Introduction

In recent years, nanotechnology and advanced nano-delivery systems have shown that can notably address some shortcomings of conventional therapeutics and the low efficacy treatments [1]. Poor solubility, low bioavailability, unexpected metabolization and drug elimination of the body before function on their desired sites, non-specific selectivity, and side effects are some of the drawbacks of traditional treatments [2, 3]. However, recent advances in nanotechnology and nanomaterials could have provided a better bio-distribution of drugs, reduced adverse effects, and delivered therapeutic agents to the targeted sites, improving bioavailability [4, 5]. In recent years, numerous organic and inorganic nanomaterials have been investigated for nanomedicine applications [6,7,8,9]. In this context, two-dimensional (2D)-carbon-based nanomaterials provide new prospects attracting the attention of many scientists due to their fascinating characteristics such as high surface area, easy functionalization, and acceptable biocompatibility [10, 11]. 2D-carbon-based materials have been differentiated based on the number of layers in their structure, oxygen-containing functional groups, and chemical composition [12]. Graphene, graphene oxide (GO), and reduced graphene oxide (rGO) are the most well-known and most frequently explored forms of 2D-carbon-based materials in the realm of cancer diagnosis and treatment [13].

Graphene is composed of a uniform monolayer of carbon atoms. Carbon atoms with hexagonal arrangements are bonded tightly in a honeycomb structure. In this structure, any carbon atoms are attached to two others with sp2 hybridization, and one out-of-plane p orbital supplies the electron network [14]. Graphene results of graphite exfoliation via chemical vapor deposition. As graphene does not contain any oxygen groups, it is considered hydrophobic. However, this unique structure leads to graphene’s interesting properties, including mechanical strength, and high thermal, electrical, optical, and magnetic features, making it a good candidate for different applications as well as cancer diagnosis and therapy [15]. To date, numerous 2D-graphene-based materials have been investigated; these materials range in size, chemical make-up, biodegradability, and compatibility with living systems [16]. 2D-graphene-based materials having such a wide range of properties hold great promise for use in biomedicine, notably in areas like biosensors, multimodal imaging, drug/gene delivery, and cancer treatment [17].

The 2D-graphene-based materials have a huge specific surface area, which makes them stand out. This property allows them to adsorb molecules efficiently via covalent or non-covalent interactions. As a result, they show more restraint in their discharge in response to environmental cues. 2D-graphene-based materials are of special interest for optical treatments including photothermal therapy (PTT) and photodynamic therapy (PDT) due to their sheet-like nanostructures. Additionally, these nanomaterials may respond to near-infrared (NIR) light. The advantages above have resulted in 2D-graphene-based materials becoming the prevailing nanoplatforms for theranostic investigations. In recent years, the primary focus of researchers has been on the fabrication or optimization of monotherapy. However, numerous preclinical and clinical studies have demonstrated that monotherapy does not yield the expected level of effectiveness. This is primarily attributed to the recurrence and metastasis of cancer. The combination of 2D-graphene-based nanocomposites with other functional moieties has the potential to create a novel category of therapeutic agents in the field of biomedicine. This integration can significantly enhance the properties of 2D-graphene-based nanocomposites, enabling synergistic applications in cancer diagnosis and therapy on the 2D-graphene-based nanomaterials [18].

This stydy provides an overview of the present status and advantages associated with the utilization of 2D-graphene-based nanomaterials in the field of cancer detection and therapy. Additionally, it explores the challenges encountered and the potential for future advancements in this area. In this discussion, we will examine three highly promising applications of 2D-graphene-based nanomaterials in the field of cancer treatment. These applications include: (1) cancers diagnosis, (2) drug delivery, (3) PTT, and (4) PDT. This study also presents a summary of the biocompatibility of 2D-graphene-based nanomaterials, offering valuable insights into this subject matter.

