Previous Article in Journal
The Expanding E3 Ligase-Ligand Landscape for PROTAC Technology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Integrating Nanotechnology and Artificial Intelligence for Early Detection and Prognostication of Glioblastoma: A Translational Perspective

by
Meghraj Vivekanand Suryawanshi
1,*,
Imtiyaz Bagban
2 and
Akshata Yashwant Patne
3,4,*
1
Department of Pharmaceutics, Sandip Institute of Pharmaceutical Sciences (SIPS), Savitribai Phule Pune University (SPPU, Pune), Nashik 422213, Maharashtra, India
2
Department of Pharmacology, Krishna School of Pharmacy and Research (KSP), KPGU University, Vadodara 391240, Gujarat, India
3
Graduate Programs, Taneja College of Pharmacy, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
4
Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
*
Authors to whom correspondence should be addressed.
Targets 2025, 3(4), 31; https://doi.org/10.3390/targets3040031
Submission received: 16 July 2025 / Revised: 30 September 2025 / Accepted: 9 October 2025 / Published: 14 October 2025

Abstract

Glioblastoma (GBM) is the most common and aggressive malignant brain tumor in adults. This review explains the connections between the genesis and progression of GBM and particular cellular tumorigenic mechanisms, such as angiogenesis, invasion, migration, growth factor overexpression, genetic instability, and apoptotic disorders, as well as possible therapeutic targets that help predict the course of the disease. Glioblastoma multiforme (GBM) diagnosis relies heavily on histopathological features, molecular markers, extracellular vesicles, neuroimaging, and biofluid-based glial tumor identification. In order to improve miRNA stability and stop the proliferation of cancer cells, nanoparticles, magnetic nanoparticles, contrast agents, gold nanoparticles, and nanoprobes are being created for use in cancer treatments, neuroimaging, and biopsy. Targeted nanoparticles can boost the strength of an MRI signal by about 28–50% when compared to healthy tissue or controls in a preclinical model like mouse lymph node metastasis. Combining the investigation of CNAs and noncoding RNAs with deep learning-driven global profiling of genes, proteins, RNAs, miRNAs, and metabolites presents exciting opportunities for creating new diagnostic markers for malignancies of the central nervous system. Artificial intelligence (AI) advances precision medicine and cancer treatment by enabling the real-time analysis of complex biological and clinical data through wearable sensors and nanosensors; optimizing drug dosages, nanomaterial design, and treatment plans; and accelerating the development of nanomedicine through high-throughput testing and predictive modeling.

Graphical Abstract

1. Introduction

Gliomas, glioneuronal tumors, and neuronal tumors are divided into six families by the World Health Organization (WHO): ependymomas, glioneuronal and neuronal tumors, circumscribed astrocytic gliomas, pediatric-type diffuse low-grade gliomas, and adult-type diffuse gliomas. These are differentiated from choroid plexus tumors by their unique epithelial characteristics [1]. Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary malignant brain tumor in adults, comprising about 14.5% of central nervous system tumors. Its incidence ranges from 0.6 to 5 per 100,000 people, more commonly affecting men, older adults (median diagnosis age 60–64), and white, non-Hispanic individuals. The rising incidence may relate to an aging population and enhanced diagnostic techniques, although specific causes are unclear. GBM is primarily sporadic with few risk factors, such as ionizing radiation exposure, and the prognosis remains poor, with a median survival of 10–15 months despite treatments. Outcomes vary, influenced by age, performance status, and tumor resection extent, with distinct primary and secondary subtypes sharing an aggressive clinical behavior [2,3,4,5,6].
An uncommon malignancy, glioblastoma (GBM) primarily affects older individuals and is characterized by a poor prognosis. Risk factors include exposure to ionizing radiation, genetic predispositions such as TP53 mutations, and inherited conditions like Li–Li-Fraumeni syndrome and neurofibromatosis type 1. No evidence links GBM to routine radiation exposure. The tumorigenesis of GBM involves cellular processes such as angiogenesis, invasion, and migration, with IDH mutations leading to cancer development due to the abnormal accumulation of the oncometabolite 2-HG [7,8,9,10].
Notch signaling is required for cell differentiation, proliferation, and death in a wide variety of cell types and tissues, including neurons in the central nervous system. The Notch-1, Notch-2, Notch-3, and Notch-4 receptors are important. Notch-1 is either a tumor suppressor or an oncogene [11]. When ceramides are broken down by the acid ceramidase enzyme, they promote senescence and cell death [12]. Angiogenesis of glioma stem cells and the restoration of oxygen supply are stimulated by vascular endothelial growth factor (VEGF) [13]. Glioblastoma tumors have altered, rearranged, or elevated PDGFRα gene expression, which contributes to oncogenesis [14].
By attaching extracellular signaling ligands to its extracellular domain, the transmembrane cell RTK known as the epidermal growth factor receptor (EGFR) starts several signal transduction cascades. Glioblastoma etiology is often linked to EGFR mutations [15]. The PI3K/AKT/mTOR pathway is essential for controlling the cell cycle in glioblastoma because overactivation lowers survival rates and increases tumor aggressiveness. The tumor suppressor PTEN is rendered inactive by mutations in glioblastomas [16,17]. The Smoothened protein is deactivated by the sonic hedgehog (SHH) glycoprotein, which causes glioblastomas to transition to adult stem cells [18,19]. The RB1 protein regulates the cell cycle transition from G1 to S-phase. In both primary and secondary GBMs, this pathway is commonly observed to be inactivated [20].
The DNA repair enzyme 6-methylguanine-DNA methyltransferase, which is effective in treating MGMT promoter hypermethylation disorders brought on by gene silencing, is said, by proponents of MGMT encoding, to shield cells from alkylating medications [21]. One intricate piece of cellular machinery, the proteasome, participates in a number of enzymatic activities that facilitate the breakdown and recycling of proteins and may weaken cancer cells [22]. Headaches, nausea, intracerebral hypertension, motor impairments, weight loss, confusion, and visual or verbal impairments are typical clinical presentations [23,24,25].
Figure 1 explains the RTK (Receptor tyrosine kinase) signaling pathway in Glioblastoma.
Treatment for glioblastoma and stopping its progression depend on early detection. Two disadvantages of current techniques, like histomolecular ones, are the absence of biomarkers and decreased BBB penetration. In contemporary medicine, it is essential to identify the causes of origin, development, and early diagnosis. To detect tumors and neurodegenerative diseases, imaging techniques such as MRI, PET, CT, PA, and FL are required [26].
Biomimetic nanosystems are made of different materials, including proteins, cell membranes, and disease-targeting components, and they resemble physiological conditions. The created affinity nanoprobes have a long lifespan, are highly biocompatible, and replicate the microenvironment of disease. They provide a novel way to penetrate the blood–brain barrier (BBB) and make it easier to develop multipurpose “all-in-one” probes for treatment and diagnosis. Despite difficulties with BBB permeability for contrast agents, these developments improve management aspects of glioblastoma, such as early detection, treatment plans, and monitoring therapy responses [27]. The use of cutting-edge nanotechnologies in the diagnosis of glioblastoma in its early stages is covered in this review, which also emphasizes the benefits of these materials for better diagnosis and treatment. It also covers the possibilities for creating sophisticated smart sensors for the treatment of glioblastoma, as well as the drawbacks and difficulties in creating nanodiagnostic techniques.

2. Literature Search

Extensive searches were carried out in pertinent databases in order to examine literature, papers, and research pertaining to the AI and Nanotechnology framework. The following search terms were used to search the websites of international organizations and conduct thorough searches on Scopus, ACM Digital Library, and PubMed:“ Glioblastoma” AND “Imaging” AND “Biomarkers” AND “Nanotechnology” AND “Artificial intelligence” AND “clinical decision” AND “Ethical OR law OR regulation”. Review demonstrates how AI may be integrated with medical sensors and devices based on nanotechnology, allowing for individualized monitoring, advanced clinical decision support, and evidence-based suggestions for optimizing treatment and managing disease. In order to shed light on the various technologies that have developed in this dynamic and quickly evolving field, this review attempts to provide a thorough overview of AI and nanotechnology integration within the healthcare industry. As a result, it aims to overcome the primary obstacles to integrating such technologies into clinical practice by offering a comprehensive review of the ethical and legal environment around the use of AI and nanotechnology in healthcare [28,29].

3. Current Diagnostic Modalities and Limitations

Glioblastomas are a type of tumor with well-known MRI features. They comprise various infiltrating intraparenchymal lesions. In contrast to cerebral edema, the core of the lesion appears hypointense. Neoangiogenesis is visible on perfusion-weighted imaging, but diffusion/perfusion sequences provide a more precise diagnosis [30]. Due to cellular activity, choline/creatinine and choline/N-acetylaspartate ratios in MR spectroscopy (MRS) are not specific enough to diagnose glioblastomas; however, a smaller myoinositol peak and a greater lactate and lipid peak are good indicators of glioblastoma. Apart from assessing the extent of peritumoral invasion, tests can differentiate glioblastomas from brain abscesses, lymphomas, and metastases [31]. The lack of efficient diagnostic techniques is a major issue with glioblastoma treatment; neurological testing and neuroimaging studies are the main methods used for diagnosis. The slow dispersion process of brain tumors causes delayed detection, which results in the missed clinical signs [32,33].
The prognosis for secondary glioblastoma multiforme (GBM), a type of glioma that affects young people, is better than that of primary GBM. It can be identified by a variety of molecular markers and is often found in the frontal lobe [34]. Biofluid glial tumor detection can improve the quality of life for patients. The process of genotyping circulating tumor nucleic acids makes it possible to classify cancers, evaluate genetic alterations, and calculate the prognosis and tumor burden. Clinically significant biomarkers, including IDH1/2 mutant status, MGMT promoter methylation, 1p/19q co-deletion, and ATRX loss, are used to analyze these genetic indications [35,36].
Although miRNA expression levels are useful for early disease detection, including cancer, their low specificity and selectivity render them unreliable markers for cancer diagnosis. MiRNAs are not appropriate for non-tumor pathologies because they can alter several genes. Clinical evaluation and additional molecular testing are necessary for an accurate diagnosis of glioblastoma [37]. Numerous proteins have been described, and alterations in their quantitative or qualitative makeup are currently linked to the development of cancer patients’ tumors. VEGF and angiogenesis-associated proteins (FGF-b, IGFBP-2, Ang2, EGF and others), extracellular matrix proteins (TSP1/2, TNC, Cyr61/CCN1, OPN, etc.), matrix metalloproteinases (MMP-2, MMP-9, AEG-1), cell line-associated proteins (GFAP), macrophage migration inhibitory factor (MIF), and functionally related proteins (DD-T, CD74, CD44, CXCR2 and CXCR4) are a few of these proteins that can be used for the diagnosis and prognostic assessment of glioblastoma development [38,39]. Cell lipids, metabolites, and monomers are tiny molecules that diffuse across cell membranes and enter cells. However, their low molecular weight and lack of specificity make them ineffective for diagnosis. Circulating tumor cells enter the bloodstream, with antibodies, genetic analysis, and cell size evaluation used. glioma tumor cells enter the bloodstream due to their invasiveness [40,41].
Conventional imaging technologies for glioblastoma diagnosis, including MRI, CT, and PET, are commonly utilized to characterize tumors and aid in treatment planning; however, they exhibit significant limitations. These imaging modalities often lack biological specificity, complicating the differentiation between active tumor tissue and treatment-induced alterations such as inflammation or necrosis, which may result in misdiagnosis and delayed treatment decisions. PET imaging, although capable of identifying specific biochemical markers, encounters challenges like nonspecific binding and the necessity for tracers that can penetrate the blood–brain barrier, reducing its efficacy in certain situations. Furthermore, these imaging techniques do not furnish molecular or genetic data critical for personalized therapy. For instance, while MRI can reveal abnormal regions that may not denote cancer, PET tracers such as [18F] fluoro-ethyl-L-tyrosine or [18F] Fluorodopa can aid in the diagnosis but still struggle with specificity and availability. Consequently, there is an increasing interest in alternative diagnostic approaches like liquid biopsies and biosensors, which can deliver molecular insights and facilitate less invasive, more frequent monitoring of glioblastoma [42,43,44,45].

4. Nanotechnology for Glioblastoma Diagnosis

The study of atomic and molecular characteristics and interactions at the nanoscale (1–100 nanometers) and the application of these properties in diverse fields is known as nanotechnology. Continuous advancements in nanotechnology over the past few decades have shown great promise for the detection and management of gliomas. Nanoparticles (NPs) have the potential to be helpful in a range of cancer treatments due to their small size, adaptable surface, and low level of detrimental side effects [46,47,48]. When exposed to magnetic fields, certain magnetic nanoparticles (MNPs) show high permeability and characteristic magnetism, which makes them valuable for biomarker detection and possibly crucial for the diagnosis of various illnesses, including gliomas [49,50,51]. Figure 2 shows some nanomaterials used in the diagnosis and treatment of glioblastoma.
Accurate diagnosis is crucial in treating gliomas, which grow invasively in the brain and are difficult to pinpoint with imaging methods. Gliomas are a target compound due to their strong affinity for specific binding sites and are used as therapeutic and diagnostic agents. Both passive and active targeting are employed to transport nanoparticles into biological barriers. The EPR effect is caused by passive mechanisms in the brain, such as the buildup of nanoparticles, which promote defects and deficiencies in endothelial cells [52].
Nanoparticles, such as liposomes and dendrimers, cross the blood–brain barrier to deliver chemotherapeutic agents to GBM cells, improving drug bioactivity and reducing systemic toxicity. Quantum dots aid in early imaging and real-time tracking of tumor progression [53].
Nanoparticles (NPs) can cause cytogenotoxicity in three ways: direct damage to cell surfaces and organelles, formation of toxic ions, and oxidative stress. Factors like size, shape, structure, aggregation state, surface properties, dosage, and material type influence NPs’ toxicity. To minimize cytotoxicity, in vitro and in vivo cytotoxicity testing is required for biomedical applications, considering cell type and exposure duration. The lack of sophisticated machinery for accurate and scalable nanomaterial production, the challenge of evaluating their safety and effectiveness, and the unique limitations of specific materials are some potential disadvantages that must be addressed before discussing the many benefits of nanoparticles and their uses. The creation of nanoparticles should be facilitated by factors including stability in physiological settings, biocompatibility, propensity to cross biological barriers, and low cost and high effectiveness [54]. Table 1 displays information on the nanotechnology approach for the diagnosis of glioblastoma.

4.1. Nanotechnology for MRI

Gadolinium-based contrast agents’ poor sensitivity, resolution, and background noise limit the use of current MRI techniques for glioma diagnosis. High magnetic relaxation and specificity nanoscale contrast agents have become viable substitutes as clinical requirements change, with the potential to improve tissue differentiation and MRI sensitivity. Iron oxide nanoparticles (IONPs) and manganese oxide nanoparticles (MnO NPs) are two examples of magnetic nanomaterials that work well for T1/T2 MRIs [64]. Superparamagnetic iron oxide can be used to identify low-grade gliomas because it is covalently decorated with interleukin-6 receptor-targeting peptide, which enables the particle to cross the blood–brain barrier [65]. By thermally breaking down manganese-based compounds, MnO NPs (nanoparticles) with excitation-dependent fluorescence have been created for use as nanoprobes in magnetic resonance imaging [66].
Using SPIONs modified with indocyanine (Cy7) molecules and peptides, a nanoprobe was created for MRI/NIR fluorescence dual-modality imaging that demonstrated improved tumor-homing and barrier penetration capabilities. In both ex vivo and in vivo imaging, this probe allowed for precise aggregation at glioma sites, which could enhance preoperative and intraoperative GBM imaging [67]. The design of paramagnetic or Gd-containing nanomaterials with optimal ratios to improve imaging contrast is necessary to better distinguish IDH1 mutation patients and treat GBM pseudoprogression. Furthermore, for better targeting and imaging resolution, Gd can be functionalized or encapsulated in liposomes [68].

4.1.1. Computerized Tomography Nanoprobe

PET, SPECT, and ECT are frequently used imaging methods for the diagnosis of gliomas. While conventional iodine agents have low specificity and resolution, contrast materials improve X-ray imaging to differentiate between gliomas and normal brain tissue. On the other hand, compared to iodine, gold nanoparticles provide imaging results that are more stable and clear [69]. Selecting polyethyleneimine as the template and modifying it successively with polyethylene glycol (PEG) and a glioma-specific peptide (chlorotoxin, CTX) is comparatively simple. The complex is then embedded onto gold nanoparticles (Au NP). In subcutaneous tumor models, these particles were effective as a nanoprobe for targeted SPECT/CT imaging and radionuclide therapy of glioma cells both in vitro and in vivo [70].
A nanoprobe was demonstrated to be able to penetrate the blood–brain barrier and specifically target glioma cells in a rat intracranial glioma model. Gold nanoparticles can be readily altered to improve targeted delivery and BBB penetration. By binding to target molecules and radioisotopes, they can increase the quantity of contrast agent that is delivered to tumor lesions. Furthermore, glioma imaging and treatment have made use of radioisotope-labeled nanomaterials [71]. In conclusion, radiation modalities based on nanotechnology might represent a useful innovation for delivering more accurate and thorough anatomical data.

