Next Article in Journal
Quorum Signaling Molecules: Interactions Between Plants and Associated Pathogens
Previous Article in Journal
Synergy of Tetracyclines and Potassium Azeloyl Diglycinate (Azeloglycine) in Hydrogels: Evaluation of Stability, Antimicrobial Activity, and Physicochemical Properties
Previous Article in Special Issue
Single-Cell RNA Sequencing Outperforms Single-Nucleus RNA Sequencing in Analyzing Pancreatic Cell Diversity and Gene Expression in Goats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Imaging: Unveiling Metabolic Abnormalities in Pancreatic Cancer

Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(11), 5242; https://doi.org/10.3390/ijms26115242
Submission received: 20 April 2025 / Revised: 21 May 2025 / Accepted: 27 May 2025 / Published: 29 May 2025
(This article belongs to the Special Issue Molecular Basis of Pancreatic Secretion and Metabolism)

Abstract

Pancreatic cancer remains one of the most aggressive malignancies globally, with a 5-year survival rate of less than 13%. This poor prognosis stems from late-stage diagnosis and intrinsic resistance to conventional therapies, including chemotherapy and radiotherapy. A hallmark of PC is oncogene-driven metabolic reprogramming—notably mediated by mutations in KRAS and other key pathways—which fuels tumor progression and undermines the efficacy of neoadjuvant treatments. Consequently, there is a pressing demand for non-invasive techniques capable of mapping metabolic alterations at both the tumor microenvironmental and systemic levels. This review will discuss molecular imaging techniques that identify metabolic changes within the tumor microenvironment. By bridging preclinical insights with clinical applications, we highlight how these innovations promise to revolutionize PC diagnosis, treatment stratification, and therapeutic monitoring, ultimately paving the way for precision oncology.

1. Introduction

Pancreatic cancer (PC) is one of the most aggressive and lethal malignancies worldwide, with a 5-year survival rate of less than 13% [1,2]. The majority of patients with this malignancy are diagnosed at an advanced stage, precluding eligibility for radical surgical resection, which is the only potentially curative treatment option at present [2]. Systematic chemotherapy regimens, including FOLFRINOX and gemcitabine in combination with nab-paclitaxel, continue to be the cornerstone of treatment for individuals with advanced disease [2]. Despite their utility, the rapid development of chemoresistance leads to poor prognosis [3].
A key driver of chemoresistance and tumor progression in PC is metabolic reprogramming, a hallmark of cancer that enables adaptation to the high energy and biosynthetic demands of rapidly proliferating cells [4,5]. PC cells rewire their metabolism to satisfy elevated energy requirements necessary for sustaining tumor progression in a nutrient-deprived and hypoxic tumor microenvironment (TME) [6]. This reprogramming encompasses alterations in the metabolism of glucose, lipids, and amino acids, in addition to dynamic interactions with stromal and immune cells within the TME [7].
The pathogenesis of pancreatic cancer is primarily driven by several critical mutations, notably including KRAS, which is present in more than 90% of cases [8]. This oncogene plays a central role in mediating metabolic reprogramming that fuels pancreatic carcinogenesis [9,10,11]. Recent advances in transcriptome profiling have enabled the molecular stratification of pancreatic adenocarcinoma (PDAC) into distinct subtypes [12]. These disparate classification schemes share fundamental commonalities. For example, the squamous (Bailey et al.), quasi-mesenchymal (Collisson et al.), and basal-like (Moffitt et al.) subtypes share many similarities at the transcriptomic level [12]. Clinically, this molecular heterogeneity correlates with prognostic divergence, where patients with basal-like subtypes exhibit significantly reduced overall survival compared to those with classical subtypes [13]. Functional validation using metastatic PC-derived patient organoids (PDOs) has unveiled subtype-specific metabolic dichotomies; basal-like PDOs demonstrate heightened oxidative phosphorylation (OXPHOS) activity and increased sensitivity to MCP1 inhibition relative to their classical counterparts [14]. In addition, PC can also be divided into different metabolic phenotypes according to its energy sources or gene expression related to metabolic processes [15,16,17]. Metabolic subtyping based on preferential energy utilization pathways reveals additional prognostic stratification. Specifically, glycolytically dominant tumors portend worse clinical outcomes than cholesterogenic phenotypes [17]. These multilayered classification frameworks—integrating molecular signatures and metabolic dependencies—provide a rationale for developing subtype-targeted therapies. The pharmacological exploitation of critical metabolic nodes represents a promising avenue for precision oncology in PC [18,19].
Molecular imaging techniques, capable of resolving spatial and temporal metabolic heterogeneity, offer unprecedented opportunities to decode the metabolic reprogramming of cancer at both molecular and cellular levels [20]. The most common molecular imaging modalities are positron emission tomography (PET) and magnetic resonance spectroscopy or imaging (MRS/MRI) [20]. PET employs radiolabeled metabolic analogs (e.g., 18F-fluorodeoxyglucose and 18F-FDG) to non-invasively quantify metabolic activity, with tracer uptake typically measured via standardized uptake value (SUV) [20,21]. In contrast, MRS/MRI exploits the spin polarization properties of atomic nuclei (notably 1H and 13C) to determine the biochemical composition and spatial distribution of metabolites [22]. While conventional 1H-MRS provides valuable metabolic information, its clinical utility is constrained by inherent sensitivity limitations arising from low metabolite concentrations and spectral peak overlap [23]. Hyperpolarized (HP)-13C-labeled probes have emerged as a breakthrough technology, amplifying signal intensity and enabling the investigation of dynamic metabolic processes in preclinical models [24]. Parallel innovations in deuterium metabolic imaging (DMI) utilize 2H-labeled substrates to achieve enhanced sensitivity for mapping specific metabolites [25]. Other techniques, including optical and near-infrared (NIR) imaging, while predominantly employed in small animal research due to limited tissue penetration depth, provide unparalleled spatiotemporal resolution for mechanistic investigations [22].
Despite these advancements, the clinical translation of metabolic imaging in PC remains limited. In this review, we systematically delineate key molecular imaging modalities and metabolic targets for use in characterizing tumor metabolism (Figure 1), with a focus on their emerging applications in guiding diagnosis, therapeutic stratification, and treatment monitoring in PC (Table 1). By synthesizing recent advances and unresolved challenges, we aim to bridge the gap between preclinical discoveries and clinical applications, thereby stimulating further research and innovation in this evolving field.

2. Molecular Imaging of Key Metabolic Pathways in Pancreatic Cancer: Glucose, Lipids, and Amino Acids

In this section, we focus on the major metabolic aberrations in glucose, lipid, and amino acid metabolism in PC and discuss how metabolic imaging can be utilized to identify these alterations.

2.1. Imaging Glucose Metabolism

The metabolic rewiring of pancreatic cancer cells toward aerobic glycolysis, first described by Otto Warburg in the 1920s, remains a defining hallmark of their malignant phenotype [26]. This metabolic reprogramming is characterized by the dramatic upregulation of glucose transporter 1 (GLUT1) and key rate-limiting glycolytic enzymes, including hexokinase 1/2 (HK1/2), phosphofructokinase 1 (PFK1), and lactate dehydrogenase (LDH). These molecular adaptations collectively enhance glycolytic flux, driving excessive lactate production, a hallmark of the Warburg effect [27,28,29].
The profound glycolytic reprogramming in PC establishes key molecular targets for metabolic imaging. 18F-FDG PET (Figure 2A), often integrated with CT or MRI for anatomical correlation, has emerged as the cornerstone technique for visualizing glucose metabolism in vivo [30]. Following GLUT-mediated cellular uptake, FDG undergoes HK-dependent phosphorylation to FDG-6-phosphate, which is structurally incapable of proceeding through glycolysis due to C-2 fluorine substitution. While theoretically reversible via glucose-6-phosphatase (G6Pase)-mediated dephosphorylation, tumor-specific G6Pase deficiency creates a metabolic trap that promotes intracellular radiotracer retention, forming the biochemical basis of 18F-FDG PET signal quantification [31].
Clinically, 18F-FDG PET demonstrates diagnostic superiority over conventional anatomical imaging techniques and provides valuable prognostic information [32,33,34,35,36]. Dual-time 18F-FDG PET/CT offers quantitative insights into intra-tumoral metabolic heterogeneity, which has been associated with cancer treatment outcomes [37,38]. Yoo et al. demonstrated that a reduction in metabolic intra-tumoral heterogeneity during chemotherapy in patients with advanced-stage PC might serve as a predictor of progression-free survival (PFS) and overall survival (OS) [38]. Emerging evidence suggests that the multimodal integration of 18F-FDG PET/MRI with contrast-enhanced computed tomography (CECT) and CA19-9 monitoring enhances the accuracy of resectability prediction in patients undergoing neoadjuvant therapy, particularly for identifying vascular involvement [39]. In addition to 18F-FDG, other PET tracers are available for imaging glucose metabolism. Pyruvate kinase (PK) catalyzes pyruvate production, and its M2 splice variant, PKM2, has been shown to contribute to upregulated glycolysis in cancer cells [40]. 18F-DASA-23, a PKM2-specific isotopologue radiotracer, has demonstrated the ability to visualize PKM2 status in glioblastoma [41]. Although this tracer has yet to be tested in PC models, aberrant PKM2 expression and activation have been identified in PC and are associated with cancer cell survival and metastasis [42,43]. These findings provide a foundation for the potential future application of such tracers in pancreatic cancer research.
Pyruvate, the final product of glycolysis, serves as a pivotal junction in cellular metabolism, with three primary metabolic fates: (1) its conversion to acetyl-CoA via pyruvate dehydrogenase in the mitochondria, enabling entry into the tricarboxylic acid (TCA) cycle; (2) LDH-mediated lactate production; and (3) alanine aminotransferase (ALT)-catalyzed synthesis of alanine, supporting amino acid metabolism [44]. LDH catalyzes the conversion of pyruvate to lactate, and elevated LDH levels have been found to be associated with multiple types of tumors, including PC [45]. Metabolic crosstalk between stromal cells and PC cells results in the altered secretion and consumption of alanine [46]. Furthermore, distinct alanine-to-lactate ratios have been observed in experimental models of PC [47].
The emerging technique of HP-13C metabolic MRI complements conventional imaging by enabling the dynamic, real-time investigation of glycolytic flux. This technique holds promise for precursor lesion detection and the monitoring of chemotherapy response. In one study, the intravenous administration of HP-13C-labeled pyruvate enabled the non-invasive identification of PC precursor lesions through the quantification of 13C-alanine/13C-lactate signal ratios, which correlate with the alanine/lactate concentration ratio and also decrease with the ALT/LDH activity ratio during disease progression [23] (Figure 2E). In addition, in patients with locally advanced or metastatic PC, alanine-to-lactate ratios observed in HP-13C-labeled pyruvate MRI before and 4 weeks after treatment demonstrated altered metabolism following chemotherapy [47].
Furthermore, this technique aids in mapping metabolic heterogeneity. In patient-derived PC model systems, HP-13C-labeled pyruvate and lactate detected via MRS demonstrated functional metabolic heterogeneity in patient-derived primary cells, particularly when compared to conventional cell lines [48]. By detecting 13C-pyruvate and 13C-lactate interconversion in vivo, highly glycolytic PC xenografts could be identified non-invasively using HP-MRS [48]. Moreover, Kishmoto et al. demonstrated that, even with a similar genetic background, 13C-glucose tracers (Figure 2C) are able to distinguish PC xenografts by localizing lactate production and glucose metabolism [49]. Moreover, in another study, the combination of hyperpolarized MRI and 18F-FDG PET also provided quantitative insights into the relationship between glycolysis and oxygenation status inside tumors [30]. This approach revealed that tumor metabolism is heterogeneous, with 18F-FDG uptake negatively correlated with pO2 in the tumor core and positively correlated in the periphery. Unlike pO2 and 18F-FDG uptake, LDH activity was relatively evenly distributed throughout the tumor [30]. In another study, real-time metabolic imaging data also showed that the overexpression of LDH-A and hypoxia-inducible factor (HIF-1α) significantly influenced the conversion kinetics of HP pyruvate-to-lactate in PDX tumors [50].
Deuterium MRI (2H-MRI) was also applied in probing tumor glucose metabolism [51]. Through alternating deuterated and non-deuterated glucose injections, it was observed that tumors maintain relentless glucose consumption and lactate production whenever glucose is available. Lactate output appeared to be constrained solely by glucose availability, vividly illustrating the significant glucose consumption driven by the Warburg effect [51].
The pentose phosphate pathway (PPP) is also enhanced in PC [7]. During radiotherapy, the metabolic flux of PPP increases, with it being recognized that combining radiotherapy with PPP inhibitors can synergistically suppress tumor growth [52]. Research results from glioblastoma studies have demonstrated that after administering [2-13C]-glucose, 13C MRI can visualize the flux of [4-13C]-glutamate (produced via PPP) and [5-13C]-glutamate (generated through glycolysis), thereby reflecting changes in PPP activity within cells [53]. In light of the above findings, in PC, 13C MRI may serve as a potential tool for monitoring treatment response and tumor recurrence during radiotherapy.