Graphene-based hybrid composites for cancer diagnosis

Nowadays, cancer is one of the human health threats in the world due to its high prevalence and death [19]. It is predicted that the early diagnosis of cancer can attenuate its death rate. Magnetic resonance imaging (MRI) and Computed Tomography (CT) scan are the most conventional methods used for cancer detection [20]. Although these techniques have been to a degree of success, the lack of effective techniques with acceptable sensitivity and inefficiency for early detection of cancer still remains a major concern. Recent studies have stated that 2D-graphene-based materials combined with many contrast agents can provide a new platform to detect tumor tissues to achieve the necessity of multimodal imaging [20]. Herein, it was focused on 2D-graphene-based materials used in different biosensing techniques and bioimaging methods.

Biosensing

Biosensors are analytical tools constructed of two parts including a receptor (biological unit) and a transducer (electronic unit) which enable to detect the small molecules and large biomolecules [22, 23]. They can qualitatively or quantitatively recognize specific types of analytes using various techniques such as spectrochemical, electrochemical, magneto-chemical, fluorescence resonance energy transfer (FRET), surface-enhanced Raman scattering (SERS), fluorescence spectroscopy, and surface plasmon resonance (SPR). They are mostly used for detecting chemical analytes and biomolecules that have a key role in the disease process, so their detection is very crucial in diagnosis and therapy [22]. Graphene-based materials have good electrochemical and optical properties for biosensing detection applications. they can interact with various molecules via p–p stacking or electrostatic interaction [24]. Additionally, their surface can be modified to allow them to interact with defined analytes. GO is more explored for biosensing among graphene derivatives due to its acceptable biocompatibility, good water dispersibility, surface functional groups, and its G-band in Raman spectra [25]. GO-based biosensors can detect lower limits, respond rapidly, have high sensitivity, and improve signal-to-noise ratios [27]. The electrochemical part consisted of screen-printed carbon electrodes (SPCEs) functionalized with rGO–carboxymethylcellulose (rGO-CMC). Single nucleotide polymorphism in cDNAs from human breast cancer cell lines was discriminated by this platform, which can be an excellent diagnostic device in clinical analysis. Furthermore, an electrochemical DNA probe immobilizedMoS2/graphene-based sensor was developed for detecting circulating tumor DNA. In comparison to other methods for the detection of circulating tumor DNA, this biosensor showed high sensitivity not needing labeling and using amplifiers [28].

As miRNAs have a key role in the early diagnosis of cancer, several graphene-based electrode materials with different structures and composites have been developed for the detection of various types of microRNAs. For instance, a disposable pencil graphite electrode modified with graphene was developed for mir-21 detection [29]. This study reported that the graphene-modified electrode had a 2.77 times lower detection limit (3.12 pmol) compared with the unmodified electrode. In addition, this graphene-modified electrode did not require for sample preparation and amplification process before the detection of real samples. Another electrochemical biosensor was fabricated for plasma miR-155 detection by M.Azimzadeh et al. [30]. They fabricated a thiolated glassy carbon electrode whose surface was modified with GO sheets decorated with gold nanorods (GNRs). They reported that for the range of 2.0 fM to 8.0 pM, the biosensor signal relationship with miRNA concentration was linear and the detection limit was 0.6 fM. In addition, this system showed high selectivity, good sensitivity, storage ability, and acceptable response, and did not need any purification and amplification process before the detection of real samples. A label-free electrochemical immune-sensor functionalized with polymer-rGO was fabricated for the detection of miRNA-29b-1 and miRNA-141 [31]. This immune-sensor displayed excellent stability, good sensitivity with a limit of quantification of 8 fM, and high selectivity as it discriminates mismatch.

