applications in various fields, including sensitive cell detection, DNA and protein analysis, and advanced imaging techniques.

4.1.2 Hydrogen peroxide detection. Hydrogen peroxide (H2O2) is a crucial oxidizing agent generated during various physiological processes. However, its potent oxidative activity poses a threat to cellular components, such as proteins and DNA. Detecting H2O2 has proven instrumental in studying disease progression, particularly for early-stage vascular diseases. An abnormal concentration level of H2O2 can lead to myocardial infarction, Alzheimer's disease, Parkinson's disease, cancer, etc.192 The graphene frameworks offer outstanding stability, expansive surface area, and superior electrical conductivity, providing a promising platform for enhanced catalytic properties. Several studies have demonstrated that these nanozymes exhibit remarkable efficiency in H2O2 detection, characterized by high sensitivity, specificity, and selectivity. Moreover, graphene-based nanozymes have been utilized to simultaneously detect H2O2 and other biomolecules by exploiting their catalytic properties.

The association between H2O2 and glucose is fascinating, as H2O2 is a byproduct of glucose oxidation mediated by glucose oxidase. Therefore, in clinical and basic research, it is common to employ compatible methodologies to detect H2O2 and glucose simultaneously. This parallel analysis enables a comprehensive investigation of the interplay between glucose metabolism, oxidative stress, and pathological conditions, facilitating the development of diagnostic and therapeutic strategies. Zhang et al. designed a catalyst showing synergistically enhanced peroxidase-like activity by employing graphene with gold & platinum MOF (AuPt/MOF-graphene) using OPD.193 They utilized it for the electrochemical detection of H2O2, achieving a detection limit of 19 nM. Jin et al. fabricated a nanozyme showing good peroxidase activity using graphene oxide and gold to detect H2O2 using the electrochemical method with an estimated detection limit of 1.9 nM.194 Furthermore, due to their vast active centers, Li et al. developed an MOF using the peroxidase activity of Fe3O4, graphene, and NH2-MIL-88B(Fe). NH2-MIL-88B(Fe)@MRGO was utilized for glucose detection.154 This mimic enzyme exhibited a high catalytic velocity (2.57 × 10−7 M s−1) and affinity (Km = 0.0091 mM) for H2O2 substrates. The limit of detection (LOD) for glucose was found to be 3.16 μM.

4.1.3 Detection of small molecules of biological importance. Small molecules include chemicals and biochemical agents, such as cholesterol, dopamine, and uric acid. Detecting these agents is helpful as they are the key biomarkers of disease. The functionalization of graphene-based nanozymes with specific receptors or ligands enables them to recognize and bind to target small molecules selectively. Upon interaction with the target molecule, the nanozymes catalyze a reaction that produces a discernible signal, such as fluorescence response, color change, or electrochemical response. As dopamine and uric acid are interlinked and produce similar signals, it is easy to detect them simultaneously. Using the hydrothermal method, Boruah et al. prepared 3D nitrogen-doped crumpled graphene nanoparticles (CGNPs).62 They used it to detect dopamine (DA) with high sensitivity in complex biological media, such as blood serum. Lu et al. synthesized a composite material consisting of polydopamine surface-functionalized cobalt oxide nanoparticles, N-doped carbon sheets, and reduced graphene oxide (CoO/N-CS–rGO) through an adsorption process followed by pyrolysis (Fig. 8). This composite exhibited oxidase-like activity. It was employed for the electrochemical detection of dopamine and uric acid. Structural characterization confirmed the presence of cobalt oxide, N-doped carbon sheets, and rGO, as indicated by XRD analysis, which revealed their characteristic peaks. SEM and TEM images further demonstrated that the N-doped carbon sheets are well-dispersed on the rGO surface, resulting in a distinctive sandwich-like morphology, as illustrated in Fig. 6.121 Novel graphene-supported ferric porphyrin and streptavidin were also investigated for their role as a peroxidase mimic for DNA.155 This method combines probe-conjugated gold nanoparticles with graphene-coated electrodes and utilizes enzyme-functionalized carbon spheres as tracers. Combining these materials enables triplex signal amplification, enhancing the sensitivity and detection limit to the attomolar level in electrochemical DNA biosensing.
image file: d5ra04765j-f8.tif
Fig. 8 (a) Schematic illustration of the synthesis of CoO/N-CS–rGO and sensing of dopamine and uric acid (b) XRD (c) SEM and (d) TEM images of CoO/N-CS–rGO. Reprinted with permission from ref. 121. Copyright 2022 American Chemical Society.

Furthermore, graphene–palladium nanowires have been utilized as an effective peroxidase mimic for electrochemical sensing. By incorporating ZnFe2O4–graphene quantum dots, the nanowires exhibit peroxidase-like activity, enabling the detection of small molecules. Glutathione (GSH), considered an essential antioxidant in biological systems, can be detected using a hemin dispersion by graphene quantum dots.195 The nanozyme-based sensor shows a linear response to GSH concentrations ranging from 0.1 to 100 μM, with a detection limit of 0.01 μM.

Ascorbic acid (AA) functions as a cofactor in various physiological and metabolic processes. Inadequate or excessive AA can cause scurvy and anaemia, as well as different psychological disorders. As a result, it is crucial to develop a straightforward and rapid method with superior selectivity and precision for identifying AA in physiological and clinical analyses.196 In recent years, several technologies for detecting AA have been developed, including fluorescence,184,185 ultra-performance liquid chromatography (UPLC) with a UV detector,187 and chemiluminescence.197 However, the nanozymes-based approach is more effective and simpler. AA may modify the peroxidase-like activity of nanomaterials, resulting in a color shift that can be visually observed. Based on the antioxidant characteristics of AA, peroxidase-based nanoprobes were reported to detect AA with reasonable accuracy. Sun et al. performed the colorimetric detection of AA using the IrO2/GO–H2O2–TMB reaction system.198 The limit of detection (LOD) for ascorbic acid (AA) was determined to be 5.3 M. The colorimetric AA detection test was also done in simulated serum, vitamin C pills, and Mizone beverages to examine the practicality of the described approach for detecting AA in actual samples. Dong Peng et al. used a synthesis method to integrate reduced graphene oxide nanosheets (p-rGO) with Mn3O4 nanoparticles (Mn3O4@p-rGO).199 In this process, Mn2+ ions were adsorbed onto GO as a precursor, and a porous p-rGO structure was produced by directly annealing in open air. Decorating the surface of p-rGO nanosheets with Mn3O4 nanoparticles significantly enhanced their oxidase-like activity, primarily due to the increased surface area, a higher density of active sites, and accelerated electron transfer efficiency. Furthermore, actual use for AA detection in real-world samples confirmed its practicality and dependability. This study demonstrates that decorating nanoparticles on specific supports is an effective technique for enhancing their enzyme-like functions, offering promising uses in pharmacy, food safety, biosensing, and clinical diagnosis.

4.1.4 Pesticide detection. Pesticides are chemical substances that are used to enhance plant growth and increase the supply of plant products. The use of pesticides leads to residues, which appear in processed commodities and eventually in the food chain. Boruah et al. synthesized Fe3O4/polydopamine functionalized graphene nanocomposites, which were established as nanozymes for detecting and degrading the pesticide simazine.200 The LOD (limit of detection) value for simazine was found to be 2.24 μM, and the nanozymes achieved 99% degradation of simazine. In another study, graphene oxide was used as an intrinsic peroxidase mimic for sensitively detecting organophosphate pesticides (OPs).201 The colorimetric assay produces a measurable signal when AChE and ChO are present. OPs lead to the inhibition of AChE, resulting in a corresponding decrease in color intensity, which is directly proportional to the concentration of OPs. It was also reported that small-sized GO showed superior catalytic activity and higher stability and was used for OP detection. The corresponding LODs for dimethoate, methyl parathion, and chlorpyrifos were all found to be lower than 2 ppb, which is below the MRL, indicating their great potential for the quantitative analysis of pesticide residues. In another groundbreaking study, Boruah et al. introduce a dual-responsive magnetic Fe3O4–TiO2/graphene nanocomposite as an enzyme mimic for both colorimetric pesticide detection and efficient photodegradation.200 The nanocomposite exhibits a synergistic enhancement in catalytic activity, yielding an impressive limit of detection (LOD) of 2.98 μg L−1 for the detection of atrazine. Moreover, it effectively photodegrades atrazine into harmless compounds, contributing to environmental safety. The magnetic properties of these nanocomposites enable easy separation, simplifying the isolation process after degradation. This magnetic separability further facilitates the reuse of nanocomposites, enabling recyclability up to 10 times.

In another study, Tai et al. prepared cerium-doped graphene oxide nanoribbons (Ce-GONRs) with a three-dimensional porous structure evincing both peroxidase and oxidase like activities, enabling synergistic oxidation of TMB.106 This study revealed that diafenthiuron and carbaryl pesticides inhibit the peroxidase and oxidase activity of Ce-GONRs, attributed to π–π stacking and hydrogen bonding between the pesticides and nanozymes. The detection limits were calculated to be 0.57 and 0.23 ng mL−1 for diafenthiuron and carbaryl, respectively. The detection was selective for these pesticides, whereas Lin et al. used the same nanocomposite for the detection of chlorpyrifos by inhibiting AChE, with a detection limit of 3.34 ng mL−1 (0.0034 ppb).202 Furthermore, another study demonstrates the synthesis of Co-histidine functionalized graphene quantum dot-graphene nanohybrid.169 It evinced outstanding oxidase-like activity, superparamagnetic behaviour, and high stability. The method was applied to detect chlorpyrifos in peaches by inhibiting AChE. The detection limit was determined to be 0.57 μg L−1.

4.1.5 Detection of heavy metal ions. Due to their toxic and nonbiodegradable properties, as well as their tendency to accumulate in ecological systems, heavy metals pose significant challenges as pollutants. Recognizing the crucial role of monitoring environmental quality amid rising pollution from industrial, agricultural, and domestic sources, it is imperative to determine the presence of heavy metals accurately. This is especially essential given their persistence and potential harm to ecosystems. Consequently, the assessment of heavy metal content assumes immense significance in maintaining ecological balance and safeguarding ecosystem health.203 Graphene-based nanozymes play an important role in the detection of heavy metals. Song et al. developed a ternary Ag–CoFe2O4/rGO material using a microwave-assisted method.204 The resulting nanocomposites displayed impressive oxidase-like activity in catalyzing the oxidation of colorless TMB to blue oxTMB, as shown in Fig. 4. The incorporation of AgNPs into the nanocomposites significantly accelerated the formation of the Ag–Hg alloy in the presence of Hg2+. A strong synergistic effect was observed among AgNPs, the large surface area of CoFe2O4, and rGO, leading to the expedited decomposition of dissolved O2 under weakly acidic conditions. As a result, in an acidic solution, O2 adsorbed onto Ag–CoFe2O4/rGO nanocomposites could be readily converted into O[2 with combining macron], which oxidized TMB to blue oxTMB. Leveraging the enhanced oxidase-like activity of Ag–CoFe2O4/rGO in the presence of Hg2+, a simple nanoenzyme-based sensor was developed for the colorimetric detection of Hg2+. The sensor exhibited a good linear range from 2 × 10−9 M to 1 × 10−8 M for Hg2+, with a low limit of detection (LOD) of 0.67 nM. Chen et al., fabricated a GO–AuNP-based colorimetric sensor for the colorimetric detection of Hg2+ and Pb2+ simultaneously.205 This hybrid was further modified by introducing ssDNA to the GO–AuNP solution to form DNA–GO–AuNP, which exhibits enzyme-like catalytic activity and a salt aggregation effect. In the absence of Hg2+ or Pb2+, the hybrids remained stable even after the addition of NaCl. However, in the presence of Hg2+ or Pb2+, the structure of the hybrid changed, leading to the aggregation of DNA–GO–AuNP hybrids and the loss of their peroxidase-like activity at high NaCl concentrations. This resulted in the inability to catalyze the oxidation of TMB to oxTMB in the presence of H2O2. This unique phenomenon enabled the development of an efficient label-free nanoenzyme-based colorimetric sensor with limits of detection (LODs) of 300 nM for Hg2+ and 500 nM for Pb2+.

In another study, Tao et al. reported that a colorimetric aptamer-based method for detecting Pb2+ was developed and validated using graphene/Fe3O4–AuNP composites as enzyme mimetics.206 The Pb2+ aptamer was anchored on magnetic beads functionalized with amine, and the presence of lead ions inhibited the catalytic activity and color reaction of the nanozyme composites. The method detected Pb2+ in the 1–300 ng mL−1 range with a low detection limit of 0.63 ng mL−1, showing potential for practical on-site analysis. This innovative approach has implications for addressing lead contamination and represents a significant advancement in colorimetric aptamer-based sensing.