2.2. Imaging Amino Acid Metabolism

Amino acid metabolism also undergoes profound transformation in PC, impacting tumorigenesis and development by influencing energy metabolism and signal transduction [54]. As the most abundant amino acid in blood plasma, glutamine (Gln) is involved in numerous important cellular processes, such as glucogenesis and the biosynthesis of proteins, and it also participates in the tricarboxylic acid (TCA) cycle [55,56,57]. Unlike most cells that utilize glutamate dehydrogenase (GLUD1) to convert glutamine-derived glutamate into α-ketoglutarate in the mitochondria for the TCA cycle, PC employs a unique cytoplasmic pathway [58]. In this process, glutamine-derived aspartate is metabolized via aspartate transaminase (GOT1) into oxaloacetate, malate, and then pyruvate, thereby enhancing the NADPH/NADP+ ratio to maintain redox balance [58]. In PC, Gln demand is increased, and glutamate ammonia ligase (GLUL), which is responsible for de novo synthesis, is upregulated in PC patient samples and mouse PC models [59].
Research indicates that single-voxel MRS could be used as a technique to measure metabolites, including Glx (glutamine and glutamate) and N-acetylaspartate (NAA) [60]. In one study, the levels of the aforementioned metabolites significantly decreased in PC when compared with a healthy pancreas [60]. Moreover, low Glx levels were found to be associated with poor PFS and OS [60]. (4S)-4-(3-18F-Fluoropropyl)-l-glutamate (FSPG) is a PET tracer designed to image the activity of a heterodimeric transporter that mediates the cellular uptake of cystine for the exchange of intracellular glutamine [61]. Compared to 18F-FDG PET, FSPG PET demonstrated higher sensitivity (95.0% vs. 90.0%), specificity (100.0% vs. 66.7%), and diagnostic accuracy (95.7% vs. 90.0%) in the detection of metastasis, particularly in the liver [61].

2.3. Imaging Lipid Metabolism

PC also demonstrates distinctive adaptive modifications in lipid metabolism that facilitate tumor progression, characterized by dual activation of exogenous uptake and dysregulated endogenous synthesis pathways [62]. Tumor cells acquire fatty acids (FAs) and cholesterol through multiple transporters. FA internalization is mediated by fatty acid transport proteins (FATPs), fatty acid translocase (CD36), and fatty acid-binding proteins (FABPs); in comparison, cholesterol is primarily acquired via low-density lipoprotein receptor (LDLR)-mediated endocytosis [63]. In head and neck squamous cell carcinoma (HNSCC), it has been reported that immunohistochemical (IHC) detection of CD36 expression makes it possible to differentiate false-negative lymph nodes in 18F-FDG-PET/CT imaging [64]. In a glioma orthotopic tumor model, a CD36-targeting NIR fluorescent probe was developed to visualize the CD36 protein and delineate tumor boundaries, expanding the toolbox for surgical navigation [65]. These studies collectively suggest a promising strategy for imaging lipid metabolism in PC.
De novo FA and cholesterol synthesis both rely on acetyl-CoA derived from glucose and glutamine metabolism [62]. Cholesterol imaging in vivo has been performed using PET probes [66,67]. However, such studies have yet to be conducted in pancreatic tumors. Cholesterol dysregulation in PC can be accessed through translocator protein (TSPO) imaging. In one study, TSPO, involved in cholesterol transport, showed elevated expression in high-grade pancreatic precancerous lesions [68]. In genetically engineered mouse models, a TSPO-targeted NIR probe was found to localize to pre-malignant pancreatic lesions and advanced-stage tumors, enabling real-time image-guided surgery [68].
Aberrant choline metabolism can also be identified through molecular imaging in PC. The use of 1H-MRSI resulted in the detection of increased levels of total choline in PC cell lines and tumors, corresponding to the elevated expression of choline kinase-α (Chkα), and the choline transporters CHT1 and CTL1 [69]. The authors of another study proposed that choline levels detected through the use of MRS were significantly higher in TNM stage 3+ PC tumors [70]. Other researchers have employed deuterium metabolic imaging (DMI) with the use of [2H9] choline and [6,6′-2H2] glucose (Figure 2B) to assess the uptake and conversion of choline and glucose [71]. The reconstruction of DMI data revealed the spatial accumulation patterns of 2H-labeled compounds, providing a more precise metabolic characterization of PC. Other choline-based tracers have also been synthesized for pre-clinical and clinical use, including 11C-choline, 18F-fluoro-choline (FCH), and 18F-fluoroethyl-choline (FEC) [72,73,74,75]. In a patient with prostate cancer, an 18F-FCH PET/CT scan revealed chronic mass-forming pancreatitis during evaluation for lymph node recurrence [76]. In another study, 11C-choline accumulation was observed in a metastatic pancreatic tumor originating from renal cell carcinoma; in comparison, 18F-FDG did not demonstrate similar uptake [77].
In human-derived PC cell lines, elevated levels of phosphocholine and phosphoethanolamine can also be detected [78]. To perform a comprehensive analysis of phosphorus metabolites, 31P chemical shift imaging is a commonly employed method [79]. Phosphorus metabolic imaging combined with 7T MR makes it possible to detect increased levels of phospholipid metabolites in tumor tissues compared to background signals, enabling non-invasive pH measurements and indicating the presence of hypoxia [80].

3. Imaging Phenotypic Consequences of Cancer Metabolism and Microenvironment

In this section, we will highlight imaging modalities designed to target key phenotypic traits of cancer metabolism, including altered redox homeostasis, chemotherapy drug metabolism, and tumor microenvironment changes such as hypoxia, desmoplasia, and vascularization.

3.1. Imaging Redox Homeostasis

While molecular oxygen (O2) serves as an indispensable substrate for ATP generation through OXPHOS in eukaryotic organisms, its metabolic processing paradoxically generates reactive oxygen species (ROS) through electron leakage. These redox-active molecules induce significant cellular damage, particularly within tumors. Therein, they induce genomic instability, promote malignant progression and metastasis, and confer resistance to chemotherapy [81]. The generation and detoxification of ROS are regulated by cellular metabolism [82,83]. The production of ROS is intricately linked with the activity of nicotinamide dinucleotide oxidases and flavin adenine dinucleotide (FAD) [84]. Nicotinamide dinucleotide (NADPH) and FAD are cycled between oxidized and reduced states to transfer electrons in the mitochondria through active TCA cycle reactions and OXPHOS [85].
Capitalizing on the inherent fluorescent properties of reduced NADPH and oxidized FAD, advanced imaging modalities enable the non-invasive monitoring of cellular metabolism [86,87]. Two-photon and multi-photon excitation microscopy techniques have been successfully implemented to quantify these autofluorescence signals in three-dimensional PC models, achieving subcellular resolution in intact tumor architectures [86,87]. In two studies, NADPH and FAD autofluorescence signals were measured at excitation and emission wavelengths of 400–480 nm and 500–600 nm, respectively [86,87]. In three-dimensional PC models, the data were integrated over a z-stack of 30 images acquired with 0.1 µm intervals. The optical redox ratio (ORR), defined as the ratio of NADPH-to-FAD fluorescence intensities, provides critical insights into cellular redox homeostasis by reflecting the relative abundance of electron donors versus acceptors. In addition, the fluorescence lifetimes of NADPH and FAD depend on their binding states to enzyme complexes, offering further metabolic insights. The composite optical metabolic imaging (OMI) index, integrating both ORR and lifetime parameters, has emerged as a robust quantitative biomarker for tracking metabolic reprogramming and therapeutic response in PC malignancies [86,87]. Recent applications in tumor organoid models have revealed critical metabolic crosstalk within the pancreatic tumor microenvironment. In their study, Datta et al. employed optical redox imaging in co-culture systems containing both neoplastic cells and stromal components, demonstrating that direct contact with pancreatic stellate cells (PSCs) significantly enhances oxidative metabolism in PC cells [88]. In complementary studies, researchers found that cancer-associated fibroblasts (CAFs) can induce redox state elevation in PC cells, correlating with acquired treatment resistance across both in vitro and in vivo models [89]. These findings underscore the importance of redox imaging as a powerful tool for elucidating tumor metabolism and its implications for therapeutic outcomes in PC.

3.2. Imaging the Metabolism of Chemotherapy Drugs

Gemcitabine, also known as 2′,2′-difluorodeoxycytidine (dFdC), is regarded as one of the first-line chemotherapy drugs for PC [2,3]. As a deoxycytidine nucleoside analog, gemcitabine effectively inhibits DNA synthesis, thereby arresting the cell cycle and suppressing cancer cell proliferation [90]. Gemcitabine relies on nucleoside transporters for cellular uptake and requires phosphorylation by deoxycytidine kinase (dCK) to exert its cytotoxic effects [3]. 18F-FAC (2′-deoxy-2′-18F-fluoro-β-d-arabinofuranosylcytosine), a PET imaging agent structurally analogous to gemcitabine, can also serve as a substrate for dCK [91] (Figure 2D). When integrated with MR, 18F-FAC PET/MR enabled the assessment of gemcitabine uptake in PDX models for PC. Notably, treatment with PEGylated recombinant human hyaluronidase (PEGPH20) was found to enhance tumor 18F-FAC uptake by 12% in these models [91].
In addition to PET imaging, the metabolism of gemcitabine can be visualized using mass spectrometry imaging (MSI), a label-free technique that provides high-resolution mapping of chemotherapy drugs and their metabolites in fresh frozen tissue sections [92]. In the LSL-KrasG12D/+, LSL-Trp53R172H/+, and Pdx-1-Cre (KPC) mouse models of PC, MSI has been successfully employed to map the spatial distribution of gemcitabine and its phosphorylated metabolites, including dFdCMP, dFdCDP, and dFdCTP, in addition to the inactive metabolite dFdU [92]. The biologically active phosphorylated metabolites were found to be related to the therapeutic function of gemcitabine [93]. This approach offers valuable insights into the pharmacokinetics and therapeutic response of gemcitabine in preclinical models.