4.1.2. Fluorescent Nanoprobe

The behavior of silicon nanoparticles (SNPs-PEG-RGD-FITC) as gliomas developed was investigated using a mouse glioma model. The study showed that these nanoparticles, which target integrin αvβ3, effectively penetrated and remained in solid gliomas after intravenous injection. This method enables the observation of the dynamics of nanomaterials in glioma targeting and infiltration in real time [72]. A new work described a glioma-targeting and redox-activatable theranostic nanoprobe (Co-NP-RGD1/1) for on-demand synergistic chemotherapy/photodynamic therapy (Chemo-PDT) of orthotopic gliomas, guided by magnetic resonance (MR) and fluorescence (FL) bimodal imaging [73].
In the presence of cRGD-targeting groups on the surface, intravenous Co-NP-RGD could be given to in situ glioma cells across the blood–brain barrier in an in vitro model, producing an increased MRI contrast signal for the localization of gliomas in the brain [73]. Based on the idea of NIR-II fluorescence imaging, a new multimodal nanoprobe of NIR-II fluorescent molecules with aggregation-induced emission (AIE) characteristics has been developed [74]. Using a mouse glioma model, they were able to do dual-modal molecular imaging of gliomas with high resolution and high signal-to-noise ratio by using NIR one-region photoacoustic imaging and NIR two-region fluorescence. Furthermore, multimodal nanoprobe guiding for identifying the intraoperative borders of gliomas has advanced significantly in other domestic and foreign investigations, offering a new guidance method for glioma resection [75].
Gold nanoparticles, Ag and Cu-doped nanoparticles, and rare Earth nanomaterials are examples of nanoparticles that have the right absorption in the infrared window and are utilized for fluorescence and NIR detection. Compared to NIR detection, QDs have exceptional chemical and photochemical stability as well as a high fluorescence quantum yield [76]. Streptavidin-PEG-CdSe/ZnS QDs coupled with biotin aptamers for in vitro and in vivo fluorescence imaging of gliomas based on particular EGFRvIII signaling [75]. Additionally, a method for real-time in vivo intraoperative visualization of GBM has been reported: plasminogen activator receptor (uPAR) linked with NIR-II nanoprobe [77].
As a preoperative marker, their findings revealed a clearly defined glioma boundary with strong fluorescence intensity. Carbon dots have garnered interest among various QDs because of their low toxicity and good biocompatibility, which makes them appropriate for use in biomedical applications. CDs generated from metformin and noted their multimodal strong fluorescence capacity to penetrate the blood–brain barrier and function as an imaging tool for brain disorders, such as the U87 cell model [78].

4.1.3. PA Imaging

PA imaging utilizes FL imaging with the spatial resolution of soft tissue imaging (ultrasound). PA imaging has been applied recently to brain illnesses, including structural visualizations, lymph node imaging, and tumor imaging [79]. Using a wide variety of nanomaterials, including Au, carbon nanomaterials, transition metal dichalcogenides 2D nanomaterials (MXenen), and semiconducting polymers, several studies examined the PA imaging of brain tumors [80]. Covalently coupled molybdenum disulfide (MoS2) nanosheets with indocyanine green (ICG) dye were able to image GBM at a depth of 3.5 mm [81].
In order to overcome the difficulties of light absorption and scattering in living tissue, the NIR-II PA imaging modules show a markedly improved signal/background ratio and improved tissue penetration. For efficient imaging of orthotopic brain tumors, a conjugated polymer (CP)-based nanoparticle (NP) has been created. Through the Stille polymerization of benzobisthiadiazole (BBT) and benzodithiophene, this NP was created using electron-deficient acceptors and donor paradigms. It was then nanoprecipitated into a DSPE-PEG2000 matrix. This design makes it possible to detect glioma tumors up to 3.4 mm below the skull with little interference from the background, indicating that NIR-II PA contrast agents may be essential for further translational studies in imaging technologies [82].

4.2. Liquid Biopsy

The diagnosis and prognosis of brain malignancies have been thoroughly investigated using neuroimaging and biopsy. A simple and minimally invasive technique is liquid biopsy (LB), which uses plasma or cerebrospinal fluid (CSF) [83]. LB is a highly invasive tissue extraction substitute that is restricted to a small tumor area and uses bodily fluids to provide information that is typically combined with a clinical diagnosis [84]. Because of the turnover and continuous release of nucleic acids into the bloodstream, LB makes it possible to monitor tumors in real time, which aids in the early detection and successful treatment of brain tumors. DNA and RNA are utilized in this context to describe malignancy, serving as biomarkers in LBs and enhancing the capacity to characterize the genetic profile of GBM. The sensitivity of these assays is dependent on the size, kind, and volume of the tumor, and LBs allow for the detection of changes in pathways [3].
Since it is close to the brain tissues and has a higher concentration of biomarkers than plasma, CSF has been studied due to the difficulties in obtaining blood fluid under optimal circumstances. According to recent research, ultrasound combined with LB raises marker concentrations and promotes noninvasive deep tissue penetration into the tumor [73,76]. Numerous microfluidics-based technologies have been studied to find events and mutations that are hard to spot using traditional imaging methods [85]. Figure 3 explained Liquid biopsy in Glioblastoma.
For cancer monitoring and early diagnosis, liquid biopsy is increasingly using a variety of biomarkers, including circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), exosomes, extracellular vesicles (EVs), and microRNAs. Clinical trials being conducted by renowned cancer centers are yielding encouraging results, especially for CTCs and ctDNA. Additionally, EVs have greatly enhanced the detection and diagnosis of a number of solid tumors [86]. These biomarkers help with disease tracking and treatment decisions by offering vital information about tumor characteristics. Liquid biopsies’ main benefits are their noninvasiveness (just blood or fluid samples are needed) and their suitability for continuous monitoring, which reduces patient risk and discomfort while enabling the identification of dynamic changes in the disease [87].
Machine learning models can identify the mutations with the highest clinical significance by classifying and assessing the discovered mutations through training. The possibility that ctDNA would be found in the plasma samples of lung cancer patients was investigated using models such as logistic regression and elastic net [88]. By analyzing specific DNA methylation segments of CpG islands, the challenge of determining the tissue from which ctDNA originates can be overcome. Since they are the first abnormalities to show up during the course of cancer development, they are reliable markers for determining the tissue origin of ctDNA. Studies that have employed machine learning to examine whole-genome sequencing data of cfDNA and other data types have found that whole-genome methylation sequences offer the highest predictive value for differentiating the tissue origin of ctDNA [89]. This does not, however, imply that brief cfDNA sequences are worthless for machine learning applications. Short sequences can greatly improve machine learning models’ diagnostic skills, according to research [90].

4.3. Extracellular Vesicles

Extracellular vehicles (EVs) are naturally occurring cellular products with a hydrophilic aqueous core and an exterior hydrophobic lipid bilayer. They are essential for maintaining cell homeostasis and play a key role in cancer development by regulating invasion, migration, drug resistance, angiogenesis induction, and proliferation. Glioma cells can produce various types of EVs, making them attractive targets for diagnostic research. EVs are crucial intercellular communication mediators and biomarkers for diagnosing illnesses and assessing treatment effectiveness. EVs are classified into various sizes, including exosomes, microvesicles, oncosomes, apoptotic bodies, and big oncosomes. They can enter bodily fluids and transport various substances [91]. They are then internalized through cellular fusion or endocytosis; several substances can enhance these internalization processes. EVs have a great ability to carry specific messages across the blood–brain barrier [92]. Since EVs can be separated from bodily fluids, including blood, urine, and saliva, they provide numerous benefits in terms of early identification and enhancing patient quality of life before a tumor develops a more serious malignant potential [93].
Extracellular vesicles (EVs) or circulating tumor nucleic acids can be used in liquid biopsies in a number of ways to increase their sensitivity and specificity. By employing dual antibody capture and photosensitizer-bead detection to target disease-specific markers like CD147, sophisticated analytical methods, including the ExoScreen assay, allow for the direct and extremely sensitive identification of cancer-derived EVs from blood without the need for intricate purification. Biomarker sensitivity and diagnostic accuracy can be further improved by combining different liquid biopsy methods (e.g., exosomes plus circulating tumor DNA) or enriching for cancer-specific exosomes. Composite signatures that improve cancer detection’s sensitivity and specificity can be found by integrating and analyzing multianalyte data from EVs and other biomarkers using machine learning [94].

4.4. MicroRNAs

MiRNAs are noncoding RNAs made up of 20–22 nucleotides that adversely control gene expression through recognition, transcription, translation, and epigenetic mechanisms. They also control the expression of numerous proteins involved in the EGFR signaling pathway. Because they are either elevated or downregulated in prognostic studies with GBM patients, many noncoding RNAs have been thought to be possible biomarkers for GBM, making them crucial diagnostic biomarkers [95]. Since miRNA is found in biofluids, stability is essential for regulation. Nanoparticles are used to carry it in order to get around this instability. Research employing miRNA in conjunction with a functionalized nanographene oxide coupled model demonstrated that it significantly improves transfection effectiveness, inhibits the growth of cancer cells more effectively, and shows promise for molecular imaging-based diagnosis [95,96]. The coupling with EVs is another tactic to increase miRNA stability, creating an effective delivery system using artificially produced nano-sized vesicles [97].

4.5. Biosensing for GBM

Although tissue and liquid biopsies are intrusive procedures, they are frequently unable to assess “real-time” tumor dynamics due to ongoing treatment. Currently, a tissue biopsy from a suspected GBM lesion, combined with histological analysis, is the gold standard diagnostic procedure for GBM [98]. In various biofluids, noncoding RNAs (ncRNAs, including microRNA, long noncoding RNA, and circular RNAs) and CNAs have been explored as potential diagnostic markers for CNS malignancies. For patients with glioma tumors, these CNAs are discharged into the biofluidic streams, such as blood and CSF, as EVs or exosomes. Because certain exosomes can be elevated in bodily fluids, they may be a noninvasive diagnostic biomarker for GBM. To do this, a variety of devices were created as biosensors for the detection of gliomas in bodily fluids, including blood and CSF, as clinical diagnostic reagents. These devices ranged from point-of-care analysis to traditional sandwich ELISA-based microarray chip technologies [99,100].
A micronuclear magnetic resonance (μNMR) functionalized microfluidic chip platform. At an early stage of GBM diagnosis, this chip may evaluate EV enrichment (EGFRvIII mRNA) using real-time RNA collection and analysis [101]. The immunomagnetic exosome RNA (iMER) setup isolates glioma-specific EVs using antibody-conjugated magnetic beads, which are lysed on chip surfaces. Before reverse transcription and polymerase chain reaction (PCR) analysis, the lysed EVs (EGFR (þ) and EGFRvIII (þ)) exosomes go through a filter that resembles a glass bead mesh. The current chip combines a custom thermocycler-based PCR setup with a portable fluorescence detector.
Another method for separating and analyzing extracellular vesicles (EVs) linked to glioblastoma multiforme (GBM) involves a fluorescence-based biosensing technique. It utilizes alternating current electrokinetic (ACE) devices to align microelectrodes with an electric field, creating dielectrophoretic (DEP) high-field zones. These zones concentrate nucleosomes and proteins, allowing for the targeted detection using fluorescent antibodies specific to exosomal proteins, such as anti-CD63. The ACE device can separate GBM EVs from raw plasma in about 30 min. Additionally, a nanoplasmonic platform employing materials like titanium nitride (TiN) has been developed to measure glioma-derived exosomes and analyze nanoparticle EVs [101,102].
Surface plasmon resonance (SPR) analysis was performed using the biotinylated antibody coupled BA-TiN plasmonic biosensor. Exosomes (30–200 nm) may be quantitatively detected and isolated from a glioma cell (U251MG) using this method. A diagnostic marker seen on the surfaces of glioma cells and exosomes produced from U251 GM is the EGFRvIII mutant protein [103]. Recently, label-free electrochemical biosensors based on Zr-based metal–organic frameworks (Zr-MOFs) were developed to collect exosomes produced from GBM. The EGFR and EGFRvIII diagnosis biomarkers were precisely targeted by the (H-C-acpacp-FALGEA-NH2) peptide ligands conjugated on the Zr-MOFs surfaces. This aids in clinical analysis and helps distinguish between normal and diseased GBM at an extremely early stage. Numerous studies have also shown that, in addition to the angiogenic proteins, GBM-derived exosomes contain mRNA, CNAs, and miRNAs, which can serve as diagnostic indicators for early-stage GBM. The CSF of both healthy individuals and GBM patients contained a variety of exosome-derived miRNAs [30,104].
The sensitivity and specificity of nanostructures, including nanoparticles, nanowires, and hybrid nanomaterials, have transformed biosensing. By manipulating these nanoengineered components, GBM-specific markers can be selectively attached, increasing detection throughput and accuracy. Data analysis, pattern recognition, and predictive modeling are all improved by artificial intelligence (AI), which raises biosensor performance. AI-TENG and other AI-assisted biosensors provide accuracy, efficiency, and self-powered functioning. Through IoT platforms, machine learning models facilitate remote monitoring and increase the accuracy of cancer detection [105,106,107].

5. AI for Multi-Omics and Radiomic Integration

AI has had a significant impact on oncology, as it has completely changed how complicated problems associated with cancer are approached. AI-powered methods have significantly improved oncologic research’s accuracy and productivity, paving the way for customized cancer therapies. Its uses are numerous and include drug development, genomic research, data mining from medical records, and analysis of cancer images [108,109].

Machine Learning Techniques in Multi-Omics Data Integration

Supervised learning in medical imaging trains models with labeled data to distinguish between benign and malignant tumors. Unsupervised learning analyzes unlabeled data to categorize patients based on genetic similarities for personalized treatments. Reinforcement learning enhances treatment strategies through trial and error. Deep learning, especially with convolutional neural networks (CNNs), excels at processing unstructured data like images for cancer analysis. For sequential data, recurrent neural networks and long short-term memory networks assist in medical record mining and genetic sequence evaluation [110]. By grouping related data points, clustering techniques like k-means and hierarchical clustering can assist in the discovery of new cancer subtypes or disease states. By projecting multi-omics data into lower-dimensional spaces, dimensionality reduction techniques like principal component analysis (PCA) and t-distributed stochastic neighbor embedding (t-SNE) make the data easier to handle for additional analysis [111].
Random Forest is an ensemble learning technique that combines several decision trees to improve prediction accuracy for classification and regression tasks on a variety of datasets. On the other hand, hierarchical clustering groups related data points into nested clusters in order to find natural data structures. Gaining insight into the main uses of these algorithms can highlight their advantages in data processing and AI-based multiomics analysis. Furthermore, through the analysis of patient omics profiles and treatment outcomes, reinforcement learning teaches models to make decisions based on environmental input, enabling the identification of the best ways to treat illnesses [112].
Multi-layer neural networks are used in deep learning, a branch of machine learning, to find intricate patterns in large, varied datasets, particularly in multiomics research. It is excellent at processing complex, non-linear data and has uses in audio, image, and natural language processing. Furthermore, deep learning can efficiently examine omics data to identify important patterns that conventional statistical techniques might overlook [113].
Deep learning architectures, particularly convolutional neural networks (CNNs) and recurrent neural networks (RNNs), are pivotal in multi-omics research. CNNs adapt omics data as one- or two-dimensional images, while RNNs handle sequential data like time-series or transcriptomes [64]. Additional models, including deep belief networks, autoencoders, and generative adversarial networks, help identify hidden patterns and correlations in omics data. This integration of deep learning may significantly enhance our understanding of complex biological systems, potentially leading to innovative diagnostic and therapeutic strategies [114].
Graph neural networks (GNNs) are increasingly utilized in node classification, especially in predicting cancer molecular subtypes through models that examine gene-gene relationships using graph convolutional networks (GCNs). Advanced technologies enable profiling various biological “omics” data, such as transcriptomics and proteomics, providing insights into biological systems and diseases. Traditional pathological diagnoses do not reveal the molecular basis of diseases, and individual omics data tend to offer correlational rather than causal insights, limiting their use in proactive processes [115,116,117].
Deep learning is used in large-scale CRISPR screening datasets to find the best target sequences, increasing editing efficiency and reducing off-target effects. By automatically recommending gRNA designs based on comprehensive CRISPR experiments, AI-based tools speed up gene editing. Furthermore, the PinPoint test is an AI-powered blood test designed to enhance cancer referral procedures. To provide precise cancer risk predictions, it makes use of a machine learning algorithm that incorporates variables like age and sex. Medical practitioners can reduce post-pandemic assessment delays by using this decision support tool to prioritize high-risk patients for prompt evaluations [118].
In precision oncology, it is crucial to forecast how patient populations will react to treatment. To predict treatment responses, machine learning methods have been used, such as deep neural network evaluations. In order to predict drug response behavior, the MOLI method, a multi-omics late mix technique that uses deep neural networks, combines expression data, copy number variations, and somatic mutations. Additionally, MOLI is used in conjunction with data on board medications that have comparable targets [119].
Significant differences between pancreatic ductal adenocarcinoma specimens and normal tissues in terms of RNA and miRNA transcriptomics data have been found using the Support Vector Machine (SVM) and Leave-One-Out Cross-Validation (LOOCV) models. To find effective diagnostic markers, these features (chosen RNAs and miRNAs) were further integrated with miRNA target expression data. These markers were then verified in other datasets and physiologically interpreted by analyzing the relevant target genes’ pathways [120]. Finding biomarkers for glioblastoma is essential to minimize invasive procedures. A new technique using differential scanning fluorimetry has been developed, allowing ML-based algorithms to distinguish cancer patients from healthy individuals with about 92% precision, based on plasma denaturation profiles. This method requires only a simple blood test and has the potential to serve as a pan-cancer diagnostic and monitoring tool [121]. Table 2 displayed information about the multiomics-based prediction of cancer prognosis.