4.2 Antimicrobial applications

Bacterial and microbial infections are on the rise, and there is a pressing need for antibacterial and antimicrobial agents to protect living organisms from disease-causing microorganisms. The common methods for combating bacterial infections primarily involve antibiotics, functionalized polymers,207 metal nanoparticles,208 metal oxide/sulfide nanostructures,11 antibacterial peptides and antibacterial enzymes.209 While these approaches have generally been effective in preventing drug resistance, they still have limitations. For example, they may not be effective in combating infections caused by multidrug-resistant bacteria. Additionally, the simultaneous use of different antibiotics has led to increased antibiotic resistance, particularly in Gram-negative bacteria like Escherichia coli (E. coli), which can impede antibiotic entry into the cell and modify the cell wall to evade recognition by the antibiotic.210

Graphene-based nanozymes have shown promising potential and catalytic behavior as the physical ability of graphene and its derivatives, such as their sharp-edge structure, to rupture bacterial membranes and their chemical properties, such as the production of oxidative stresses that deactivate bacterial proteins and lipids and disrupt the proliferation process, are the leading causes of their antimicrobial activity. Graphene-based nanozymes have unique catalytic properties that effectively kill bacteria by generating hydroxyl radicals, as summarized in Table 4. By using a simple hydrothermal technique, Wang et al. developed copper sulfide nanozymes attached to graphene nanosheets (CuS/GO NC), which exhibited potent oxidase and peroxidase-like activity.120 CuS/GO NC exhibited physical and chemical activity against methicillin-resistant Staphylococcus aureus bacteria. Due to the needle-like morphology of CuS/GO NC, they punctured bacterial membranes, causing leakage of the intracellular matrix that rendered the bacteria inactive. They also caused damage to the bacterial cell by producing large amounts of ROS, leading to oxidative stress. In another study, hemin–reduced graphene oxide doped with gold nanoflakes (hemin–rGO@Au NFs) was made by Liu et al. and exhibited vigorous peroxidase-like catalytic activity, outstanding photothermal conversion efficiency in the NIR range, and quick electron transfer capability, due to which it showed synergistic antibacterial activity to Gram-negative drug-resistant bacteria (E. coli).211 Model bacteria, drug-resistant Gram-negative E. coli, were subjected to a solution containing the nanozyme composite (hemin–rGO@Au NFs) and a small amount of H2O2, which caused the generation of singlet oxygen (1O2), a reactive oxygen species with potent antibacterial properties. The singlet oxygen exhibited remarkable antibacterial activity following further NIR irradiation treatment, leveraging the composite's high photothermal conversion efficiency in the NIR range.

Table 4 Some graphene-based nanozymes for antibacterial applicationsa
Nanoparticle Enzymatic activity Bacterial types and application Antibacterial mechanism References
a GQDs – graphene quantum dots; Au/g-C3N4 – gold nanoparticle with graphitic carbon nitride; GQD/AgNP – graphene quantum dots and gold nanoparticles; PtCo@G – platinum and cobalt@graphene; GQD/TiO2 – graphene quantum dots-sensitized TiO2; MoS2/rGO VHS – molybdenum disulfide/rGO vertical heterostructures; CuS/GO NC – copper sulfide anchored to graphene oxide nanosheets; CoPt@G@GOx – CoPt@graphene@glucose oxidase; GO@Ag–Pt – graphene oxide loaded silver–platinum hybrid nanoparticles.
GQDs Peroxidase E. coli, S. aureus Generation of OH˙ radical 46
Au/g-C3N4 Peroxidase E. coli, S. aureus Generation of OH˙ radical 47
GQD/AgNP hybrids Peroxidase and oxidase E. coli, S. aureus Generation of OH˙ radical 187
PtCo@G Oxidase H. pylori Generation of OH˙ radical 212
GQD/TiO2−x Peroxidase S. aureus, E. coli, MRSA Sonodynamic nano catalytic therapy 213
MoS2/rGO VHS Peroxidase, oxidase and catalase E. coli and S. aureus Generation of OH˙ radical 148
CuS/GO NC Oxidase and peroxidase MRSA Generation of OH˙ radical 120
CoPt@G@GOx Peroxidase Streptococcus mutans biofilms Generation of toxic OH˙ radicals in acidic environment 214
GO@Ag–Pt loaded nanozyme hydrogels Superoxide dismutase S. aureus, E. coli, wound healing Antioxidant and ROS scavenger 48


Another study investigated the antibacterial properties of graphene quantum dots (GQDs) and their potential to enhance the efficacy of hydrogen peroxide (H2O2) in combating bacterial infections.46 GQDs exhibited superior enzymatic activity compared to GO due to their excellent electron transport properties, which enabled them to convert H2O2 into highly reactive hydroxyl radicals [( with combining macron]OH), making them significantly more effective at killing bacteria. Importantly, GQDs demonstrated no significant toxicity in both in vitro and in vivo, making them a safer alternative for antibacterial applications. The combination of GQDs with low concentrations of H2O2 resulted in over 90% lethality against Escherichia coli and Staphylococcus aureus, using H2O2 concentrations 100 times lower than those required when used alone. The mechanism of action involved the adherence of GQDs to bacterial membranes, facilitating the localized generation of OH radicals, which natural bacterial enzymes cannot easily neutralize. Additionally, the research led to the development of GQD-Band-Aids, which effectively prevented wound infections in mice; after 72 hours of treatment with these Band-Aids combined with H2O2, treated wounds showed no signs of erythema or edema (Fig. 9).46 Overall, this work highlighted the potential of GQDs as a biocompatible and effective antibacterial agent, particularly in wound care applications where traditional antibiotics might be ineffective due to the development of resistance.


image file: d5ra04765j-f9.tif
Fig. 9 (a) Schematic illustration of the GQD-assisted antibacterial system. (b) Picture of fabric absorbed GQDs. (c) The obtained GQD-Band-Aid. (d) The mouse treated with GQD-Band-Aid. Reprinted with permission from ref. 46. Copyright 2014 American Chemical Society.

4.3 Application in wound healing

Graphene-based nanozymes have shown promising results in wound healing due to their excellent biocompatibility, catalytically active sites, mechanical strength, and electrical conductivity, making them suitable for biomedical applications. The antibacterial properties of graphene-based materials stem from their ability to disrupt bacterial cell membranes, resulting in cell lysis and improved wound healing outcomes. These materials can effectively inhibit the growth of pathogens, eliminate biofilm formations, and promote processes such as cell differentiation, re-epithelialization, and collagen deposition, all of which contribute to enhanced wound healing. For instance, graphene oxide nanoparticles (GONPs) have been found to significantly improve wound healing in animals, reaching up to 99% healed animals within 15 days.215 Additionally, graphene-based nanozymes, such as Fe3C/N-doped graphitic carbon, have shown efficient peroxidase-like activity, enabling broad-spectrum antimicrobial effects and accelerated wound healing in vivo by decomposing H2O2 to generate hydroxyl radicals, enhancing their antimicrobial efficacy.216

The combination of graphene-based materials with other biocompatible components, such as polysaccharides and biological scaffolds, further improves their wound-healing potential. Chen et al. developed a multifunctional hydrogel by cross-linking polyvinyl alcohol and sodium alginate with GO that is loaded with silver–platinum hybrid nanoparticles (GO@Ag–Pt).48 This advanced hydrogel exhibited remarkable abilities to scavenge ROS, produce oxygen, and demonstrate antibacterial activity against various bacteria in vitro. Adding silver–platinum hybrid nanoparticles significantly enhanced the hydrogel's antibacterial effectiveness against Escherichia coli and Staphylococcus aureus compared to conventional silver nanoparticles. Moreover, the GO@Ag–Pt loaded hydrogel successfully treated S. aureus infections, notably accelerating wound healing during the inflammatory phase by lowering ROS levels and reducing inflammation. This research highlighted the potential of ROS-scavenging and antibacterial hydrogels as viable treatments for various wound types, including challenging diabetic wounds complicated by bacterial infections, suggesting a promising approach for chronic wound healing through effective ROS management and bacteriostatic properties.48

In recent years, antimicrobial resistance (AMR) has emerged as a significant threat to global health, necessitating the development of novel antibacterial agents. A study introduced a new nanozyme platform, Cu, N-GQDs@Ru–NO, which combined copper and nitrogen-doped graphene quantum dots (Cu, N-GQDs) with nitric oxide (NO) and ruthenium nitrosyl (Ru–NO).217 When exposed to 808 nm near-infrared (NIR) light, this novel nanozyme exhibited remarkable NADH dehydrogenase-like activity, efficiently photo-oxidizing NADH to NAD+, thereby disrupting the redox balance within bacterial cells and ultimately leading to bacterial death. In addition to effectively eradicating methicillin-resistant Staphylococcus aureus (MRSA) and its associated biofilms, the NIR light also activated the NO donor, which facilitated wound healing. Excellent photothermal effects were another feature of the nanozyme, which enhanced its antibacterial effectiveness. By combining photothermal treatment, NO gas therapy, and NADH dehydrogenase activity, Cu, N-GQDs@Ru–NO exhibited remarkable in vitro and in vivo efficacy against bacterial resistant infections and biofilm eradication. This research presents a promising therapeutic strategy for treating MRSA-related inflammatory wounds through a multifaceted approach that targets bacterial survival and wound healing. This study aligns with recent advancements in nanozyme research, where various metal-based nanozymes have demonstrated promise in combating antimicrobial resistance (AMR). For instance, other studies have explored the use of metal-free nanozymes, such as graphene quantum dots, for similar applications in cancer therapy and the management of oxidative stress. The unique combination of photothermal therapy and NO gas delivery distinguishes it from traditional therapies that may rely solely on one mechanism.217 Additionally, integrating multiple therapeutic strategies into a single platform represents a significant advancement over conventional antibiotic treatments that often face challenges due to resistance mechanisms. Overall, this innovative approach highlights the potential of nanozymes as versatile tools in combating antimicrobial resistance (AMR) infections and promoting wound healing and tissue regeneration. Graphene-based nanozymes represent a frontier in wound healing technology, combining antimicrobial action with enhanced cellular proliferation and tissue regeneration capabilities. Continued exploration into their mechanisms and applications will likely yield innovative solutions for effective wound management.

4.4 Utilization as antioxidants

Graphene-based nanozymes have garnered considerable interest due to their exceptional antioxidant properties, making them promising candidates for addressing oxidative stress-related diseases, including neurodegenerative disorders, cardiovascular diseases, and cancer. These nanomaterials, particularly graphene oxide and reduced graphene oxide (rGO), exhibit significant free radical scavenging capabilities, effectively neutralizing ROS, such as hydroxyl radicals and hydrogen peroxide (H2O2). Research indicates that GO can outperform traditional antioxidants in protecting biomolecules from oxidative damage, showcasing its potential as a potent hydroxyl radical scavenger. The antioxidant efficacy of graphene materials is attributed to their unique structural characteristics, including high surface area and sp2 carbon domains that facilitate electron transfer and radical adduct formation. Studies on graphene oxide quantum dots (GOQDs) have revealed their ability to significantly reduce ROS levels in neurotoxic environments, thereby preserving neuronal integrity.218 A novel and highly efficient graphene oxide–selenium (GO–Se) nanozyme with outstanding antioxidative properties, designed to protect cellular components from oxidative damage.219 This hybrid nanomaterial exhibited unique glutathione peroxidase (GPx)-like activity, effectively catalyzing the decomposition of hydrogen peroxide (H2O2) into harmless byproducts. Cell-based experiments confirmed its exceptional capacity to scavenge ROS, demonstrating its potential as a powerful antioxidant. These findings suggest that the GO–Se nanozyme could broaden the exploration of innovative selenium-based nanozymes.219 Overall, the multifunctional nature of graphene-based nanozymes positions them as valuable tools in the development of antioxidant therapies to mitigate oxidative stress across various medical applications. In-depth research is essential to optimize their design and fully understand their interactions within biological systems.

4.5 Graphene nanozymes in catalytic therapy

Catalytic therapy is a promising strategy for treating various diseases, including cancer and Alzheimer's, by using catalytic agents to trigger chemical reactions that either destroy cancer cells or break down toxic proteins associated with Alzheimer's disease. Because of their unique physicochemical properties, graphene-based nanozymes are particularly attractive as catalytic agents. They have high surface area-to-volume ratios, high electrical conductivity, and are highly biocompatible. These properties make them excellent candidates for use in catalytic therapy.

Graphene-based nanozymes possess unique properties that make them excellent candidates for targeted cancer treatment (Fig. 10).220 For instance, their high surface area enables optimal drug loading and delivery, while their catalytic activity facilitates the generation of ROS through oxidase and peroxidase activities. This ROS generation can selectively target and destroy cancer cells, offering a potential solution for the eradication of tumors. Furthermore, graphene-based nanozymes exhibit exceptional biocompatibility and can be easily functionalized to enhance their targeting capabilities.221 They have demonstrated outstanding potential in overcoming limitations associated with traditional cancer therapies, such as drug resistance and off-target effects. Additionally, these nanozymes can be combined with other therapeutic strategies, such as chemo-photothermal therapy, immunotherapy, and chemodynamic therapy, to enhance their efficacy further. These nanozymes have been the subject of extensive research owing to their intrinsic anticancer properties. Notably, they have demonstrated the ability to improve cell adhesion, capture cancer cells, and induce tumor cell death through an oxidative stress mechanism.


image file: d5ra04765j-f10.tif
Fig. 10 Properties of graphene-based nanozymes useful in catalytic cancer therapy.

Furthermore, studies have elucidated that graphene-based nanomaterials can reduce macrophage activity, thereby inhibiting cancer cell migration and invasion, ultimately suppressing tumor growth and metastasis.222 Moreover, the versatility of these nanomaterials extends to their application in diverse modalities, such as drug delivery, gene therapy, phototherapy, and magnetothermal therapy in the context of breast cancer treatment. The multifaceted nature of graphene-based nanomaterials positions them as promising candidates for revolutionary cancer therapeutic strategies, leveraging their unique ability to regulate ROS and contribute to the development of advanced cancer therapies.