Proteins are other types of biomarkers that can be measured in tissues and blood serum for early diagnosis of cancer. Any changes in protein levels indicate an alteration in their structure or post-translation process related to the existence and development of cancer cells. So, the detection of protein biomarkers is vital for diagnosis, drug screening, and treatment approach evaluation [32]. In this context, various graphene-based biosensors were established to detect a protein biomarker related to cancer cells [33]. For example, a biosensing platform was generated of porphyrinnon- functionalized graphene-modified glassy carbon electrode for clinical diagnosis of cyclin A2 which is a prognostic indicator in early-stage cancers [34]. Using graphene in this platform improves the sensor function due to its instinct conductivity and providing oxygen-containing functional groups for the following immobility of peptide. A chitosan-modified-rGO-based biosensor was fabricated for the VEGFR2 detection with acceptable susceptibility and a detection limit of 0.28 pM [35]. It could directly measure the total amount of VEGFR2 in cell lysates and also monitor VEGFR2 expression changes stimulated by different inhibitor treatments.

Reactive oxygen species (ROS) are essential small biomolecules for various physiological processes and signaling pathways. In abnormal conditions such as cancer, ROS level is changed caused by oxidative stress [36]. One of the reactive oxygen species is H2O2 molecules that are directly related to protein production, apoptosis, and DNA damage [189]. In this concept, a glassy carbon electrode modifying with a graphene-based nanocomposite (rGO-Au-poly(toluidine blue O) films) was fabricated to detect H2O2 [37]. This platform was sensitive to H2O2 with a detection limit of 0.2 µM considered a potential biosensor for various cancer cell detection. Pt–MnO2-rGO-based biosensor is another example that was fabricated for the detection of H2O2 molecules from cells with high stability and sensitivity with a detection limit of 1.0 µM [113]. In this context, poly (N-isopropylacrylamide) (PNIPAM) is one of the common thermo-sensitive polymers with a lower critical solution temperature (LCST) of 32 °C in water [114]. For this purpose, the poly(L, D-lactide)- block-poly(N-isopropylacrylamide-rand-acrylic acid) conjugated from rGO (rGO-graftPDLA-block-P(NIPAAm-rand-AAc)) was fabricated with LCST of 39 °C [115]. The DOX was loaded in this delivery system with a loading capacity of 99%. It was reported that DOX release was thermo-responsiveness as it increased in response to an increasing temperature higher than LCST. Based on the in vitro cytotoxicity, DOX-loaded rGO-g-PDLA-bP(NIPAAm-rand-AAc) systems had a more lethal effect in the HepG2 cell line compared to the free DOX. In recent study, a Janus GO nanosheets modified with PNIPAM/PCL were designed as a dual DDS [116]. This novel nano-system was able to load simultaneously both of the hydrophobic (quercetin) and hydrophilic (5-FU) anti-cancer agents with a thermos-responsive release.

In the over mentioned studies, thermos-responsive graphene-based DDS was provided via GO modification with thermo-responsive polymers. Apart from this strategy, GO photothermal property was also used to development of a nanoplatform with pH/Thermal Dual-responsive drug release, tumor-targeting, and synergistic therapeutic simultaneously [117]. To this aim, a targeted nanocomplexes of GO modified with FA (NCGOFA) was developed and loaded with DOX (NCGO@DOX-FA) (Fig. 2). The results of this study demonstrated that the NCGO-FA nanocomplexes have a high-load drug capacity, targeting specificity, photostability, and photothermal conversion efficiency leading to triggered drug release by heat and in acidic conditions. Furthermore, this dual-responsive DDS exhibits a notable synergistic anticancer effect compared with individually applied chemotherapy, PTT, or PDT, or chemotherapy.

Fig. 2
figure 2

Schematic Illustrating the Fabrication of the NCGO@DOX-FA as pH and Thermal Dual-Responsive Targeted DDS for Photothermal-Chemo/Photodynamic Synergetic Therapies [117]. Note: Copyright ©2019. ACS