6. Bridging Between AI and Nanotechnology

AI and nanotechnology are being combined to create new materials, devices, and systems with unprecedented capabilities. This integration accelerates the identification of new materials, enhances conductivity, reactivity, and strength, and facilitates precise control and supervision in nanofabrication and manufacturing. AI-driven techniques monitor and optimize processes, while robotic systems automate high-throughput production, enhancing productivity and efficiency [157]. AI and nanotechnology are revolutionizing drug delivery, diagnostic tools, and treatments in nanomedicine, promoting adaptable treatment regimens, real-time remedial optimization, predictive models, and data-driven insights in renewable energy production and sustainability [158].
AI uses data-driven methods, virtual screening, and predictive modeling to identify new pharmaceutical targets and therapeutic approaches. It improves patient outcomes and pharmaceutical efficacy. AI plays a critical role in nanofabrication and manufacturing, improving scalability, precision, and efficiency. Techniques use sensor data to optimize performance and track process factors in real-time [159,160].
AI uses simulation and predictive modeling to help design and optimize nanofabrication processes. By analyzing data from prior manufacturing cycles, machine learning algorithms forecast how changes to the process would affect the properties of the nanostructure. AI-powered simulations speed up the development of nanomaterials and devices by enabling virtual experimentation [161]. Through the analysis of enormous databases of attributes and performance metrics, AI greatly assists in the design and optimization of biomaterials. It anticipates traits, finds desirable materials fast, and optimizes processing parameters and mixtures [162]. Biomaterial behavior at the molecular and nanoscale levels can be studied, and biocompatibility, immunogenicity, and bioactivity can be predicted using AI-driven computer modeling and simulation. By comprehending host response and tissue regeneration mechanisms, these models direct the creation of biomaterials for biomedical applications [163].
AI enhances biomaterials for tissue engineering and drug delivery applications and helps with material design. To develop effective drug delivery systems, machine learning algorithms examine drug release kinetics, diffusion mechanisms, and cellular absorption paths. In order to guarantee reliability, consistency, and security for clinical usage, AI also assists in characterizing and monitoring the quality of biomaterials at every stage of production [164,165]. Through image analysis, disease early warning, biomarker search, precision medicine, and accelerating drug discovery and protein structure prediction, artificial intelligence is transforming the medical field [166]. Through surface adsorption, chemical interactions, and optical transduction, nanomaterials such as metal nanoparticles, graphene, and carbon nanotubes enable the selective detection of pollutants, resulting in highly sensitive and specific detection [167]. Table 3 display information on types of nanoparticles, AI methods, and clinical uses in cancer treatment, and Table 4 display information on the AI-integrated nanoparticle system for enhanced Glioma diagnosis and therapy.

7. Translational Considerations

7.1. Regulatory Landscape for AI in Genomic Medicine

As different jurisdictional regulations currently govern AI-driven multi-omics research, regulatory frameworks for AI in genetic medicine must change to ensure clinical validity, data security, and patient safety. In the US, the FDA and NIH provide oversight and have set rules for medical software that uses AI. However, because self-learning AI systems are dynamic, there are substantial regulatory challenges [187,188].
The European Union enforces the General Data Protection Regulation (GDPR), which requires informed consent and robust data protection for genetic research. High-risk AI applications, such as genomics and personalized medicine, are intended to be regulated by the EU’s Artificial Intelligence Act [189]. Asian nations like China and Japan, on the other hand, prioritize developing AI in healthcare while abiding by regional data protection regulations. China has imposed stringent data governance through the Personal Information Protection Law (PIPL), while Japan has developed ethical AI principles in line with its genomic research [190].
Despite these rules, cross-border AI-driven genomic research faces difficulties due to a lack of uniformity among international regulatory organizations. To promote international research collaboration, efforts are underway to establish international standards for the validation of AI models, the ethical application of AI, and the interoperability of multi-omics data [191]. Policymakers must strive toward standardized AI validation frameworks and create flexible regulatory laws that keep up with technological changes to promote AI-driven multi-omics research while maintaining ethical compliance and patient safety [192]. Table 5 displayed information on the regulatory framework for AI-driven genomics research across different regions.
The EMA and FDA’s current protocols are inadequate for the complexity of nanomedicine and AI-driven technologies, necessitating the development of new assessment and regulatory frameworks. The varied characteristics of nanoparticles, including size, shape, surface charge, and composition, pose significant challenges because they have a substantial impact on their pharmacokinetics, safety, and efficacy when compared to traditional medications [193].
Because AI-based algorithms need to process patient-specific data in real-time, they add complexity to nanomedicine. Regulatory bodies need to establish precise rules for AI-driven nanomedicine that cover both the computational logic of AI and the characteristics of nanoparticles. Adaptive clinical trial designs that enable ongoing optimization based on real-time data are one example of this [194]. Preclinical development, where AI predicts toxicity and optimizes designs; early clinical trials using data from wearable sensors enabled by AI; safety and efficacy evaluations in a variety of populations; and post-marketing surveillance, where AI analytics are used to guarantee long-term safety and compliance, are the four phases of the clinical translation roadmap. To handle discrepancies and set precise guidelines, regulatory participation is crucial at every stage [171,177].
Cancer treatment AI frameworks are still in their infancy, which leaves developers and physicians in the dark. Transparency of AI algorithms, which are frequently viewed as “black boxes,” raises ethical questions about explainability, which is important for important decisions. There are also concerns that AI may exacerbate socioeconomic, racial, and geographic healthcare disparities. Particularly with complex treatments like novel nanomedicines, concerns regarding patient autonomy and the informed consent process must be addressed as AI becomes more prevalent in decision-making [195,196].

7.2. Challenges in Clinical Implementation and Data Standardization

Data standardization, model reproducibility, and clinician adoption are just a few of the logistical and technical obstacles that must be overcome for AI-driven multi-omics to be successfully incorporated into clinical practice. Harmonization across datasets is challenging because multi-omics datasets are generated from various experimental platforms, each with its own formats, resolutions, and noise levels [197]. Since genomic, transcriptomic, and proteomic datasets frequently lack standardized formats and metadata annotations, data interoperability is a significant problem in clinical deployment. Although data integration has improved with the introduction of international bioinformatics standards like the FAIR (Findable, Accessible, Interoperable, and Reusable) principles, disparities remain between healthcare systems and research institutes [198].
The repeatability of AI models in actual healthcare settings is another major obstacle. Although a lot of AI-driven multi-omics models work well on research datasets, sample bias and overfitting frequently limit their capacity to be applied to a variety of patient groups. Researchers must use cross-cohort validation techniques and provide reliable benchmarking datasets to evaluate AI model performance in various clinical settings in order to increase reproducibility [199]. Another obstacle to clinician adoption of AI-driven multi-omics technologies is the lack of technical proficiency among many medical practitioners to decipher AI-generated findings. The integration of AI into healthcare operations is made more difficult by the absence of user-friendly interfaces and decision-support tools. AI developers need to focus on developing clinically interpretable models and providing medical practitioners with sufficient training on how to utilize AI-driven multi-omics platforms to address this [200].
AI-driven genomic medicine implementation in low-resource healthcare settings is further complicated by issues with computational infrastructure and cost. Processing large-scale multi-omics datasets requires high-performance computer resources, which limits the availability of AI-driven solutions in developing nations. Although cloud-based AI systems have surfaced as a viable remedy, issues with data security and legal compliance need to be resolved before they can be widely used [201]. Notwithstanding these obstacles, multi-omics research powered by AI has enormous promise to revolutionize precision medicine. AI-powered genomic medicine has the potential to become a common clinical tool that improves patient outcomes, therapy tailoring, and disease diagnostics by tackling problems with data standardization, model repeatability, and clinician acceptance [202]. Table 6 display information on the bridging the translation gap between the AI and Nanotechnology in Cancer Therapy.
To expedite the transition of nanotechnology and artificial intelligence methods from preclinical studies to therapeutic applications, urgent research is required. Important tasks include creating standardized datasets, creating explainable AI algorithms, and giving multidisciplinary cooperation top priority. Future clinical studies should evaluate the practical performance, safety, and efficacy of AI-guided nanomedicine platforms. AI optimization for the design and functionalization of nanomaterials can enhance their medicinal efficacy and biocompatibility. By facilitating smooth clinical translation and commercialization, regulatory frameworks and guidelines can help close the gap between precision oncology laboratory innovation and patient-centered treatments [172,197].
Glioblastoma (GBM) patient outcomes are greatly improved by early detection and timely surgical intervention, according to recent clinical evidence. In particular, a better overall survival rate—28.4 months for early intervention groups versus 18.7 months for delayed treatment groups—correlates with a GBM diagnosis made within two weeks of symptom onset and surgery performed within three weeks. Higher Karnofsky performance scores indicate that patients who receive early intervention also typically maintain better functional status [212,213]. Furthermore, compared to conventional histology-based methods, prompt detection of tumor progression or recurrence via advanced imaging or molecular profiling enables more aggressive and customized management strategies, improving clinical outcomes. However, because GBM is aggressive, has a high recurrence rate, and the available treatments are ineffective, the overall prognosis is still bleak [214,215]. Although it does not significantly alter the course of the disease for the majority of patients, early detection enables patients to receive adjuvant and surgical treatments while their neurological condition is better, extending their survival and improving their quality of life. Therefore, the lack of cures for GBM limits the benefits of early detection, even though it enhances outcomes within the current therapeutic framework [212,213].

8. Current Challenges and Future Perspectives

The physical and chemical characteristics of nanoparticles that impact imaging quality, clinical viability, and diagnostic efficacy influence the selection of these particles for imaging. Gold nanoparticles are preferred for their optical and biocompatible qualities, while superparamagnetic iron oxide nanoparticles are necessary for magnetic resonance imaging because of their powerful magnetic characteristics. Other nanoparticles with paramagnetic and photoluminescent properties, such as quantum dots and manganese oxide, are selected. Regulatory approval, circulation time, and toxicity all have an impact on clinical viability. Biocompatibility, targeted delivery, quality, reproducibility, signal attenuation, low penetration depth, and separating signals from background noise are among the difficulties; clinical implementation is further complicated by regulatory barriers [213,214,215,216,217,218].
Large-scale multi-omics data integration is anticipated to be greatly improved by AI, opening up new avenues for drug development, personalized medicine, and disease etiology research. In order to address current computational and privacy issues, future developments will concentrate on federated learning, edge computing, and quantum AI. The combination of edge AI and omics research also enables real-time processing on portable diagnostics and sequencing platforms, which speeds up and lowers the cost of genomic analysis, especially in healthcare settings with limited resources [201,219].
Quantum AI is projected to revolutionize large-scale omics data processing by overcoming computational limitations in multi-omics integration, protein structure prediction, and genomic variant interpretation. Quantum-enhanced deep learning models are expected to significantly accelerate complex biological computations, thereby advancing genomic research. Additionally, AI-driven data harmonization will facilitate the integration of diverse datasets from multiple cohorts, enhancing collaboration in research. The standardization of AI-powered data pipelines will ensure replicable and clinically actionable multi-omics insights, promoting interoperability across different omics platforms [220,221,222].
Inaccurate model representation can result from an uneven distribution of data in medical AI. Methods like transfer learning and synthetic data generation are suggested to improve accuracy. Techniques like cross-institutional data collection and reinforcement learning are used to improve generalization. To lessen bias in training datasets, future studies should concentrate on interpretability, data privacy, and ethical issues. Training on a variety of datasets, worldwide validation, and real-world data testing are necessary to guarantee the robustness of AI diagnostic models. Standardized reporting, explainable AI, federated learning, and continuous monitoring to spot biases and performance problems are some examples of evaluation protocols [223,224,225].
Traditional diagnostic techniques still require specialized equipment and trained personnel, even with developments in microfluidics, nanotechnology, and artificial intelligence. This might do away with the necessity for centralized labs and pathologists. Cost-effectiveness, limited processing power, data protection, and regulatory approval are some of the difficulties with nano-AI proof-of-concept tools. Particularly in environments with limited infrastructure, interdisciplinary collaboration is required to develop affordable instruments for individualized treatment and speedy diagnostics. To fully maximize this potential, more innovation and validation are required [226].

9. Conclusions

The commercialization of nanoparticles faces FDA approval challenges due to time-consuming and labor-intensive processes. Artificial intelligence and computer-aided nanoparticle design have been significant in clinical practice. Although nanoparticles have many advantages, problems such as a lack of sophisticated equipment, safety concerns, and material limitations must be resolved. When producing nanoparticles, factors including cost, efficacy, biocompatibility, propensity to cross biological barriers, and physiological stability should be taken into account. The combination of artificial intelligence (AI) with multi-omics research has transformed precision medicine by facilitating thorough data analysis, the identification of biomarkers, and genomics-driven treatment approaches. These advancements have improved early disease diagnosis, individualized treatment planning, and drug discovery. Notable innovations include AI-powered genomic variant interpretation using deep learning to detect disease-linked mutations more accurately and machine learning-based biomarker discovery that enhances predictive and prognostic capabilities for targeted therapies. AI also facilitates systems-level understanding through multi-omics network analysis, refining risk prediction and patient stratification. Despite these strides, challenges remain in clinical translation, data harmonization, ethical concerns, and the transparency of AI models. AI’s capacity to manage vast, heterogeneous datasets enables superior predictive modeling and therapeutic targeting, particularly in complex diseases such as cancer, neurodegenerative, and metabolic disorders. Nevertheless, ensuring model interpretability, reproducibility, and fairness across diverse populations is crucial for widespread adoption in clinical practice. The responsible use of AI in healthcare requires global regulatory standards and ethical frameworks, which require collaboration among researchers, regulatory bodies, and industry stakeholders. Policymakers must be proactive, adaptable, and able to predict developments, regulate with foresight, and modify policies as needed. Ultimately, AI-powered nanotechnology research represents a transformative force in healthcare, fostering interdisciplinary collaboration and catalyzing advancements in precision medicine that promise global patient benefits.

Author Contributions

Conceptualization, M.V.S. and A.Y.P.; methodology, M.V.S.; software, A.Y.P.; validation, M.V.S., I.B. and A.Y.P.; formal analysis, M.V.S. and I.B.; investigation, M.V.S.; resources, I.B.; data curation, A.Y.P.; writing—original draft preparation, M.V.S. and I.B.; writing—review and editing, M.V.S., I.B. and A.Y.P.; visualization, A.Y.P.; supervision, M.V.S. and I.B.; project administration, M.V.S. and I.B.; funding acquisition, I.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
GBMGlioblastoma Multiform
IDHIsocitrate dehydrogenase
2-HG2-hydroxyglutarate
VEGFVascular endothelial growth factor
PDGFPlatelet-derived growth factor
EGFREpidermal growth factor receptor
PTENThe phosphate and tensin homolog
SHHThe Sonic Hedgehog
MGMT6-methylguanine-DNA methyltransferase
TMZTemozolomide
CCNU1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea
TP53Tumor protein p53
MRIMagnetic resonance imaging
ddPCRDroplet digital PCR
FLAIRFluid-attenuated inversion recovery
PWIPerfusion-weighted imaging
EVsExtracellular vesicles
MNPsMagnetic nanoparticles
IONPsIron oxide nanoparticles
PETPositron emission tomography
SPECTSingle-photon emission computed tomography
ECTElectron Computed Tomography
AIEAggregation-induced emission
LBLiquid biopsy
TiNTitanium nitride
μNMRMicronuclear magnetic resonance
iMERImmunomagnetic exosome RNA
ACEAlternating current electrokinetic
GDPR General Data Protection Regulation
CNNsConvolutional neural networks
SPRSurface plasmon resonance
PCAprincipal component analysis
t-SNEt-distributed stochastic neighbor embedding
GNNsGraph neural networks
SVMSupport Vector Machine
LOOCVLeave-One-Out Cross-Validation
NIHNational Institutes of Health
FDAFood and Drug Administration
RNN Recurrent neural network
PIPLPersonal Information Protection Law
BBTBenzobisthiadiazole
MoS2 molybdenum disulfide
ICGIndocyanine green