By harnessing the unique properties of graphene-based nanozymes, catalytic cancer therapy offers a promising avenue for targeted and effective treatment of tumors. The integration of graphene-based nanozymes in cancer therapy represents a significant advancement in the field, opening up new possibilities for more personalized and efficient treatment options with reduced side effects. The remarkable potential of these nanomaterials has sparked a wave of optimism within the scientific and medical communities, prompting further exploration and innovation in the realm of cancer treatment.223 As research continues to unveil the intricate mechanisms and capabilities of graphene-based nanomaterials, the prospects for groundbreaking advancements in cancer therapy appear increasingly promising. One notable application of graphene-based nanozymes is in photothermal therapy (PTT), which utilizes light-absorbing materials to convert light energy into heat, selectively destroying cancer cells (Fig. 11). Graphene quantum dots (GQDs) and graphene oxide quantum dots (GOQDs) are two types of graphene-based nanozymes that have recently attracted intense focus due to their high catalytic activity, similar to natural enzymes.224 These nanomaterials act as photothermal agents when exposed to near-infrared light, generating local heat and damaging cancer cells. The exceptional electrical, thermal, size-dependent optical, and magnetic properties of GQDs make them suitable for PTT applications, which can be further enhanced by incorporating them into nanocomposites.225


image file: d5ra04765j-f11.tif
Fig. 11 Schematic representation of photothermal therapy in affected mouse model.

The potential of graphene-based nanozymes in PTT extends beyond their photothermal effect. These nanocomposites can also be utilized for drug loading and photodynamic impact, making them versatile tools in cancer treatment. Combining PTT with other therapies, such as chemotherapy or immunotherapy, holds great potential for improving cancer treatment outcomes. For instance, Hu et al. synthesized a graphene quantum dots/semiconducting polymer nanocomposite (GQD–SPN) that serves as both a temperature-sensitive nanozyme and a photothermal conversion agent.125 GQD–SPNs, upon NIR laser irradiation, not only generate a massive concentration of 1O2 but can also convert most of the optical energy into heat, thus raising the temperature within tumor regions locally. This enhanced photothermal effect further increases the peroxidase-like (POD) activity of the GQDs in the nanocomposite. The enhanced POD-like activity enables the GQD–SPNs to catalyze the endogenous hydrogen peroxide (H2O2) present in the tumor environment, leading to an abundance of hydroxyl radicals [( with combining macron]OH). The synergistic action of the generated 1O2, heat, and OH radicals produced by GQD–SPN nanocomposite under NIR irradiation resulted in enhanced outcomes in cancer therapy. This multifunctional nano platform further demonstrated the potential of rational graphene quantum dot-based nanomaterials for enhanced photodynamic and photothermal cancer treatment.125

The photothermal effect generated by graphene-based nanozymes can exert a therapeutic effect on cells, thereby enhancing the effectiveness of anticancer agents. However, further research is needed to fully understand the in vivo behaviors of graphene-based nanomaterials and their long-term toxicology. In addition to PTT, graphene-based nanozymes have also shown promise in photodynamic therapy (PDT). This advanced two-stage therapeutic strategy utilizes light energy in combination with a drug (photosensitizer) to selectively eliminate cancer cells. The photosensitizer is nontoxic until it is activated by light.49 Graphene-based nanozymes can be utilized as photosensitizers in PDT to generate ROS, such as singlet oxygen, which can induce the death of cancerous cells or pre-cancerous cells. Graphene nanozymes in PDT offer several advantages, including their excellent photothermal properties, large surface area for loading photosensitizers, and the ability to be easily functionalized for specific delivery to cancer cells.

In a study conducted by Maji et al., a novel nanostructured hybrid, GSF@AuNPs, was synthesized to serve as a mimetic enzyme for in vitro diagnostic and therapeutic interventions in cancer cells.45 The mixed material was created by attaching AuNPs to tiny pieces of reduced graphene oxide that had mesoporous silica on top. This nanozyme combines peroxidase-like activity and unique intrinsic properties of nanomaterials, making it suitable for imaging applications and the generation of toxic ROS under hypoxic conditions in the tumor microenvironment.45 In another study by Alizadeh et al., a CuO/WOx-NPs decorated graphene oxide nanosheet was fabricated, showing enhanced peroxidase activity.156 The eradication of cancer cells has been achieved by generating O[H with combining macron] radicals from both externally supplied and internally generated H2O2, facilitated by CuO/WO3-GO. Furthermore, Lin et al. synthesized graphene oxide nanoparticles (N-GO) by the chemical oxidation method, which augmented the ROS level in acidic pH conditions.226 In the acidic tumor microenvironment with high H2O2 levels, N-GOs catalyzed the oxidation of H2O2 into highly toxic hydroxyl radicals (HŌ), resulting in the necrosis of tumor cells. Conversely, in a normal cell microenvironment with neutral pH, the N-GOs exhibit catalase-like activity, effectively scavenging ROS and causing no harm to normal cells. This selective behavior of N-GOs makes them highly promising for targeted action in the tumor microenvironment.226 Another study focusing on ferroptosis and apoptosis investigated a novel metal-free inducer based on boron and nitrogen co-doped graphdiyne (BN-GDY).144 This innovative nanozyme exhibited efficient glutathione (GSH) depletion, leading to the induction of ferroptosis by inactivating GSH-dependent peroxidases (GPX4) and promoting apoptosis by downregulating Bcl-2. The high catalytic activity of BN-GDY was supported by kinetic experiments and density functional theory (DFT) calculations. Notably, the research identified a unique Bi–Bi mechanism for the nanozyme's action, as shown in Fig. 12, which differed from the conventional ping-pong Bi–Bi mechanism observed in most peroxidase mimics and natural enzymes. The findings suggested that BN-GDY could serve as a promising therapeutic agent in cancer treatment by leveraging its dual ability to induce both ferroptosis and apoptosis, thereby enhancing cell death in cancer cells.144 This nonmetal approach to inducing cell death through graphene-based nanozymes offers a novel strategy for nanocatalytic medicine, potentially paving the way for innovative treatments in oncology.


image file: d5ra04765j-f12.tif
Fig. 12 Illustration of BN-GDY regulation of ferroptosis and apoptosis signal pathways divided into three categories (a) membrane impairment, (b) lysosome dysfunction and (c) mitochondrial damage. Reprinted with permission from ref. 144. Copyright 2022 American Chemical Society.

Graphene-based nanozymes represent a compelling frontier in the advancement of cancer therapy. Their multifaceted capabilities, ranging from serving as effective drug delivery vehicles to addressing the challenges posed by the tumor microenvironment, underscore their potential to revolutionize the landscape of cancer therapeutic strategies. As research in this field continues to evolve, graphene-based nanozymes are poised to play a pivotal role in shaping the future of cancer treatment modalities.

4.6 Miscellaneous biomedical applications

Graphene-based nanozymes are gaining attention for their diverse applications beyond their well-documented biomedical and environmental uses. To illustrate, alcohol intoxication, also known as alcohol poisoning or alcohol overdose, occurs when a person consumes a large amount of alcohol in a short period, leading to a dangerous and potentially life-threatening situation such as steatosis, alcoholic hepatitis, and cirrhosis. Alcohol is a depressant that affects the central nervous system, leading to impaired judgment, coordination, and cognitive function. Alcohol poisoning is a medical emergency that requires immediate attention. It can lead to serious complications, including dehydration, brain damage, and even death. Sun et al. developed graphene oxide quantum dots (GOQDs) that effectively mitigated the decline in cell viability caused by ethanol, serving as nanozymes to expedite ethanol metabolism and prevent the buildup of harmful byproducts within cells.227 GOQDs alleviated the detrimental effects on mitochondria and the excessive production of free radicals. This discovery suggests that GOQDs can mitigate the adverse effects caused by ethanol.227

A recent study explored the neuroprotective effects of graphene oxide quantum dots (GOQDs) compared to larger graphene oxide (GO) nanosheets, explicitly focusing on their ability to combat oxidative stress and neurotoxicity.218 The researchers demonstrated that GOQDs significantly reduced levels of ROS and hydrogen peroxide in PC12 cells exposed to 1-methyl-4-phenyl-pyridinium ion, a commonly used model for neurotoxicity. In vivo experiments using zebrafish models revealed that GOQDs diminished ROS and apoptosis, enhanced locomotive activity, and preserved neuronal structures, indicating superior neuroprotective capabilities. The underlying mechanisms for these effects involved the modulation of metabolic pathways related to antioxidation and neurotransmission, as well as catalase-like activity that aided in the decomposition of H2O2. The study highlighted the potential of biocompatible GOQDs as effective agents for reducing oxidative stress and neurotoxicity, suggesting promising therapeutic applications in neurological health. Overall, the findings emphasized the advantages of using GOQDs over larger GO nanosheets in developing strategies to mitigate neurodegenerative conditions associated with oxidative damage.218

5. Challenges and future perspectives

Graphene-based nanozymes, while promising, face several limitations that hinder their practical application. Key challenges include issues with stability and reproducibility, particularly in environments with unstable substrates, such as hydrogen peroxide (H2O2), which can lead to inconsistent performance. Additionally, these nanozymes are susceptible to interference from other substances in complex biochemical environments, complicating their use in biosensing applications. Functionalization limitations also pose a challenge, as achieving optimal performance without compromising the unique properties of graphene is crucial. Furthermore, the long-term biocompatibility of these nanozymes remains uncertain, necessitating further research to ensure their safe use in biomedical applications.

To address these challenges, future research should focus on developing more robust formulations and improving functionalization techniques to enhance stability and performance. Comprehensive studies on biocompatibility will be crucial to ensure the safe application of these technologies in clinical settings. Additionally, prioritizing regulatory compliance and safety assessments will facilitate broader acceptance in various applications. Further exploration of graphene-based nanozymes could lead to the development of even more sensitive and specific diagnostic tools in biosensing. Integrating these nanozymes with advanced technologies, such as microfluidics and wearable devices, could revolutionize point-of-care diagnostics, enabling rapid and accurate testing in various settings. In the realm of catalytic cancer therapy, ongoing studies on graphene-based nanozymes aim to enhance their efficacy and improve their delivery systems. This includes exploring novel targeted delivery methods and synergistic effects combined with other therapeutic approaches. These advancements could eventually lead to more effective and personalized cancer treatments that minimize side effects and improve patient outcomes. The rise of graphene-based nanozymes in biosensing, anti-bacterial, wound healing, and catalytic cancer therapy holds immense potential for advancements in healthcare. Continued research and innovation in this field will likely pave the way for transformative diagnostic techniques and more effective treatments, improving the lives of countless individuals in the future.

6. Conclusion

In conclusion, the emergence of nanozymes, particularly graphene-based nanozymes, has opened up exciting possibilities in the fields of biosensing and catalytic cancer therapy. These innovative nanomaterials exhibit enzyme-like properties, enabling them to mimic the functions of natural enzymes and overcome the limitations of natural enzymes. This review summarizes the synthesis and catalytic properties of graphene-based nanozymes, as well as their applications in biosensing and cancer therapy. Graphene-based nanozymes in biosensing offer great potential for improved diagnostic techniques. Their high sensitivity, selectivity, and stability enable the detection and quantification of various biomarkers with high precision. This has significant implications for early disease detection and the development of personalized medicine. Furthermore, the application of graphene-based nanozymes in catalytic cancer therapy shows promise in revolutionizing cancer treatment. With the ability to target specific cancer cells and trigger catalytic reactions, these nanozymes offer a targeted and efficient approach for destroying cancer cells while minimizing damage to healthy tissues. This could potentially revolutionize cancer treatment by providing more effective and less invasive therapies.

Conflicts of interest

The authors declare that there is no conflict of interest.

Data availability

No primary research results have been included, and no new data has been generated as part of this review.

Acknowledgements

The authors are grateful to the Principal of Deshbandhu College, University of Delhi, New Delhi, for providing research support.