Nanodrugs with high molecular weight and larger particle sizes (10–500 nm)have many advantages such as: high drug loading, specific targeting, and the ability to protect the cargo from degradation and release the drug in a controlled or sustained manner. Their leak from blood vessels are more slowly than most chemotherapy drugs, which allows access to tumor tissue by leaky tumor vasculature, which is a major feature of solid tumor vasculature [118]. Nano drugs remain in the tumor bed due to the reduction of lymphatic drainage. This phenomenon has been named as the “enhanced permeability and retention” (EPR) effect to enhance the delivery of nanodrugs in tumors. Nanodrug delivery is based on drug accumulation in tumors due to the EPR effect and subsequent release. To enhance this effect in tumors, however, high interstitial fluid pressure (IFP), dense extracellular matrix (ECM), and blood vessels can be used. Tumor blood clot or embolization indicated. If the EPR effect is insufficient, the drug may be released and introduce more toxicity to normal tissues. Therefore, there is an urgent need to identify the physiological barriers that affect the EPR effect of tumors. Macromolecules between 10 and 500 nm (e.g., macromolecular anticancer agent, albumin, immunoglobulin, micelles, liposomes and protein-polymer conjugates, protein and carbon nanostructures) can selectively leak from the vascular bed and accumulate in the interstitial space. to find Tumors show different EPR effects regardless of type and size, patients or growth stages. Tumors with high blood vessel density (such as hepatocellular carcinoma) show a strong EPR effect, while tumors with low vascular density (such as pancreatic cancer) show a weak EPR effect. Nanomedicines based on EPR effect are promising to improve the effectiveness of treatment with systemic anticancer drugs. Figure 3 shows several pharmacological strategies for vascular regulation.

Fig. 3
figure 3

Increase the enhanced permeability and retention of effect nanodrug delivery systems in modulating tumor vessels [118]. Note: Copyright ©2021. MPOI

Magnetic graphene-based DDS

Recently, the combination design of graphene-based materials with magnetic nanoparticle modifications has revealed notable performance for targeted drug delivery, photothermal lethal effect on tumor cells, and magnetic-responsive drug release [7]. In magnetic-sensitive DDS, magnetic NPs (e.g., Fe, Co, Mn, or Ni derivatives) generate heating energy to increase temperature under the alternating magnetic field that leads to drug release. In an effort, a magnetic-responsive DDS was designed for DOX delivery in cancer therapy. This system was constructed of β-cyclodextrin (β-CD)/nickel NP-modified GO (GONiCD) and mitochondrial ion-targeting peptide (MitP)-conjugated HA (HAMitP) [119]. The (GONiCD) attachment to the (HAMitP) was based on host-guest interaction between β-CD and the cyclohexyl groups on MitP that lead to forming of a supramolecular assembly (Fig. 4). This multi-component DDS could enhance the DOX-loading capacity with a stimuli-controllable release in response to alternating magnetic fields and target the tumor cell mitochondria, leading to cell apoptosis via damaging both the mitochondria and the nuclei.

Fig. 4
figure 4

Schematic Illustration of construction of the AMF-sensitive multi-component DDS for inducing tumor cell apoptosis [119]. Note: Copyright ©2020. Wiley. CH

In addition, magnetic NPs are used to accumulate and locally deliver drugs through a magnetic external field generated by a magnet located near the chosen treatment site. Therefore, combining magnetic NPs with graphene-based materials can indirectly enhance their therapeutic efficiency by increasing the accumulation of DDS in the desired site or synergistically enhancing the lethal effect of magnetic hyperthermia effect [89, 120,

Fig. 6
figure 6

Schematic illustration of PTX@GO-PEG-OSA performance as a desirable strategy with dual pH and thermal-responsive drug release for chemo/photothermal/photodynamic therapy in reverse PTX’s resistance [70]. Note: Copyright ©2021. BMC

Although the graphene-based materials showed promising potential for in vivo PTT, their self-aggregation and accumulation in the target site remain challenging. To address these challenges, poloxamer-modified rGOs (prGOs)- loaded in human mesenchymal stem cells (hMSCs) were developed for targeted PTT [139]. pRGOs did not demonstrate any self-aggregation compared to rGOs. In vivo results revealed that pRGOs-loaded hMSCs remarkably improved tumor-targeting efficiency and generated higher heat than bare rGOs under laser irradiation.