References

  1. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  2. Grochans, S.; Cybulska, A.M.; Simińska, D.; Korbecki, J.; Kojder, K.; Chlubek, D.; Baranowska-Bosiacka, I. Epidemiology of Glioblastoma Multiforme–Literature Review. Cancers 2022, 14, 2412. [Google Scholar] [CrossRef] [PubMed]
  3. Thakkar, J.; Dolecek, T.; Horbinski, C.; Ostrom, Q.; Lightner, D.; Barnholtz-Sloan, J.; Villano, J. Epidemiologic and Molecular Prognostic Review of Glioblastoma. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef]
  4. Grech, N.; Dalli, T.; Mizzi, S.; Meilak, L.; Calleja, N.; Zrinzo, A. Rising Incidence of Glioblastoma Multiforme in a Well-Defined Population. Cureus 2020, 12, e8195. [Google Scholar] [CrossRef]
  5. Alexopoulos, G.; Zhang, J.; Karampelas, I.; Patel, M.; Kemp, J.; Coppens, J.; Mattei, T.A.; Mercier, P. Long-Term Time Series Forecasting and Updates on Survival Analysis of Glioblastoma Multiforme: A 1975–2018 Population-Based Study. Neuroepidemiology 2022, 56, 75–89. [Google Scholar] [CrossRef]
  6. Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.; Simjee, S. Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment. Asian Pac. J. Cancer Prev. APJCP 2017, 18, 3–9. [Google Scholar] [CrossRef] [PubMed]
  7. Khabibov, M.; Garifullin, A.; Boumber, Y.; Khaddour, K.; Fernandez, M.; Khamitov, F.; Khalikova, L.; Kuznetsova, N.; Kit, O.; Kharin, L. Signaling pathways and therapeutic approaches in glioblastoma multiforme (Review). Int. J. Oncol. 2022, 60, 69. [Google Scholar] [CrossRef]
  8. Yalamarty, S.S.K.; Filipczak, N.; Li, X.; Subhan, M.A.; Parveen, F.; Ataide, J.A.; Rajmalani, B.A.; Torchilin, V.P. Mechanisms of resistance and current treatment options for glioblastoma multiforme (GBM). Cancers 2023, 15, 2116. [Google Scholar] [CrossRef]
  9. Nakada, M.; Kita, D.; Watanabe, T.; Hayashi, Y.; Teng, L.; Pyko, I.V.; Hamada, J.-I. Aberrant Signaling Pathways in Glioma. Cancers 2011, 3, 3242–3278. [Google Scholar] [CrossRef]
  10. Turkalp, Z.; Karamchandani, J.; Das, S. IDH mutation in glioma: New insights and promises for the future. JAMA Neurol. 2014, 71, 1319–1325. [Google Scholar] [CrossRef]
  11. Du, Y.; Chen, H.; Li, X.-F.; Lu, Y.-C.; Feng, Y.-B.; Zhang, L. Molecular mechanism of Notch signaling with special emphasis on microRNAs: Implications for glioma. J. Cell. Physiol. 2018, 234, 158–170. [Google Scholar] [CrossRef]
  12. Morad, S.A.; Cabot, M.C. Ceramide-orchestrated signalling in cancer cells. Nat. Rev. Cancer 2013, 13, 51–65. [Google Scholar] [CrossRef]
  13. Wick, W.; Weller, M.; Weiler, M.; Batchelor, T.; Yung, A.W.; Platten, M. Pathway inhibition: Emerging molecular targets for treating glioblastoma. Neuro Oncol. 2011, 13, 566–579. [Google Scholar] [CrossRef]
  14. Westermark, B. Platelet-derived growth factor in glioblastoma-driver or biomarker? Ups. J. Med. Sci. 2014, 119, 298–305. [Google Scholar] [CrossRef]
  15. Ohgaki, H.; Kleihues, P. Genetic pathways to primary and secondary glioblastoma. Am. J. Pathol. 2007, 170, 1445–1453. [Google Scholar] [CrossRef] [PubMed]
  16. Mantamadiotis, T. Towards Targeting PI3K-Dependent Regulation of Gene Expression in Brain Cancer. Cancers 2017, 9, 60. [Google Scholar] [CrossRef] [PubMed]
  17. Janbazian, L.; Karamchandani, J.; Das, S. Mouse models of glioblastoma: Lessons learned and questions to be answered. J. Neurooncol. 2014, 118, 1–8. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, Y.; Liu, X.; Chen, L.; Du, W.; Cui, Y.; Piao, X.; Li, Y.; Jiang, C. Targeting glioma stem cells via the Hedgehog signaling pathway. Neuroimmunol. Neuroinflamm. 2014, 1, 9. [Google Scholar]
  19. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008, 455, 1061–1068. [Google Scholar] [CrossRef]
  20. Appin, C.L.; Brat, D.J. Molecular pathways in gliomagenesis and their relevance to neuropathologic diagnosis. Adv. Anat. Pathol. 2015, 22, 50–58. [Google Scholar] [CrossRef]
  21. Hegi, M.E.; Diserens, A.C.; Gorlia, T.; Hamou, M.F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef]
  22. Kong, X.-T.; Nguyen, N.T.; Choi, Y.J.; Zhang, G.; Nguyen, H.N.; Filka, E.; Green, S.; Yong, W.H.; Liau, L.M.; Green, R.M.; et al. Phase 2 Study of Bortezomib Combined with Temozolomide and Regional Radiation Therapy for Upfront Treatment of Patients with Newly Diagnosed Glioblastoma Multiforme: Safety and Efficacy Assessment. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 1195–1203. [Google Scholar] [CrossRef]
  23. Obrador, E.; Moreno-Murciano, P.; Oriol-Caballo, M.; López-Blanch, R.; Pineda, B.; Gutiérrez-Arroyo, J.; Loras, A.; González-Bonet, L.; Martínez-Cadenas, C.; Estrela, J.; et al. Glioblastoma Therapy: Past, Present, and Future. Int. J. Mol. Sci. 2024, 25, 2529. [Google Scholar] [CrossRef]
  24. McKinnon, C.; Nandhabalan, M.; Murray, S.A.; Plaha, P. Glioblastoma: Clinical presentation, diagnosis, and management. BMJ 2021, 374, n1560. [Google Scholar] [CrossRef]
  25. Szopa, W.; Burley, T.A.; Kramer-Marek, G.; Kaspera, W. Diagnostic and therapeutic biomarkers in glioblastoma: Current status and future perspectives. BioMed Res. Int. 2017, 2017, 8013575. [Google Scholar] [CrossRef]
  26. Cheng, Y.; Morshed, R.A.; Auffinger, B.; Tobias, A.L.; Lesniak, M.S. Multifunctional nanoparticles for brain tumor imaging and therapy. Adv. Drug Deliv. Rev. 2014, 66, 42. [Google Scholar] [CrossRef] [PubMed]
  27. Rauf, M.A.; Rehman, F.U.; Zheng, M.; Shi, B. Nanomedicine in Brain Diseases: Principles and Application; Xue, X., Ed.; Springer: Singapore, 2019; p. 29. [Google Scholar]
  28. Mennella, C.; Maniscalco, U.; De Pietro, G.; Esposito, M. Ethical and regulatory challenges of AI technologies in healthcare: A narrative review. Heliyon 2024, 10, e26297. [Google Scholar] [CrossRef] [PubMed]
  29. Kopkova, J.; Sana, P.; Fadrus, O.; Slaby, O. Cerebrospinal fluid microRNAs as diagnostic biomarkers in brain tumors. Clin. Chem. Lab. Med. 2018, 56, 869. [Google Scholar] [CrossRef]
  30. Hosseini, A.; Ashraf, H.; Rahimi, F.; Alipourfard, I.; Alivirdiloo, V.; Hashemi, B.; Yazdani, Y.; Ghazi, F.; Eslami, M.; Reza, M.A.S.; et al. Recent advances in the detection of glioblastoma, from imaging-based methods to proteomics and biosensors: A narrative review. Cancer Cell Int. 2023, 23, 98. [Google Scholar] [CrossRef] [PubMed]
  31. Kleihues, P.; Ohgaki, H. Primary and secondary glioblastomas: From concept to clinical diagnosis. Neuro Oncol. 1999, 1, 44–51. [Google Scholar] [CrossRef]
  32. Sizoo, E.M.; Braam, L.; Postma, T.J.; Pasman, H.R.; Heimans, J.J.; Klein, M.; Reijneveld, J.C.; Taphoorn, M.J. Symptoms and problems in the end-of-life phase of high-grade glioma patients. Neuro Oncol. 2010, 12, 1162–1166. [Google Scholar] [CrossRef] [PubMed]
  33. Peeken, J.C.; Goldberg, T.; Pyka, T.; Bernhofer, M.; Wiestler, B.; Kessel, K.A.; Tafti, P.D.; Nusslin, F.; Braun, A.E.; Zimmer, C.; et al. Combining multimodal imaging and treatment features improves machine learning-based prognostic assessment in patients with glioblastoma multiforme. Cancer Med. 2019, 8, 128–136. [Google Scholar] [CrossRef]
  34. Price, S.J.; Young, A.M.; Scotton, W.J.; Ching, J.; Mohsen, L.A.; Boonzaier, N.R.; Lupson, V.C.; Griffiths, J.R.; McLean, M.A.; Larkin, T.J. Multimodal MRI can identify perfusion and metabolic changes in the invasive margin of glioblastomas. J. Magn. Reson. Imaging 2016, 43, 487–494. [Google Scholar] [CrossRef]
  35. Aoki, T.; Kondo, Y.; Karakida, K.; Naito, H.; Kajiwara, H.; Ota, Y. A mucinous adenocarcinoma of the lip with elevated serum carcinoembryonic antigen levels: A case report. Oral Maxillofac Surg. 2020, 24, 127–132. [Google Scholar] [CrossRef]
  36. Herman, A.; Gruden, K.; Blejec, A.; Podpečan, V.; Motaln, H.; Rožman, P.; Hren, M.; Zupančič, K.; Veber, M.; Verbovšek, U.; et al. Analysis of Glioblastoma Patients’ Plasma Revealed the Presence of MicroRNAs with a Prognostic Impact on Survival and Those of Viral Origin. PLoS ONE 2015, 10, e0125791. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, H.; Peng, R.; Wang, J.; Qin, Z.; Xue, L. Circulating microRNAs as potential cancer biomarkers: The advantages and disadvantage. Clin. Epigenet. 2018, 10, 59. [Google Scholar] [CrossRef]
  38. Hegi, M.E.; Liu, L.; Herman, J.G.; Stupp, R.; Wick, W.; Weller, M.; Mehta, M.P.; Gilbert, M.R. Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylation with clinical outcomes in glioblastoma and clinical strategies to modulate MGMT activity. J. Clin. Oncol. 2008, 26, 4189–4199. [Google Scholar] [CrossRef]
  39. Mangano, K.; Mazzon, E.; Basile, M.S.; Di Marco, R.; Bramanti, P.; Mammana, S.; Petralia, M.C.; Fagone, P.; Nicoletti, F. Pathogenic role for macrophage migration inhibitory factor in glioblastoma and its targeting with specific inhibitors as novel tailored therapeutic approach. Oncotarget 2018, 9, 17951–17970. [Google Scholar] [CrossRef] [PubMed]
  40. Spalding, K.; Board, R.; Dawson, T.; Jenkinson, M.D.; Baker, M.J. A review of novel analytical diagnostics for liquid biopsies: Spectroscopic and spectrometric serum profiling of primary and secondary brain tumors. Brain Behav. 2016, 6, e00502. [Google Scholar] [CrossRef] [PubMed]
  41. Krol, I.; Castro-Giner, F.; Maurer, M.; Gkountela, S.; Szczerba, B.M.; Scherrer, R.; Coleman, N.; Carreira, S.; Bachmann, F.; Anderson, S.; et al. Detection of circulating tumour cell clusters in human glioblastoma. Br. J. Cancer 2018, 119, 487–491. [Google Scholar] [CrossRef]
  42. Thenuwara, G.; Curtin, J.; Tian, F. Advances in Diagnostic Tools and Therapeutic Approaches for Gliomas: A Comprehensive Review. Sensors 2023, 23, 9842. [Google Scholar] [CrossRef] [PubMed]
  43. Seyhan, A. Circulating Liquid Biopsy Biomarkers in Glioblastoma: Advances and Challenges. Int. J. Mol. Sci. 2024, 25, 7974. [Google Scholar] [CrossRef]
  44. Drake, L.; Hillmer, A.; Cai, Z. Approaches to PET Imaging of Glioblastoma. Molecules 2020, 25, 568. [Google Scholar] [CrossRef] [PubMed]
  45. Ronvaux, L.; Riva, M.; Coosemans, A.; Herzog, M.; Rommelaere, G.; Donis, N.; D’Hondt, L.; Douxfils, J. Liquid Biopsy in Glioblastoma. Cancers 2022, 14, 3394. [Google Scholar] [CrossRef]
  46. Zhang, Y.; Gu, X.; Huang, L.; Yang, Y.; He, J. Enhancing precision medicine: Bispecific antibody-mediated targeted delivery of lipid nanoparticles for potential cancer therapy. Int. J. Pharm. 2024, 654, 123990. [Google Scholar] [CrossRef]
  47. Choi, Y.; Son, W.; Han, Y.; Chae, J.; Yang, C.S.; Choi, J. Glycan-targeting nanoparticle for photodynamic immunotherapy of melanoma. Acta Pharm. Sin. B 2023, 13, 1903–1918. [Google Scholar] [CrossRef]
  48. Daraee, H.; Eatemadi, A.; Abbasi, E.; Fekri Aval, S.; Kouhi, M.; Akbarzadeh, A. Application of gold nanoparticles in biomedical and drug delivery. Artif. Cells Nanomed. Biotechnol. 2016, 44, 410–422. [Google Scholar] [PubMed]
  49. Chen, Z.; Wu, C.; Yuan, Y.; Xie, Z.; Li, T.; Huang, H.; Li, S.; Deng, J.; Lin, H.; Shi, Z.; et al. CRISPR-Cas13a-powered electrochemical biosensor for the detection of the L452R mutation in clinical samples of SARS-CoV-2 variants. J. Nanobiotechnol. 2023, 21, 141. [Google Scholar]
  50. Xue, T.; Liang, W.; Li, Y.; Sun, Y.; Xiang, Y.; Zhang, Y.; Dai, Z.; Duo, Y.; Wu, L.; Qi, K.; et al. Ultrasensitive detection of miRNA with an antimonene-based surface plasmon resonance sensor. Nat Commun. 2019, 10, 28. [Google Scholar] [CrossRef]
  51. Zheng, F.; Chen, Z.; Li, J.; Wu, R.; Zhang, B.; Nie, G.; Xie, Z.; Zhang, H. A Highly Sensitive CRISPR-Empowered Surface Plasmon Resonance Sensor for Diagnosis of Inherited Diseases with Femtomolar-Level Real-Time Quantification. Adv. Sci. 2022, 9, e2105231. [Google Scholar] [CrossRef]
  52. Gusmão, L.A.; Matsuo, F.S.; Barbosa, H.F.G.; Tedesco, A.C. Advances in nano-based materials for glioblastoma multiforme diagnosis: A mini-review. Front. Nanotechnol. 2022, 4, 836802. [Google Scholar] [CrossRef]
  53. Shabani, L.; Abbasi, M.; Amini, M.; Amani, A.; Vaez, A. The brilliance of nanoscience over cancer therapy: Novel, promising nanotechnology-based methods for eradicating glioblastoma. J. Neurol. Sci. 2022, 440, 120316. [Google Scholar] [CrossRef]
  54. Abbasi, R.; Shineh, G.; Mobaraki, M.; Doughty, S.; Tayebi, L. Structural parameters of nanoparticles affecting their toxicity for biomedical applications: A review. J. Nanoparticle Res. 2023, 25, 43. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, Y.; Lakshmi, B. Modernistic and Emerging Developments of Nanotechnology in Glioblastoma-Targeted Theranostic Applications. Int. J. Mol. Sci. 2022, 23, 1641. [Google Scholar] [CrossRef] [PubMed]
  56. Irfan, M.; Rahdar, A.; Kharaba, Z.; Fathi-Karkan, S.; Aboudzadeh, M.; Aliahmad, M.; Ullah, S.; Qindeel, M. Nanotechnology in Glioblastoma Therapy: Advances in Drug Delivery Systems and Diagnostic Approaches. J. Drug Deliv. Sci. Technol. 2024, 102, 106322. [Google Scholar] [CrossRef]
  57. Qian, H.; Yu, Y.; Song, X. Nanoparticles Mediated the Diagnosis and Therapy of Glioblastoma: Bypass or Cross the Blood-Brain Barrier. Small 2023, 19, e2302613. [Google Scholar] [CrossRef]
  58. Zottel, A.; Šamec, N.; Jovčevska, I.; Paska, V. Nanomedicine and Immunotherapy: A Step Further towards Precision Medicine for Glioblastoma. Molecules 2020, 25, 490. [Google Scholar] [CrossRef]
  59. Wu, L.; Han, I.; Zeng, X.; Glaser, T. Targeted Nanotechnology in Glioblastoma Multiforme. Front. Pharmacol. 2017, 8, 166. [Google Scholar] [CrossRef]
  60. Ghaznavi, H.; Sheervalilou, R.; Sargazi, S.; Shirvalilou, S.; Afzalipour, R.; Khoei, S. New insights into targeted therapy of glioblastoma using smart nanoparticles. Cancer Cell Int. 2024, 24, 160. [Google Scholar] [CrossRef]
  61. Van Straten, D.; Préat, V.; Broekman, M.; Schiffelers, R.; Zhao, M. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics 2020, 10, 1355–1372. [Google Scholar] [CrossRef]
  62. Yu, S.; Wei, W.; Jiang, J.; Long, S.; Niu, X.; Chen, L.; Xu, H.; Li, X. Application of nanomaterials in the diagnosis and treatment of glioblastoma. Front. Chem. 2022, 10, 1063152. [Google Scholar] [CrossRef]
  63. Wang, H.; Liao, C.; Hsu, J.; Wang, Y.; Tsai, M.; Chu, S.; Wang, C.; Huang, H.; Lai, M. Nanotechnology and Nanocarrier-Based Drug Delivery as the Potential Therapeutic Strategy for Glioblastoma Multiforme: An Update. Cancers 2021, 13, 195. [Google Scholar] [CrossRef]
  64. Vallabani, N.V.; Singh, S.; Karakoti, A.S. Magnetic Nanoparticles: Current Trends and Future Aspects in Diagnostics and Nanomedicine. Curr. Drug Metab. 2019, 20, 457–472. [Google Scholar] [CrossRef]
  65. Tan, J.; Sun, W.; Lu, L.; Xiao, Z.; Wei, H.; Shi, W.; Wang, Y.; Han, S.; Shuai, X. I6P7 peptide modified superparamagnetic iron oxide nanoparticles for magnetic resonance imaging detection of low-grade brain gliomas. J. Mater. Chem. B 2019, 7, 6139–6147. [Google Scholar] [CrossRef]