References

  1. H. Y. Shin, T. J. Park and M. I. Kim, Recent Research Trends and Future Prospects in Nanozymes, J. Nanomater., 2015, 2015, 756278 CrossRef.
  2. W. Xiao-Yu and W. Hui, Advance in Peroxidase-like Nanozymes and Their Analytical Applications, Chin. J. Anal. Chem., 2023, 51(5), 666–680 Search PubMed.
  3. X. Y. Wang, Y. H. Hu and H. Wei, Nanozymes in bionanotechnology: from sensing to therapeutics and beyond, Inorg. Chem. Front., 2016, 3, 41–60 RSC.
  4. H. Wei and E. Wang, Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes, Chem. Soc. Rev., 2013, 42, 6060–6093 RSC.
  5. Y. Huang, J. Ren and X. Qu, Nanozymes: classification, catalytic mechanisms, activity regulation, and applications, Chem. Rev., 2019, 119, 4357–4412 CrossRef CAS PubMed.
  6. K. Yang, L. Feng, X. Shi and Z. Liu, Nano-graphene in biomedicine: theranostic applications, Chem. Soc. Rev., 2013, 42, 530–547 RSC.
  7. M. Liang and X. Yan, Nanozymes: from new concepts, mechanisms, and standards to applications, Acc. Chem. Res., 2019, 52, 2190–2200 CrossRef CAS PubMed.
  8. B. Liu, Y. Wang, Y. Chen, L. Guo and G. Wei, Biomimetic two-dimensional nanozymes: Synthesis, hybridization, functional tailoring, and biosensor applications, J. Mater. Chem. B, 2020, 8, 10065–10086 RSC.
  9. Y. F. Liu, X. Y. Wang and H. Wei, Light-responsive nanozymes for biosensing, Analyst, 2020, 145, 4388–4397 RSC.
  10. J. Wu, X. Wang, Q. Wang, Z. Lou, S. Li, Y. Zhu, L. Qin and H. Wei, Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes (II), Chem. Soc. Rev., 2019, 48, 1004–1076 RSC.
  11. Q. Liu, A. Zhang, R. Wang, Q. Zhang and D. Cui, A review on metal-and metal oxide-based nanozymes: properties, mechanisms, and applications, Nano-Micro Lett., 2021, 13, 1–53 CrossRef PubMed.
  12. L. Gao, J. Zhuang, L. Nie, J. Zhang, Y. Zhang, N. Gu, T. Wang, J. Feng, D. Yang, S. Perrett and X. Yan, Intrinsic peroxidase-like activity of ferromagnetic nanoparticles, Nat. Nanotechnol., 2007, 2, 577–583 CrossRef CAS PubMed.
  13. X. Zhang, G. Li, D. Wu, X. Li, N. Hu, J. Chen, G. Chen and Y. Wu, Recent progress in the design fabrication of metal-organic frameworks-based nanozymes and their applications to sensing and cancer therapy, Biosens. Bioelectron., 2019, 137, 178–198 CrossRef CAS PubMed.
  14. S. F. Cai and R. Yang, Two-Dimensional Nanomaterials With Enzyme-Like Properties for Biomedical Applications, Front. Chem., 2020, 8, 565940 CrossRef CAS PubMed.
  15. S. Jin, C. Wu, Z. Ye and Y. Ying, Designed inorganic nanomaterials for intrinsic peroxidase mimics: A review, Sens. Actuators, B, 2019, 283, 18–34 CrossRef CAS.
  16. H. Chen, C. Wenqiang, F. Linhui, Z. Sufeng and L. Chen, Covalent organic frameworks (COFs) materials in enzyme immobilization and mimic enzymes, Prog. Chem., 2020, 32, 895 Search PubMed.
  17. D. Zhang, Q. Chen, Q. Ren, W. Zhong, H. Zhang, G. Wang and Y. Zhang, Transition metal-based nanozymes: Classification, catalytic mechanisms and emerging biomedical applications, Coord. Chem. Rev., 2024, 508, 215771 CrossRef CAS.
  18. Y. F. Chen, Y. Zhang, L. Jiao, H. Y. Yan, W. L. Gu and C. Z. Zhu, Research Progress of Carbon-based Nanozymes for Biosensing, Chin. J. Anal. Chem., 2021, 49, 907–921 CAS.
  19. L. Zhang, H. Wang and X. Qu, Biosystem-inspired engineering of nanozymes for biomedical applications, Adv. Mater., 2024, 36, 2211147 CrossRef CAS PubMed.
  20. J. Han and J. Yoon, Supramolecular nanozyme-based cancer catalytic therapy, ACS Appl. Bio Mater., 2020, 3, 7344–7351 CrossRef CAS PubMed.
  21. Z. Xu, P. Sun, J. Zhang, X. Lu, L. Fan, J. Xi, J. Han and R. Guo, High-efficiency platinum–carbon nanozyme for photodynamic and catalytic synergistic tumor therapy, Chem. Eng. J., 2020, 399, 125797 CrossRef CAS.
  22. Z. Gao, Y. Li, Y. Zhang, K. Cheng, P. An, F. Chen, J. Chen, C. You, Q. Zhu and B. Sun, Biomimetic platinum nanozyme immobilized on 2D metal–organic frameworks for mitochondrion-targeting and oxygen self-supply photodynamic therapy, ACS Appl. Mater. Interfaces, 2019, 12, 1963–1972 CrossRef PubMed.
  23. Y. Liu, Y. Qing, L. Jing, W. Zou and R. Guo, Platinum–copper bimetallic colloid nanoparticle cluster nanozymes with multiple enzyme-like activities for scavenging reactive oxygen species, Langmuir, 2021, 37, 7364–7372 CrossRef CAS PubMed.
  24. Y. Lin, X. Liu, Z. Liu and Y. Xu, Visible-light-driven photocatalysis-enhanced nanozyme of TiO2 nanotubes@ MoS2 nanoflowers for efficient wound healing infected with multidrug-resistant bacteria, Small, 2021, 17, 2103348 CrossRef CAS PubMed.
  25. A. Ali, J. Liu, H. Zhou, T. Liu, M. Ovais, H. Liu, Y. Rui and C. Chen, Graphdiyne–hemin-mediated catalytic system for wound disinfection and accelerated wound healing, Mater. Chem. Front., 2021, 5, 6041–6051 RSC.
  26. G.-L. Wang, L.-Y. Jin, X.-M. Wu, Y.-M. Dong and Z.-J. Li, Label-free colorimetric sensor for mercury (II) and DNA on the basis of mercury (II) switched-on the oxidase-mimicking activity of silver nanoclusters, Anal. Chim. Acta, 2015, 871, 1–8 CrossRef CAS PubMed.
  27. J.-G. You, Y.-W. Liu, C.-Y. Lu, W.-L. Tseng and C.-J. Yu, Colorimetric assay of heparin in plasma based on the inhibition of oxidase-like activity of citrate-capped platinum nanoparticles, Biosens. Bioelectron., 2017, 92, 442–448 CrossRef CAS PubMed.
  28. R. Long, K. Mao, X. Ye, W. Yan, Y. Huang, J. Wang, Y. Fu, X. Wang, X. Wu and Y. Xie, Surface facet of palladium nanocrystals: a key parameter to the activation of molecular oxygen for organic catalysis and cancer treatment, J. Am. Chem. Soc., 2013, 135, 3200–3207 CrossRef CAS PubMed.
  29. W. He, Y. Liu, J. Yuan, J.-J. Yin, X. Wu, X. Hu, K. Zhang, J. Liu, C. Chen and Y. Ji, Au@ Pt nanostructures as oxidase and peroxidase mimetics for use in immunoassays, Biomaterials, 2011, 32, 1139–1147 CrossRef CAS PubMed.
  30. L. Su, W. Dong, C. Wu, Y. Gong, Y. Zhang, L. Li, G. Mao and S. Feng, The peroxidase and oxidase-like activity of NiCo2O4 mesoporous spheres: mechanistic understanding and colorimetric biosensing, Anal. Chim. Acta, 2017, 951, 124–132 CrossRef CAS PubMed.
  31. S. G. Liu, L. Han, N. Li, N. Xiao, Y. J. Ju, N. B. Li and H. Q. Luo, A fluorescence and colorimetric dual-mode assay of alkaline phosphatase activity via destroying oxidase-like CoOOH nanoflakes, J. Mater. Chem. B, 2018, 6, 2843–2850 RSC.
  32. Z. Lin, X. Zhang, S. Liu, L. Zheng, Y. Bu, H. Deng, R. Chen, H. Peng, X. Lin and W. Chen, Colorimetric acid phosphatase sensor based on MoO3 nanozyme, Anal. Chim. Acta, 2020, 1105, 162–168 CrossRef CAS PubMed.
  33. A. B. Ganganboina and R.-a. Doong, The biomimic oxidase activity of layered V2O5 nanozyme for rapid and sensitive nanomolar detection of glutathione, Sens. Actuators, B, 2018, 273, 1179–1186 CrossRef CAS.
  34. X. Zhang and Y. Huang, Evaluation of the antioxidant activity of phenols and tannic acid determination with Mn 3 O 4 nano-octahedrons as an oxidase mimic, Anal. Methods, 2015, 7, 8640–8646 RSC.
  35. W. Qin, L. Su, C. Yang, Y. Ma, H. Zhang and X. Chen, Colorimetric detection of sulfite in foods by a TMB–O2–Co3O4 nanoparticles detection system, J. Agric. Food Chem., 2014, 62, 5827–5834 CrossRef CAS PubMed.
  36. W. Lu, J. Shu, Z. Wang, N. Huang and W. Song, The intrinsic oxidase-like activity of Ag2O nanoparticles and its application for colorimetric detection of sulfite, Mater. Lett., 2015, 154, 33–36 CrossRef CAS.
  37. C. Li, Y. Sun, X. Li, S. Fan, Y. Liu, X. Jiang, M. D. Boudreau, Y. Pan, X. Tian and J.-J. Yin, Bactericidal effects and accelerated wound healing using Tb 4 O 7 nanoparticles with intrinsic oxidase-like activity, J. Nanobiotechnol., 2019, 17, 1–10 CrossRef PubMed.
  38. W. Zhen, Y. Liu, W. Wang, M. Zhang, W. Hu, X. Jia, C. Wang and X. Jiang, Specific “unlocking” of a nanozyme-based butterfly effect to break the evolutionary fitness of chaotic tumors, Angew. Chem., 2020, 132, 9578–9584 CrossRef.
  39. Y. Xiong, S. Chen, F. Ye, L. Su, C. Zhang, S. Shen and S. Zhao, Synthesis of a mixed valence state Ce-MOF as an oxidase mimetic for the colorimetric detection of biothiols, Chem. Commun., 2015, 51, 4635–4638 RSC.
  40. J. Yu, X. Ma, W. Yin and Z. Gu, Synthesis of PVP-functionalized ultra-small MoS 2 nanoparticles with intrinsic peroxidase-like activity for H 2 O 2 and glucose detection, RSC Adv., 2016, 6, 81174–81183 RSC.
  41. S. Chen, M. Chi, Z. Yang, M. Gao, C. Wang and X. Lu, Carbon dots/Fe 3 O 4 hybrid nanofibers as efficient peroxidase mimics for sensitive detection of H 2 O 2 and ascorbic acid, Inorg. Chem. Front., 2017, 4, 1621–1627 RSC.
  42. L. Huang, W. Zhu, W. Zhang, K. Chen, J. Wang, R. Wang, Q. Yang, N. Hu, Y. Suo and J. Wang, Layered vanadium (IV) disulfide nanosheets as a peroxidase-like nanozyme for colorimetric detection of glucose, Microchim. Acta, 2018, 185, 1–8 CrossRef CAS PubMed.
  43. L. Zuo, H. King, M. A. Hossain, F. Farhana, M. M. Kist, R. L. Stratton, J. Chen and H. Shen, Single-Molecule Spectroscopy Reveals the Plasmon-Assisted Nanozyme Catalysis on AuNR@TiO2, Chem. Biomed. Imaging, 2023, 1, 760–766 CrossRef CAS PubMed.
  44. N. Bellier, P. Baipaywad, N. Ryu, J. Y. Lee and H. Park, Recent biomedical advancements in graphene oxide-and reduced graphene oxide-based nanocomposite nanocarriers, Biomater. Res., 2022, 26, 65 CrossRef CAS PubMed.
  45. S. K. Maji, A. K. Mandal, K. T. Nguyen, P. Borah and Y. Zhao, Cancer cell detection and therapeutics using peroxidase-active nanohybrid of gold nanoparticle-loaded mesoporous silica-coated graphene, ACS Appl. Mater. Interfaces, 2015, 7, 9807–9816 CrossRef CAS PubMed.
  46. H. Sun, N. Gao, K. Dong, J. Ren and X. Qu, Graphene quantum dots-band-aids used for wound disinfection, ACS Nano, 2014, 8, 6202–6210 CrossRef CAS PubMed.
  47. Z. Wang, K. Dong, Z. Liu, Y. Zhang, Z. Chen, H. Sun, J. Ren and X. Qu, Activation of biologically relevant levels of reactive oxygen species by Au/g-C3N4 hybrid nanozyme for bacteria killing and wound disinfection, Biomaterials, 2017, 113, 145–157 CrossRef CAS PubMed.
  48. L.-P. Chen, X.-Y. Wang, M.-J. Ren, Y. Wang, J.-M. Zhao, T.-T. Qiang, L.-Y. Dong and X.-H. Wang, Promoting the healing of infected diabetic wound by nanozyme-containing hydrogel with anti-bacterial inflammation suppressing, ROS-scavenging and oxygen-generating properties, J. Biomed. Mater. Res., Part B, 2024, 112, e35458 CrossRef CAS PubMed.
  49. P. Kumari, R. Gautam and A. Milhotra, Application of Porphyrin nanomaterials in Photodynamic therapy, Chem. Biol. Lett., 2016, 3, 32–37 Search PubMed.
  50. S. Kumar, P. Kumari and R. Singh, Emerging Nanomaterials for Cancer Therapy, in Nanoparticles in Medicine, ed. A. K. Shukla, Springer Singapore, Singapore, 2020, pp. 25–54 Search PubMed.