The integration of various nanomaterials into a unique platform lets to bring their advantages together in a synergistic manner in cancer treatment. For example, polydopamine-modified rGO was coated with mesoporous silica (MS) and further conjugated with HA (pRGO@MS-HA) and used as a multifunctional nano-platform for targeted chemo-PTT in cancer treatment (Fig. 7) [140]. In addition to heat-generating of rGO, polydopamine modification could enhance rGO biocompatibility while MS employed for DOX loading, and HA acted as a targeting ligand. It was reported that DOX release was pH and NIR-responsive, which could improve the efficiency of this system. In vitro results revealed that pRGO@MS(DOX)-HA showed excellent photothermal effect along with a notable lethal effect on tumor cells with good specificity toward target cells. In vivo study confirmed that pRGO@MS(DOX)-HA possessed a synergistic antitumor effect more than individual chemo or photothermal effect.

Fig. 7
figure 7

Schematic illustration of multifunctional pRGO@MS(DOX)-HA for targeted chemo-PTT in cancer treatment [140]. Note: Copyright ©2017. ACS

In the same way, a targeted dual pH/thermal-responsive DDS with chemo-photothermal effect was fabricated by integrating a DNA aptamer with polydopamine-rGO nanosheets (rGO-PDA) nanosheets [141]. The rGO-PDA nanosheets simultaneously possessed the photothermal properties to generate hyperthermia, served as a nano-carrier for DOX loading. In addition, DNA aptamer modification provided a supplementary carrier for drug loading, served as a targeting ligand to recognize specific cell, and was able to control DOX release. All these advantages led to achieve an effective multifunctional DDS for chemo-PTT in a synergistic manner.

In recent years, graphene-based materials have been employed to construct hydrogels for achieving a multimodal DDS. For instance, GO-hybridized nanogels were developed to deliver chemotherapeutic agents and simultaneously presented PTT against cancer cells [142]. These nanogels were obtained by in situ combining GO nanosheets into alginate through a double emulsion method using disulfide bonds as crosslinkers. In the following, DOX was loaded by electrostatic interactions. The controllability of the PH/redox-sensitive DOX release improved the DOX uptake and long-term accumulation in cancer cells which along with GO photothermal property, led to an excellent anticancer effect, representing their potential for anticancer combination therapy. Additionally, the GO photothermal effect was employed for controlling drug release and PTT simultaneously. For example, a targeted light-responsive DDS was developed by a Cy5.5-AS1411 aptamer-conjugated GO wrap** on the surface of the DOX-loaded mesoporous silica NPS (MSN-Dox@GO-Apt) [143]. In this system, GO acts as a gatekeeper to control the Dox release in response to NIR irradiation and convert NIR to heat to induce PTT. The results showed that a combination of chemotherapy and PTT in a single platform resulted in a synergic effect much more effective than monotherapies, which can be considered a new strategy for cancer treatment.

PDT

PDT is a therapeutic modality employed with the purpose of eradicating malignant cells within tumors that involves the utilization of light with a certain wavelength to initiate the activation of molecules known as photosensitizers (PS). Subsequently, these molecules generate reactive oxygen species (ROSs) that possess the ability to induce apoptosis in the tumor cells [144]. In order to optimize the targeted delivery of PDT agents to tumor sites by reducing the doses to be effective, allowing easy cell entrance, and reducing skin sensitivity to light, the utilization of nanocarriers is imperative [145]. In recent times, researchers have conducted investigations on the optical loading properties of G, with a particular focus on its potential applications in the medical domain. Notably, G has been recognized as a crucial element in the development of miniature technological platforms for healthcare purposes. The exploration of theranostics started by employing G-based materials as carriers for both therapeutic substances and imaging agents [146]. Consequently, this development prompted subsequent investigations into the utilization of nanotechnology-based PDT for the purpose of achieving more targeted and precise cancer treatment. G has the capability to effectively catch light inside the NIR band. This enables the investigation of its potential application in cancer treatment by the utilization of light therapy, both internally and externally [147].