  66. Lai, J.; Wang, T.; Wang, H.; Shi, F.; Gu, W.; Ye, L. MnO nanoparticles with unique excitation-dependent fluorescence for multicolor cellular imaging and MR imaging of brain glioma. Microchim. Acta 2018, 185, 244. [Google Scholar] [CrossRef]
  67. Xie, R.; Wu, Z.; Zeng, F.; Cai, H.; Wang, D.; Gu, L.; Zhu, H.; Lui, S.; Guo, G.; Song, B.; et al. Retro-enantio isomer of angiopep-2 assists nanoprobes across the blood-brain barrier for targeted magnetic resonance/fluorescence imaging of glioblastoma. Signal Transduct. Target. Ther. 2021, 6, 309. [Google Scholar] [CrossRef] [PubMed]
  68. Mukhtar, M.; Bilal, M.; Rahdar, A.; Barani, M.; Arshad, R.; Behl, T.; Brisc, C.; Banica, F.; Bungau, S. Nanomaterials for diagnosis and treatment of brain cancer: Recent updates. Chemosensors 2020, 8, 117. [Google Scholar] [CrossRef]
  69. Jackson, P.A.; Rahman, W.N.; Wong, C.J.; Ackerly, T.; Geso, M. Potential dependent superiority of gold nanoparticles in comparison to iodinated contrast agents. Eur. J. Radiol. 2010, 75, 104–109. [Google Scholar] [CrossRef]
  70. Zhao, L.; Li, Y.; Zhu, J.; Sun, N.; Song, N.; Xing, Y.; Huang, H.; Zhao, J. Chlorotoxin peptide-functionalized polyethylenimine-entrapped gold nanoparticles for glioma SPECT/CT imaging and radionuclide therapy. J. Nanobiotechnol. 2019, 17, 30. [Google Scholar] [CrossRef] [PubMed]
  71. Silindir, M.; Erdoğan, S.; Özer, A.Y.; Doğan, A.L.; Tuncel, M.; Uğur, Ö.; Torchilin, V.P. Nanosized multifunctional liposomes for tumor diagnosis and molecular imaging by SPECT/CT. J. Liposome Res. 2013, 23, 20–27. [Google Scholar] [CrossRef]
  72. Zhang, W.; Chen, X.; Ding, D.; Zhang, G.; Zhu, Z.; Yang, X.; Li, M.; Liang, L.; Shi, X.; Wang, T.; et al. Real-time in vivo imaging reveals specific nanoparticle target binding in a syngeneic glioma mouse model. Nanoscale 2022, 14, 5678–5688. [Google Scholar] [CrossRef]
  73. An, R.; Liu, L.; Wei, S.; Huang, Z.; Qiu, L.; Lin, J.; Liu, H.; Ye, D. Controlling Disassembly of Paramagnetic Prodrug and Photosensitizer Nanoassemblies for On-Demand Orthotopic Glioma Theranostics. ACS Nano 2022, 16, 20607–20621. [Google Scholar] [CrossRef] [PubMed]
  74. Sheng, Z.; Guo, B.; Hu, D.; Xu, S.; Wu, W.; Liew, W.H.; Yao, K.; Jiang, J.; Liu, C.; Zheng, H.; et al. Bright Aggregation-Induced-Emission Dots for Targeted Synergetic NIR-II Fluorescence and NIR-I Photoacoustic Imaging of Orthotopic Brain Tumors. Adv. Mater. 2018, 28, e1800766. [Google Scholar] [CrossRef]
  75. Tang, T.; Chang, B.; Zhang, M.; Sun, T. Nanoprobe-mediated precise imaging and therapy of glioma. Nanoscale Horiz. 2021, 6, 634–650. [Google Scholar] [CrossRef]
  76. Liu, Z.; Ren, F.; Zhang, H.; Yuan, Q.; Jiang, Z.; Liu, H.; Sun, Q.; Li, Z. Boosting often overlooked long-wavelength emissions of rare-earth nanoparticles for NIR-II fluorescence imaging of orthotopic glioblastoma. Biomaterials 2019, 219, 119364. [Google Scholar] [CrossRef] [PubMed]
  77. Kurbegovic, S.; Juhl, K.; Chen, H.; Qu, C.; Ding, B.; Leth, J.M.; Drzewiecki, K.T.; Kjaer, A.; Cheng, Z. Molecular-targeted NIR-II probes for image-guided brain tumor surgery. Bioconjug. Chem. 2018, 29, 3833–3840. [Google Scholar] [CrossRef] [PubMed]
  78. Kirbas Cilingir, E.; Seven, E.S.; Zhou, Y.; Walters, B.M.; Mintz, K.J.; Pandey, R.R.; Wikramanayake, A.H.; Chusuei, C.C.; Vanni, S.; Graham, R.M.; et al. Metformin-derived carbon dots: Highly biocompatible fluorescent nanomaterials as mitochondrial targeting and blood-brain barrier penetrating biomarkers. J. Colloid Interface Sci. 2021, 592, 485–497. [Google Scholar] [CrossRef]
  79. Fu, Q.; Zhu, R.; Song, J.; Yang, H.; Chen, X. Photoacoustic Imaging: Contrast Agents and Their Biomedical Applications. Adv. Mater. 2019, 31, e1805875. [Google Scholar] [CrossRef]
  80. Fan, Q.; Cheng, K.; Yang, Z.; Zhang, R.; Yang, M.; Hu, X.; Ma, X.; Bu, L.; Lu, X.; Xiong, X. Perylene-Diimide-Based Nanoparticles as Highly Efficient Photoacoustic Agents for Deep Brain Tumor Imaging in Living Mice. Adv. Mater. 2015, 27, 843. [Google Scholar] [CrossRef]
  81. Liu, C.; Chen, J.; Zhu, Y.; Gong, X.; Zheng, R.; Chen, N.; Chen, D.; Yan, H.; Zhang, P.; Zheng, H.; et al. Highly Sensitive MoS2–Indocyanine Green Hybrid for Photoacoustic Imaging of Orthotopic Brain Glioma at Deep Site. Nano-Micro Lett. 2018, 10, 48. [Google Scholar] [CrossRef]
  82. Guo, B.; Sheng, Z.; Hu, D.; Lin, X.; Xu, S.; Liu, C.; Zheng, H.; Liu, B. Molecular engineering of conjugated polymers for biophotonic imaging and therapy. Mater. Hor. 2017, 4, 1151. [Google Scholar]
  83. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic: Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef] [PubMed]
  84. Saenz-Antoñanzas, A.; Auzmendi-Iriarte, J.; Carrasco-Garcia, E.; Moreno-Cugnon, L.; Ruiz, I.; Villanua, J.; Egaña, L.; Otaegui, D.; Samprón, N.; Matheu, A. Liquid biopsy in glioblastoma: Opportunities, applications, and challenges. Cancers 2019, 11, 950. [Google Scholar] [CrossRef] [PubMed]
  85. Xu, Z.; Qiao, Y.; Tu, J. Microfluidic technologies for cfDNA isolation and analysis. Micromachines 2019, 10, 672. [Google Scholar] [CrossRef]
  86. Su, X.; Shan, Z.; Duan, S. Harnessing extracellular vesicles using liquid for cancer diagnosis and monitoring: Highlights from AACR Annual Meeting 2024. J. Hematol. Oncol. 2024, 17, 55. [Google Scholar] [CrossRef]
  87. Yin, H.; Zhang, M.; Zhang, Y.; Zhang, X.; Zhang, X.; Zhang, B. Liquid biopsies in cancer. Mol. Biomed. 2025, 6, 18. [Google Scholar] [CrossRef]
  88. Chabon, J.J.; Hamilton, E.G.; Kurtz, D.M.; Esfahani, M.S.; Moding, E.J.; Stehr, H.; Schroers-Martin, J.; Nabet, B.Y.; Chen, B.; Chaudhuri, A.A.; et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 2020, 580, 245–251. [Google Scholar] [CrossRef]
  89. Jamshidi, A.; Liu, M.C.; Klein, E.A.; Venn, O.; Hubbell, E.; Beausang, J.F.; Gross, S.; Melton, C.; Fields, A.P.; Liu, Q.; et al. Evaluation of cell-free DNA approaches for multi-cancer early detection. Cancer Cell 2022, 40, 1537–1549.e12. [Google Scholar] [CrossRef]
  90. Mouliere, F.; Chandrananda, D.; Piskorz, A.M.; Moore, E.K.; Morris, J.; Ahlborn, L.B.; Mair, R.; Goranova, T.; Marass, F.; Heider, K.; et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl. Med. 2018, 10, eaat4921. [Google Scholar] [CrossRef]
  91. Zottel, A.; Paska, A.V.; Jovcevska, I. Nanotechnology meets oncology: Nanomaterials in brain cancer research, diagnosis, and therapy. Materials 2019, 12, 1588. [Google Scholar] [CrossRef]
  92. Schuurmans, C.; Balakrishnan, A.; Roy, S.; Fleming, T.; Leong, H.S. The emerging role of extracellular vesicles in the glioma microenvironment: Biogenesis and clinical relevance. Cancers 2020, 12, 1964. [Google Scholar] [CrossRef]
  93. Figueroa, J.M.; Carter, B.S. Detection of glioblastoma in biofluids. J. Neurosurg. 2018, 129, 334–340. [Google Scholar] [CrossRef] [PubMed]
  94. Lin, A.; Nimgaonkar, V.; Issadore, D.; Carpenter, E. Extracellular Vesicle-Based Multianalyte Liquid Biopsy as a Diagnostic for Cancer. Annu. Rev. Biomed. Data Sci. 2022, 5, 269–292. [Google Scholar] [CrossRef] [PubMed]
  95. Cellot, G.; Franceschi Biagioni, A.; Ballerini, L. Nanomedicine and graphene-based materials: Advanced technologies for potential treatments of diseases in the developing nervous system. Pediatr. Res. 2021, 92, 71–79. [Google Scholar] [CrossRef]
  96. Kutwin, M.; Sosnowska, M.E.; Strojny-Cieślak, B.; Jaworski, S.; Trzaskowski, M.; Wierzbicki, M.; Chwalibog, A.; Sawosz, E. MicroRNA delivery by graphene-based complexes into glioblastoma cells. Molecules 2021, 26, 5804. [Google Scholar] [CrossRef] [PubMed]
  97. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  98. Verduin, M.; Compter, I.; Steijvers, D.; Postma, A.A.; Eekers, D.B.P.; Anten, M.M.; Ackermans, L.; Ter Laan, M.; Leijenaar, R.T.H.; van de Weijer, T.; et al. Noninvasive Glioblastoma Testing: Multimodal Approach to Monitoring and Predicting Treatment Response. Dis. Markers 2018, 2018, 2908609. [Google Scholar] [CrossRef]
  99. Shao, H.; Im, H.; Castro, C.M.; Breakefield, X.; Weissleder, R.; Lee, H. New technologies for analysis of extracellular vesicles. Chem. Rev. 2018, 118, 1917–1950. [Google Scholar] [CrossRef]
  100. Xu, R.; Greening, D.W.; Zhu, H.J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef]
  101. Ibsen, D.; Wright, J.; Lewis, J.M.; Kim, S.; Ko, S.-Y.; Ong, J.; Manouchehri, S.; Vyas, A.; Akers, J.; Chen, C.C. Rapid Isolation and Detection of Exosomes and Associated Biomarkers from Plasma. ACS Nano 2017, 11, 6641. [Google Scholar] [CrossRef]
  102. Qiu, G.; Thakur, A.; Xu, C.; Ng, S.P.; Lee, Y.; Wu, C.M.L. Detection of Glioma-Derived Exosomes with the Biotinylated Antibody-Functionalized Titanium Nitride Plasmonic Biosensor. Adv. Funct. Mater. 2019, 29, 1806761. [Google Scholar] [CrossRef]
  103. Figueroa, J.M.; Skog, J.; Akers, J.; Li, H.; Komotar, R.; Jensen, R.; Ringel, F.; Yang, I.; Kalkanis, S.; Thompson, R. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro Oncol. 2017, 19, 1494. [Google Scholar] [CrossRef] [PubMed]
  104. Drusco, A.; Bottoni, A.; Lagana, M.; Acunzo, M.; Fassan, L.; Cascione, A.; Antenucci, P.; Kumchala, C.; Vicentini, M.P.; Gardiman, M.P. A differentially expressed set of microRNAs in cerebro-spinal fluid (CSF) can diagnose CNS malignancies. Oncotarget 2015, 6, 20829. [Google Scholar] [CrossRef]
  105. Jiang, M.; Zheng, S.; Zhu, Z. What can AI-TENG do for Low Abundance Biosensing? Front. Bioeng. Biotechnol. 2022, 10, 899858. [Google Scholar] [CrossRef]
  106. Banerjee, A.; Maity, S.; Mastrangelo, C. Nanostructures for Biosensing, with a Brief Overview on Cancer Detection, IoT, and the Role of Machine Learning in Smart Biosensors. Sensors 2021, 21, 1253. [Google Scholar] [CrossRef]
  107. Kasture, K.; Shende, P. Amalgamation of Artificial Intelligence with Nanoscience for Biomedical Applications. Arch. Comput. Methods Eng. 2023, 30, 4667–4685. [Google Scholar] [CrossRef]
  108. Hanif, S.; Muhammad, P.; Niu, Z.; Ismail, M.; Morsch, M.; Zhang, X.; Li, M.; Shi, B. Nanotechnology-based strategies for early diagnosis of central nervous system disorders. Adv. NanoBiomed Res. 2021, 1, 2100008. [Google Scholar] [CrossRef]
  109. Ozaki, Y.; Broughton, P.; Abdollahi, H.; Valafar, H.; Blenda, A.V. Integrating omics data and AI for cancer diagnosis and prognosis. Cancers 2024, 16, 2448. [Google Scholar] [CrossRef] [PubMed]
  110. Shiri, F.M.; Perumal, T.; Mustapha, N.; Mohamed, R. A comprehensive overview and comparative analysis on deep learning models: CNN, RNN, LSTM, GRU. arXiv 2023, arXiv:2305.17473. [Google Scholar] [CrossRef]
  111. Tan, K.; Huang, W.; Hu, J.; Dong, S. A multi-omics supervised autoencoder for pan-cancer clinical outcome endpoints prediction. BMC Med. Inform. Decis. Mak. 2020, 20, 129. [Google Scholar] [CrossRef]
  112. Ali, M.; Aittokallio, T. Machine learning and feature selection for drug response prediction in precision oncology applications. Biophys Rev. 2019, 11, 31–39. [Google Scholar] [CrossRef] [PubMed]
  113. Picard, M.; Scott-Boyer, M.P.; Bodein, A.; Périn, O.; Droit, A. Integration strategies of multi-omics data for machine learning analysis. Comput. Struct. Biotechnol. J. 2021, 19, 3735–3746. [Google Scholar] [CrossRef]
  114. Bian, Y.; Xie, X.Q. Generative chemistry: Drug discovery with deep learning generative models. J. Mol. Model. 2021, 27, 1–18. [Google Scholar] [CrossRef]
  115. Rhee, S.; Seo, S.; Kim, S. Hybrid Approach of Relation Network and Localized Graph Convolutional Filtering for Breast Cancer Subtype Classification. arXiv 2018, arXiv:1711.05859. [Google Scholar] [CrossRef]
  116. Hasin, Y.; Seldin, M.; Lusis, A. Multi-omics approaches to disease. Genome Biol. 2017, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  117. McCague, C.; Ramlee, S.; Reinius, M.; Selby, I.; Hulse, D.; Piyatissa, P.; Bura, V.; Crispin-Ortuzar, M.; Sala, E.; Woitek, R. Introduction to radiomics for a clinical audience. Clin. Radiol. 2023, 78, 83–98. [Google Scholar] [CrossRef]
  118. Fawaz, A.; Ferraresi, A.; Isidoro, C. Systems biology in cancer diagnosis: Integrating omics technologies and artificial intelligence to support physician decision making. J. Pers. Med. 2023, 13, 1590. [Google Scholar] [CrossRef]
  119. Sharifi-Noghabi, H.; Zolotareva, O.; Collins, C.C.; Ester, M. MOLI: Multi-Omics Late Integration with Deep Neural Networks for Drug Response Prediction. Bioinformatics 2019, 35, i501–i509. [Google Scholar] [CrossRef]
  120. Kwon, M.-S.; Kim, Y.; Lee, S.; Namkung, J.; Yun, T.; Yi, S.G.; Han, S.; Kang, M.; Kim, S.W.; Jang, J.-Y.; et al. Integrative Analysis of Multi-Omics Data for Identifying Multi-Markers for Diagnosing Pancreatic Cancer. BMC Genom. 2015, 16, S4. [Google Scholar] [CrossRef]
  121. Ali, H. Artificial intelligence in multi-omics data integration: Advancing precision medicine, biomarker discovery, and genomic-driven disease interventions. Int. J. Sci. Res. Arch. 2023, 8, 1012–1030. [Google Scholar] [CrossRef]
  122. Henderson, S.; Perez-Gil, D.; Caulfield, M.; Rendon, A.; Chubb, D.; Hing, S.; Bentley, D.; Chan, G.; Fowler, T.; Siddiq, A.; et al. Insights for precision oncology from the integration of genomic and clinical data of 13,880 tumors from the 100,000 Genomes Cancer Programme. Nat. Med. 2024, 30, 279–289. [Google Scholar] [CrossRef] [PubMed]
  123. Jacquet, A.; Lacroix, L.; Mège, A.; Isambert, N.; Mosele, F.; André, F.; Filleron, T.; Hajjaji, N.; Jacot, W.; Rouleau, E.; et al. Genomics to select treatment for patients with metastatic breast cancer. Nature 2022, 610, 343–348. [Google Scholar] [CrossRef] [PubMed]
  124. Xie, Y.; Ma, S.; Zhao, Q.; Shia, B.; Huang, J.; Shi, X. Combining multidimensional genomic measurements for predicting cancer prognosis: Observations from TCGA. Brief. Bioinform. 2015, 16, 291–303. [Google Scholar] [CrossRef]
  125. Rao, J.; Zhao, H.; Zhou, X.; Zhang, Z.; Yang, Y.; Chai, H. Integrating multi-omics data through deep learning for accurate cancer prognosis prediction. Comput. Biol. Med. 2021, 134, 104481. [Google Scholar] [CrossRef]
  126. Wang, J.; Williamson, D.; Lindeman, N.; Lu, M.; Mahmood, F.; Chen, R.; Rodig, S. Pathomic Fusion: An Integrated Framework for Fusing Histopathology and Genomic Features for Cancer Diagnosis and Prognosis. IEEE Trans. Med. Imaging 2022, 41, 757–770. [Google Scholar] [CrossRef]
  127. Shady, M.; Joo, B.; Shaban, M.; Williamson, D.; Lipková, J.; Lu, M.; Williams, M.; Mahmood, F.; Chen, R.; Chen, T.; et al. Pan-cancer integrative histology-genomic analysis via multimodal deep learning. Cancer Cell 2022, 40, 865–878.e6. [Google Scholar] [CrossRef]
  128. Durand, S.; Corcos, L.; Jossic-Corcos, C.; Badic, B.; Visvikis, D.; Hatt, M.; Simon, B. Radiogenomics-based cancer prognosis in colorectal cancer. Sci. Rep. 2019, 9, 9743. [Google Scholar] [CrossRef]
  129. Tabernero, J.; Mouillet-Richard, S.; Agueeff, N.; Laurent-Puig, P.; De Reyniès, A.; Sroussi, M.; Dourthe, L.; Lécaille, C.; Bachet, J.; Coutzac, C.; et al. Prognostic Models from Transcriptomic Signatures of the Tumor Microenvironment and Cell Cycle in Stage III Colon Cancer From PETACC-8 and IDEA-France Trials. J. Clin. Oncol. 2025, 43, 1765–1776. [Google Scholar] [CrossRef]