  51. S. Campuzano, M. Pedrero, P. Yáñez-Sedeño and J. M. Pingarrón, Nanozymes in electrochemical affinity biosensing, Microchim. Acta, 2020, 187, 1–16 CrossRef PubMed.
  52. L. A. Dinu and S. Kurbanoglu, Enhancing electrochemical sensing through the use of functionalized graphene composites as nanozymes, Nanoscale, 2023, 15, 16514–16538 RSC.
  53. B. Garg, T. Bisht and Y.-C. Ling, Graphene-based nanomaterials as efficient peroxidase mimetic catalysts for biosensing applications: an overview, Molecules, 2015, 20, 14155–14190 CrossRef CAS PubMed.
  54. E. Sánchez-Tirado, P. Yáñez-Sedeño and J. M. Pingarrón, Carbon-based enzyme mimetics for electrochemical biosensing, Micromachines, 2023, 14, 1746 CrossRef.
  55. H. Wang, C. Liu, Z. Liu, J. Ren and X. Qu, Specific Oxygenated Groups Enriched Graphene Quantum Dots as Highly Efficient Enzyme Mimics, Small, 2018, 14, 1703710 CrossRef PubMed.
  56. Y. Song, K. Qu, C. Zhao, J. Ren and X. Qu, Graphene oxide: intrinsic peroxidase catalytic activity and its application to glucose detection, Adv. Mater., 2010, 22, 2206–2210 CrossRef CAS PubMed.
  57. D. C. Marcano, D. V. Kosynkin, J. M. Berlin, A. Sinitskii, Z. Sun, A. Slesarev, L. B. Alemany, W. Lu and J. M. Tour, Improved synthesis of graphene oxide, ACS Nano, 2010, 4, 4806–4814 CrossRef CAS PubMed.
  58. M. S. Kim, J. Lee, H. S. Kim, A. Cho, K. H. Shim, T. N. Le, S. S. A. An, J. W. Han, M. I. Kim and J. Lee, Heme Cofactor-Resembling Fe–N Single Site Embedded Graphene as Nanozymes to Selectively Detect H2O2 with High Sensitivity, Adv. Funct. Mater., 2020, 30, 1905410 CrossRef CAS.
  59. Raveena, Alka, N. Gandhi and P. Kumari, Efficacious Removal of Flonicamid Insecticide from Water by GO@functionalized Calix[4]pyrrole: Synergistic Effect in Adsorption, ChemistrySelect, 2023, 8, e202203431 CrossRef CAS.
  60. A. Singhal, P. Kumari and K. Nisa, Facile One-Pot Friedlander Synthesis of Functionalized Quinolines using Graphene Oxide Carbocatalyst, Curr. Org. Synth., 2019, 16, 154–159 CrossRef CAS PubMed.
  61. V. Panwar, A. Chattree and K. Pal, A new facile route for synthesizing of graphene oxide using mixture of sulfuric–nitric–phosphoric acids as intercalating agent, Phys. E, 2015, 73, 235–241 CrossRef CAS.
  62. P. K. Boruah, P. Borthakur, G. Neog, B. Le Ouay, N. U. Afzal, P. Manna and M. R. Das, Porous Nitrogen-Doped Crumpled Graphene Nanoparticles: A Metal-Free Nanozyme for Selective Detection of Dopamine in Aqueous Medium and Human Serum, ACS Appl. Nano Mater., 2023, 6, 1667–1677 CrossRef CAS.
  63. N. N. Lu, Y. Liu, X. Y. Yan, Z. Q. Xu, Y. Xing, Y. Song, P. Y. Zhao, M. H. Liu, Y. Gu, Z. Q. Zhang and S. Y. Zhai, Bioinspired Surface Modification of Graphene-Based Hybrids as Nanozyme Sensors for Simultaneous Detection of Dopamine and Uric Acid, ACS Appl. Nano Mater., 2022, 5, 11361–11370 CrossRef CAS.
  64. E. Vermisoglou, T. Giannakopoulou, M. Giannouri, N. Boukos, C. Lekakou and C. Trapalis, Single-step hydrothermal synthesis of reduced graphene oxide (rGO) and Fe2O3/rGO composites for supercapacitor applications, 16th European Conference on Composite Materials, ECCM 2014, 2014 Search PubMed.
  65. J. Zhao, Z. Shi, M. Chen and F. Xi, Highly active nanozyme based on nitrogen-doped graphene quantum dots and iron ion nanocomposite for selective colorimetric detection of hydroquinone, Talanta, 2025, 281, 126817 CrossRef CAS PubMed.
  66. X. Deng, J. Zhao, Y. Ding, H. Tang and F. Xi, Iron and nitrogen co-doped graphene quantum dots as highly active peroxidases for the sensitive detection of l-cysteine, New J. Chem., 2021, 45, 19056–19064 RSC.
  67. Q. Li, D. S. Yu, C. Y. Fan, Q. L. Huang, Y. H. Tang, R. R. Guo, Y. F. Huang, H. Wang, C. X. Lin and Y. H. Lin, Gold Nanoparticles Adsorbed on Graphene as Nanozymes for the Efficient Elimination of Dye Pollutants, ACS Appl. Nano Mater., 2022, 5, 94–100 CrossRef CAS.
  68. C. Liu, Y. M. Zhao, D. Xu, X. X. Zheng and Q. Huang, A green and facile approach to a graphene-based peroxidase-like nanozyme and its application in sensitive colorimetric detection of l-cysteine, Anal. Bioanal. Chem., 2021, 413, 4013–4022 CrossRef CAS PubMed.
  69. F. Liu, C. Wang, X. Sui, M. A. Riaz, M. Xu, L. Wei and Y. Chen, Synthesis of graphene materials by electrochemical exfoliation: Recent progress and future potential, Carbon Energy, 2019, 1, 173–199 CrossRef.
  70. X. Li, G. Lin, L. Zhou, O. Prosser, M. H. Malakooti and M. Zhang, Green synthesis of iron-doped graphene quantum dots: an efficient nanozyme for glucose sensing, Nanoscale Horiz., 2024, 9, 976–989 RSC.
  71. P. Borthakur, P. K. Boruah and M. R. Das, CuS and NiS Nanoparticle-Decorated Porous-Reduced Graphene Oxide Sheets as Efficient Peroxidase Nanozymes for Easy Colorimetric Detection of Hg(II) Ions in a Water Medium and Using a Paper Strip, ACS Sustainable Chem. Eng., 2021, 9, 13245–13255 CrossRef CAS.
  72. M. Bahri, S. H. Gebre, M. A. Elaguech, F. T. Dajan, M. G. Sendeku, C. Tlili and D. Wang, Recent advances in chemical vapour deposition techniques for graphene-based nanoarchitectures: From synthesis to contemporary applications, Coord. Chem. Rev., 2023, 475, 214910 CrossRef CAS.
  73. G. Deokar, J. Jin, U. Schwingenschlögl and P. M. F. J. Costa, Chemical vapor deposition-grown nitrogen-doped graphene's synthesis, characterization and applications, npj 2D Mater. Appl., 2022, 6, 14 CrossRef CAS.
  74. M. Saeed, Y. Alshammari, S. A. Majeed and E. Al-Nasrallah, Chemical Vapour Deposition of Graphene-Synthesis, Characterisation, and Applications: A Review, Molecules, 2020, 25(17), 3856 CrossRef CAS PubMed.
  75. R. Yang, Z. Liu, H. Chen, X. Zhang, Q. Sun, H. S. El-Mesery, W. Lu, X. Dai and R. Xu, Advances in Nanozyme Catalysis for Food Safety Detection: A Comprehensive Review on Progress and Challenges, Foods, 2025, 14, 2580 CrossRef PubMed.
  76. J. He, Y. Hou, Z. Zhang, J. Zhang, X. Yan, K. Fan and M. Liang, Carbon-based Nanozymes: How Structure Affects Performance, Nano Biomed. Eng., 2024, 16, 28–47 CrossRef CAS.
  77. P. G. Le, D. Kim, J.-P. Chung and S. Cho, Peroxidase-Mimicking Nanozymes of Nitrogen Heteroatom-Containing Graphene Oxide for Biomedical Applications, Biosensors, 2025, 15, 435 CrossRef PubMed.
  78. N. Reckinger, A. Felten, C. N. Santos, B. Hackens and J.-F. Colomer, The influence of residual oxidizing impurities on the synthesis of graphene by atmospheric pressure chemical vapor deposition, Carbon, 2013, 63, 84–91 CrossRef CAS.
  79. X. Yan, Reproducible chemical vapor deposition of high quality graphene, Doctoral dissertation, Columbia University, 2025.
  80. M. Shams, L. M. Guiney, L. Huang, M. Ramesh, X. Yang, M. C. Hersam and I. Chowdhury, Influence of functional groups on the degradation of graphene oxide nanomaterials, Environ. Sci.:Nano, 2019, 6, 2203–2214 RSC.
  81. R. Li, X. Yang, M. Ma and J. Zhang, Hydrogen-Enhanced Catalytic Conversion of Amorphous Carbon to Graphene for Achieving Superlubricity, Small, 2023, 19, 2206580 CrossRef CAS PubMed.
  82. D. Wang, X. Song, P. Li, X. J. Gao and X. Gao, Origins of the peroxidase mimicking activities of graphene oxide from first principles, J. Mater. Chem., 2020, 8, 9028–9034 CAS.
  83. Y. Xu, J. Xue, Q. Zhou, Y. Zheng, X. Chen, S. Liu, Y. Shen and Y. Zhang, The Fe-N-C Nanozyme with Both Accelerated and Inhibited Biocatalytic Activities Capable of Accessing Drug–Drug Interactions, Angew. Chem., Int. Ed., 2020, 59, 14498–14503 CrossRef CAS PubMed.
  84. A. Eckmann, A. Felten, I. Verzhbitskiy, R. Davey and C. Casiraghi, Raman study on defective graphene: Effect of the excitation energy, type, and amount of defects, Phys. Rev. B:Condens. Matter Mater. Phys., 2013, 88, 035426 CrossRef.
  85. H. Fan, R. Zhang, K. Fan, L. Gao and X. Yan, Exploring the Specificity of Nanozymes, ACS Nano, 2024, 18, 2533–2540 CrossRef CAS PubMed.
  86. Y. Su, M. Lv, Z. Huang, N. An, Y. Chen, H. Wang, Z. Li, S. Wu, F. Ye, J. Shen and A. Li, Defect engineering to tailor structure-activity relationship in biodegradable nanozymes for tumor therapy by dual-channel death strategies, J. Controlled Release, 2024, 367, 557–571 CrossRef CAS PubMed.
  87. V. Garg, M. Kaur, M. K. Sangha and M. Javed, Ternary CTAB@Co3O4@GO nanocomposite as a promising superoxide dismutase mimic, Bull. Mater. Sci., 2022, 45, 145 CrossRef CAS.
  88. K. Kim, K. N. Chaudhari, S. Kim, Y. Kim and K. S. Shin, Facile single-step synthesis of Cu-rGO nanocomposite through simultaneous reduction process and its peroxidase mimic activity, J. Ind. Eng. Chem., 2021, 95, 388–396 CrossRef CAS.
  89. P. Kumari, S. Kumar, S. Gupta, A. Mishra and A. Kumar, Efficacious and selective oxidation of atrazine with hydrogen peroxide catalyzed by magnetite nanoparticles: influence of reaction media, ChemistrySelect, 2018, 3, 2135–2139 CrossRef CAS.
  90. P. Kumari, R. Gautam, H. Yadav, V. Kushwaha, A. Mishra, S. Gupta and V. Arora, Efficient reduction of C–N multiple bonds catalyzed by magnetically retrievable magnetite nanoparticles with sodium borohydride, Catal. Lett., 2016, 146, 2149–2156 CrossRef CAS.
  91. S. Kumar, Alka, Tarun, J. Saxena, C. Bansal and P. Kumari, Visible light-assisted photodegradation by silver tungstate-modified magnetite nanocomposite material for enhanced mineralization of organic water contaminants, Appl. Nanosci., 2020, 10, 1555–1569 CrossRef CAS.
  92. S. M. Chauhan and P. Kumari, Biomimetic oxidation of metribuzin with hydrogen peroxide catalyzed by 5, 10, 15, 20-tetraarylporphyrinatoiron (III) chlorides, Tetrahedron Lett., 2007, 48, 5035–5038 CrossRef CAS.
  93. P. Kumari, R. Nagpal and S. M. Chauhan, Efficient oxidation of polycyclic aromatic hydrocarbons with H2O2 catalyzed by 5, 10, 15-triarylcorrolatoiron (IV) chloride in ionic liquids, Catal. Commun., 2012, 29, 15–20 CrossRef CAS.
  94. P. Kumari, N. Sinha, P. Chauhan and S. M. S. Chauhan, Isolation, synthesis and biomimetic reactions of metalloporphyrinoids in ionic liquids, Curr. Org. Synth., 2011, 8, 393–437 CrossRef CAS.
  95. P. Kumari and S. Chauhan, Efficient cobalt (II) phthalocyanine-catalyzed reduction of flavones with sodium borohydride, Chem. Commun., 2009, 42, 6397–6399 RSC.
  96. Z. Guo, S. Chakraborty, F. A. Monikh, D. D. Varsou, A. J. Chetwynd, A. Afantitis, I. Lynch and P. Zhang, Surface functionalization of graphene-based materials: Biological behavior, toxicology, and safe-by-design aspects, Adv. Biol., 2021, 5, 2100637 CrossRef CAS.
  97. K. Li, W. Liu, Y. Ni, D. Li, D. Lin, Z. Su and G. Wei, Technical synthesis and biomedical applications of graphene quantum dots, J. Mater. Chem., 2017, 5, 4811–4826 CAS.