3.3. Imaging Tumor Microenvironment: Hypoxia, Desmoplasia, and Vascularization

The complex TME, characterized by hypoxia and desmoplasia, interacts with tumor metabolism and plays a crucial role in the malignant progression of PC [94,95]. This interaction between the TME and tumor metabolism underscores the importance of understanding the interplay between these factors in disease progression. Molecular imaging offers a powerful tool to explore the TME by targeting key features such as hypoxia, desmoplasia, and vascularization. One approach to imaging hypoxia involves the use of nitroimidazole-based agents, such as 18F-FMISO and 18F-FAZA [96,97]. These agents, when employed in PET/CT imaging, provided valuable prognostic information for PC patients and aid in identifying suitable candidates for hypoxia-targeted therapy trials [96,97]. Furthermore, in orthotopic xenograft models of PC, dual-modality ultrasound (US) and photoacoustic (PA) imaging techniques have demonstrated the ability to measure relative tissue oxygenation [98]. This US/PA dual-modality imaging technique facilitates the acquisition of both anatomical and functional data, enhancing our understanding of tumor growth dynamics.
One hallmark of PC is the extensive desmoplastic reaction, which is primarily driven by the activation of CAFs [99]. Advancements in single-cell RNA sequencing (scRNAseq) have revealed the heterogeneity of CAF subpopulations, providing deeper insights into their roles in tumor progression [100]. Furthermore, the spatial distribution and temporal dynamics of CAFs underscore the importance of imaging techniques in understanding their contributions to tumor biology [101]. Fibroblast activation protein (FAP), which is highly and selectively expressed on CAFs, serves as a key imaging target [101]. To target FAP, various FAP inhibitor (FAPI) tracers have been designed and labeled with radionuclides including Gallium-68 (Ga68) and 18F for both diagnostic imaging and therapeutic applications [102,103]. There are currently numerous clinical studies underway to explore in greater detail the potential of these imaging techniques in PC diagnosis and treatment. The association between 68Ga-FAPI PET SUVmax and FAP expression, as determined using IHC, has been confirmed [104,105]. The results of a number of studies have demonstrated that 68Ga-FAPI PET results in superior or equivalent detection rates and diagnostic accuracy compared to 18F-FDG PET-CT [104,105,106,107,108,109]. In addition, the results of some studies suggest that 68Ga-FAPI-04 PET may outperform 18F-FDG PET in detecting suspicious lymph node metastases [110]. In terms of therapeutic management, 68Ga-FAPI PET-CT demonstrated a superior ability to identify distant metastases, particularly in advanced or recurrent cases, resulting in TNM stage reclassification when compared to CECT, which may lead to major treatment alterations [111,112]. Furthermore, in patients with PC following first-line treatment, a higher target-to-background ratio (TBR-blood) was associated with poor treatment response [113]. Collectively, these advances position FAPI-based imaging as a transformative tool for precision oncology in PC, enabling the non-invasive investigation of stromal biology and personalized therapeutic strategies. Moreover, CAFs have also been reported to be targeted by platelet-derived growth factor receptor beta (PDGFRβ) [114]. Following radiolabeling with 64Cu, PDGFRβ affibody ZPDGFRβ exhibited ideal imaging capabilities when combined with PET/CT in mouse models of PC [114].
The vascularization of PC can be effectively visualized using contrast-enhanced ultrasound (CEUS) with vascular endothelial growth factor receptor type 2 (VEGFR2)-targeted microbubbles [115] (Figure 2F). In transgenic mice with PC, VEGFR2-targeted ultrasound demonstrated significantly higher signal intensities in PC compared to the pancreas of wild-type mice, particularly in tumors smaller than 3 mm [115]. The above results highlight the potential of VEGFR2-targeted microbubbles for the earlier detection of small PC lesions [115]. Furthermore, clinical trials on the VEGFR2-targeted microbubble BR55 have been conducted in breast and ovarian lesions, demonstrating more pronounced targeted signals in malignant lesions compared to benign ones, thereby laying a solid foundation for its clinical application in oncology [116]. After being combined with the NIR probe IRDye800CW, the VEGFR2 monoclonal antibody bevacizumab could also be used for surgery guidance [117].

4. The Role of Artificial Intelligence (AI) in Enhancing Metabolic Imaging Interpretation

AI has undergone remarkable evolution in medical imaging, fundamentally transforming disease diagnosis and clinical management [118]. In PC research, significant efforts have focused on leveraging AI for predictive modeling, risk stratification, and diagnostic applications [119]. In the field of AI-assisted PC image analysis, key research directions include tumor growth modeling, organ/multi-organ segmentation and edge detection, pancreatic cancer diagnosis and risk assessment, prognostic prediction, and image and annotation quality optimization [119,120]. Cao et al.’s landmark study introduced PANDA, a deep learning system outperforming conventional non-contrast CT in pancreatic lesion detection and classification accuracy [121]. Placido et al. developed a machine learning model utilizing clinical history data for the risk prediction of PC [122]. Moreover, a 3D Contrast-Enhanced Convolutional Long Short-Term Memory network (CE-ConvLSTM) using CECT data was developed by researchers to evaluate tumor–vascular relationships, enabling survival prediction in patients with primarily resectable PC [123]. As discussed above, metabolic imaging modalities provide critical insights into the unique metabolic profile of pancreatic malignancies, offering essential information for both diagnosis and therapeutic planning. The integration of AI with metabolic imaging technologies presents a transformative opportunity to enhance diagnostic precision, enable earlier tumor detection, and potentially reveal novel metabolic biomarkers. A machine learning model was developed by researchers based on 18F-FDG PET/CT data combined with clinical features to predict tumor grade and prognosis in pancreatic neuroendocrine tumor patients [124]. Radiomics and the deep learning features of 18F-FDG PET/CT were also used to distinguish PC and autoimmune pancreatitis with an accuracy of 90.1% (95% CI: 88.7–91.5%) [125]. A deep learning model based on 18F-FDG-PET/CT examination before surgery was also developed by another group of researchers for pathological grading. Following integration with clinical data, the AUC of the model reached 0.77 [126]. Current investigative paradigms predominantly utilize PET/CT-based models that synergize radiological signatures with clinical parameters for pancreatic lesion classification and prognosis prediction. The future integration of AI with diverse metabolic imaging modalities and multi-omics data, including genomic sequencing in addition to spatial transcriptomics, holds significant potential for refining metabolic subtyping in PC.
AI applications in pancreatic cancer research currently face multiple challenges, with insufficient sample size being a particularly prominent issue. This limitation is especially acute in metabolic imaging—the inherent difficulties in data acquisition hinder the accumulation of adequately sized datasets, creating substantial barriers to reliable model training and validation, which ultimately results in unstable algorithmic performance. Moreover, the intrinsic ‘black-box’ nature of deep learning models and their lack of interpretability significantly impede the clinical translation and further development of these technologies [127].

5. Discussion and Conclusions

In conclusion, metabolic imaging has emerged as a revolutionary tool in PC research, significantly contributing to the elucidation of tumor metabolic abnormalities and offering critical insights into the TME.
Emerging evidence from molecular imaging studies demonstrates that tumor metabolic profiling plays potential roles across the pancreatic cancer management spectrum, spanning early detection and precise staging to therapeutic monitoring and prognostic stratification (Figure 3). These advancements pave the way for novel therapeutic approaches tailored to individual patient needs. In PC management, real-time treatment monitoring is critically important. Although existing studies have established correlative associations between metabolic imaging findings and therapeutic response [112,128], there exists a paucity of direct evidence demonstrating how real-time metabolic changes detected through molecular imaging during treatment can reliably predict clinical outcomes.
Notably, in this review, we focus on established clinical applications from existing studies, though some potential uses remain unexplored. The clinical utility of metabolic imaging depends on both biological targets and imaging modalities. For example, PET/CT and PET/MRI are well suited for detecting distant metastases due to their whole-body scanning capability. NIR imaging offers real-time intraoperative navigation, making it ideal for surgical guidance. US/CEUS provides a non-invasive, radiation-free, and cost-effective approach, rendering it highly suitable for longitudinal treatment monitoring. The same molecular target can address different clinical needs when paired with appropriate imaging techniques. For instance, TSPO-targeted NIR probes assist in real-time surgery, whereas 18F-labeled TSPO tracers can aid in the early identification of early-stage pancreatic cancer lesions [68].
Metabolic imaging can also be combined with conventional serological biomarkers (e.g., CA19-9) to enhance diagnostic performance. Although they operate through fundamentally distinct mechanisms—metabolic imaging visualizes in vivo metabolic activity, whereas CA19-9 detects circulating tumor-derived carbohydrate antigens—their combined use enhances diagnostic confidence. When results are concordant, this integrated approach significantly strengthens either the confirmation or exclusion of PC. Combining CECT, PET, and CA19-9 may improve the assessment of resectability and recurrence prediction [39,129]. Furthermore, metabolic adaptations in PC significantly influence the levels of CA19-9. An integrated analysis of clinical and proteomic data from 128 resectable PC cases revealed that tumors with elevated CA19-9 levels exhibit enhanced glycolytic activity and demonstrate significant metabolic heterogeneity across multiple pathways—including carbohydrate, protein, amino acid, lipid, and nucleic acid metabolism—when compared to low-CA19-9-level tumors [130].
Beyond targeting metabolic products, processes, and phenotypic consequences as discussed herein, the imaging of upstream genetic mutations can also indicate potential metabolic abnormalities in tumors. For instance, KRAS plays a pivotal role in metabolic reprogramming in PC. Through the coordinated regulation of hexosamine, PPP, and glutamine pathways, oncogenic KRAS orchestrates a metabolic network supporting biosynthesis, redox homeostasis, and tumor proliferation [131]. The structural alterations induced by the G12C mutation enable inhibitor molecules to bind the allosteric switch II pocket (S-IIP) [132]. These small-molecule inhibitors can be conjugated with fluorescent probes (e.g., Alexa Fluor 647) via click chemistry, enabling the specific fluorescence imaging of KRAS G12C [133].
In addition to target identification and analytical methodologies, innovations in molecular probes and imaging materials also play important roles in the advancement of metabolic imaging. Nanotechnology-derived nanoparticles (NPs) are emerging as a transformative platform that may revolutionize molecular imaging through enhanced imaging performance and integrated diagnostic–therapeutic capabilities. A representative example is radiolabeled iron oxide (IO) nanoparticles, which enable the synergistic combination of high-sensitivity radionuclide imaging (SPECT/PET) with MRI, which offers outstanding spatial resolution [134]. CAF-targeted, controlled-release nanodroplets FH-V9302-siGLUL-NDs have also been developed to deliver the amino acid transporter ASCT2 (SLC1A5) inhibitor (V9302) and siRNA for GLUL (siGLUL) [128]. In one instance, FH-V9302-siGLUL-NDs serve as a CEUS imaging agent for tumor visualization. Conversely, through ultrasound-targeted microbubble destruction (UTMD), they enable the spatially controlled co-delivery of V9302 and siGLUL to disrupt the glutamine crosstalk between CAFs and tumor cells [128].
Together, advancements in functional imaging modalities and exploratory target selection are set to significantly enhance the identification of early-stage PC and the assessment of treatment responses, effectively tackling critical clinical challenges in disease management.