Researchers are trying to find more efficient methods of treating cancer. They are looking at how the nanoplatforms may complement existing treatments to better handle cancer as presented in Table 1. Using π-π stacking of GO, Tian’s group used PEG-GO and Ce6 as PS. The chemical was taken up by the cervical cancer cells, and when exposed to light, it triggered the generation of ROS. When compared to Ce6, GO-PEG-Ce6 proved more successful in the therapy of cancer [128]. GO was proposed by Huang and Collagenous as a Ce6 carrier. Ce6 was bound to folic acid-functionalized GO through π-π stacking binding, which is consistent with prior research. Researchers showed that when exposed to radiation, stomach cancer cells were killed by the system [148]. Zhou and coworkers used π-π stacking interaction to successfully add hypocrellin B (HB) as a PS to GO. When subjected to radiation, they showed that the material produced ROSs [127, 149]. Hypocrellin A and 7-ethyl-10-hydroxycamptothecin were combined on GO for this study. More lung cancer cells died when this mixture was exposed to light. This demonstrates the synergistic benefits of combining chemotherapy with PDT [149]. Increasing cellular sensitivity to ROS by inhibiting the DNA oxidative damage repair enzyme MTH1 may increase the efficacy of PDT. In order to combine PDT with DOX, Huang and coworkers used a GO-based nanocarrier to transport TH287 (an MTH1 inhibitor) and DOX to cancer cells [150]. They grafted PEG, FA, the photosensitizer indocyanine green (ICG), TH287, and DOX onto the GO nanocarrier. Proliferation and migration were inhibited, and endoplasmic reticulum (ER) stress-induced apoptosis and autophagy were augmented, in MNNG/HOS, MG63, U2OS, and SaOS-2 (osteosarcoma cancer) cells thanks to the efficient transport of DOX and TH287 with the PEG-GO-FA/ICG carrier. The distinctive characteristics of G have enhanced the efficacy and expediency of cancer treatment when utilizing G-based nanocarriers for the delivery of PDT and chemotherapy medications, surpassing the individual effectiveness of these methods.

Recently, G-derived nanocarriers with triple capacities have been conceived and produced to increase the efficacy of cancer therapy approaches. The development of a GO-based nanocarrier that is pH- and temperature-sensitive makes cancer treatment that combines PTT, PDT, and chemotherapy easier. Nanocomplexes GO (NCGO) NCGO@DOX-FA, and NCGO@methylene-FA are complexes formed when GO is grafted with the medicines DOX and methylene blue, respectively. This nanocarrier is distinguished from others in a number of ways, including its large surface area, photostability, and capacity to transport medications to specific locations. The FA receptors effectively facilitate the targeted delivery of the complexes into the cancer cells, leading to the subsequent release of the medication triggered by either the acidic pH or heat conditions [117]. A subsequent investigation by Ding et al. resulted in the development of a GO-involved nanoparticle with the specific goal of targeting cancer [151]. The nanoparticle was noncovalently functionalized with cucurbit [7] uril (CB [7]) in order to serve as a potential agent for drug delivery. Hence, the NGO was painted with carbon black (CB) and became known as GO-CB. Chlorin e6, a PS, and AQ4N (banoxantrone dihydrochloride), a hypoxia-responsive prodrug, were subsequently included in this composite material to alter its properties. Next, we added a CB [7] (more precisely, oxaliplatin, abbreviated as OX) guest molecule and a CD44-targeting chemical, ADA-hyaluronic acid (ADA-HA), that is well-known for improving biocompatibility. In the presence of OX and AQ4N, this nanoplatform has the potential to act as a PTT-PDT agent for the treatment of L02 (human fetal hepatocyte line) and B16 (murine melanoma) cells. This research demonstrates a drug delivery platform that has the potential to be used in a variety of different ways for cancer therapy, both in vivo and in vitro. In summary, it can be inferred that the combination of PDT or PTT with chemotherapy has the potential to successfully eliminate cancer cells through the utilization of graphene’s natural NIR absorption properties. The utilization of G-based vehicles has considerable potential in the field of cancer treatment.