  98. H. Sun, A. Zhao, N. Gao, K. Li, J. Ren and X. Qu, Deciphering a Nanocarbon-Based Artificial Peroxidase: Chemical Identification of the Catalytically Active and Substrate-Binding Sites on Graphene Quantum Dots, Angew. Chem., Int. Ed., 2015, 54, 7176–7180 CrossRef CAS PubMed.
  99. E. M. Hamed, F. M. Fung and S. F. Li, Unleashing the Potential of Single-Atom Nanozymes: Catalysts for the Future, ACS Sens., 2024, 9, 3840–3847 CrossRef CAS PubMed.
  100. M.-H. Chan, B.-G. Chen, W.-T. Huang, T.-Y. Su, M. Hsiao and R.-S. Liu, Tunable single-atom nanozyme catalytic system for biological applications of therapy and diagnosis, Mater. Today Adv., 2023, 17, 100342 CrossRef CAS.
  101. Z. Guo, J. Hong, N. Song and M. Liang, Single-Atom Nanozymes: From Precisely Engineering to Extensive Applications, Acc. Mater. Res., 2024, 5, 347–357 CrossRef CAS.
  102. Y.-S. Park, B. U. Park and H.-J. Jeon, Advances in machine learning-enhanced nanozymes, Front. Chem., 2024, 12, 1483986 CrossRef CAS PubMed.
  103. N. Chen, C. He and W. Zhu, Lightweight machine-learning model for efficient design of graphene-based microwave metasurfaces for versatile absorption performance, Nanomaterials, 2023, 13, 329 CrossRef CAS PubMed.
  104. T. S. Alahmari and K. Arif, Machine learning approaches to predict the strength of graphene nanoplatelets concrete: Optimization and hyper tuning with graphical user interface, Mater. Today Commun., 2024, 40, 109946 CrossRef CAS.
  105. W. Huang, Y. Xu and Y. Sun, Functionalized Graphene Fiber Modified With MOF-Derived Rime-Like Hierarchical Nanozyme for Electrochemical Biosensing of H2O2 in Cancer Cells, Front. Chem., 2022, 10, 873187 CrossRef CAS.
  106. S. M. Tai, Q. L. Pan, X. J. Chen, C. F. Peng, C. Z. Zhang and Z. P. Wang, Selective inhibition toward the enzyme-like activity of 3D porous cerium-doped graphene oxide nanoribbons for highly sensitive and enzyme-free colorimetric detection of pesticides, Sens. Actuators, B, 2023, 378, 133130 CrossRef CAS.
  107. Z. Li, J. Zhang, G. Dai, F. Luo, Z. Chu, X. Geng, P. He, F. Zhang and Q. Wang, A ratiometric electrochemical biosensor for glycated albumin detection based on enhanced nanozyme catalysis of cuprous oxide-modified reduced graphene oxide nanocomposites, J. Mater. Chem. B, 2021, 9, 9324–9332 RSC.
  108. L. Liu, J. Du, W.-e. Liu, Y. Guo, G. Wu, W. Qi and X. Lu, Enhanced His@ AuNCs oxidase-like activity by reduced graphene oxide and its application for colorimetric and electrochemical detection of nitrite, Anal. Bioanal. Chem., 2019, 411, 2189–2200 CrossRef CAS PubMed.
  109. H. V. Tran, T. A. Le, B. L. Giang, B. Piro and L. Dai Tran, Silver nanoparticles on graphene quantum dots as nanozyme for efficient H2O2 reduction in a glucose biosensor, Mater. Res. Express, 2019, 6, 115403 CrossRef CAS.
  110. R. K. Sindhu, A. Najda, P. Kaur, M. D. S. Shah, H. Singh, P. Kaur, S. Cavalu, M. Jaroszuk-Sierocinska and M. H. Rahman, Potentiality of Nanoenzymes for Cancer Treatment and Other Diseases: Current Status and Future Challenges, Materials, 2021, 14, 5965 CrossRef CAS PubMed.
  111. M. Devi, P. Das, P. K. Boruah, M. J. Deka, R. Duarah, A. Gogoi, D. Neog, H. S. Dutta and M. R. Das, Fluorescent graphitic carbon nitride and graphene oxide quantum dots as efficient nanozymes: Colorimetric detection of fluoride ion in water by graphitic carbon nitride quantum dots, J. Environ. Chem. Eng., 2021, 9, 104803 CrossRef CAS.
  112. N. Salarizadeh, A. F. Kalurazi, S. S. Shahangian, E. Hadipour and B. Rasti, A Highly Stable Graphene-Based Metal Oxide Nano Enzyme with the Peroxidase-Mimetic Activity for Equipment-Free Diagnosis of Diabetic Patients, Chemistryselect, 2023, 8, e202300211 CrossRef CAS.
  113. R. Geetha Bai, K. Muthoosamy, S. Manickam and A. Hilal-Alnaqbi, Graphene-based 3D scaffolds in tissue engineering: fabrication, applications, and future scope in liver tissue engineering, Int. J. Nanomed., 2019, 14, 5753–5783 CrossRef PubMed.
  114. C. Chen, Y. Xi and Y. Weng, Progress in the development of graphene-based biomaterials for tissue engineering and regeneration, Materials, 2022, 15, 2164 CrossRef CAS PubMed.
  115. X. Wu, S.-J. Ding, K. Lin and J. Su, A review on the biocompatibility and potential applications of graphene in inducing cell differentiation and tissue regeneration, J. Mater. Chem. B, 2017, 5, 3084–3102 RSC.
  116. X. Z. Mou, Q. Y. Wu, Z. Zhang, Y. H. Liu, J. G. Zhang, C. W. Zhang, X. Y. Chen, K. L. Fan and H. Y. Liu, Nanozymes for Regenerative Medicine, Small Methods, 2022, 6, 2200997 CrossRef PubMed.
  117. M. Maleki, R. Zarezadeh, M. Nouri, A. R. Sadigh, F. Pouremamali, Z. Asemi, H. S. Kafil, F. Alemi and B. Yousefi, Graphene oxide: a promising material for regenerative medicine and tissue engineering, Biomol. Concepts, 2020, 11, 182–200 CAS.
  118. R. G. Bai, N. Ninan, K. Muthoosamy and S. Manickam, Graphene: A versatile platform for nanotheranostics and tissue engineering, Prog. Mater. Sci., 2018, 91, 24–69 CrossRef.
  119. N. Chakraborty, S. Gandhi, R. Verma and I. Roy, Emerging Prospects of Nanozymes for Antibacterial and Anticancer Applications, Biomedicines, 2022, 10, 1378 CrossRef CAS PubMed.
  120. W. Wang, B. Li, H. Yang, Z. Lin, L. Chen, Z. Li, J. Ge, T. Zhang, H. Xia and L. Li, Efficient elimination of multidrug-resistant bacteria using copper sulfide nanozymes anchored to graphene oxide nanosheets, Nano Res., 2020, 13, 2156–2164 CrossRef CAS.
  121. N. Lu, Y. Liu, X. Yan, Z. Xu, Y. Xing, Y. Song, P. Zhao, M. Liu, Y. Gu and Z. Zhang, Bioinspired Surface Modification of Graphene-Based Hybrids as Nanozyme Sensors for Simultaneous Detection of Dopamine and Uric Acid, ACS Appl. Nano Mater., 2022, 5, 11361–11370 CrossRef CAS.
  122. L. A. D. Gugoasa, F. Pogacean, S. Kurbanoglu, L.-B. Tudoran, A. B. Serban, I. Kacso and S. Pruneanu, Graphene-gold nanoparticles nanozyme-based electrochemical sensor with enhanced laccase-like activity for determination of phenolic substrates, J. Electrochem. Soc., 2021, 168, 067523 CrossRef CAS.
  123. Q. Wang, X. Zhang, L. Huang, Z. Zhang and S. Dong, One-pot synthesis of Fe3O4 nanoparticle loaded 3D porous graphene nanocomposites with enhanced nanozyme activity for glucose detection, ACS Appl. Mater. Interfaces, 2017, 9, 7465–7471 CrossRef CAS PubMed.
  124. N. Jahandar Shamami, S. Shahangiyan, S. S Afghahi, N. Salarizadeh and H. Zamani, Kinetic properties of a novel graphene/carbon-based peroxidase mimic nanozyme and its application for detection of glutathione in drugs, Nanoscale, 2021, 8, 68–79 Search PubMed.
  125. L. Hu, W. Sun, Y. Tang, S. Li, B. Zhang, X. Sun, W. Ji, L. Ma, H. Deng and S. Han, Photothermal effect enhancing graphene quantum dots/semiconducting polymer/nanozyme-mediated cancer catalytic therapy, Carbon, 2021, 176, 148–156 CrossRef CAS.
  126. H. Liu, Z. Deng, Z. Zhang, W. Lin, M. Zhang and H. Wang, Graphene quantum dots as metal-free nanozymes for chemodynamic therapy of cancer, Matter, 2024, 7, 977–990 CrossRef CAS.
  127. C. Zhou, R. Cheng, B. Liu, Y. Fang, K. Nan, W. Wu and Y. Xu, Cascade selective recognition of H2O2 and ascorbic acid in living cells using carbon-based nanozymes with peroxidase-like activity, Sens. Actuators, B, 2024, 402, 135118 CrossRef CAS.
  128. L. Chen, T. Hou, Y. Tan, C. Guo, B. Wang, L. Ge and F. Li, Laser-induced N-and B-codoped graphene nanozymes with intrinsic peroxidase-like activities for bactericidal application, ACS Sustainable Chem. Eng., 2022, 10, 2750–2760 CrossRef CAS.
  129. T. Zhang, Y. Liu, J. Pi, N. Lu, R. Zhang, W. Chen, Z. Zhang and D. Xing, A novel artificial peroxisome candidate based on nanozyme with excellent catalytic performance for biosensing, Biosens. Bioelectron., 2022, 196, 113686 CrossRef CAS PubMed.
  130. D.-D. Liu, F.-F. Zhang, M. Gao, J.-C. Zhou, Y.-F. Wang and Y.-Z. Lu, Pt nanoparticle/N-doped graphene nanozymes for colorimetric detection of acetylcholinesterase activity and inhibition, Chin. J. Anal. Chem., 2022, 50, 100177 Search PubMed.
  131. Q. J. Guo, Z. Y. Pan, C. Men, W. Y. Lv, H. Y. Zou and C. Z. Huang, Visual detection of cancer cells by using in situ grown functional Cu 2− x Se/reduced graphene oxide hybrids acting as an efficient nanozyme, Analyst, 2019, 144, 716–721 RSC.
  132. F. Honarasa, R. Mokhtare, A. Mokhtare and S. Yousefinejad, High performance nanozymatic assay-based CuO nanocluster supported by reduced graphene oxide for determination of hydrogen peroxide and ascorbic acid, Process Biochem., 2021, 111, 256–261 CrossRef CAS.
  133. X. Li, S. Li, Q. Lv, C. Wang, J. Liang, Z. Zhou and G. Li, Colorimetric biosensor for visual determination of Golgi protein 73 based on reduced graphene oxide-carboxymethyl chitosan-Hemin/platinum@palladium nanozyme with peroxidase-like activity, Microchim. Acta, 2022, 189, 392 CrossRef CAS PubMed.
  134. Y. Liu, N. Li, K. Su, J. Du and R. Guo, Arginine-rich peptide–rhodium nanocluster@ reduced graphene oxide composite as a highly selective and active uricase-like nanozyme for the degradation of uric acid and inhibition of urate crystal, Inorg. Chem., 2024, 63, 13602–13612 CrossRef CAS PubMed.
  135. S. Tai, H. Cao, A. O. Barimah, Y. Gao, C. Peng, J. Xu and Z. Wang, Highly sensitive colorimetric and paper-based detection for sildenafil in functional food based on monodispersed spherical magnetic graphene composite nanozyme, Anal. Chim. Acta, 2024, 1329, 343260 CrossRef CAS.
  136. Q. Zhao, W. Gou, X. Zhang, M. Zhang, Y. Bu, L. Wang, L. Hu, W. Yao and Z. Yan, Hg2+-activated oxidase-like activity of Ag2S@graphene oxide nanozyme and its naked-eye monitoring Hg2+ application with obvious hyperchromic effect, Appl. Surf. Sci., 2021, 545, 148973 CrossRef CAS.
  137. Z. Song, C. Jiang, F. Wang, L. Yu, S. Ye, P. Dramou and H. He, Nanozyme based on graphene oxide modified with Fe3O4, CuO, and cucurbit[6]uril for colorimetric determination of homocysteine, Microchim. Acta, 2021, 188, 207 CrossRef CAS PubMed.
  138. J. Jia, X. Du, X. Lv, F. Xie, J. Zhou and J. Cai, DNA-Mediated Graphene Oxide/Platinum Nanozymes for Label-Free Colorimetric Detection of Hg2+, ACS Appl. Nano Mater., 2023, 6, 15224–15233 CrossRef CAS.
  139. P.-C. Lee, N.-S. Li, Y.-P. Hsu, C. Peng and H.-W. Yang, Direct glucose detection in whole blood by colorimetric assay based on glucose oxidase-conjugated graphene oxide/MnO2 nanozymes, Analyst, 2019, 144, 3038–3044 RSC.
  140. P. G. Le, X. A. Le, H. S. Duong, S. H. Jung, T. Kim and M. I. Kim, Ultrahigh peroxidase-like catalytic performance of Cu–N4 and Cu–N4S active sites-containing reduced graphene oxide for sensitive electrochemical biosensing, Biosens. Bioelectron., 2024, 255, 116259 CrossRef CAS PubMed.