Funding

This study was supported by the National High Level Hospital Clinical Research Funding 2022-PUMCH-D-001.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
18F-FAC2′-deoxy-2′-18F-fluoro-β-d-arabinofuranosylcytosine
AIArtificial intelligence
CAFsCancer-associated fibroblasts
CB2RCannabinoid receptor type 2
CD36Fatty acid translocase
CECTContrast-enhanced computed tomography
CEUSContrast-enhanced ultrasound
ChkαCholine kinase-α
dCKDeoxycytidine kinase
dFdC2′,2′-difluorodeoxycytidine
DMIDeuterium metabolic imaging
FAFatty acid
FABPsFatty acid-binding proteins
FADFlavin adenine dinucleotide
FAPIFAP inhibitor
FATPsFatty acid transport proteins
FCH18F-fluoro-choline
FEC18F-fluoroethyl-choline
FSPG(4S)-4-(3-18F-Fluoropropyl)-l-glutamate
G6PaseGlucose-6-phosphatase
Ga68Gallium-68
GlnGlutamine
GLUD1Glutamate dehydrogenase
GLULGlutamate ammonia ligase
GLUT1Glucose transporter1
GOT1Aspartate transaminase
HK1/2Hexokinase 1/2
HNSCCHead and neck squamous cell carcinoma
HPHyperpolarized
IHCImmunohistochemical
LDHLactate dehydrogenase
LDLRLow-density lipoprotein receptor
MRIMagnetic resonance imaging
MRSMagnetic resonance spectroscopy
MSIMass spectrometry imaging
NAAN-acetylaspartate
NADPHNicotinamide dinucleotide
NIRNear-infrared
O2Oxygen
OMIOptical metabolic imaging
ORROptical redox ratio
OSOverall survival
OXPHOSOxidative phosphorylation
PAPhotoacoustic
PCPancreatic cancer
PDACPancreatic adenocarcinoma
PDGFRβPlatelet-derived growth factor receptor beta
PDOPDAC-derived patient organoids
PEGPH20PEGylated recombinant human hyaluronidase
PETPositron emission tomography
PFK1Phosphofructokinase1
PFSProgression-free survival
PPPPentose phosphate pathway
PSCPancreatic stellate cells
ROSReactive oxygen species
scRNAseqSingle-cell RNA sequencing
SUVStandardized uptake value
TCATricarboxylic acid
TMETumor microenvironment
TSPOTranslocator protein
USUltrasound
VEGFR2Vascular endothelial growth factor receptor type 2