  130. Tsimberidou, A.; Kurzrock, R.; Fountzilas, E.; Bleris, L. Transcriptomics and Solid Tumors: The Next Frontier in Precision Cancer Medicine. Semin. Cancer Biol. 2020, 84, 50–59. [Google Scholar] [CrossRef]
  131. Chapman, L.; Rubin, E.; Srinivasan, A.; Dinstag, G.; Wang, K.; Cha, H.; Schperberg, A.; Kim, D.; Berger, R.; Chung, Y.; et al. Synthetic lethality-mediated precision oncology via the tumor transcriptome. Cell 2021, 184, 2487–2502.e13. [Google Scholar] [CrossRef]
  132. Pellegrini, B.; Yip, S.; Taylor, S.; Reisle, C.; Nelson, J.; Jones, M.; Pleasance, E.; Feng, X.; Tinker, A.; Sadeghi, S.; et al. Whole genome and transcriptome analysis enhances precision cancer treatment options. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2022, 33, 939–949. [Google Scholar] [CrossRef]
  133. Moore, R.; Wee, K.; Mungall, K.; Titmuss, E.; Zhao, Y.; Rassekh, S.; Marra, M.; Jones, S.; Nelson, J.; Ch’ng, C.; et al. Whole-genome and transcriptome integrated analyses guide clinical care of pediatric poor-prognosis cancers. Nat. Commun. 2024, 15, 4165. [Google Scholar] [CrossRef]
  134. Blakely, C.; Benes, C.; Schäffer, A.; Bivona, T.; Sinha, S.; Yosef, N.; Kammula, A.; Jiang, P.; Dhruba, S.; Ruppin, E.; et al. PERCEPTION predicts patient response and resistance to treatment using single-cell transcriptomics of their tumors. Nat. Cancer 2024, 5, 938–952. [Google Scholar] [CrossRef]
  135. Zhang, C.; Pontén, F.; Micke, P.; Uhlén, M.; Lundberg, E.; Lindskog, C.; Sjöblom, T.; Edfors, F.; Bidkhori, G.; Arif, M.; et al. A pathology atlas of the human cancer transcriptome. Science 2017, 357, 2507. [Google Scholar] [CrossRef] [PubMed]
  136. Li, X.; Liu, N.; Zhuang, L.; Li, J.; Li, J. Development and validation of epigenetic modification-related signals for the diagnosis and prognosis of colorectal cancer. BMC Genom. 2024, 25, 51. [Google Scholar] [CrossRef]
  137. Jin, S.; Dai, R.; Xu, Q.; Zhang, R. Exploring the role of epigenetic regulation in cancer prognosis with the epigenetic score. Front. Pharmacol. 2025, 16, 1538205. [Google Scholar] [CrossRef] [PubMed]
  138. Fan, M.; Zhang, K.; Wang, W.; Wu, Z.; Lin, R. Epigenetic and Tumor Microenvironment for Prognosis of Patients with Gastric Cancer. Biomolecules 2023, 13, 736. [Google Scholar] [CrossRef] [PubMed]
  139. Kujan, O.; Alam, A.; Tan, J.; Lim, I.; Della Coletta, R.; Idrees, M.; Brenan, P. Epigenetics in the diagnosis and prognosis of head and neck cancer: A systematic review. J. Oral Pathol. Med. 2024, 53, 90–106. [Google Scholar] [CrossRef]
  140. Bolt, I.; Dillner, J.; Beaufort, I.; Rebitschek, F.; Joly, Y.; Tassé, A.; Colombo, N.; Knoppers, B.; Jones, A.; Sroczynski, G.; et al. Epigenome-based cancer risk prediction: Rationale, opportunities and challenges. Nat. Rev. Clin. Oncol. 2018, 15, 292–309. [Google Scholar] [CrossRef]
  141. Grady, W.; Markowitz, S.; Yu, M. Epigenetic alterations in the gastrointestinal tract: Current and emerging use for biomarkers of cancer. Gastroenterology 2020, 160, 690–709. [Google Scholar] [CrossRef]
  142. Eritja, N.; Matias-Guiu, X.; Gatius, S.; Megino-Luque, C.; Albertí-Valls, M.; Macià, A. Metabolomic-Based Approaches for Endometrial Cancer Diagnosis and Prognosis: A Review. Cancers 2023, 16, 185. [Google Scholar] [CrossRef]
  143. Werner, H.; Rižner, T.; Schröder, C.; Lowy, C.; Fishman, D.; Semczuk, A.; Griesbeck, A.; Tokarz, J.; Romano, A.; Adamski, J. Endometrial cancer diagnostic and prognostic algorithms based on proteomics, metabolomics, and clinical data: A systematic review. Front. Oncol. 2023, 13, 1120178. [Google Scholar] [CrossRef]
  144. Srivastava, A.; Creek, D. Discovery and Validation of Clinical Biomarkers of Cancer: A Review Combining Metabolomics and Proteomics. Proteomics 2018, 19, e1700448. [Google Scholar] [CrossRef]
  145. Shi, H.; Weng, W.; Huang, W.; Shan, Y.; Chen, J.; Xiang, Y. Development of cancer prognostic signature based on pan-cancer proteomics. Bioengineered 2020, 11, 1368–1381. [Google Scholar] [CrossRef] [PubMed]
  146. Zhang, X.; Yin, Z.; Feng, Y.; Ye, H.; Lou, H.; Zhang, H.; Tian, J.; Wang, L.; Wang, Z.; Ni, J.; et al. Plasma-based proteomic and metabolomic characterization of lung and lymph node metastases in cervical cancer patients. J. Pharm. Biomed. Anal. 2024, 253, 116521. [Google Scholar] [CrossRef] [PubMed]
  147. Han, P.; Yang, L.; Song, H.; Jin, P.; Tian, Y.; Yang, T.; Wang, B.; Lou, S.; Wang, M.; Chen, Y.; et al. Metabolomic machine learning predictor for diagnosis and prognosis of gastric cancer. Nat. Commun. 2024, 15, 1657. [Google Scholar] [CrossRef]
  148. Haider, M.; Khalvati, F.; Zhang, Y.; Wong, A.; Oikonomou, A. Radiomics-based Prognosis Analysis for Non-Small Cell Lung Cancer. Sci. Rep. 2017, 7, srep46349. [Google Scholar] [CrossRef]
  149. Vallières, M.; Naqa, I.; Kay-Rivest, E.; Perrin, L.; Liem, X.; Khaouam, N.; Aerts, H.; Sultanem, K.; Seuntjens, J.; Wang, C.; et al. Radiomics strategies for risk assessment of tumour failure in head-and-neck cancer. Sci. Rep. 2017, 7, 10117. [Google Scholar] [CrossRef] [PubMed]
  150. Li, X.; Wang, K.; Xia, Y.; Zhang, Y.; Zhu, F.; Zhang, Y.; Jiang, W.; Wang, X.; Zhang, H.; Ji, G. Biliary Tract Cancer at CT: A Radiomics-based Model to Predict Lymph Node Metastasis and Survival Outcomes. Radiology 2019, 290, 90–98. [Google Scholar] [CrossRef]
  151. Nguyen, V.H.; Kha, Q.H.; Le, V.H.; Le, N.Q.K.; Le, V.L.; Minh, T.N.T. Development and Validation of CT-Based Radiomics Signature for Overall Survival Prediction in Multi-organ Cancer. J. Digit. Imaging 2023, 36, 911–922. [Google Scholar] [CrossRef]
  152. Wang, W.; Cai, D.; Zhong, M.; Wu, X.; Kou, W.; Gao, F.; Li, C.; Zhu, Q.; Huang, Z.; Duan, X.; et al. A Metabolism-Related Radiomics Signature for Predicting the Prognosis of Colorectal Cancer. Front. Mol. Biosci. 2021, 7, 613918. [Google Scholar] [CrossRef] [PubMed]
  153. Deng, J.; Sun, X.; Chen, S.; Zhong, Y.; Chen, C.; Xie, D.; Zhao, M.; Wang, T.; Yang, Y.; She, Y.; et al. Radiomics for Survival Risk Stratification of Clinical and Pathologic Stage IA Pure-Solid Non-Small Cell Lung Cancer. Radiology 2021, 302, 425–434. [Google Scholar] [CrossRef]
  154. Gomez, D.; Liao, Z.; Followill, D.; Balter, P.; Yang, J.; Fave, X.; Court, L.; Zhang, L.; Stingo, F.; Mackin, D.; et al. Delta-radiomics features for the prediction of patient outcomes in non–small cell lung cancer. Sci. Rep. 2017, 7, 588. [Google Scholar] [CrossRef]
  155. Lehrer, E.; Kothari, G.; Korte, J.; Hardcastle, N.; Kron, T.; Zaorsky, N.; Lazarakis, S.; Siva, S. A systematic review and meta-analysis of the prognostic value of radiomics-based models in non-small cell lung cancer treated with curative radiotherapy. Radiother. Oncol. 2020, 155, 188–203. [Google Scholar] [CrossRef]
  156. Liu, Y.; Xi, J.; Qiu, X.; Luo, J.; Luo, Y.; Kang, W.; Yang, Z.; Li, X. Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosis. J. Transl. Med. 2023, 21, 598. [Google Scholar] [CrossRef]
  157. Nandipati, M.; Fatoki, O.; Desai, S. Bridging nanomanufacturing and artificial intelligence—A comprehensive review. Materials 2024, 17, 1621. [Google Scholar] [CrossRef]
  158. Badini, S.; Regondi, S.; Pugliese, R. Unleashing the power of Artificial Intelligence in materials design. Materials 2023, 16, 5927. [Google Scholar] [CrossRef]
  159. Visan, A.I.; Negut, I. Integrating Artificial Intelligence for drug discovery in the context of revolutionizing drug delivery. Life 2024, 14, 233. [Google Scholar] [CrossRef]
  160. Nayak, M.R.; Bayannavar, P.K.; Kamble, R.R. Design of Medicinally Pertinent Multifunctional Inorganic Nanomaterials Using Artificial Intelligence. In Multifunctional Inorganic Nanomaterials for Energy Applications; CRC Press: Boca Raton, FL, USA, 2024. [Google Scholar]
  161. Wang, Z.; Zhu, X. Enhancing nanocrystal synthesis: A comparative study of online artificial intelligence optimization and offline high-throughput experimentation in chemical material discovery. ACS Appl. Nano Mater. 2024, 7, 6499–6505. [Google Scholar] [CrossRef]
  162. Putra, R.V.W.; Marchisio, A.; Zayer, F.; Dias, J.; Shafique, M. Embodied Neuromorphic Artificial Intelligence for Robotics: Perspectives, Challenges, and Research Development Stack. In Proceedings of the 2024 18th International Conference on Control, Automation, Robotics and Vision (ICARCV), Dubai, United Arab Emirates, 12–15 December 2024; pp. 612–619. [Google Scholar]
  163. Kumar, A.; Panda, D.; Gangawane, K. Computational modeling on the design of the morphology of aerogels. In Hybrid Aerogels: Energy and Environmental Applications; De Gruyter: Berlin, Germany, 2024; pp. 269–290. [Google Scholar]
  164. Vijaya, R.; Lincy, B.C.; RamanG, G.; Kirubakaran, N.; Chacko, L.; Bhupathyraaj, M.; Kiruba, M.; Alharbi, H.F. Applications of Artificial Intelligence in Drug Delivery Systems. In Artificial intelligence in Pharmaceutical Sciences; CRC Press: Boca Raton, FL, USA, 2023. [Google Scholar]
  165. Deori, C.; Hujuri, L.; Sarma, G. Artificial Intelligence (AI): It’s Role in Drug Discovery and Novel Drug Delivery System. Int. J. Sci. Res. 2024, 13, 1434–1439. [Google Scholar] [CrossRef]
  166. Xu, Y.; Liu, X.; Cao, X.; Huang, C.; Liu, E.; Qian, S.; Liu, X.; Wu, Y.; Dong, F.; Qiu, C.-W.; et al. Artificial intelligence: A powerful paradigm for scientific research. Innovation 2021, 2, 100179. [Google Scholar] [CrossRef] [PubMed]
  167. Vidal, C.L.A.; Govan, J.E. Machine learning techniques for improving nanosensors in agroenvironmental applications. Agronomy 2024, 14, 341. [Google Scholar] [CrossRef]
  168. Das, K.; J, C. Nanoparticles and convergence of artificial intelligence for targeted drug delivery for cancer therapy: Current progress and challenges. Front. Med. Technol. 2023, 4, 1067144. [Google Scholar] [CrossRef] [PubMed]
  169. Sun, L.; Liu, H.; Ye, Y.; Lei, Y.; Islam, R.; Tan, S.; Tong, R.; Miao, Y.; Cai, L. Smart nanoparticles for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 418. [Google Scholar] [CrossRef]
  170. Kapoor, D.; Sharma, J.; Gandhi, S.; Prajapati, B.; Thanawuth, K.; Limmatvapirat, S.; Sriamornsak, P. AI-driven design and optimization of nanoparticle-based drug delivery systems. Science. Eng. Health Stud. 2024, 18, 24010003. [Google Scholar] [CrossRef]
  171. Akhtar, M.; Nehal, N.; Gull, A.; Parveen, R.; Khan, S.; Khan, S.; Ali, J.; Hamdard, J. Explicating the transformative role of artificial intelligence in designing targeted nanomedicine. Expert Opin. Drug Deliv. 2025, 22, 971–991. [Google Scholar] [CrossRef] [PubMed]
  172. Alafeef, M.; Srivastava, I.; Pan, D. Machine-learning for Precision Breast Cancer Diagnosis and Prediction of the Nanoparticles Cellular internalization. ACS Sens. 2020, 5, 1689–1698. [Google Scholar] [CrossRef]
  173. Esmaeilpour, D.; Ghomi, M.; Zare, E.; Sillanpää, M. Nanotechnology-Enhanced siRNA Delivery: Revolutionizing Cancer Therapy. ACS Appl. Bio. Mater. 2025, 8, 4549–4579. [Google Scholar] [CrossRef]
  174. Chou, W.; Chen, Q.; Yuan, L.; Cheng, Y.; He, C.; Monteiro-Riviere, N.; Riviere, J.; Lin, Z. An artificial intelligence-assisted physiologically-based pharmacokinetic model to predict nanoparticle delivery to tumors in mice. J. Control. Release 2023, 361, 53–63. [Google Scholar] [CrossRef]
  175. Moore, J.; Chow, J. Recent progress and applications of gold nanotechnology in medical biophysics using artificial intelligence and mathematical modeling. Nano Express 2021, 2, 22001. [Google Scholar] [CrossRef]
  176. Ho, D.; Wang, P.; Kee, T. Artificial intelligence in nanomedicine. Nanoscale Horiz. 2019, 4, 365–377. [Google Scholar] [CrossRef] [PubMed]
  177. Adir, O.; Poley, M.; Chen, G.; Froim, S.; Krinsky, N.; Shklover, J.; Shainsky-Roitman, J.; Lammers, T.; Schroeder, A. Integrating Artificial Intelligence and Nanotechnology for Precision Cancer Medicine. Adv. Mater. 2019, 32, e1901989. [Google Scholar] [CrossRef] [PubMed]
  178. Luo, J.; Pan, M.; Mo, K.; Mao, Y.; Zou, D. Emerging Role of Artificial Intelligence in Diagnosis, Classification, and Clinical Management of Glioma. Semin. Cancer Biol. 2023, 91, 110–123. [Google Scholar] [CrossRef]
  179. Rudie, M.; Rauschecker, M.; Bryan, M.; Davatzikos, P.; Mohan, M. Emerging Applications of Artificial Intelligence in Neuro-Oncology. Radiology 2019, 290, 607–618. [Google Scholar] [CrossRef]
  180. Khalighi, S.; Reddy, K.; Midya, A.; Pandav, K.; Madabhushi, A.; Abedalthagafi, M. Artificial intelligence in neuro-oncology: Advances and challenges in brain tumor diagnosis, prognosis, and precision treatment. NPJ Precis. Oncol. 2024, 8, 80. [Google Scholar] [CrossRef]
  181. Razek, A.; Alksas, A.; Shehata, M.; Abdelkhalek, A.; Baky, K.; El-Baz, A.; Helmy, E. Clinical applications of artificial intelligence and radiomics in neuro-oncology imaging. Insights Into Imaging 2021, 12, 152. [Google Scholar] [CrossRef] [PubMed]
  182. Jin, L.; Shi, F.; Chun, Q.; Chen, H.; Ma, Y.; Wu, S.; Hameed, N.; Mei, C.; Lu, J.; Zhang, J.; et al. Artificial Intelligence Neuropathologist for Glioma Classification using Deep Learning on Hematoxylin and Eosin-Stained Slide images and Molecular Markers. Neuro Oncol. 2020, 23, 44–52. [Google Scholar] [CrossRef]
  183. Zhou, K.; Xiao, Z.; Liu, Q.; Wang, X.; Huo, J.; Wu, X.; Zhao, X.; Feng, X.; Fu, B.; Xu, P.; et al. Comprehensive application of AI algorithms with TCR NGS data for glioma diagnosis. Sci. Rep. 2024, 14, 15361. [Google Scholar] [CrossRef]
  184. Li, T.; Li, J.; Chen, Z.; Zhang, S.; Li, S.; Wageh, S.; Al-Hartomy, O.; Al-Sehemi, A.; Xie, Z.; Kankala, R.; et al. Glioma diagnosis and therapy: Current challenges and nanomaterial-based solutions. J. Control. Release 2022, 352, 338–370. [Google Scholar] [CrossRef]
  185. Palkar, A.; Dias, C.; Chadaga, K.; Sampathila, N. Empowering Glioma Prognosis with Transparent Machine Learning and Interpretative Insights Using Explainable AI. IEEE Access 2024, 12, 31697–31718. [Google Scholar] [CrossRef]
  186. Tan, P.; Chen, X.; Zhang, H.; Wei, Q.; Luo, K. Artificial Intelligence Aids in the Development of Nanomedicines for Cancer Management. Semin. Cancer Biol. 2023, 89, 61–75. [Google Scholar] [CrossRef]
  187. Ameh, B. Advancing national security and economic prosperity through resilient and technology-driven supply chains. World J. Adv. Res. Rev. 2024, 24, 483–500. [Google Scholar] [CrossRef]
  188. Marima, R.; Hull, R.; Mbeje, M.; Molefi, T.; Mathabe, K.; Elbagory, A.M.; Demetriou, D.; Dlamini, Z. Role of precision oncology in type II endometrial and prostate cancers in the African population: Global cancer genomics disparities. Int. J. Mol. Sci. 2022, 23, 628. [Google Scholar] [CrossRef] [PubMed]