  141. H. Tian, J. Liu, J. Guo, L. Cao and J. He, L-Cysteine functionalized graphene oxide nanoarchitectonics: A metal-free Hg2+ nanosensor with peroxidase-like activity boosted by competitive adsorption, Talanta, 2022, 242, 123320 CrossRef CAS PubMed.
  142. L. Zhang, L. Zhang, H. Deng, H. Li, W. Tang, L. Guan, Y. Qiu, M. J. Donovan, Z. Chen and W. Tan, In vivo activation of pH-responsive oxidase-like graphitic nanozymes for selective killing of Helicobacter pylori, Nat. Commun., 2021, 12, 2002 CrossRef CAS PubMed.
  143. L. Tian, J. Qi, K. Qian, O. Oderinde, Y. Cai, C. Yao, W. Song and Y. Wang, An ultrasensitive electrochemical cytosensor based on the magnetic field assisted binanozymes synergistic catalysis of Fe3O4 nanozyme and reduced graphene oxide/molybdenum disulfide nanozyme, Sens. Actuators, B, 2018, 260, 676–684 CrossRef CAS.
  144. C. Zhang, L. Chen, Q. Bai, L. Wang, S. Li, N. Sui, D. Yang and Z. Zhu, Nonmetal graphdiyne nanozyme-based ferroptosis–apoptosis strategy for colon cancer therapy, ACS Appl. Mater. Interfaces, 2022, 14, 27720–27732 CrossRef CAS.
  145. J. Hu, X. He, J. Cai, Z. Zhang, X. Bai, S. Zhang, B. Geng, D. Pan and L. Shen, Biodegradable CoSnO3 nanozymes modulate pH-responsive graphene quantum dot release for synergistic chemo-sonodynamic-nanocatalytic cancer therapy, Chem. Eng. J., 2024, 481, 148561 CrossRef CAS.
  146. F. Meng, M. Peng, Y. Chen, X. Cai, F. Huang, L. Yang, X. Liu, T. Li, X. Wen, N. Wang, D. Xiao, H. Jiang, L. Xia, H. Liu and D. Ma, Defect-rich graphene stabilized atomically dispersed Cu3 clusters with enhanced oxidase-like activity for antibacterial applications, Appl. Catal., B, 2022, 301, 120826 CrossRef CAS.
  147. X. Guo, X. Zhang, M. Yu, Z. Cheng, Y. Feng and B. Chen, Iron decoration in binary graphene oxide and copper iron sulfide nanocomposites boosting catalytic antibacterial activity in acidic microenvironment against antimicrobial resistance, J. Colloid Interface Sci., 2024, 661, 802–814 CrossRef CAS PubMed.
  148. L. Wang, F. Gao, A. Wang, X. Chen, H. Li, X. Zhang, H. Zheng, R. Ji, B. Li, X. Yu, J. Liu, Z. Gu, F. Chen and C. Chen, Defect-Rich Adhesive Molybdenum Disulfide/rGO Vertical Heterostructures with Enhanced Nanozyme Activity for Smart Bacterial Killing Application, Adv. Mater., 2020, 32, 2005423 CrossRef CAS.
  149. M. Lu, J. Wang, G. Ren, F. Qin, Z. Zhao, K. Li, W. Chen and Y. Lin, Superoxide-like Cu/GO single-atom catalysts nanozyme with high specificity and activity for removing superoxide free radicals, Nano Res., 2022, 15, 8804–8809 CrossRef CAS.
  150. D. Liang, Y. Yang, G. Li, Q. Wang, H. Chen and X. Deng, Endogenous H(2)O(2)-Sensitive and Weak Acidic pH-Triggered Nitrogen-Doped Graphene Nanoparticles (N-GNMs) in the Tumor Microenvironment Serve as Peroxidase-Mimicking Nanozymes for Tumor-Specific Treatment, Materials, 2021, 14, 1933 CrossRef CAS PubMed.
  151. L. Li, C. Zeng, L. Ai and J. Jiang, Synthesis of reduced graphene oxide-iron nanoparticles with superior enzyme-mimetic activity for biosensing application, J. Alloys Compd., 2015, 639, 470–477 CrossRef CAS.
  152. L. Yuan, D. X. Lu and J. H. Li, Application of Colorimetric Biosensor Based on Nanozyme in Biomedical Detection, Prog. Biochem. Biophys., 2023, 50, 1638–1650 CAS.
  153. A. T. Lawal, Progress in utilisation of graphene for electrochemical biosensors, Biosens. Bioelectron., 2018, 106, 149–178 CrossRef CAS PubMed.
  154. Y. Li, Q. Wang, Z. Ding, D. Wan, X. Nie and C. Zhong, A functionalized magnetic graphene-based MOFs platform as the heterogeneous mimic enzyme sensor for glucose detection, Catal. Lett., 2022, 1–11 Search PubMed.
  155. Q. Wang, J. Lei, S. Deng, L. Zhang and H. Ju, Graphene-supported ferric porphyrin as a peroxidase mimic for electrochemical DNA biosensing, Chem. Commun., 2013, 49, 916–918 RSC.
  156. N. Alizadeh, A. Salimi, R. Hallaj, F. Fathi and F. Soleimani, CuO/WO3 nanoparticles decorated graphene oxide nanosheets with enhanced peroxidase-like activity for electrochemical cancer cell detection and targeted therapeutics, Mater. Sci. Eng., C, 2019, 99, 1374–1383 CrossRef CAS PubMed.
  157. J. Xi, C. Xie, Y. Zhang, L. Wang, J. Xiao, X. Duan, J. Ren, F. Xiao and S. Wang, Pd Nanoparticles Decorated N-Doped Graphene Quantum Dots@N-Doped Carbon Hollow Nanospheres with High Electrochemical Sensing Performance in Cancer Detection, ACS Appl. Mater. Interfaces, 2016, 8, 22563–22573 CrossRef CAS PubMed.
  158. M. Wu, S. Meng, Q. Wang, W. Si, W. Huang and X. Dong, Nickel–Cobalt Oxide Decorated Three-Dimensional Graphene as an Enzyme Mimic for Glucose and Calcium Detection, ACS Appl. Mater. Interfaces, 2015, 7, 21089–21094 CrossRef CAS PubMed.
  159. H. Wang, S. Li, Y. Si, Z. Sun, S. Li and Y. Lin, Recyclable enzyme mimic of cubic Fe3O4 nanoparticles loaded on graphene oxide-dispersed carbon nanotubes with enhanced peroxidase-like catalysis and electrocatalysis, J. Mater. Chem. B, 2014, 2, 4442–4448 RSC.
  160. M. S. Kim, S. Cho, S. H. Joo, J. Lee, S. K. Kwak, M. I. Kim and J. Lee, N- and B-Codoped Graphene: A Strong Candidate To Replace Natural Peroxidase in Sensitive and Selective Bioassays, ACS Nano, 2019, 13, 4312–4321 CrossRef CAS PubMed.
  161. X. Zheng, Q. Zhu, H. Song, X. Zhao, T. Yi, H. Chen and X. Chen, In Situ Synthesis of Self-Assembled Three-Dimensional Graphene–Magnetic Palladium Nanohybrids with Dual-Enzyme Activity through One-Pot Strategy and Its Application in Glucose Probe, ACS Appl. Mater. Interfaces, 2015, 7, 3480–3491 CrossRef CAS PubMed.
  162. Y. Yan, Y. Ge, H. Mao, Q. Wang, Q. Li and H. Li, A γ-Fe2O3/thiophene-sulfur-doped graphene nanozymes-based colorimetric sensor for visual and quantitative determination of chlorpyrifos, J. Environ. Chem. Eng., 2024, 12, 113558 CrossRef CAS.
  163. H. Fan, W. Yang, Y. Dai, L. Huang, Q. Zhang, H. Zhang, J. Liu, W. Zhu and J. Hong, Hydroxyl radical-mediated synthesis of carbonyl functionalized graphene quantum dots-like as enzyme mimics with tunable fluorescence emission, Anal. Chim. Acta, 2024, 1318, 342931 CrossRef CAS PubMed.
  164. D. D. Liu, F. F. Zhang, M. Gao, J. C. Zhou, Y. F. Wang and Y. Z. Lu, Pt nanoparticle/N-doped graphene nanozymes for colorimetric detection of acetylcholinesterase activity and inhibition, Chin. J. Anal. Chem., 2022, 50, 100177 Search PubMed.
  165. P. Das, P. K. Boruah, P. Sarmah, R. Dutta, R. Boukherroub and M. R. Das, A Facile Preparation of Reduced Graphene Oxide Capped AuAg Bimetallic Nanoparticles: A Selective Nanozyme for Glutathione Detection, ChemistrySelect, 2022, 7, e202203415 CrossRef CAS.
  166. A. S. Siddiqui, M. A. Ahmad, M. H. Nawaz, A. Hayat and M. Nasir, Decorating Zirconium on Graphene Oxide to Design a Multifunctional Nanozyme for Eco-Friendly Detection of Hydrogen Peroxide, Catalysts, 2022, 12, 1105 CrossRef CAS.
  167. H. Li, H. Sun, J. Ding, T. Wu, S. Cai, C. Wang and R. Yang, RuO2/rGO heterostructures as mimic peroxidases for colorimetric detection of glucose, Microchim. Acta, 2022, 189, 261 CrossRef CAS PubMed.
  168. S. Keerthana, A. Rajapriya, C. Viswanathan and N. Ponpandian, Enzyme like-colorimetric sensing of H2O2 based on intrinsic peroxidase mimic activity of WS2 nanosheets anchored reduced graphene oxide, J. Alloys Compd., 2022, 889, 161669 CrossRef CAS.
  169. L. Nana, L. Ruiyi, W. Qinsheng, Y. Yongqiang, S. Xiulan, W. Guangli and L. Zaijun, Colorimetric detection of chlorpyrifos in peach based on cobalt-graphene nanohybrid with excellent oxidase-like activity and reusability, J. Hazard. Mater., 2021, 415, 125752 CrossRef PubMed.
  170. M. X. Liu, H. Zhang, S. Chen, Y. L. Yu and J. H. Wang, MnO2-graphene oxide hybrid nanomaterial with oxidase-like activity for ultrasensitive colorimetric detection of cancer cells, Anal. Bioanal. Chem., 2021, 413, 4451–4458 CrossRef CAS PubMed.
  171. S. Gajendar, K. Amisha and S. Manu, Mildly acidic pH and room temperature triggered peroxidase-mimics of rGO-Cu3(OH)2(MoO4)2cuboidal nanostructures: An effective colorimetric detection of neurotransmitter dopamine in blood serum and urine samples, CrystEngComm, 2021, 23, 599–616 RSC.
  172. J. Hassanzadeh and A. Khataee, Ultrasensitive chemiluminescent biosensor for the detection of cholesterol based on synergetic peroxidase-like activity of MoS2 and graphene quantum dots, Talanta, 2018, 178, 992–1000 CrossRef CAS PubMed.
  173. L. Guo, H. Zheng, C. Zhang, L. Qu and L. Yu, A novel molecularly imprinted sensor based on PtCu bimetallic nanoparticle deposited on PSS functionalized graphene with peroxidase-like activity for selective determination of puerarin, Talanta, 2020, 210, 120621 CrossRef CAS PubMed.
  174. M. Bilal, N. Khaliq, M. Ashraf, N. Hussain, Z. Baqar, J. Zdarta, T. Jesionowski and H. M. Iqbal, Enzyme mimic nanomaterials as nanozymes with catalytic attributes, Colloids Surf., B, 2023, 221, 112950 CrossRef CAS PubMed.
  175. S. Zhang, H. Li, Z. Wang, J. Liu, H. Zhang, B. Wang and Z. Yang, A strongly coupled Au/Fe 3 O 4/GO hybrid material with enhanced nanozyme activity for highly sensitive colorimetric detection, and rapid and efficient removal of Hg 2+ in aqueous solutions, Nanoscale, 2015, 7, 8495–8502 RSC.
  176. D. J. Baruah, A. Thakur, E. Roy, K. Roy, S. Basak, D. Neog, H. K. Bora, R. Konwar, V. Chaturvedi and M. V. Shelke, Atomically Dispersed Manganese on Graphene Nanosheets as Biocompatible Nanozyme for Glutathione Detection in Liver Tissue Lysate Using Microfluidic Paper-based Analytical Devices, ACS Appl. Mater. Interfaces, 2023, 15, 47902–47920 CrossRef CAS PubMed.
  177. N. Nashat and Z. Haider, Therapeutic applications of nanozymes and their role in cardiovascular disease, International Journal of Nanomaterials, Nanotechnology and Nanomedicine, 2021, 7, 009–018 Search PubMed.
  178. S. Khan, M. Sharifi, S. H. Bloukh, Z. Edis, R. Siddique and M. Falahati, In vivo guiding inorganic nanozymes for biosensing and therapeutic potential in cancer, inflammation and microbial infections, Talanta, 2021, 224, 121805 CrossRef CAS PubMed.
  179. D. Crosby, S. Bhatia, K. M. Brindle, L. M. Coussens, C. Dive, M. Emberton, S. Esener, R. C. Fitzgerald, S. S. Gambhir and P. Kuhn, Early detection of cancer, Science, 2022, 375, eaay9040 CrossRef CAS PubMed.
  180. R. C. Fitzgerald, A. C. Antoniou, L. Fruk and N. Rosenfeld, The future of early cancer detection, Nat. Med., 2022, 28, 666–677 CrossRef CAS PubMed.
  181. M. Perfézou, A. Turner and A. Merkoçi, Cancer detection using nanoparticle-based sensors, Chem. Soc. Rev., 2012, 41, 2606–2622 RSC.
  182. J. A. Barreto, W. O'Malley, M. Kubeil, B. Graham, H. Stephan and L. Spiccia, Nanomaterials: applications in cancer imaging and therapy, Adv. Mater., 2011, 23, H18–H40 CAS.
  183. S. J. Mambou, P. Maresova, O. Krejcar, A. Selamat and K. Kuca, Breast cancer detection using infrared thermal imaging and a deep learning model, Sensors, 2018, 18, 2799 CrossRef PubMed.