References

  1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef] [PubMed]
  2. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef] [PubMed]
  3. Zeng, S.; Pöttler, M.; Lan, B.; Grützmann, R.; Pilarsky, C.; Yang, H. Chemoresistance in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 4504. [Google Scholar] [CrossRef] [PubMed]
  4. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  5. Park, W.; Chawla, A.; O’Reilly, E.M. Pancreatic Cancer: A Review. JAMA 2021, 326, 851–862. [Google Scholar] [CrossRef]
  6. De Santis, M.C.; Bockorny, B.; Hirsch, E.; Cappello, P.; Martini, M. Exploiting pancreatic cancer metabolism: Challenges and opportunities. Trends Mol. Med. 2024, 30, 592–604. [Google Scholar] [CrossRef]
  7. Qin, C.; Yang, G.; Yang, J.; Ren, B.; Wang, H.; Chen, G.; Zhao, F.; You, L.; Wang, W.; Zhao, Y. Metabolism of pancreatic cancer: Paving the way to better anticancer strategies. Mol. Cancer 2020, 19, 50. [Google Scholar] [CrossRef]
  8. Biankin, A.V.; Waddell, N.; Kassahn, K.S.; Gingras, M.C.; Muthuswamy, L.B.; Johns, A.L.; Miller, D.K.; Wilson, P.J.; Patch, A.M.; Wu, J.; et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012, 491, 399–405. [Google Scholar] [CrossRef]
  9. Humpton, T.J.; Alagesan, B.; DeNicola, G.M.; Lu, D.; Yordanov, G.N.; Leonhardt, C.S.; Yao, M.A.; Alagesan, P.; Zaatari, M.N.; Park, Y.; et al. Oncogenic KRAS Induces NIX-Mediated Mitophagy to Promote Pancreatic Cancer. Cancer Discov. 2019, 9, 1268–1287. [Google Scholar] [CrossRef]
  10. Ying, H.; Kimmelman, A.C.; Lyssiotis, C.A.; Hua, S.; Chu, G.C.; Fletcher-Sananikone, E.; Locasale, J.W.; Son, J.; Zhang, H.; Coloff, J.L.; et al. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell 2012, 149, 656–670. [Google Scholar] [CrossRef]
  11. Dey, P.; Li, J.; Zhang, J.; Chaurasiya, S.; Strom, A.; Wang, H.; Liao, W.T.; Cavallaro, F.; Denz, P.; Bernard, V.; et al. Oncogenic KRAS-Driven Metabolic Reprogramming in Pancreatic Cancer Cells Utilizes Cytokines from the Tumor Microenvironment. Cancer Discov. 2020, 10, 608–625. [Google Scholar] [CrossRef] [PubMed]
  12. Collisson, E.A.; Bailey, P.; Chang, D.K.; Biankin, A.V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 207–220. [Google Scholar] [CrossRef] [PubMed]
  13. Suurmeijer, J.A.; Soer, E.C.; Dings, M.P.G.; Kim, Y.; Strijker, M.; Bonsing, B.A.; Brosens, L.A.A.; Busch, O.R.; Groen, J.V.; Halfwerk, J.B.G.; et al. Impact of classical basal-like molecular subtypes on overall survival in resected pancreatic cancer in the SPACIOUS-2 multicentre study. Br. J. Surg. 2022, 109, 1150–1155. [Google Scholar] [CrossRef] [PubMed]
  14. Ali, H.A.; Karasinska, J.M.; Topham, J.T.; Johal, D.; Kalloger, S.; Metcalfe, A.; Warren, C.S.; Miyagi, A.; Tao, L.V.; Kevorkova, M.; et al. Pancreatic cancer tumor organoids exhibit subtype-specific differences in metabolic profiles. Cancer Metab. 2024, 12, 28. [Google Scholar] [CrossRef]
  15. Liang, C.; Qin, Y.; Zhang, B.; Ji, S.; Shi, S.; Xu, W.; Liu, J.; Xiang, J.; Liang, D.; Hu, Q.; et al. Energy sources identify metabolic phenotypes in pancreatic cancer. Acta Biochim. Biophys. Sin. 2016, 48, 969–979. [Google Scholar] [CrossRef]
  16. Rong, Z.; Yang, J.; Liu, J.; Meng, Q.; Hua, J.; Tan, Z.; Zhang, B.; Liu, Y.; Du, Q.; Wang, W.; et al. Dense stroma activates the TGF-β1/FBW7 axis to induce metabolic subtype switching in pancreatic cancer. Int. J. Surg. 2025, 111, 1891–1903. [Google Scholar] [CrossRef]
  17. Karasinska, J.M.; Topham, J.T.; Kalloger, S.E.; Jang, G.H.; Denroche, R.E.; Culibrk, L.; Williamson, L.M.; Wong, H.L.; Lee, M.K.C.; O’Kane, G.M.; et al. Altered Gene Expression along the Glycolysis-Cholesterol Synthesis Axis Is Associated with Outcome in Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 135–146. [Google Scholar] [CrossRef]
  18. Mehla, K.; Singh, P.K. Metabolic Subtyping for Novel Personalized Therapies Against Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 6–8. [Google Scholar] [CrossRef]
  19. Vaezi, M.A.; Nekoufar, S.; Robati, A.K.; Salimi, V.; Tavakoli-Yaraki, M. Therapeutic potential of β-hydroxybutyrate in the management of pancreatic neoplasms: Exploring novel diagnostic and treatment strategies. Lipids Health Dis. 2024, 23, 376. [Google Scholar] [CrossRef]
  20. Ruan, T.; Keshari, K.R. Imaging Tumor Metabolism. Cold Spring Harb. Perspect. Med. 2024, 15, a041551. [Google Scholar] [CrossRef]
  21. Wartski, M.; Sauvanet, A. 18F-FDG PET/CT in pancreatic adenocarcinoma: A role at initial imaging staging? Diagn. Interv. Imaging 2019, 100, 735–741. [Google Scholar] [CrossRef] [PubMed]
  22. Rowe, S.P.; Pomper, M.G. Molecular imaging in oncology: Current impact future directions. CA Cancer J. Clin. 2022, 72, 333–352. [Google Scholar] [CrossRef] [PubMed]
  23. Serrao, E.M.; Kettunen, M.I.; Rodrigues, T.B.; Dzien, P.; Wright, A.J.; Gopinathan, A.; Gallagher, F.A.; Lewis, D.Y.; Frese, K.K.; Almeida, J.; et al. MRI with hyperpolarised [1–13C]pyruvate detects advanced pancreatic preneoplasia prior to invasive disease in a mouse model. Gut 2016, 65, 465–475. [Google Scholar] [CrossRef]
  24. Wang, Z.J.; Ohliger, M.A.; Larson, P.E.Z.; Gordon, J.W.; Bok, R.A.; Slater, J.; Villanueva-Meyer, J.E.; Hess, C.P.; Kurhanewicz, J.; Vigneron, D.B. Hyperpolarized (13)C MRI: State of the Art and Future Directions. Radiology 2019, 291, 273–284. [Google Scholar] [CrossRef]
  25. Zhang, G.; Keshari, K.R. Deuterium Metabolic Imaging of Pancreatic Cancer. NMR Biomed. 2021, 34, e4603. [Google Scholar] [CrossRef]
  26. Koppenol, W.H.; Bounds, P.L.; Dang, C.V. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 2011, 11, 325–337. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, Y.H.; Hu, C.M.; Hsu, Y.S.; Lee, W.H. Interplays of glucose metabolism and KRAS mutation in pancreatic ductal adenocarcinoma. Cell Death Dis. 2022, 13, 817. [Google Scholar] [CrossRef]
  28. Yan, L.; Raj, P.; Yao, W.; Ying, H. Glucose Metabolism in Pancreatic Cancer. Cancers 2019, 11, 1460. [Google Scholar] [CrossRef]
  29. Tanner, L.B.; Goglia, A.G.; Wei, M.H.; Sehgal, T.; Parsons, L.R.; Park, J.O.; White, E.; Toettcher, J.E.; Rabinowitz, J.D. Four Key Steps Control Glycolytic Flux in Mammalian Cells. Cell Syst. 2018, 7, 49–62.e8. [Google Scholar] [CrossRef]
  30. Yamamoto, K.; Brender, J.R.; Seki, T.; Kishimoto, S.; Oshima, N.; Choudhuri, R.; Adler, S.S.; Jagoda, E.M.; Saito, K.; Devasahayam, N.; et al. Molecular Imaging of the Tumor Microenvironment Reveals the Relationship between Tumor Oxygenation, Glucose Uptake, and Glycolysis in Pancreatic Ductal Adenocarcinoma. Cancer Res. 2020, 80, 2087–2093. [Google Scholar] [CrossRef]
  31. Gallagher, B.M.; Ansari, A.; Atkins, H.; Casella, V.; Christman, D.R.; Fowler, J.S.; Ido, T.; MacGregor, R.R.; Som, P.; Wan, C.N.; et al. Radiopharmaceuticals, X.X.V.I.I. 18F-labeled 2-deoxy-2-fluoro-d-glucose as a radiopharmaceutical for measuring regional myocardial glucose metabolism in vivo: Tissue distribution and imaging studies in animals. J. Nucl. Med. 1977, 18, 990–996. [Google Scholar] [PubMed]
  32. Huang, S.; Chong, H.; Sun, X.; Wu, Z.; Jia, Q.; Zhang, Y.; Lan, X. The Value of (18)F-FDGPET/CTin Diagnosing Pancreatic Lesions: Comparison With, C.A.1.9.–.9.; Enhanced CTor Enhanced, M.R. Front. Med. 2021, 8, 668697. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, X.Y.; Yang, F.; Jin, C.; Fu, D.L. Utility of PET/CTin diagnosis staging assessment of resectability metabolic response of pancreatic cancer. World J. Gastroenterol. 2014, 20, 15580–15589. [Google Scholar] [CrossRef] [PubMed]
  34. Pimiento, J.M.; Davis-Yadley, A.H.; Kim, R.D.; Chen, D.T.; Eikman, E.A.; Berman, C.G.; Malafa, M.P. Metabolic Activity by 18F-FDG-PET/CT Is Prognostic for Stage I and II Pancreatic Cancer. Clin. Nucl. Med. 2016, 41, 177–181. [Google Scholar] [CrossRef]
  35. Smeets, E.M.M.; Withaar, D.S.; Grootjans, W.; Hermans, J.J.; van Laarhoven, K.; de Geus-Oei, L.F.; Gotthardt, M.; Aarntzen, E. Optimal respiratory-gated [(18)F]FDG PET/CT significantly impacts the quantification of metabolic parameters and their correlation with overall survival in patients with pancreatic ductal adenocarcinoma. EJNMMI Res. 2019, 9, 24. [Google Scholar] [CrossRef]
  36. Chen, B.B.; Tien, Y.W.; Chang, M.C.; Cheng, M.F.; Chang, Y.T.; Yang, S.H.; Wu, C.H.; Kuo, T.C.; Shih, I.L.; Yen, R.F.; et al. Multiparametric PET/MRimaging biomarkers are associated with overall survival in patients with pancreatic cancer. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1205–1217. [Google Scholar] [CrossRef]
  37. Jia, G.; Zhang, J.; Li, R.; Yan, J.; Zuo, C. The exploration of quantitative intra-tumoral metabolic heterogeneity in dual-time (18)F-FDG PET/CT of pancreatic cancer. Abdom. Radiol. 2021, 46, 4218–4225. [Google Scholar] [CrossRef]
  38. Yoo, S.H.; Kang, S.Y.; Cheon, G.J.; Oh, D.Y.; Bang, Y.J. Predictive Role of Temporal Changes in Intratumoral Metabolic Heterogeneity During Palliative Chemotherapy in Patients with Advanced Pancreatic Cancer: AProspective Cohort Study. J. Nucl. Med. 2020, 61, 33–39. [Google Scholar] [CrossRef]
  39. Yoo, J.; Lee, J.M.; Joo, I.; Lee, D.H.; Yoon, J.H.; Yu, M.H.; Jang, J.Y.; Lee, S.H. Post-neoadjuvant treatment pancreatic cancer resectability and outcome prediction using CT, (18)F-FDG PET/MRI and CA 19–9. Cancer Imaging 2023, 23, 49. [Google Scholar] [CrossRef]
  40. Christofk, H.R.; Vander Heiden, M.G.; Harris, M.H.; Ramanathan, A.; Gerszten, R.E.; Wei, R.; Fleming, M.D.; Schreiber, S.L.; Cantley, L.C. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008, 452, 230–233. [Google Scholar] [CrossRef]
  41. Beinat, C.; Patel, C.B.; Haywood, T.; Murty, S.; Naya, L.; Castillo, J.B.; Reyes, S.T.; Phillips, M.; Buccino, P.; Shen, B.; et al. A Clinical PETImaging Tracer ([(18)F]DASA-23) to Monitor Pyruvate Kinase M2-Induced Glycolytic Reprogramming in Glioblastoma. Clin. Cancer Res. 2021, 27, 6467–6478. [Google Scholar] [CrossRef]
  42. Li, X.; Deng, S.; Liu, M.; Jin, Y.; Zhu, S.; Deng, S.; Chen, J.; He, C.; Qin, Q.; Wang, C.; et al. The responsively decreased PKM2 facilitates the survival of pancreatic cancer cells in hypoglucose. Cell Death Dis. 2018, 9, 133. [Google Scholar] [CrossRef]
  43. He, D.; Feng, H.; Sundberg, B.; Yang, J.; Powers, J.; Christian, A.H.; Wilkinson, J.E.; Monnin, C.; Avizonis, D.; Thomas, C.J.; et al. Methionine oxidation activates pyruvate kinase M2 to promote pancreatic cancer metastasis. Mol. Cell. 2022, 82, 3045–3060.e3011. [Google Scholar] [CrossRef] [PubMed]