  189. Pauly, R.; Schwartz, C.E. The future of clinical diagnosis: Moving functional genomics approaches to the bedside. Adv. Mol. Pathol. 2019, 40, 221–230. [Google Scholar]
  190. Pauly, R.; Ziats, C.A.; Abenavoli, L.; Schwartz, C.E.; Boccuto, L. New strategies for clinical trials in autism spectrum disorder. Rev. Recent Clin. Trials 2021, 16, 131–137. [Google Scholar] [CrossRef] [PubMed]
  191. Krentel, F.; Singer, F.; Rosano-Gonzalez, M.L.; Gibb, E.A.; Liu, Y.; Davicioni, E.; Keller, N.; Stekhoven, D.J.; Kruithof-de Julio, M.; Seiler, R. A showcase study on personalized in silico drug response prediction based on the genetic landscape of muscle invasive bladder cancer. Sci. Rep. 2021, 11, 5849. [Google Scholar] [CrossRef] [PubMed]
  192. Chen, C.; Chen, X.; Hu, Y.; Pan, B.; Huang, Q.; Dong, Q.; Xue, X.; Shen, X.; Chen, X. Utilizing machine learning to integrate single-cell and bulk RNA sequencing data for constructing and validating a novel cell adhesion molecules-related prognostic model in gastric cancer. Comput. Biol. Med. 2024, 180, 108998. [Google Scholar] [CrossRef]
  193. Dai, E.; Han, L.; Liu, J.; Xie, Y.; Zeh, H.J.; Kang, R.; Bai, L.; Tang, D. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat. Commun. 2020, 11, 6339. [Google Scholar] [CrossRef]
  194. Sun, S.; Shen, J.; Jiang, J.; Wang, F.; Min, J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Sign. Transduct. Target Ther. 2023, 8, 372. [Google Scholar] [CrossRef]
  195. Zhao, Y.Y.; Lian, J.X.; Lan, Z.; Zou, K.L.; Wang, W.M.; Yu, G.T. Ferroptosis promotes anti-tumor immune response by inducing immunogenic exposure in HNSCC. Oral Dis. 2023, 29, 933–941. [Google Scholar] [CrossRef]
  196. Dixon, S.J.; Patel, D.N.; Welsch, M.; Skouta, R.; Lee, E.D.; Hayano, M. Pharmacological inhibition of cystine-glutamate exchange induces endo plasmic reticulum stress and ferroptosis. eLife 2014, 3, e02523. [Google Scholar] [CrossRef]
  197. Pitini, E.; Baccolini, V.; Isonne, C.; Maran, P.; Marzuillo, C.; Villari, P.; Galeone, D.; Vaia, F. Public health genomics research in Italy: An overview of ongoing projects. Front. Public Health 2024, 12, 1343509. [Google Scholar] [CrossRef] [PubMed]
  198. Palmisano, A.; Zhao, Y.; Li, M.C.; Polley, E.C.; Simon, R.M. OpenGeneMed: A portable, flexible, and customizable informatics hub for the coordination of next-generation sequencing studies in support of precision medicine trials. Brief. Bioinform. 2017, 18, 723–734. [Google Scholar] [CrossRef]
  199. Majumder, M.M. Improving Precision in Therapies for Hematological Malignancies. Diss. Sch. Dr. Ad Sanit. Investig. Univ. Hels. 2018, 66, 2018. [Google Scholar]
  200. De Mattos-Arruda, L.; Siravegna, G. How to use liquid biopsies to treat patients with cancer. ESMO Open 2021, 6, 100060. [Google Scholar] [CrossRef] [PubMed]
  201. Klocker, E.V.; Hasenleithner, S.; Bartsch, R.; Gampenrieder, S.P.; Egle, D.; Singer, C.F.; Rinnerthaler, G.; Hubalek, M.; Schmitz, K.; Bago-Horvath, Z.; et al. Clinical applications of next-generation sequencing-based ctDNA analyses in breast cancer: Defining treatment targets and dynamic changes during disease progression. Mol. Oncol. 2024, 19, 1897–1917. [Google Scholar] [CrossRef]
  202. Kawauchi, D.; Ohno, M.; Miyakita, Y.; Takahashi, M.; Yanagisawa, S.; Omura, T.; Yoshida, A.; Kubo, Y.; Igaki, H.; Ichimura, K.; et al. Early Diagnosis and Surgical Intervention Within 3 Weeks from Symptom Onset Are Associated with Prolonged Survival of Patients with Glioblastoma. Neurosurgery 2022, 91, 741–748. [Google Scholar] [CrossRef]
  203. Gavas, S.; Quazi, S.; Karpiński, T. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef]
  204. Chehelgerdi, M.; Goldberg Allela, O.; Pecho, R.; Jayasankar, N.; Rao, D.; Thamaraikani, T.; Vasanthan, M.; Viktor, P.; Lakshmaiya, N.; Saadh, M.; et al. Progressing nanotechnology to improve targeted cancer treatment: Overcoming hurdles in its clinical implementation. Mol. Cancer 2023, 22, 169. [Google Scholar] [CrossRef]
  205. Fan, D.; Cao, Y.; Cao, M.; Wang, Y.; Cao, Y.; Gong, T. Nanomedicine in cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 293. [Google Scholar] [CrossRef]
  206. Khan, M.; Alshahrani, M.; Wahab, S.; Gupta, G.; Kesharwani, P. Evolution of artificial intelligence as a modern technology in advanced cancer therapy. J. Drug Deliv. Sci. Technol. 2024, 98, 105892. [Google Scholar] [CrossRef]
  207. Pandurangan, P.; Dinesh, R.; MohanaSundaram, A.; Samrat, A.; Meenambika, S.; Vedanarayanan, V.; Meena, R.; Namasivayam, S.; Moovendhan, M. Integrating cutting-edge technologies: AI, IoT, blockchain and nanotechnology for enhanced diagnosis and treatment of colorectal cancer—A review. J. Drug Deliv. Sci. Technol. 2023, 91, 105197. [Google Scholar] [CrossRef]
  208. Goldberg, M. Improving cancer immunotherapy through nanotechnology. Nat. Rev. Cancer 2019, 19, 587–602. [Google Scholar] [CrossRef]
  209. Song, W.; Anselmo, A.; Huang, L. Nanotechnology intervention of the microbiome for cancer therapy. Nat. Nanotechnol. 2019, 14, 1093–1103. [Google Scholar] [CrossRef]
  210. Zhu, J.; Lee, H.; Huang, R.; Zhou, J.; Zhang, J.; Yang, X.; Zhou, W.; Jiang, W.; Chen, S. Harnessing nanotechnology for cancer treatment. Front. Bioeng. Biotechnol. 2025, 12, 1514890. [Google Scholar] [CrossRef] [PubMed]
  211. Nie, S.; Xing, Y.; Kim, G.; Simons, J. Nanotechnology applications in cancer. Annu. Rev. Biomed. Eng. 2007, 9, 257–288. [Google Scholar] [CrossRef] [PubMed]
  212. Narita, Y.; Kawauchi, D.; Ohno, M.; Takahashi, M.; Yanagisawa, S.; Omura, T.; Miyakita, Y. AB023. Early diagnosis and treatment lead to improved survival of glioblastoma patients. Chin. Clin. Oncol. 2024, 13 (Suppl. S1), AB023. [Google Scholar] [CrossRef]
  213. Zhang, Y.; Lucas, C.; Young, J.; Morshed, R.; McCoy, L.; Bush, O.; Taylor, J.; Daras, M.; Butowski, N.; Villanueva-Meyer, J.; et al. Prospective genomically guided identification of “early/evolving” and “undersampled” IDH-wildtype glioblastoma leads to improved clinical outcomes. Neuro Oncol. 2022, 24, 1749–1762. [Google Scholar] [CrossRef]
  214. Qi, D.; Li, J.; Quarles, C.; Fonkem, E.; Wu, E. Assessment and prediction of glioblastoma therapy response: Challenges and opportunities. Brain 2022, 146, 1281–1298. [Google Scholar] [CrossRef]
  215. Jiang, S.; Liu, Y.; Xu, Y.; Sang, X.; Lu, X. Research on liquid biopsy for cancer: A bibliometric analysis. Heliyon 2023, 9, e14145. [Google Scholar] [CrossRef]
  216. Luo, D.; Wang, X.; Burda, C.; Basilion, J. Recent Development of Gold Nanoparticles as Contrast Agents for Cancer Diagnosis. Cancers 2021, 13, 1825. [Google Scholar] [CrossRef]
  217. Padmanabhan, P.; Kumar, A.; Kumar, S.; Chaudhary, R.; Gulyás, B. Nanoparticles in practice for molecular-imaging applications: An overview. Acta Biomater. 2016, 41, 1–16. [Google Scholar] [CrossRef]
  218. Crist, R.; Dasa, S.; Liu, C.; Clogston, J.; Dobrovolskaia, M.; Stern, S. Challenges in the development of nanoparticle-based imaging agents: Characterization and biology. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 13, e1665. [Google Scholar] [CrossRef]
  219. Appenzeller, B.M.; Chadeau-Hyam, M.; Aguilar, L. Skin exposome science in practice: Current evidence on hair biomonitoring and future perspectives. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 26–30. [Google Scholar] [CrossRef]
  220. Chen, Y.; Liu, S.; Papageorgiou, L.G.; Theofilatos, K.; Tsoka, S. Optimisation models for pathway activity inference in cancer. Cancers 2023, 15, 1787. [Google Scholar] [CrossRef]
  221. Park, I.; Kim, N.; Lee, S.; Park, K.; Son, M.Y.; Cho, H.S.; Kim, D.S. Characterization of signature trends across the spectrum of non-alcoholic fatty liver disease using a deep learning method. Life Sci. 2023, 314, 121195. [Google Scholar] [CrossRef] [PubMed]
  222. Li, Y.; Wang, X.; Zhang, J.; Zhang, S.; Jiao, J. Applications of artificial intelligence (AI) in research on non-alcoholic fatty liver disease (NAFLD): A systematic review. Rev. Endocr. Metab. Disord. 2022, 23, 387–400. [Google Scholar] [CrossRef] [PubMed]
  223. Xu, X.; Li, J.; Zhu, Z.; Zhao, L.; Wang, H.; Song, C.; Chen, Y.; Zhao, Q.; Yang, J.; Pei, Y. A comprehensive review on the synergy of multi-modal data and ai technologies in medical diagnosis. Bioengineering 2024, 11, 219. [Google Scholar] [CrossRef] [PubMed]
  224. Yala, A.; Mikhael, P.; Strand, F.; Lin, G.; Satuluru, S.; Kim, T.; Banerjee, I.; Gichoya, J.; Trivedi, H.; Lehman, C.; et al. Multi-Institutional Validation of a Mammography-Based Breast Cancer Risk Model. J. Clin. Oncol. 2021, 40, 1732–1740. [Google Scholar] [CrossRef]
  225. Lipková, J.; Chen, R.; Chen, B.; Lu, M.; Barbieri, M.; Shao, D.; Vaidya, A.; Chen, C.; Zhuang, L.; Williamson, D.; et al. Artificial intelligence for multimodal data integration in oncology. Cancer Cell 2022, 40, 1095–1110. [Google Scholar] [CrossRef]
  226. Arya, S.; Dias, S.; Jelinek, H.; Hadjileontiadis, L.; Pappa, A. The convergence of traditional and digital biomarkers through AI-assisted biosensing: A new era in translational diagnostics? Biosens. Bioelectron. 2023, 235, 115387. [Google Scholar] [CrossRef] [PubMed]
Figure 1. RTK (Receptor tyrosine kinase) signaling pathway in Glioblastoma.
Figure 1. RTK (Receptor tyrosine kinase) signaling pathway in Glioblastoma.
Targets 03 00031 g001
Figure 2. Nanomaterials in the diagnosis and treatment of glioblastoma.
Figure 2. Nanomaterials in the diagnosis and treatment of glioblastoma.
Targets 03 00031 g002
Figure 3. Liquid biopsy in Glioblastoma.
Figure 3. Liquid biopsy in Glioblastoma.
Targets 03 00031 g003
Table 1. Nanotechnology approach for the diagnosis of glioblastoma [55,56,57,58,59,60,61,62,63].
Table 1. Nanotechnology approach for the diagnosis of glioblastoma [55,56,57,58,59,60,61,62,63].
Nanotechnology ApproachDiagnostic ApplicationAdvantagesChallenges/LimitationsReferences
Nanoparticle-based MRI contrast agentsEnhanced tumor imaging, intraoperative guidanceImproved sensitivity, real-time trackingBBB penetration, potential toxicity[55,56,57,58]
Fluorescent nanoparticlesTumor visualization during surgeryIncreased tumor cell visibility, precisionLimited clinical translation[55,56,58]
Gold and iron oxide nanoparticlesMRI, PET, photoacoustic imagingMultifunctional, high contrast, targetingBiocompatibility, clearance[55,56,59,60]
Liposomes, dendrimers, micellesDrug delivery + imaging (theranostics)Dual function (diagnosis + therapy), tunabilityStability, manufacturing complexity[59,61,62,63]
Smart/inorganic nanoparticlesTargeted molecular imagingSpecificity, surface modificationOff-target effects, immune response[55,56,60]
Nanocarriers for liquid biopsyDetection of circulating tumor DNA/cellsNon-invasive, early detectionSensitivity, standardization[56,58,62]
Nanomedicine-enabled PET/MRI agentsMultimodal imagingComprehensive tumor characterizationCost, regulatory hurdles[55,56,57]
Extracellular vesicle analysisBiomarker discovery, monitoringPersonalized diagnosis, prognosisIsolation techniques, reproducibility[56,58,62]
Polymer-based nanoparticlesTargeted imaging and drug deliveryBBB penetration, controlled releaseLong-term safety, scalability[56,59,61,63]
CRISPR/Cas9 delivery via nanoparticlesMolecular diagnostics, gene editingPrecision, potential for personalized medicineDelivery efficiency, ethical concerns[56,59]
Table 2. Multiomics-based prediction of cancer prognosis [122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156].
Table 2. Multiomics-based prediction of cancer prognosis [122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156].
Genomics-Based Prediction of Cancer Prognosis
Study TitleCancer Type(s)ApproachKey FindingsReference
Insights for precision oncology from the integration of genomic and clinical data of 13,880 tumors from the 100,000 Genomes Cancer Programme33 solid tumor typesWhole-genome sequencing (WGS) integrated with clinical dataLinking WGS and clinical outcomes enables survival analysis and identification of prognostic cancer genes, supporting precision oncology[122]
Genomics to select treatment for patients with metastatic breast cancer (SAFIR02-BREAST trial)Metastatic breast cancerGenomic profiling for therapy selectionTargeted therapies matched to actionable genomic alterations improve progression-free survival; benefit depends on actionability level[123]
Combining multidimensional genomic measurements for predicting cancer prognosis: observations from TCGABreast, glioblastoma, AML, lung SCCmRNA, DNA methylation, miRNA, copy numberMultidimensional genomic data can improve prognosis prediction, but gene expression and clinical data are most predictive[124]
Integrating multi-omics data through deep learning for accurate cancer prognosis prediction15 cancer types (TCGA)Multi-omics (genomics, transcriptomics, etc.) with deep learningDenoising autoencoder improves prognostic accuracy; robust integration of multi-omics data identifies prognostic markers[125]
Pathomic Fusion: An Integrated Framework for Fusing Histopathology and Genomic Features for Cancer Diagnosis and PrognosisGlioma, renal cell carcinomaHistology + genomics (mutations, CNV, RNA-Seq)Multimodal deep learning fusion improves survival prediction over unimodal models[126]
Pan-cancer integrative histology-genomic analysis via multimodal deep learning14 cancer typesHistology + genomicsMultimodal deep learning predicts outcomes and discovers prognostic features across cancers[127]
Radiogenomics-based cancer prognosis in colorectal cancerColorectal cancerRadiomics + gene expressionCombining imaging and gene expression enhances prognostic stratification[128]
Transcriptomic-Based Prediction of Cancer Prognosis.
PETACC-8 & IDEA-France (2025)Stage III colon cancer3′RNA sequencing, TME, and cell cycle signaturesDeveloped prognostic models integrating transcriptomic signatures, improving risk stratification for recurrence[129]
WINTHER Trial (2020)Diverse solid tumorsRNA expression profilingCombined genomics and transcriptomics increased actionable targets; improved patient matching to therapies[130,131]
POG Program (2022)Advanced/metastatic cancersWhole genome and transcriptome analysis (WGTA)RNA data informed 67% of treatments; 46% of WGTA-informed treatments led to clinical benefit[132]
Pediatric Poor Prognosis Study (2024)Pediatric cancersWGTAIntegrating transcriptome data identified actionable variants in 96% of cases, guiding therapy[133]
PERCEPTION (2024)Multiple myeloma, breast, and lung cancerSingle-cell transcriptomicsscRNA-seq-based models outperformed bulk predictors in clinical response prediction[134]
SELECT (2021)10 cancer typesSynthetic lethality via transcriptomePredicted therapy response in 80% of 35 clinical trials, including WINTHER[131]
Pathology Atlas (2017)17 cancer typesGenome-wide transcriptomicsIdentified prognostic genes and created an open-access atlas for survival prediction[135]
Epigenomics-Based Prediction of Cancer Prognosis.
Development and validation of epigenetic modification-related signals for the diagnosis and prognosis of colorectal cancerColorectal cancerPredictive model using gene expression and epigenetic-related genes, validated on patient cohortsAn 8-gene epigenetic signature effectively predicts prognosis and may guide targeted therapies[136]
Exploring the role of epigenetic regulation in cancer prognosis with the epigenetic scorePan-cancer (TCGA datasets)LASSO Cox model to create an epigenetic score, a nomogram integrating clinical featuresEpigenetic score correlates with cancer hallmarks and predicts survival across cancer types[137]
Epigenetic and Tumor Microenvironment for Prognosis of Patients with Gastric CancerGastric cancerMachine learning (NMF, LASSO, SVM) to identify prognostic gene signaturesIdentified hub genes for prognosis; signatures performed well in survival prediction and immunotherapy response[138]
Epigenetics in the diagnosis and prognosis of head and neck cancer: A systematic reviewHead and neck squamous cell carcinomaSystematic review of 25 studies on DNA methylation and histone modificationsSeveral biomarkers (e.g., DAPK, TIMP3) show promise for early detection, but more robust trials are needed[139]
Epigenome-based cancer risk prediction: rationale, opportunities and challengesGeneral/Multiple cancersReview of DNA methylation-based risk prediction testsDNA methylation tests are promising for risk prediction, but challenges include cell-type specificity and implementation[140]
Epigenetic alterations in the gastrointestinal tract: Current and emerging use for biomarkers of cancerGI cancers (colorectal, liver, etc.)Review of clinical and emerging epigenetic biomarkersEpigenetic alterations are robust biomarkers for prognosis and are being integrated into clinical tests[141]
Proteomics and Metabolomics-Based Prediction of Cancer Prognosis.
Metabolomic-Based Approaches for Endometrial Cancer Diagnosis and Prognosis: A ReviewEndometrialMetabolomicsIdentifies metabolite biomarkers for improved diagnosis, prognosis, and monitoring[142]
Endometrial cancer diagnostic and prognostic algorithms based on proteomics, metabolomics, and clinical data: a systematic reviewEndometrialProteomics and MetabolomicsReviews diagnostic/prognostic biomarker discovery using omics and clinical data[143]
Discovery and Validation of Clinical Biomarkers of Cancer: A Review Combining Metabolomics and ProteomicsVariousProteomics and MetabolomicsHighlights advance in biomarker discovery and clinical translation[144]
Development of a cancer prognostic signature based on pan-cancer proteomicsMultiple (pan-cancer)ProteomicsProteomics-based model accurately predicts survival across cancers[145]
Plasma-based proteomic and metabolomic characterization of lung and lymph node metastases in cervical cancer patientsCervicalProteomics and MetabolomicsIdentifies biomarker panels for predicting lung and lymph node metastasis[146]
Metabolomic machine learning predictor for diagnosis and prognosis of gastric cancerGastricMetabolomics and Machine LearningMachine learning model outperforms traditional markers for diagnosis and prognosis[147]
Radiomic-Based Prediction of Cancer Prognosis.
Radiomics-based Prognosis Analysis for Non-Small Cell Lung CancerNSCLC112 patients, CT-based radiomics, various modeling strategiesRandom Forest and PCA improved the prediction of recurrence and survival; addressing data imbalance increased accuracy[148]
Radiomics strategies for risk assessment of tumour failure in head-and-neck cancerHead and Neck300 patients, multi-cohort, PET/CT radiomicsRadiomics plus clinical data predicted recurrence/metastasis risk; validated across cohorts[149]
Biliary Tract Cancer at CT: A Radiomics-based Model to Predict Lymph Node Metastasis and Survival OutcomesBiliary Tract247 patients, CT-based, retrospectiveRadiomics model predicted lymph node metastasis and survival; the high-risk group had worse outcomes[150]
Development and Validation of CT-Based Radiomics Signature for Overall Survival Prediction in Multi-organ CancerLung, Kidney, Head and Neck905 patients, multi-organ, CT-basedRadiomics signature predicted survival across cancer types; the combined model improved accuracy[151]
A Metabolism-Related Radiomics Signature for Predicting the Prognosis of Colorectal CancerColorectal381 patients, CT-based, LASSO regressionRadiomics score independently predicted disease-free survival; the nomogram outperformed the TNM stage[152]
Radiomics for Survival Risk Stratification of Clinical and Pathologic Stage IA Pure-Solid Non-Small Cell Lung CancerNSCLC (Stage IA)592 patients, multi-region radiomicsMultiregional radiomics signature stratified survival risk, improved over clinical predictors[153]
Delta-radiomics features for the prediction of patient outcomes in non–small cell lung cancerNSCLC107 patients, longitudinal CTRadiomics features changed during therapy; end-of-treatment features predicted response[154]
Radiogenomics-based cancer prognosis in colorectal cancerColorectal64 patients, CT radiomics + gene expressionCombined radiomics and genomics improved prognostic stratification[128]
Systematic review and meta-analysis of radiomics-based models in NSCLCNSCLC40 studies, 6223 patientsRadiomics models showed modest prognostic value; need for standardization[155]
Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosisVariousReviewMultiomics (radiomics, pathomics, genomics) enhances TME assessment and prognosis prediction[156]
Table 3. Types of Nanoparticles, AI Methods, and Clinical Uses in Cancer Treatment [168,169,170,171,172,173,174,175,176,177].
Table 3. Types of Nanoparticles, AI Methods, and Clinical Uses in Cancer Treatment [168,169,170,171,172,173,174,175,176,177].
Nanoparticle TypeAI Algorithm/ApproachClinical Outcome/Use CaseCitations
Polymeric nanoparticlesMachine learningEnhanced targeted drug delivery in cancer[168,169,170]
DendrimersNeural networksImproved drug loading and release profiles[169,170]
MicellesOptimization algorithmsControlled drug release, reduced toxicity[169,170]
LiposomesDeep learningPersonalized dosing, improved therapeutic index[169,170,171]
Protein nanoparticlesPattern recognitionTumor-specific targeting, better diagnostics[169,172]
Cell membrane nanoparticlesAI-driven designEnhanced immune evasion, longer circulation[169,173]
Mesoporous silica nanoparticlesPredictive modelingIncreased delivery efficiency to tumors[169,174]
Gold nanoparticlesMathematical modeling, MLOptimized photothermal therapy, improved imaging[169,175]
Iron oxide nanoparticlesAI-assisted PBPK modelingAccurate prediction of biodistribution[169,174]
Quantum dotsClassification algorithmsImproved cancer cell identification[169,172]
Carbon nanotubesANN, MLHigh-accuracy cancer cell classification[169,172]
Black phosphorusAI-based optimizationEnhanced drug delivery, reduced side effects[169,170]
MOF nanoparticlesQSAR, MLOptimized structure for drug delivery[169,171]
Exosome-mimicking nanoparticlesAI-driven nanocarrier designImproved biocompatibility, targeted siRNA delivery[169,173]
Multifunctional nanoparticlesML for synergy predictionCombined chemo/immunotherapy, reduced resistance[173,176]
Biomimetic nanocarriersML, optimizationEnhanced tumor targeting, immune evasion[169,173]
Smart nanoparticles (stimuli-responsive)AI-powered designPrecision therapy, individualized treatment[168,169]
Carbon nanoparticles (CNPs)ANN, MLSubclassification of breast cancer, >98% diagnostic accuracy[172]
Nanoparticle-modified drugsAI for dose optimizationImproved combination therapy outcomes[176]
Nanoparticle-based imaging agentsAI for image analysisEnhanced diagnostic accuracy, better treatment planning[169,177]
Table 4. Integration of Nanotechnology and AI Models in Glioma Diagnosis and Therapy: Applications, Clinical Status, and Examples [178,179,180,181,182,183,184,185,186].
Table 4. Integration of Nanotechnology and AI Models in Glioma Diagnosis and Therapy: Applications, Clinical Status, and Examples [178,179,180,181,182,183,184,185,186].
Technology/
Model
Nanotechnology Type and ExampleApplication (What It Does)Example (Specific Implementation)Clinical StatusReferences
Deep learning on MRI/PathologyMetal nanoparticles (e.g., gold, iron oxide) for enhanced imaging contrast and targeted deliveryTumor segmentation, grading, and molecular subtypingCNNs and transformer-based models for classifying glioma subtypes from histopathology and MRIClinically validated[178,179,180,181,182]
AI on TCR NGS dataPersistent luminescence nanoparticles (e.g., TRZD: ZnGa2O4:Cr3+, Sn4+) for long-term NIR imaging and therapyImmune repertoire-based glioma diagnosisAI models using T-cell receptor sequencing to classify glioma with an AUC of up to 96.7%Early clinical[183,184]
AI model optimizationChlorotoxin peptide-functionalized gold nanoparticles (CTX-PEI-AuNPs) for targeted SPECT/CT imaging/therapyDeploying AI in low-resource clinical settingsPost-training optimization of ResUNet for tumor delineation, reducing latency and memory usageEarly clinical[177,184]
Explainable AI (XAI) for prognosisAI-guided nanomedicine design for optimizing nanoparticle properties for diagnosis and therapyTransparent, interpretable prediction of glioma outcomesXGBoost, SHAP, LIME, and other XAI tools for feature importance and model explanationPreclinical[177,185,186]
Deep learning for digital pathologyAlbumin-based nanotheranostic probes (e.g., ICG/AuNR@BCNP) for multimodal imaging and phototherapyAutomated histopathological subtype classificationSD-Net_WCE (DenseNet variant) for classifying five glioma subtypes from H&E slidesClinically validated[179,182]
Radiomics and radiogenomicsMultifunctional metal nanoparticles for targeted drug delivery, imaging, and therapyLinking imaging features to molecular/
genomic profiles
AI models predicting IDH mutation and 1p/19q codeletion from MRI featuresClinically validated[179,180,184]
AI for treatment response predictionSurface-modified nanoparticles (e.g., PEGylated, ligand-targeted) for BBB penetration and targeted deliveryPredicting
therapy outcomes and recurrence
Machine learning models integrating imaging, genomics, and clinical dataPreclinical[179,184,186]
AI-enabled nanomedicine designAI-enabled design of nanomedicines for personalized dosing and reduced nanotoxicityOptimizing nanomaterial properties for diagnosis/therapyAI algorithms predicting nanomaterial interactions for improved efficacy and safetyPreclinical[177,186]
Nanoparticle-based imaging agentsIron oxide nanoparticles (IONPs) for MRI and therapy; gold nanoparticles for SPECT/CT imaging and therapyEnhanced
Imaging for glioma detection
Iron oxide nanoparticles (IONPs) for MRI contrast, cell tracking, and magnetic hyperthermiaMostly preclinical[177,186]
Table 5. Regulatory framework for AI-driven genomics research across different regions.
Table 5. Regulatory framework for AI-driven genomics research across different regions.
RegionKey Regulatory FrameworksFocus Areas
United StatesFDA, NIH, GINAAI software validation, genetic privacy laws
European UnionGDPR, AI ActData protection, high-risk AI regulation
ChinaPIPL, AI Governance InitiativesNational AI strategies, genomic data security
JapanEthical AI Guidelines, Genome Research LawsAI in genomics, patient data ethics
Table 6. Clinical Translation Gaps for Cancer Diagnosis and Treatment Using Nanotechnology and AI [203,204,205,206,207,208,209,210,211].
Table 6. Clinical Translation Gaps for Cancer Diagnosis and Treatment Using Nanotechnology and AI [203,204,205,206,207,208,209,210,211].
Translation GapNanotechnology Example(s)AI Example(s)Citations
Limited Clinical ApprovalsLiposomal doxorubicin (Doxil), albumin-bound paclitaxel (Abraxane); most nanomedicines remain preclinicalAI-driven design of nanocarriers for siRNA delivery, but few AI-optimized nanomedicines have clinical approval[173,177,203,204]
Biological BarriersNanoparticles for siRNA/chemotherapy delivery face rapid degradation, poor tumor accumulation, and immune clearanceAI models predict nanoparticle–biological interactions to optimize delivery, but translation to humans is limited[173,177,204,205]
Tumor HeterogeneityMultifunctional nanoparticles for combined therapy (e.g., chemo-immuno- or photothermal therapy) to address tumor diversityAI analyzes patient omics/imaging data to tailor nanomedicine, but heterogeneity complicates universal solutions[177,206,207,208]
Safety and Toxicity ConcernsBiomimetic nanocarriers (e.g., exosome-mimicking) improve biocompatibility, but long-term human safety data lackingAI used to predict and minimize nanotoxicity, but real-world validation is limited[173,186,206,209]
Regulatory and Manufacturing ChallengesFew standardized protocols for nanomedicine production; scalability and reproducibility issuesAI can optimize manufacturing processes, but regulatory pathways for AI-designed nanomedicines are unclear[177,205,210,211]
Data Integration and Patient StratificationNanodiagnostics and nano-imaging generate large datasets for patient profilingAI integrates multi-omics, imaging, and clinical data for personalized therapy, but clinical implementation is rare[177,207,208]
Gap Between Preclinical and ClinicalMost nanoformulations (e.g., targeted nanoparticles, nano-theranostics) show efficacy in animals but not in humansAI-optimized nanomedicines are often validated only in silico or in animals, not in clinical trials[173,177,203,206]
Incomplete Understanding of Cancer BiologyNanoparticles are designed for targeted delivery, but incomplete knowledge of the tumor microenvironment limits successAI helps uncover new biomarkers and drug targets, but translation to effective therapies is ongoing[177,206,210,211]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Suryawanshi, M.V.; Bagban, I.; Patne, A.Y. Integrating Nanotechnology and Artificial Intelligence for Early Detection and Prognostication of Glioblastoma: A Translational Perspective. Targets 2025, 3, 31. https://doi.org/10.3390/targets3040031

AMA Style

Suryawanshi MV, Bagban I, Patne AY. Integrating Nanotechnology and Artificial Intelligence for Early Detection and Prognostication of Glioblastoma: A Translational Perspective. Targets. 2025; 3(4):31. https://doi.org/10.3390/targets3040031

Chicago/Turabian Style

Suryawanshi, Meghraj Vivekanand, Imtiyaz Bagban, and Akshata Yashwant Patne. 2025. "Integrating Nanotechnology and Artificial Intelligence for Early Detection and Prognostication of Glioblastoma: A Translational Perspective" Targets 3, no. 4: 31. https://doi.org/10.3390/targets3040031

APA Style

Suryawanshi, M. V., Bagban, I., & Patne, A. Y. (2025). Integrating Nanotechnology and Artificial Intelligence for Early Detection and Prognostication of Glioblastoma: A Translational Perspective. Targets, 3(4), 31. https://doi.org/10.3390/targets3040031

Article Metrics

Back to TopTop