  184. T. Hristozova, R. Konschak, V. Budach and I. Tinhofer, A simple multicolor flow cytometry protocol for detection and molecular characterization of circulating tumor cells in epithelial cancers, Cytometry, Part A, 2012, 81, 489–495 CrossRef PubMed.
  185. B. Davidson, H. P. Dong, A. Holth, A. Berner and B. Risberg, Flow cytometric immunophenotyping of cancer cells in effusion specimens: diagnostic and research applications, Diagn. Cytopathol., 2007, 35, 568–578 CrossRef PubMed.
  186. H. Zhang, S. Chen, Y. L. Yu and J. H. Wang, MnO2-graphene oxide hybrid nanomaterial with oxidase-like activity for ultrasensitive colorimetric detection of cancer cells, Anal. Bioanal. Chem., 2021, 413(17), 4451–4458 CrossRef PubMed.
  187. Y. Tao, Y. Lin, Z. Huang, J. Ren and X. Qu, Incorporating graphene oxide and gold nanoclusters: a synergistic catalyst with surprisingly high peroxidase-like activity over a broad pH range and its application for cancer cell detection, Adv. Mater., 2013, 25, 2594–2599 CrossRef CAS PubMed.
  188. M. I. Kim, M. S. Kim, M.-A. Woo, Y. Ye, K. S. Kang, J. Lee and H. G. Park, Highly efficient colorimetric detection of target cancer cells utilizing superior catalytic activity of graphene oxide–magnetic-platinum nanohybrids, Nanoscale, 2014, 6, 1529–1536 RSC.
  189. L.-N. Zhang, H.-H. Deng, F.-L. Lin, X.-W. Xu, S.-H. Weng, A.-L. Liu, X.-H. Lin, X.-H. Xia and W. Chen, In situ growth of porous platinum nanoparticles on graphene oxide for colorimetric detection of cancer cells, Anal. Chem., 2014, 86, 2711–2718 CrossRef CAS PubMed.
  190. J. Liu, M. Cui, L. Niu, H. Zhou and S. Zhang, Enhanced peroxidase-like properties of graphene–hemin-composite decorated with Au nanoflowers as electrochemical aptamer biosensor for the detection of K562 leukemia cancer cells, Chem.–Eur. J., 2016, 22, 18001–18008 CrossRef CAS.
  191. Y. Song, Y. Chen, L. Feng, J. Ren and X. Qu, Selective and quantitative cancer cell detection using target-directed functionalized graphene and its synergetic peroxidase-like activity, Chem. Commun., 2011, 47, 4436–4438 RSC.
  192. J. Pravda, Hydrogen peroxide and disease: towards a unified system of pathogenesis and therapeutics, Mol. Med., 2020, 26, 41 CAS.
  193. T. Zhang, Y. Xing, Y. Song, Y. Gu, X. Yan, N. Lu, H. Liu, Z. Xu, H. Xu and Z. Zhang, Aupt/mof–graphene: a synergistic catalyst with surprisingly high peroxidase-like activity and its application for H2O2 detection, Anal. Chem., 2019, 91, 10589–10595 CrossRef CAS PubMed.
  194. G. H. Jin, E. Ko, M. K. Kim, V.-K. Tran, S. E. Son, Y. Geng, W. Hur and G. H. Seong, Graphene oxide-gold nanozyme for highly sensitive electrochemical detection of hydrogen peroxide, Sens. Actuators, B, 2018, 274, 201–209 CrossRef CAS.
  195. Z. S. Li, X. C. Deng, X. P. Hong and S. F. Zhao, Nanozyme Based on Dispersion of Hemin by Graphene Quantum Dots for Colorimetric Detection of Glutathione, Molecules, 2022, 27, 6779 CrossRef CAS PubMed.
  196. R. Wu, M. Sun, X. Liu, F. Qin, X. Zhang, Z. Qian, J. Huang, Y. Li, T. Tan and W. Chen, Oxidase-like ZnCoFe three-atom nanozyme as a colorimetric platform for ascorbic acid sensing, Anal. Chem., 2022, 94, 14308–14316 CrossRef CAS PubMed.
  197. H. Chen, Q. Wang, Q. Shen, X. Liu, W. Li, Z. Nie and S. Yao, Nitrogen doped graphene quantum dots based long-persistent chemiluminescence system for ascorbic acid imaging, Biosens. Bioelectron., 2017, 91, 878–884 CrossRef CAS PubMed.
  198. H. Sun, X. Liu, X. Wang, Q. Han, C. Qi, Y. Li, C. Wang, Y. Chen and R. Yang, Colorimetric determination of ascorbic acid using a polyallylamine-stabilized IrO 2/graphene oxide nanozyme as a peroxidase mimic, Microchim. Acta, 2020, 187, 1–9 CrossRef PubMed.
  199. D. Peng, M. Que, X. Deng, Q. He, Y. Zhao, S. Liao, X. Li and H. Qiu, Mn3O4 nanoparticles decorated porous reduced graphene oxide with excellent oxidase-like activity for fast colorimetric detection of ascorbic acid, Microchim. Acta, 2023, 190, 243 CrossRef CAS PubMed.
  200. P. K. Boruah, G. Darabdhara and M. R. Das, Polydopamine functionalized graphene sheets decorated with magnetic metal oxide nanoparticles as efficient nanozyme for the detection and degradation of harmful triazine pesticides, Chemosphere, 2021, 268, 129328 CrossRef CAS.
  201. S. Chu, W. Huang, F. Shen, T. Li, S. Li, W. Xu, C. Lv, Q. Luo and J. Liu, Graphene oxide-based colorimetric detection of organophosphorus pesticides via a multi-enzyme cascade reaction, Nanoscale, 2020, 12, 5829–5833 RSC.
  202. L. Lin, H. Ma, C. Yang, W. Chen, S. Zeng and Y. Hu, A colorimetric sensing platform based on self-assembled 3D porous CeGONR nanozymes for label-free visual detection of organophosphate pesticides, Mater. Adv., 2020, 1, 2789–2796 RSC.
  203. Z. Q. Yan, H. Yuan, Q. Zhao, L. Xing, X. Y. Zheng, W. G. Wang, Y. L. Zhao, Y. Yu, L. Hu and W. L. Yao, Recent developments of nanoenzyme-based colorimetric sensors for heavy metal detection and the interaction mechanism, Analyst, 2020, 145, 3173–3187 RSC.
  204. Y. Guo, Y. Tao, X. Ma, J. Jin, S. Wen, W. Ji, W. Song, B. Zhao and Y. Ozaki, A dual colorimetric and SERS detection of Hg2+ based on the stimulus of intrinsic oxidase-like catalytic activity of Ag-CoFe2O4/reduced graphene oxide nanocomposites, Chem. Eng. J., 2018, 350, 120–130 CrossRef CAS.
  205. X. Chen, N. Zhai, J. H. Snyder, Q. Chen, P. Liu, L. Jin, Q. Zheng, F. Lin, J. Hu and H. Zhou, Colorimetric detection of Hg 2+ and Pb 2+ based on peroxidase-like activity of graphene oxide–gold nanohybrids, Anal. Methods, 2015, 7, 1951–1957 RSC.
  206. Z. Tao, Y. Zhou, N. Duan and Z. Wang, A colorimetric aptamer sensor based on the enhanced peroxidase activity of functionalized graphene/Fe3O4-AuNPs for detection of lead (II) ions, Catalysts, 2020, 10, 600 CrossRef CAS.
  207. B. Hisey, P. J. Ragogna and E. R. Gillies, Phosphonium-functionalized polymer micelles with intrinsic antibacterial activity, Biomacromolecules, 2017, 18, 914–923 CrossRef CAS PubMed.
  208. J. Zhu, Q. Li, X. Li, X. Wu, T. Yuan and Y. Yang, Simulated enzyme activity and efficient antibacterial activity of copper-doped single-atom nanozymes, Langmuir, 2022, 38, 6860–6870 CrossRef CAS PubMed.
  209. M. M. Anjos, E. H. Endo, F. V. Leimann, O. H. Gonçalves, B. P. Dias-Filho and B. A. de Abreu Filho, Preservation of the antibacterial activity of enzymes against Alicyclobacillus spp. through microencapsulation, LWT--Food Sci. Technol., 2018, 88, 18–25 CrossRef CAS.
  210. J. Davies and D. Davies, Origins and evolution of antibiotic resistance, Microbiol. Mol. Biol. Rev., 2010, 74, 417–433 CrossRef CAS PubMed.
  211. J. Liu, R. Wang, F. Zhai, W. Zhang, B. Wang and H. Zhou, A nanozyme composite with high near-infrared photothermal ability for synergistic bacterial elimination, Mater. Chem. Front., 2023, 7, 1642–1649 RSC.
  212. L. F. Zhang, L. Zhang, H. Deng, H. Li, W. T. Tang, L. Y. Guan, Y. Qiu, M. J. Donovan, Z. Chen and W. H. Tan, In vivo activation of pH-responsive oxidase-like graphitic nanozymes for selective killing of Helicobacter pylori, Nat. Commun., 2021, 12, 2002 CrossRef CAS PubMed.
  213. Y. Qian, J. Wang, X. Geng, B. Jia, L. Wang, Y.-Q. Li, B. Geng and W. Huang, Graphene Quantum Dots Nanoantibiotic-Sensitized TiO2− Heterojunctions for Sonodynamic-Nanocatalytic Therapy of Multidrug-Resistant Bacterial Infections, Adv. Healthcare Mater., 2024, 13, 2400659 CrossRef CAS PubMed.
  214. Q. Dong, Z. Li, J. Xu, Q. Yuan, L. Chen and Z. Chen, Versatile graphitic nanozymes for magneto actuated cascade reaction-enhanced treatment of S. mutans biofilms, Nano Res., 2022, 15, 9800–9808 CrossRef CAS.
  215. W. Fadhil, I. Jabbar, E. Ali, G. Sulaiman, R. Khan and H. Mohammed, Freshly Prepared Graphene Oxide Nanoparticles' Wound-Healing Potential and Antibacterial Activity Specifically Against Staphylococcus aureus: In Vivo Efficacy and Clinical Isolate Evaluation, Plasmonics, 2024, 20, 763–773 CrossRef.
  216. Y. Li, W. Ma, J. Sun, M. Lin, Y. Niu, X. Yang and Y. Xu, Electrochemical generation of Fe3C/N-doped graphitic carbon nanozyme for efficient wound healing in vivo, Carbon, 2020, 159, 149–160 CrossRef CAS.
  217. Y. Wang, H. D. Shi, H. L. Zhang, Y. Yu Chen, B. Ren, Q. Tang, Q. Sun, Q. L. Zhang and J. G. Liu, A Multifunctional Nanozyme with NADH Dehydrogenase-Like Activity and Nitric Oxide Release under Near-Infrared Light Irradiation as an Efficient Therapeutic for Antimicrobial Resistance Infection and Wound Healing, Adv. Healthcare Mater., 2023, 12, e2300568 CrossRef PubMed.
  218. C. Ren, X. Hu and Q. Zhou, Graphene Oxide Quantum Dots Reduce Oxidative Stress and Inhibit Neurotoxicity In Vitro and In Vivo through Catalase-Like Activity and Metabolic Regulation, Adv. Sci., 2018, 5, 1700595 CrossRef.
  219. Y. Huang, C. Liu, F. Pu, Z. Liu, J. Ren and X. Qu, A GO–Se nanocomposite as an antioxidant nanozyme for cytoprotection, Chem. Commun., 2017, 53, 3082–3085 RSC.
  220. M. Asadi, S. H. Ghorbani, L. Mahdavian and M. Aghamohammadi, Graphene-based hybrid composites for cancer diagnostic and therapy, J. Transl. Med., 2024, 22, 611 CrossRef PubMed.
  221. T. P. Dasari Shareena, D. McShan, A. K. Dasmahapatra and P. B. Tchounwou, A Review on Graphene-Based Nanomaterials in Biomedical Applications and Risks in Environment and Health, Nano-Micro Lett., 2018, 10, 53 CrossRef PubMed.
  222. H. Sachdeva, A. R. Khandelwal, R. Meena, K. Sharma and N. Khatik, Graphene-based nanomaterials for cancer therapy, Mater. Today: Proc., 2021, 43, 2954–2957 CAS.
  223. A. Singhal, N. Sinha, P. Kumari and M. Purkayastha, Synthesis and Applications of Hydrogels in Cancer Therapy, Anti-Cancer Agents Med. Chem., 2020, 20, 1431–1446 CrossRef CAS PubMed.
  224. K. Arab, A. Jafari and F. Shahi, The role of graphene quantum dots in cutting-edge medical therapies, Polym. Adv. Technol., 2024, 35, e6571 CrossRef CAS.
  225. A. Zarepour, A. Khosravi, N. Yücel Ayten, P. Çakır Hatır, S. Iravani and A. Zarrabi, Innovative approaches for cancer treatment: graphene quantum dots for photodynamic and photothermal therapies, J. Mater. Chem. B, 2024, 12, 4307–4334 RSC.
  226. B. Lin, H. Chen, D. Liang, W. Lin, X. Qi, H. Liu and X. Deng, Acidic pH and high-H2O2 dual tumor microenvironment-responsive nanocatalytic graphene oxide for cancer selective therapy and recognition, ACS Appl. Mater. Interfaces, 2019, 11, 11157–11166 CrossRef CAS.
  227. A. Sun, L. Mu and X. Hu, Graphene oxide quantum dots as novel nanozymes for alcohol intoxication, ACS Appl. Mater. Interfaces, 2017, 9, 12241–12252 CrossRef CAS PubMed.

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.