  44. Otto, A.M. Metabolic Constants and Plasticity of Cancer Cells in a Limiting Glucose and Glutamine Microenvironment-A Pyruvate Perspective. Front. Oncol. 2020, 10, 596197. [Google Scholar] [CrossRef]
  45. Comandatore, A.; Franczak, M.; Smolenski, R.T.; Morelli, L.; Peters, G.J.; Giovannetti, E. Lactate Dehydrogenase and its clinical significance in pancreatic and thoracic cancers. Semin. Cancer Biol. 2022, 86, 93–100. [Google Scholar] [CrossRef] [PubMed]
  46. Biancur, D.E.; Kimmelman, A.C. The plasticity of pancreatic cancer metabolism in tumor progression and therapeutic resistance. Biochim. Biophys. Acta Rev. Cancer 2018, 1870, 67–75. [Google Scholar] [CrossRef] [PubMed]
  47. Gordon, J.W.; Chen, H.Y.; Nickles, T.; Lee, P.M.; Bok, R.; Ohliger, M.A.; Okamoto, K.; Ko, A.H.; Larson, P.E.Z.; Wang, Z.J. Hyperpolarized (13)CMetabolic MRIof Patients with Pancreatic Ductal Adenocarcinoma, J. Magn. Reson. Imaging 2024, 60, 741–749. [Google Scholar] [CrossRef]
  48. Heid, I.; Munch, C.; Karakaya, S.; Lueong, S.S.; Winkelkotte, A.M.; Liffers, S.T.; Godfrey, L.; Cheung, P.F.Y.; Savvatakis, K.; Topping, G.J.; et al. Functional noninvasive detection of glycolytic pancreatic ductal adenocarcinoma. Cancer Metab. 2022, 10, 24. [Google Scholar] [CrossRef]
  49. Kishimoto, S.; Brender, J.R.; Crooks, D.R.; Matsumoto, S.; Seki, T.; Oshima, N.; Merkle, H.; Lin, P.; Reed, G.; Chen, A.P.; et al. Imaging of glucose metabolism by 13C-MRI distinguishes pancreatic cancer subtypes in mice. eLife 2019, 8, e46312. [Google Scholar] [CrossRef]
  50. Dutta, P.; Perez, M.R.; Lee, J.; Kang, Y.; Pratt, M.; Salzillo, T.C.; Weygand, J.; Zacharias, N.M.; Gammon, S.T.; Koay, E.J.; et al. Combining Hyperpolarized Real-Time Metabolic Imaging NMR Spectroscopy To Identify Metabolic Biomarkers in Pancreatic Cancer. J. Proteome Res. 2019, 18, 2826–2834. [Google Scholar] [CrossRef]
  51. Montrazi, E.T.; Sasson, K.; Agemy, L.; Scherz, A.; Frydman, L. Molecular imaging of tumor metabolism: Insight from pyruvate- and glucose-based deuterium MRI studies. Sci. Adv. 2024, 10, eadm8600. [Google Scholar] [CrossRef]
  52. Shimoni-Sebag, A.; Abramovich, I.; Agranovich, B.; Massri, R.; Stossel, C.; Atias, D.; Raites-Gurevich, M.; Yizhak, K.; Golan, T.; Gottlieb, E.; et al. A metabolic switch to the pentose-phosphate pathway induces radiation resistance in pancreatic cancer. Radiother. Oncol. 2025, 202, 110606. [Google Scholar] [CrossRef] [PubMed]
  53. Viswanath, P.; Batsios, G.; Ayyappan, V.; Taglang, C.; Gillespie, A.M.; Larson, P.E.Z.; Luchman, H.A.; Costello, J.F.; Pieper, R.O.; Ronen, S.M. Metabolic imaging detects elevated glucose flux through the pentose phosphate pathway associated with TERT expression in low-grade gliomas. Neuro. Oncol. 2021, 23, 1509–1522. [Google Scholar] [CrossRef]
  54. Fu, S.; Xu, S.; Zhang, S. The role of amino acid metabolism alterations in pancreatic cancer: From mechanism to application. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188893. [Google Scholar] [CrossRef]
  55. Dos Santos, K.; Bertho, G.; Baudin, M.; Giraud, N. Glutamine: A key player in human metabolism as revealed by hyperpolarized magnetic resonance. Prog. Nucl. Magn. Reson. Spectrosc. 2024, 144–145, 15–39. [Google Scholar] [CrossRef] [PubMed]
  56. Stumvoll, M.; Perriello, G.; Meyer, C.; Gerich, J. Role of glutamine in human carbohydrate metabolism in kidney and other tissues. Kidney Int. 1999, 55, 778–792. [Google Scholar] [CrossRef]
  57. Ren, L.L.; Mao, T.; Meng, P.; Zhang, L.; Wei, H.Y.; Tian, Z.B. Glutamine addiction therapeutic strategies in pancreatic cancer. World J. Gastrointest. Oncol. 2023, 15, 1852–1863. [Google Scholar] [CrossRef] [PubMed]
  58. Son, J.; Lyssiotis, C.A.; Ying, H.; Wang, X.; Hua, S.; Ligorio, M.; Perera, R.M.; Ferrone, C.R.; Mullarky, E.; Shyh-Chang, N.; et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 2013, 496, 101–105. [Google Scholar] [CrossRef]
  59. Bott, A.J.; Shen, J.; Tonelli, C.; Zhan, L.; Sivaram, N.; Jiang, Y.P.; Yu, X.; Bhatt, V.; Chiles, E.; Zhong, H.; et al. Glutamine Anabolism Plays a Critical Role in Pancreatic Cancer by Coupling Carbon and Nitrogen Metabolism. Cell Rep. 2019, 29, 1287–1298.e6. [Google Scholar] [CrossRef]
  60. Chang, C.K.; Shih, T.T.; Tien, Y.W.; Chang, M.C.; Chang, Y.T.; Yang, S.H.; Cheng, M.F.; Chen, B.B. Metabolic Alterations in Pancreatic Cancer Detected by In Vivo (1)H-MR Spectroscopy: Correlation with Normal Pancreas, PET Metabolic Activity, Clinical Stages, and Survival Outcome. Diagnostics 2021, 11, 1541. [Google Scholar] [CrossRef]
  61. Cheng, M.F.; Huang, Y.Y.; Ho, B.Y.; Kuo, T.C.; Hsin, L.W.; Shiue, C.Y.; Kuo, H.C.; Jeng, Y.M.; Yen, R.F.; Tien, Y.W. Prospective comparison of (4S)-4-(3-(18)F-fluoropropyl)-L-glutamate versus (18)F-fluorodeoxyglucose PET/CTfor detecting metastases from pancreatic ductal adenocarcinoma: Aproof-of-concept study. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 810–820. [Google Scholar] [CrossRef] [PubMed]
  62. Yin, X.; Xu, R.; Song, J.; Ruze, R.; Chen, Y.; Wang, C.; Xu, Q. Lipid metabolism in pancreatic cancer: Emerging roles and potential targets. Cancer Commun. 2022, 42, 1234–1256. [Google Scholar] [CrossRef]
  63. Wu, H.; Fu, M.; Wu, M.; Cao, Z.; Zhang, Q.; Liu, Z. Emerging mechanisms and promising approaches in pancreatic cancer metabolism. Cell Death Dis. 2024, 15, 553. [Google Scholar] [CrossRef]
  64. Meng, X.; Sun, J.; Xu, F.; Liu, Z.; He, Y. Lipid metabolism marker CD36 is associated with (18)FDG-PET/CT false negative lymph nodes in head and neck squamous cell carcinoma. Front. Oncol. 2023, 13, 1156527. [Google Scholar] [CrossRef]
  65. Tao, T.; Li, G.; Zhou, K.; Pan, Q.; Wu, D.; Lai, L.; Gao, M.; Li, S.; Chen, L.; Han, R.P.S.; et al. Discovery of Fatty Acid Translocase CD36-Targeting Near-Infrared Fluorescent Probe Enables Visualization and Imaging-Guided Surgery for Glioma. Anal. Chem. 2025, 97, 3687–3695. [Google Scholar] [CrossRef]
  66. Brooks, A.F.; Winton, W.P.; Stauff, J.; Arteaga, J.; Henderson, B.; Niedbala, J.; Scott, P.J.H.; Viglianti, B.L. Development of Fluorinated NP-59: ARevival of Cholesterol Use Imaging with, PET. J. Nucl. Med. 2022, 63, 1949–1955. [Google Scholar] [CrossRef]
  67. Ahmed, H.; Wang, Y.; Griffiths, W.J.; Levey, A.I.; Pikuleva, I.; Liang, S.H.; Haider, A. Brain cholesterol and Alzheimer’s disease: Challenges and opportunities in probe and drug development. Brain 2024, 147, 1622–1635. [Google Scholar] [CrossRef] [PubMed]
  68. Cohen, A.S.; Li, J.; Hight, M.R.; McKinley, E.; Fu, A.; Payne, A.; Liu, Y.; Zhang, D.; Xie, Q.; Bai, M.; et al. TSPO-targeted PET and Optical Probes for the Detection Localization of Premalignant Malignant Pancreatic Lesions. Clin. Cancer Res. 2020, 26, 5914–5925. [Google Scholar] [CrossRef] [PubMed]
  69. Penet, M.F.; Shah, T.; Bharti, S.; Krishnamachary, B.; Artemov, D.; Mironchik, Y.; Wildes, F.; Maitra, A.; Bhujwalla, Z.M. Metabolic imaging of pancreatic ductal adenocarcinoma detects altered choline metabolism. Clin. Cancer Res. 2015, 21, 386–395. [Google Scholar] [CrossRef]
  70. Chen, B.B.; Tien, Y.W.; Chang, M.C.; Cheng, M.F.; Chang, Y.T.; Wu, C.H.; Chen, X.J.; Kuo, T.C.; Yang, S.H.; Shih, I.L.; et al. PET/MRIin pancreatic periampullary cancer: Correlating diffusion-weighted imaging MRspectroscopy glucose metabolic activity with clinical stage prognosis. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 1753–1764. [Google Scholar] [CrossRef]
  71. Veltien, A.; van Asten, J.; Ravichandran, N.; de Graaf, R.A.; De Feyter, H.M.; Oosterwijk, E.; Heerschap, A. Simultaneous Recording of the Uptake and Conversion of Glucose and Choline in Tumors by Deuterium Metabolic Imaging. Cancers 2021, 13, 4034. [Google Scholar] [CrossRef] [PubMed]
  72. Donadon, M.; Lopci, E.; Galvanin, J.; Giudici, S.; Del Fabbro, D.; Lanza, E.; Pedicini, V.; Chiti, A.; Torzilli, G. Prognostic Value of Metabolic Imaging Data of (11)C-choline PET/CT in Patients Undergoing Hepatectomy for Hepatocellular Carcinoma. Cancers 2021, 13, 472. [Google Scholar] [CrossRef]
  73. Caroli, P.; De Giorgi, U.; Scarpi, E.; Fantini, L.; Moretti, A.; Galassi, R.; Celli, M.; Conteduca, V.; Rossi, L.; Bianchi, E.; et al. Prognostic value of 18F-choline PET/CTmetabolic parameters in patients with metastatic castration-resistant prostate cancer treated with abiraterone or enzalutamide. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 348–354. [Google Scholar] [CrossRef]
  74. Treglia, G.; Rizzo, A.; Piccardo, A. Expanding the clinical indications of [(18)F]fluorocholine PET/CTin primary hyperparathyroidism: The evidence cannot be evaded. Eur. J. Nucl. Med. Mol. Imaging 2024, 51, 1345–1348. [Google Scholar] [CrossRef] [PubMed]
  75. Clauser, P.; Rasul, S.; Kapetas, P.; Fueger, B.J.; Milos, R.I.; Balber, T.; Berroterán-Infante, N.; Hacker, M.; Helbich, T.H.; Baltzer, P.A.T. Prospective validation of 18F-Fluoroethylcholine as a tracer in PET/MRI for the evaluation of breast lesions and prediction of lymph node status. Radiol. Med. 2023, 128, 689–698. [Google Scholar] [CrossRef] [PubMed]
  76. Evangelista, L.; De Pellegrin, A.; Girometti, R.; Cassarino, G.; Giacomuzzi, F.; Rensi, M. An Incidental Pancreatic Finding at 18F-Choline PET/CT: Chronic Mass-Forming Pancreatitis. Diagnostics 2021, 11, 1490. [Google Scholar] [CrossRef]
  77. Kitajima, K.; Fukushima, K.; Yamamoto, S.; Kato, T.; Odawara, S.; Takaki, H.; Kobayashi, K.; Yamano, T.; Yamakado, K.; Nakanishi, Y.; et al. (11)C-Choline positive but (18)F-FDGnegative pancreatic metastasis from renal cell carcinoma on, P.E.T. Nagoya J. Med. Sci. 2017, 79, 273–277. [Google Scholar]
  78. Shah, T.; Krishnamachary, B.; Wildes, F.; Wijnen, J.P.; Glunde, K.; Bhujwalla, Z.M. Molecular causes of elevated phosphoethanolamine in breast and pancreatic cancer cells. NMR Biomed. 2018, 31, e3936. [Google Scholar] [CrossRef]
  79. Buchthal, S.D.; Thoma, W.J.; Taylor, J.S.; Nelson, S.J.; Brown, T.R. In vivo T1 values of phosphorus metabolites in human liver and muscle determined at 1.5 T by chemical shift imaging. NMR Biomed. 1989, 2, 298–304. [Google Scholar] [CrossRef]
  80. Rivera, D. Emerging Role for 7T MRI and Metabolic Imaging for Pancreatic and Liver Cancer. Metabolites 2022, 12, 409. [Google Scholar] [CrossRef]
  81. Cheung, E.C.; Vousden, K.H. The role of ROS in tumour development and progression. Nat. Rev. Cancer 2022, 22, 280–297. [Google Scholar] [CrossRef] [PubMed]
  82. Purohit, V.; Simeone, D.M.; Lyssiotis, C.A. Metabolic Regulation of Redox Balance in Cancer. Cancers 2019, 11, 955. [Google Scholar] [CrossRef] [PubMed]
  83. Xing, F.; Hu, Q.; Qin, Y.; Xu, J.; Zhang, B.; Yu, X.; Wang, W. The Relationship of Redox with Hallmarks of Cancer: The Importance of Homeostasis and Context. Front. Oncol. 2022, 12, 862743. [Google Scholar] [CrossRef]
  84. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef]
  85. Martínez-Reyes, I.; Chandel, N.S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 2020, 11, 102. [Google Scholar] [CrossRef]
  86. Broekgaarden, M.; Bulin, A.L.; Frederick, J.; Mai, Z.; Hasan, T. Tracking, Photodynamic and Chemotherapy-Induced Redox-State Perturbations in 3DCulture Models of Pancreatic Cancer: A Tool for Identifying Therapy-Induced Metabolic Changes. J. Clin. Med. 2019, 8, 1399. [Google Scholar] [CrossRef] [PubMed]
  87. Walsh, A.J.; Castellanos, J.A.; Nagathihalli, N.S.; Merchant, N.B.; Skala, M.C. Optical Imaging of Drug-Induced Metabolism Changes in Murine and Human Pancreatic Cancer Organoids Reveals Heterogeneous Drug Response. Pancreas 2016, 45, 863–869. [Google Scholar] [CrossRef]
  88. Datta, R.; Sivanand, S.; Lau, A.N.; Florek, L.V.; Barbeau, A.M.; Wyckoff, J.; Skala, M.C.; Vander Heiden, M.G. Interactions with stromal cells promote a more oxidized cancer cell redox state in pancreatic tumors. Sci. Adv. 2022, 8, eabg6383. [Google Scholar] [CrossRef]
  89. Broekgaarden, M.; Anbil, S.; Bulin, A.L.; Obaid, G.; Mai, Z.; Baglo, Y.; Rizvi, I.; Hasan, T. Modulation of redox metabolism negates cancer-associated fibroblasts-induced treatment resistance in a heterotypic 3D culture platform of pancreatic cancer. Biomaterials 2019, 222, 119421. [Google Scholar] [CrossRef]
  90. Huang, P.; Chubb, S.; Hertel, L.W.; Grindey, G.B.; Plunkett, W. Action of 2′,2′-difluorodeoxycytidine on DNA synthesis. Cancer Res. 1991, 51, 6110–6117. [Google Scholar]
  91. Russell, J.; Grkovski, M.; O’Donoghue, I.J.; Kalidindi, T.M.; Pillarsetty, N.; Burnazi, E.M.; Kulick, A.; Bahr, A.; Chang, Q.; LeKaye, H.C.; et al. Predicting Gemcitabine Delivery by (18)F-FACPETin Murine Models of Pancreatic Cancer. J. Nucl. Med. 2021, 62, 195–200. [Google Scholar] [CrossRef] [PubMed]
  92. Strittmatter, N.; Richards, F.M.; Race, A.M.; Ling, S.; Sutton, D.; Nilsson, A.; Wallez, Y.; Barnes, J.; Maglennon, G.; Gopinathan, A.; et al. Method To Visualize the Intratumor Distribution and Impact of Gemcitabine in Pancreatic Ductal Adenocarcinoma by Multimodal Imaging. Anal. Chem. 2022, 94, 1795–1803. [Google Scholar] [CrossRef] [PubMed]
  93. Bjånes, T.K.; Jordheim, L.P.; Schjøtt, J.; Kamceva, T.; Cros-Perrial, E.; Langer, A.; Ruiz de Garibay, G.; Kotopoulis, S.; McCormack, E.; Riedel, B. Intracellular Cytidine Deaminase Regulates Gemcitabine Metabolism in Pancreatic Cancer Cell Lines. Drug Metab. Dispos. 2020, 48, 153–158. [Google Scholar] [CrossRef] [PubMed]
  94. Yamamoto, K.; Iwadate, D.; Kato, H.; Nakai, Y.; Tateishi, K.; Fujishiro, M. Targeting the Metabolic Rewiring in Pancreatic Cancer and Its Tumor Microenvironment. Cancers 2022, 14, 4351. [Google Scholar] [CrossRef]
  95. Li, Y.; Zhang, J.; Xu, J.; Liu, S. The Metabolism Symbiosis Between Pancreatic Cancer and Tumor Microenvironment. Front. Oncol. 2021, 11, 759376. [Google Scholar] [CrossRef]
  96. Yamane, T.; Aikawa, M.; Yasuda, M.; Fukushima, K.; Seto, A.; Okamoto, K.; Koyama, I.; Kuji, I. [(18)F]FMISO PET/CT as a preoperative prognostic factor in patients with pancreatic cancer. EJNMMI Res. 2019, 9, 39. [Google Scholar] [CrossRef]
  97. Metran-Nascente, C.; Yeung, I.; Vines, D.C.; Metser, U.; Dhani, N.C.; Green, D.; Milosevic, M.; Jaffray, D.; Hedley, D.W. Measurement of Tumor Hypoxia in Patients with Advanced Pancreatic Cancer Based on 18F-Fluoroazomyin Arabinoside Uptake. J. Nucl. Med. 2016, 57, 361–366. [Google Scholar] [CrossRef]
  98. Wang, Y.; Jhang, D.F.; Tsai, C.H.; Chiang, N.J.; Tsao, C.H.; Chuang, C.C.; Chen, L.T.; Chang, W.W.; Liao, L.D. In Vivo Assessment of Hypoxia Levels in Pancreatic Tumors Using a Dual-Modality Ultrasound/Photoacoustic Imaging System. Micromachines 2021, 12, 668. [Google Scholar] [CrossRef]
  99. Pereira, B.A.; Vennin, C.; Papanicolaou, M.; Chambers, C.R.; Herrmann, D.; Morton, J.P.; Cox, T.R.; Timpson, P. CAF Subpopulations: A New Reservoir of Stromal Targets in Pancreatic Cancer. Trends Cancer 2019, 5, 724–741. [Google Scholar] [CrossRef]
  100. Lavie, D.; Ben-Shmuel, A.; Erez, N.; Scherz-Shouval, R. Cancer-associated fibroblasts in the single-cell era. Nat. Cancer 2022, 3, 793–807. [Google Scholar] [CrossRef] [PubMed]
  101. Saúde-Conde, R.; Arçay Öztürk, A.; Stosic, K.; Azurmendi Senar, O.; Navez, J.; Bouchart, C.; Arsenijevic, T.; Flamen, P.; Van Laethem, J.L. Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma or a Metaphor for Heterogeneity: From Single-Cell Analysis to Whole-Body Imaging. Biomedicines 2024, 12, 591. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, Q.; Shi, S.; Liu, S.; Xu, X.; Hu, S.; Zhang, J.; Wang, C.; Yu, X.; Song, S. The added value of [(68)Ga]Ga-DOTA-FAPI-04 PET/CTin pancreatic cancer: Acomparison to, [.(.1.8.).F.].F.-F.D.G. Eur. Radiol. 2023, 33, 5007–5016. [Google Scholar] [CrossRef]
  103. Kaghazchi, F.; Divband, G.; Amini, H.; Adinehpour, Z.; Akbarian Aghdam, R. 68 Ga-FAPI-46 and 18 F-FDG in Advanced Metastatic Pancreatic Cancer. Clin. Nucl. Med. 2022, 47, e666–e669. [Google Scholar] [CrossRef]
  104. Kessler, L.; Hirmas, N.; Pabst, K.M.; Hamacher, R.; Ferdinandus, J.; Schaarschmidt, B.M.; Milosevic, A.; Nader, M.; Umutlu, L.; Uhl, W.; et al. (68)Ga-Labeled Fibroblast Activation Protein Inhibitor ((68)Ga-FAPI) PET for Pancreatic Adenocarcinoma: Data from the (68)Ga-FAPI PET Observational Trial. J. Nucl. Med. 2023, 64, 1910–1917. [Google Scholar] [CrossRef]
  105. Spektor, A.M.; Gutjahr, E.; Lang, M.; Glatting, F.M.; Hackert, T.; Pausch, T.; Tjaden, C.; Schreckenberger, M.; Haberkorn, U.; Röhrich, M. Immunohistochemical FAP Expression Reflects (68)Ga-FAPI PET Imaging Properties of Low- and High-Grade Intraductal Papillary Mucinous Neoplasms and Pancreatic Ductal Adenocarcinoma. J. Nucl. Med. 2024, 65, 52–58. [Google Scholar] [CrossRef] [PubMed]
  106. Rasinski, P.; Af Burén, S.; Holstensson, M.; Nilsson, T.; Loizou, L.; Tran, T.A.; Sparrelid, E.; Löhr, J.M.; Axelsson, R. Tumor Characterization by [(68)Ga]FAPI-46 PET/CT Can Improve Treatment Selection for Pancreatic Cancer Patients: An Interim Analysis of a Prospective Clinical Trial. J. Nucl. Med. 2023, 64, 1232–1237. [Google Scholar] [CrossRef] [PubMed]
  107. Ouyang, J.; Ding, P.; Zhang, R.; Lu, Y. Head-to-head comparison of (68)Ga-FAPI-04 PET/CT and (18)F-FDG PET/CT in the evaluation of primary digestive system cancer: A systematic review and meta-analysis. Front. Oncol. 2023, 13, 1202505. [Google Scholar] [CrossRef]
  108. Ding, J.; Qiu, J.; Hao, Z.; Huang, H.; Liu, Q.; Liu, W.; Ren, C.; Hacker, M.; Zhang, T.; Wu, W.; et al. Comparing the clinical value of baseline [(68) Ga]Ga-FAPI-04 PET/CT and [(18)F]F-FDG PET/CT in pancreatic ductal adenocarcinoma: Additional prognostic value of the distal pancreatitis. Eur. J. Nucl. Med. Mol. Imaging 2023, 50, 4036–4050. [Google Scholar] [CrossRef]
  109. Ding, J.; Qiu, J.; Hao, Z.; Huang, H.; Liu, Q.; Liu, W.; Ren, C.; Hacker, M.; Zhang, T.; Wu, W.; et al. Prognostic value of preoperative [(68) Ga]Ga-FAPI-04 PET/CT in patients with resectable pancreatic ductal adenocarcinoma in correlation with immunohistological characteristics. Eur. J. Nucl. Med. Mol. Imaging 2023, 50, 1780–1791. [Google Scholar] [CrossRef]
  110. Zhang, Z.; Jia, G.; Pan, G.; Cao, K.; Yang, Q.; Meng, H.; Yang, J.; Zhang, L.; Wang, T.; Cheng, C.; et al. Comparison of the diagnostic efficacy of (68) Ga-FAPI-04 PET/MR and (18)F-FDG PET/CT in patients with pancreatic cancer. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 2877–2888. [Google Scholar] [CrossRef]
  111. Röhrich, M.; Naumann, P.; Giesel, F.L.; Choyke, P.L.; Staudinger, F.; Wefers, A.; Liew, D.P.; Kratochwil, C.; Rathke, H.; Liermann, J.; et al. Impact of (68)Ga-FAPI PET/CT Imaging on the Therapeutic Management of Primary and Recurrent Pancreatic Ductal Adenocarcinomas. J. Nucl. Med. 2021, 62, 779–786. [Google Scholar] [CrossRef] [PubMed]
  112. Metzger, G.; Bayerl, C.; Rogasch, J.M.; Furth, C.; Wetz, C.; Beck, M.; Mehrhof, F.; Amthauer, H.; Ghadjar, P.; Neumann, C.; et al. (68)Ga-labeled fibroblast activation protein inhibitor (FAPI) PET/CT for locally advanced or recurrent pancreatic cancer staging and restaging after chemoradiotherapy. Theranostics 2024, 14, 4184–4197. [Google Scholar] [CrossRef] [PubMed]
  113. Jia, R.; Hao, X.-Z.; Si, H.-Y.; Zhu, W.-J.; Wang, Z.-K.; Huo, L.; Dai, G.-H. Predictive value of baseline68Ga-FAPI PET/CT in the first-line therapy of pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2024, 42, e16300. [Google Scholar] [CrossRef]
  114. Li, Z.; Geng, R.; Zhan, Y.; Liu, R.; Li, M.; Ke, N.; Yang, H.; Lu, X.; Li, L.; Li, S.; et al. (64)Cu Radiolabeled PDGFRβ-Targeting Affibody for PET Imaging in Pancreatic Cancer. Mol. Pharm. 2025, 22, 1633–1640. [Google Scholar] [CrossRef]
  115. Pysz, M.A.; Machtaler, S.B.; Seeley, E.S.; Lee, J.J.; Brentnall, T.A.; Rosenberg, J.; Tranquart, F.; Willmann, J.K. Vascular Endothelial Growth Factor Receptor Type 2–targeted Contrast-enhanced US of Pancreatic Cancer Neovasculature in a Genetically Engineered Mouse Model: Potential for Earlier Detection. Radiology 2015, 274, 790–799. [Google Scholar] [CrossRef]
  116. Willmann, J.K.; Bonomo, L.; Testa, A.C.; Rinaldi, P.; Rindi, G.; Valluru, K.S.; Petrone, G.; Martini, M.; Lutz, A.M.; Gambhir, S.S. Ultrasound Molecular Imaging with BR55 in Patients with Breast Ovarian Lesions: First-in-Human Results. J. Clin. Oncol. 2017, 35, 2133–2140. [Google Scholar] [CrossRef]
  117. Mulder, B.G.S.; Koller, M.; Duiker, E.W.; Sarasqueta, A.F.; Burggraaf, J.; Meijer, V.E.; Vahrmeijer, A.L.; Hoogwater, F.J.H.; Bonsing, B.A.; van Dam, G.M.; et al. Intraoperative Molecular Fluorescence Imaging of Pancreatic Cancer by Targeting Vascular Endothelial Growth Factor: A Multicenter Feasibility Dose-Escalation Study. J. Nucl. Med. 2023, 64, 82–89. [Google Scholar] [CrossRef]
  118. Tripathi, S.; Tabari, A.; Mansur, A.; Dabbara, H.; Bridge, C.P.; Daye, D. From Machine Learning to Patient Outcomes: A Comprehensive Review of AI in Pancreatic Cancer. Diagnostics 2024, 14, 174. [Google Scholar] [CrossRef]
  119. Liu, W.; Zhang, B.; Liu, T.; Jiang, J.; Liu, Y. Artificial Intelligence in Pancreatic Image Analysis: A Review. Sensors 2024, 24, 4749. [Google Scholar] [CrossRef]
  120. Enriquez, J.S.; Chu, Y.; Pudakalakatti, S.; Hsieh, K.L.; Salmon, D.; Dutta, P.; Millward, N.Z.; Lurie, E.; Millward, S.; McAllister, F.; et al. Hyperpolarized Magnetic Resonance and Artificial Intelligence: Frontiers of Imaging in Pancreatic Cancer. JMIR Med. Inform. 2021, 9, e26601. [Google Scholar] [CrossRef]
  121. Cao, K.; Xia, Y.; Yao, J.; Han, X.; Lambert, L.; Zhang, T.; Tang, W.; Jin, G.; Jiang, H.; Fang, X.; et al. Large-scale pancreatic cancer detection via non-contrast CT and deep learning. Nat. Med. 2023, 29, 3033–3043. [Google Scholar] [CrossRef] [PubMed]
  122. Placido, D.; Yuan, B.; Hjaltelin, J.X.; Zheng, C.; Haue, A.D.; Chmura, P.J.; Yuan, C.; Kim, J.; Umeton, R.; Antell, G.; et al. A deep learning algorithm to predict risk of pancreatic cancer from disease trajectories. Nat. Med. 2023, 29, 1113–1122. [Google Scholar] [CrossRef] [PubMed]
  123. Yao, J.; Shi, Y.; Cao, K.; Lu, L.; Lu, J.; Song, Q.; Jin, G.; Xiao, J.; Hou, Y.; Zhang, L. DeepPrognosis: Preoperative prediction of pancreatic cancer survival and surgical margin via comprehensive understanding of dynamic contrast-enhanced CT imaging and tumor-vascular contact parsing. Med. Image Anal. 2021, 73, 102150. [Google Scholar] [CrossRef] [PubMed]
  124. Park, Y.J.; Park, Y.S.; Kim, S.T.; Hyun, S.H. A Machine Learning Approach Using [(18)F]FDG PET-Based Radiomics for Prediction of Tumor Grade and Prognosis in Pancreatic Neuroendocrine Tumor. Mol. Imaging Biol. 2023, 25, 897–910. [Google Scholar] [CrossRef]
  125. Wei, W.; Jia, G.; Wu, Z.; Wang, T.; Wang, H.; Wei, K.; Cheng, C.; Liu, Z.; Zuo, C. A multidomain fusion model of radiomics and deep learning to discriminate between PDAC and AIP based on 18F-FDG PET/CT images. Jpn. J. Radiol. 2023, 41, 417–427. [Google Scholar] [CrossRef]
  126. Zhang, G.; Bao, C.; Liu, Y.; Wang, Z.; Du, L.; Zhang, Y.; Wang, F.; Xu, B.; Zhou, S.K.; Liu, R. (18)F-FDG-PET/CT-based deep learning model for fully automated prediction of pathological grading for pancreatic ductal adenocarcinoma before surgery. EJNMMI Res. 2023, 13, 49. [Google Scholar] [CrossRef]
  127. Zhao, G.; Chen, X.; Zhu, M.; Liu, Y.; Wang, Y. Exploring the application and future outlook of Artificial intelligence in pancreatic cancer. Front. Oncol. 2024, 14, 1345810. [Google Scholar] [CrossRef]
  128. Ai, C.; Sun, X.; Xiao, S.; Guo, L.; Shang, M.; Shi, D.; Meng, D.; Zhao, Y.; Wang, X.; Li, J. CAFs targeted ultrasound-responsive nanodroplets loaded V9302 and GLULsiRNA to inhibit melanoma growth via glutamine metabolic reprogramming and tumor microenvironment remodeling. J. Nanobiotechnology 2023, 21, 214. [Google Scholar] [CrossRef] [PubMed]
  129. Kobayashi, K.; Einama, T.; Tsunenari, T.; Yonamine, N.; Takao, M.; Takihata, Y.; Tsujimoto, H.; Ueno, H.; Tamura, K.; Ishida, J.; et al. Preoperative CA19-9 level and dual time point FDG-PET/CT as strong biological indicators of borderline resectability in pancreatic cancer: A retrospective study. Oncol. Lett. 2024, 27, 279. [Google Scholar] [CrossRef]
  130. Kanda, T.; Wakiya, T.; Ishido, K.; Kimura, N.; Fujita, H.; Yoshizawa, T.; Goto, S.; Tatara, Y.; Kijima, H.; Hakamada, K. Heterogeneity of metabolic adaptive capacity affects the prognosis among pancreatic ductal adenocarcinomas. J. Gastroenterol. 2022, 57, 798–811. [Google Scholar] [CrossRef]
  131. Mukhopadhyay, S.; Vander Heiden, M.G.; McCormick, F. The Metabolic Landscape of RAS-Driven Cancers from biology to therapy. Nat. Cancer 2021, 2, 271–283. [Google Scholar] [CrossRef] [PubMed]
  132. De Jesus, V.H.F.; Mathias-Machado, M.C.; de Farias, J.P.F.; Aruquipa, M.P.S.; Jácome, A.A.; Peixoto, R.D. Targeting KRAS in Pancreatic Ductal Adenocarcinoma: The Long Road to Cure. Cancers 2023, 15, 5015. [Google Scholar] [CrossRef] [PubMed]
  133. Koch, P.D.; Quintana, J.; Ahmed, M.; Kohler, R.H.; Weissleder, R. Small Molecule Imaging Agent for Mutant KRAS G12C. Adv. Ther. 2021, 4, 2000290. [Google Scholar] [CrossRef]
  134. Karageorgou, M.-A.; Bouziotis, P.; Stiliaris, E.; Stamopoulos, D. Radiolabeled Iron Oxide Nanoparticles as Dual Modality Contrast Agents in SPECT/MRI and PET/MRI. Nanomaterials 2023, 13, 503. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The landscape of key components in the metabolic imaging of PC. (Created in BioRender. Wang, A. (2025) https://BioRender.com/dekq39y). Key metabolites and targets for metabolic imaging and their metabolic processes are shown in the figure, with exogenous molecules highlighted in red boxes; 3-PG: 3-bisphosphoglycerate, ALT: alanine transaminase, CDA: cytidine deaminase, CoA: coenzyme A, CTL1: choline transporter like-protein 1, dCK: deoxycytidine kinase, DHAP: dihydroxyacetone phosphate, FAD: flavin adenine dinucleotide, fructose-6P: fructose 6-phosphate, fructose-1,6-biP: fructose 1,6-bisphosphate, R-NO2: NO2-containing molecular imaging probes, GA3P: glyceraldehyde 3-phosphate, GLUT1: glucose transporter 1, glucose-6P: glucose 6-phosphate, HK1/2: hexokinase 1/2, LDH: lactate dehydrogenase, MPC: mitochondrial pyruvate carrier, NAD: nicotinamide adenine dinucleotide, NADPH: nicotinamide adenine dinucleotide phosphate, OAA: oxaloacetate, TCA: tricarboxylic acid.
Figure 1. The landscape of key components in the metabolic imaging of PC. (Created in BioRender. Wang, A. (2025) https://BioRender.com/dekq39y). Key metabolites and targets for metabolic imaging and their metabolic processes are shown in the figure, with exogenous molecules highlighted in red boxes; 3-PG: 3-bisphosphoglycerate, ALT: alanine transaminase, CDA: cytidine deaminase, CoA: coenzyme A, CTL1: choline transporter like-protein 1, dCK: deoxycytidine kinase, DHAP: dihydroxyacetone phosphate, FAD: flavin adenine dinucleotide, fructose-6P: fructose 6-phosphate, fructose-1,6-biP: fructose 1,6-bisphosphate, R-NO2: NO2-containing molecular imaging probes, GA3P: glyceraldehyde 3-phosphate, GLUT1: glucose transporter 1, glucose-6P: glucose 6-phosphate, HK1/2: hexokinase 1/2, LDH: lactate dehydrogenase, MPC: mitochondrial pyruvate carrier, NAD: nicotinamide adenine dinucleotide, NADPH: nicotinamide adenine dinucleotide phosphate, OAA: oxaloacetate, TCA: tricarboxylic acid.
Ijms 26 05242 g001
Figure 2. Probe structures or schematic diagrams of the probes for metabolic imaging. (Created in BioRender. Wang, A. (2025) https://BioRender.com/iihhl3v). (A) 18F-FDG; (B) [6,6-2H2] glucose; (C) 13C-Glucose; (D) gemcitabine and 18F-FAC; (E) the metabolism of 13C-Pyruvate in cancer; (F) schematic diagrams of VEGFR2-targeted microbubbles.
Figure 2. Probe structures or schematic diagrams of the probes for metabolic imaging. (Created in BioRender. Wang, A. (2025) https://BioRender.com/iihhl3v). (A) 18F-FDG; (B) [6,6-2H2] glucose; (C) 13C-Glucose; (D) gemcitabine and 18F-FAC; (E) the metabolism of 13C-Pyruvate in cancer; (F) schematic diagrams of VEGFR2-targeted microbubbles.
Ijms 26 05242 g002
Figure 3. Potential role of metabolic imaging in clinical decision-making. (Created in BioRender. Wang, A. (2025) https://BioRender.com/pcrjp4k).
Figure 3. Potential role of metabolic imaging in clinical decision-making. (Created in BioRender. Wang, A. (2025) https://BioRender.com/pcrjp4k).
Ijms 26 05242 g003
Table 1. Key molecular imaging modalities in pancreatic cancer related to metabolism.
Table 1. Key molecular imaging modalities in pancreatic cancer related to metabolism.
Imaging TechniqueTracersTargeted Molecules or ProcessesApplicationsStage 1
PET18F-FDGGlucose uptakeDiagnosis, staging, recurrence detection, resectability prediction, prognosisLicensed tracers used in the clinic
18F-FSPGCystine/glutamate transporterDetection of metastasisClinical evaluation
18F-FACdCK activityGemcitabine metabolismExperimental modalities tested in animals
68Ga-FAPIFAPStromal mapping, metastasis detection, therapy responseClinical evaluation
64Cu- ZPDGFRβPDGFRβNAExperimental modalities tested in animals
18F-V-1008TSPODistinguish early diseaseExperimental modalities tested in animals
18F-FMISO and 18F-FAZAHypoxiaPrognosisClinical evaluation
MRI/MRSNAGlx, NAA, cholineDiagnosisClinical evaluation
13C-glucoseGlucose metabolismMetabolic flux analysis, therapy responseClinical evaluation
13C-pyruvate
2H-labeled glucose/cholineGlucose/cholineMulti-metabolite mappingExperimental modalities tested in animals
MSINAGemcitabine and its metabolitesGemcitabine metabolismExperimental modalities tested in animals
NIR imagingV-1520TSPOImage-guided surgeryExperimental modalities tested in animals
IRDye800CWVEGFR2Image-guided surgeryClinical evaluation
Optical metabolic imagingNANADPH and FADTherapy responseExperimental modalities tested in animals
US/PA/CEUSNAOxygenationTherapy responseExperimental modalities tested in animals
VEGFR2-targeted microbubblesVEGFR2DiagnosisClinical evaluation
1 The stage of application in pancreatic cancer. PET: positron emission tomography; MRI: magnetic resonance imaging; MRS: magnetic resonance spectroscopy; MSI: mass spectrometry imaging; NIR: near-infrared; US: ultrasound; PA: photoacoustic; CEUS: contrast-enhanced ultrasound.
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

Wang, H.; Gui, Y.; Lv, K. Molecular Imaging: Unveiling Metabolic Abnormalities in Pancreatic Cancer. Int. J. Mol. Sci. 2025, 26, 5242. https://doi.org/10.3390/ijms26115242

AMA Style

Wang H, Gui Y, Lv K. Molecular Imaging: Unveiling Metabolic Abnormalities in Pancreatic Cancer. International Journal of Molecular Sciences. 2025; 26(11):5242. https://doi.org/10.3390/ijms26115242

Chicago/Turabian Style

Wang, Huanyu, Yang Gui, and Ke Lv. 2025. "Molecular Imaging: Unveiling Metabolic Abnormalities in Pancreatic Cancer" International Journal of Molecular Sciences 26, no. 11: 5242. https://doi.org/10.3390/ijms26115242

APA Style

Wang, H., Gui, Y., & Lv, K. (2025). Molecular Imaging: Unveiling Metabolic Abnormalities in Pancreatic Cancer. International Journal of Molecular Sciences, 26(11), 5242. https://doi.org/10.3390/ijms26115242

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop