Next Article in Journal
New Benzofuran–Pyrazole-Based Compounds as Promising Antimicrobial Agents: Design, Synthesis, DNA Gyrase B Inhibition, and In Silico Studies
Previous Article in Journal
Effects of Alnus japonica Hot Water Extract and Oregonin on Muscle Loss and Muscle Atrophy in C2C12 Murine Skeletal Muscle Cells
Previous Article in Special Issue
Characterization and Comparison of Contrast Imaging Properties of Naturally Isolated and Heterologously Expressed Gas Vesicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells

1
Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan
2
Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
3
Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2024, 17(12), 1663; https://doi.org/10.3390/ph17121663
Submission received: 20 November 2024 / Revised: 8 December 2024 / Accepted: 9 December 2024 / Published: 10 December 2024
(This article belongs to the Special Issue Molecular Imaging of the Tumor Microenvironment)

Abstract

:
The tumor microenvironment (TME) is a critical factor in cancer progression, driving tumor growth, immune evasion, therapeutic resistance, and metastasis. Understanding the dynamic interactions within the TME is essential for advancing cancer management. Molecular imaging provides a non-invasive, real-time, and longitudinal approach to studying the TME, with techniques such as positron emission tomography (PET), magnetic resonance imaging (MRI), and fluorescence imaging offering complementary strengths, including high sensitivity, spatial resolution, and intraoperative precision. Recent advances in imaging probe development have enhanced the ability to target and monitor specific components of the TME, facilitating early cancer diagnosis, therapeutic monitoring, and deeper insights into tumor biology. By integrating these innovations, molecular imaging offers transformative potential for precision oncology, improving diagnostic accuracy and treatment outcomes through a comprehensive assessment of TME dynamics.

1. Introduction

Cancer remains the leading cause of death worldwide, claiming millions of lives annually. Mounting evidence reveals that tumor progression is driven not solely by cancer cell proliferation but also by intricate interactions within the tumor microenvironment (TME) [1]. The concept of the TME was first proposed by Stephen Paget in his seminal “seed and soil” theory in 1889 [2,3], suggesting that the success of metastasis relies on both the inherent characteristics of cancer cells (seed) and the supportive microenvironment (soil). Various non-cancerous components can be found in the TME [4] (Figure 1), including cancer-associated fibroblasts (CAFs), immune cells, endothelial cells, and extracellular matrix elements (ECM), which collectively contribute to cancer survival, proliferation, immune evasion, therapeutic resistance, and metastasis [5,6].
Targeting specific TME components presents an ideal strategy for improving cancer treatment and diagnosis. The TME is highly dynamic and continues to evolve throughout cancer progression. In other words, technologies that can identify the early changes in the TME, such as liquid biopsy [7,8] and molecular imaging [9,10], are invaluable tools for diagnosing cancer and monitoring these changes throughout different stages.
Molecular imaging techniques, including positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), computed tomography (CT), ultrasound, and fluorescence imaging, are widely used in clinical and preclinical cancer research [10]. Each imaging modality offers unique strengths tailored to specific requirements, yet they also present inherent challenges that must be addressed when evaluating their utility for tumor imaging.
PET and SPECT are highly sensitive techniques that detect biochemical and functional changes within the TME. PET imaging excels in quantitative molecular imaging, providing high sensitivity and temporal resolution for tracking specific molecules, including metabolic activity, receptor dynamics, and enzyme activity [11,12]. Similarly, SPECT enables the imaging of molecules labeled with longer-lived isotopes. Both modalities lack anatomical information, often requiring integration with CT or MRI for precise localization. Additionally, using radiotracers with short half-lives can present challenges, particularly for facilities without an on-site cyclotron.
MRI provides exceptional soft tissue contrast and high spatial resolution, making it particularly effective for delineating tumor margins and TME [13]. By adjusting sequencing parameters, MRI can generate images with different signal weightings, such as T1-weighted (T1WI) and T2-weighted (T2WI) imaging, allowing for the monitoring of anatomical and molecular information regarding lesions. Furthermore, MRI does not involve ionizing radiation, making it safer for repeated use in longitudinal studies and treatment monitoring. However, significant drawbacks include its relatively high cost, longer acquisition times, and limited availability. Although MRI can provide some molecular information, its sensitivity is lower than that of PET or SPECT. New contrast agents and pulse sequences need to be developed to enhance its capabilities.
CT is a widely available modality known for its exceptional anatomical detail and fast acquisition times, making it particularly valuable in emergencies and dynamic imaging settings. Its clinical applications include assessing perfusion in ischemic stroke, evaluating coronary calcification, and detecting lung nodules for early cancer diagnosis. However, CT relies on ionizing radiation, which limits the rationality of using it for repeated imaging.
Ultrasound (US) is an accessible and cost-effective imaging method recognized for its ability to provide real-time images without the use of ionizing radiation. It helps guide interventional procedures and monitor fetal development. However, its effectiveness is highly operator-dependent, and its limited penetration depth and resolution reduces its utility for imaging deep-seated tumors or providing detailed insights into the TME. Additionally, the current US contrast agents mainly enhance signal intensity and cannot help to provide molecular details unless labeled with specific ligands.
Fluorescence imaging has gained attention for its applications in intraoperative guidance and preclinical studies [14,15]. It offers non-radiative, real-time visualization of specific molecular targets, significantly enhancing surgical precision. However, its limited tissue penetration and susceptibility to autofluorescence restrict its use to superficial or near-surface applications.
Each imaging technique offers specific advantages tailored to distinct clinical needs (listed in Table 1), and their combination enhances the overall effectiveness of tumor imaging by addressing individual limitations. These technologies are invaluable for early disease detection, disease progression monitoring, therapeutic response assessment, and deepening our understanding of TME interactions at cellular and molecular levels.
The potential clinical impact of these imaging technologies goes beyond diagnostics. Their ability to provide detailed, patient-specific information about TME composition and dynamics plays a crucial role in the era of personalized medicine. By identifying specific molecular and cellular characteristics of the tumors, these techniques facilitate tailored treatment selection, such as choosing patients eligible for immunotherapies, anti-angiogenic agents, or tumor stromal-targeting drugs. Additionally, molecular imaging offers real-time monitoring of therapeutic responses, allowing for adjustments to treatment plans at early time points and ultimately improving patient outcomes.
Specialized imaging probes further enhance the utility of these modalities. Probes targeting key TME components, such as immune cell markers or ECM elements, provide deeper insights into tumor biology and therapeutic resistance. Their application enables precise disease characterization, promoting a shift toward predictive and adaptive cancer management.
In summary, integrating advanced molecular imaging technologies into clinical practice has transformative potential for cancer care. By bridging the gap between tumor biology and treatment, these tools offer a comprehensive approach to diagnosing, monitoring, and personalizing therapy, ultimately leading to improved patient outcomes in oncology. This review summarizes recent advancements in designing innovative probes that target the TME for cancer diagnostics and treatment.

2. Imaging the Extracellular Matrix

The extracellular matrix (ECM) is a complex, non-cellular network composed of collagens, proteoglycans, elastin, fibronectin, oxidases, and proteases [16]. The ECM offers several essential physical functions [17], including structural support, maintaining tissue integrity, water retention, mechanical resilience, and protection barriers. Beyond these functions, ECM remodeling plays a critical role in tissue repair [18], regulating cell behavior [19], tissue homeostasis [20], and signal transduction activation [21].
ECM remodeling is a multi-step process involving (1) ECM deposition and modification, which adjusts the biochemical and mechanical properties of ECM by altering its composition; (2) ECM degradation, mediated by proteolytic enzymes such as matrix metalloproteinases (MMPs), leading to the release of ECM fragments and ECM-bound cytokines and ultimately facilitating cell migration; (3) mechanics-mediated ECM remodeling, where integrin-ECM interactions induce deformation and alignment of ECM fibers, thereby modulating the overall ECM structure [16,22].
Cancer cells strategically manipulate ECM remodeling to create a more supportive TME at various stages of tumor progression [20]. This remodeling involves several processes: the differentiation of cancer-associated fibroblasts (CAFs) through stromal cell activation [23], crosslinking CAFs by ECM-modifying enzymes to increase matrix stiffness [24], and the alignment of collagen fibers to establish physical barriers that prevent cytotoxic immune cell infiltration [25,26]. Additionally, ECM-bound cytokines released during ECM remodeling further stimulate angiogenesis and support tumor outgrowth [27]. Both basement membrane degradation and integrin-mediated mechanical forces are critical drivers of cancer invasion and distal metastasis [27,28].
Beyond establishing the tumor niche, ECM remodeling also contributes to treatment resistance [29,30]. Developing ECM-targeting imaging probes can significantly improve cancer diagnosis, guide treatment selection, and predict metastatic potential. This section focuses on molecular imaging probes targeting specific ECM components, including collagen, elastin, and fibronectin.

2.1. Collagen

Collagen is the most abundant ECM component in tissues [31], and its abnormal accumulation is associated with increased cancer risk [32]. As a result, collagen has emerged as a potential prognostic marker for cancer patients [33], and collagen-targeted therapies may help slow tumor progression and prevent metastasis.
Desogere et al. developed a series of collagen-binding probes (CBPs) for imaging pulmonary fibrosis [34,35]. Among all the CBPs, [68Ga]Ga-CBP8 was later used to visualize collagen deposition in pancreatic ductal adenocarcinoma (PDAC) patients undergoing FOLFIRINOX treatment using PET/MRI. In PDAC-bearing mice, PET revealed a strong positive correlation between tumor collagen deposition and [68Ga]Ga-CBP8 uptake, mirroring the results detected in a PDAC patient with FOLFIRINOX-induced collagen accumulation. These results suggest the potential of [68Ga]Ga-CBP8 for monitoring chemotherapy response non-invasively [36].
The same group also developed EP-3533, a gadolinium (Gd)-labeled collagen-detecting probe for MRI [37]. Enhanced MRI signals indicating collagen deposition in LNCaP prostate tumors were validated through Picrosirius Red staining at the histological level [38]. Additionally, Guo et al. introduced a collagen-responsive indocyanine green (ICG) probe, the collagen-adhesive probe (CA-P), for detecting collagen distribution in bladder cancer via a sol–gel transformation triggered by the interactions between CA-P and collagen [39]. The probe utilizes aldehyde groups on a hydrogel to form a stable gel with amine-rich regions in collagen through a Schiff base reaction [40]. CA-P effectively helped delineate tumor boundaries and increase surgical precision [34]. These imaging probes demonstrate the potential of targeting collagen for cancer diagnosis, treatment monitoring, and image-guided surgery, underscoring its value in advancing precision oncology.

2.2. Elastin

Elastin provides essential elasticity and resilience to various tissues such as skin, lungs, and blood vessels [41]. Recent studies reveal that elevated elastin and collagen fiber accumulation correlates with increased incidence and poorer prognosis in cancers such as hepatocellular carcinoma and gastric cancer [42,43].
Keller et al. developed a Gd-labeled elastin-specific molecular agent (Gd-ESMA), a low-molecular-weight MRI contrast agent, to visualize the TME in VX2 HCC-bearing rabbits. Compared to a conventional Gd-based MRI contrast agent gadobutrol, elastin-specific ESMA resulted in significantly stronger signal enhancements in the tumor center, tumor margins, and peritumoral regions [44]. Gd-ESMA has also been applied to monitor the dynamic changes in elastin deposition in VX2 tumor-bearing rabbits following radiofrequency ablation, offering insight into TME remodeling post-treatment [45]. Additionally, in PC3 prostate cancer-bearing mice, higher Gd-ESMA signals were detected with smaller tumors, compared to the ones with larger tumors, indicating more elastin deposition within tumors and possibly increased metastatic potential [46].
A radiolabeled elastin-targeting probe was also developed, using elastin-like polypeptides labeled with iodine-131. This theranostic agent leverages the γ-ray and β-particles emitted by iodine-131, combining SPECT imaging for elastin-specific visualization with 131I-mediated brachytherapy that significantly inhibits tumor growth in VX2 tumor-bearing rabbits [47].
Since elastin deposition influences TME remodeling and correlates to distal metastasis, elastin-targeting probes offer a promising cancer imaging and treatment approach. These probes enhance our understanding of tumor biology, facilitate early cancer detection, enable non-invasive monitoring of tumor progression, and have potential applications in theranostics for precision oncology through targeting elastin.

2.3. Fibronectin

Fibronectin is a glycoprotein vital for regulating cell adhesion, wound healing, and tissue repair and providing a scaffold for cell attachment and migration [48]. The above physiological processes are crucial for cell growth, differentiation, and survival. However, abnormal fibronectin production and reorganization are frequently observed in cancer, where they are closely linked to tumor progression, invasion, and metastasis [49,50]. Changes in fibronectin play a critical role in establishing the “pre-metastatic niche”, a microenvironment that enhances colonization and survival of circulating tumor cells, thereby promoting distant metastasis [51]. Fibronectin alterations also contribute to angiogenesis, facilitating new blood vessel formation that nourishes cancer cells and sustains their growth [52]. Accordingly, fibrin–fibronectin complexes have emerged as important imaging biomarkers [53], enabling precise cancer detection with specific imaging probes.
Zhou et al. developed a tumor-homing pentapeptide, Cys-Arg-Glu-Lys-Ala (CREKA), which targets fibrin–fibronectin complexes in the TME. By labeling it with gadolinium, they synthesized CREKA-Tris(Gd-DOTA)3 for MRI. Compared to the conventional MRI agent Prohance™, CREKA-Tris(Gd-DOTA)3 doubled the contrast-to-noise ratio and maintained tumor signal visibility for at least 30 min [54]. The detection limit of the probe was further tested in a 4T1 breast cancer mouse model of spontaneous metastasis. CREKA-Tris(Gd-DOTA)3; successfully identified bone micrometastases smaller than 0.5 mm in diameter, with a sensitivity of 79% for tumors as small as 0.125 mm3 [55].
Yang et al. modified the CREKA probe by conjugating it with a near-infrared fluorophore (Cy7), creating a dual-contrast agent, CREKA-Cy7(Gd-DOTA)3. This modified probe improved the visualization of HGC-27 gastric cancers in vivo, providing enhanced sensitivity and reduced imaging time [56]. Additional modifications to fibronectin-targeting probes have further increased tumor specificity and precision. For example, Cheng et al. developed an MMP-activatable probe, CREKA-GK8-QC, which combined fibronectin specificity with MMP-9 responsiveness. This probe demonstrated superior tumor retention and specificity compared to its non-fibronectin-targeting counterparts, enabling better identification of microscopic cancer lesions in 4T1 tumor-bearing models [53].
Fibronectin-targeting probes have also been engineered to focus on the extra domain B (ED-B) fibronectin variant, which is selectively expressed during angiogenesis and tumorigenesis [57,58]. The ED-B variant accumulates around neovascular structures in aggressive tumors and remodeling tissues, making it a conserved and highly specific target. Han et al. synthesized ZD2, an ED-B-binding peptide, for near-infrared fluorescence (NIRF) imaging [59]. To overcome the limited tissue penetration of NIRF imaging, Ye et al. radiolabeled ZD2 with technetium-99m, creating [99mTc]Tc-HYNIC-ZD2 for SPECT, thereby enhancing its clinical applicability [60].
In addition to peptides, recombinant antibody fragments targeting ED-B have been demonstrated to have promising applications in clinical trials. Radretumab (L19SIP), an ED-B-specific antibody fragment, has been successfully used for both imaging and radioimmunotherapy in patients with lung, colorectal, or brain cancer. Using [123I]I-L19(scFv)2/SPECT imaging, liver metastases from colorectal cancer and small-cell lung carcinoma with mammary involvement were effectively visualized [61]. Furthermore, 131I-L19SIP has been applied as a radioimmunotherapy agent, while [124I]I-L19SIP has been utilized for high-resolution PET imaging and accurate dosimetry, particularly for red bone marrow [62]. Tijink et al. reported that [131I]I-L19SIP exhibited significantly lower accumulation in the liver, spleen, and kidneys compared to 177Lu-labeled L19SIP [58]. Furthermore, [131I]I-L19SIP demonstrated superior stability, whereas [79Br]Br-L19SIP faced challenges due to debromination, compromising its radiochemical purity. This instability increases the risk of off-target activity accumulation in non-target organs during in vivo applications [63,64].
Fibronectin-targeting probes, including CREKA-based and ED-B-specific agents, hold significant potential in advancing cancer diagnostics and treatment. These imaging probes have demonstrated exceptional precision in tumor detection, intraoperative guidance, and theranostic applications, underscoring their transformative role in enhancing cancer management.

3. Molecular Imaging of Stromal Cells

In addition to the ECM and fibronectin, the TME encompasses various stromal cell types recruited from neighboring non-cancerous tissues or derived through the transformation of cancer cells themselves [65]. These stromal cells secrete diverse signaling molecules and exosomes, which play crucial roles in triggering angiogenesis and modulating immune responses—key processes in tumor initiation, progression, and drug resistance [66]. As highlighted, stromal cells, including cancer-associated fibroblasts (CAFs), tumor-endothelial cells, and immune cells [67,68], actively remodel the ECM, and influence critical aspects of tumor biology such as metabolism, growth, metastasis, and immune evasion. Their continuous and dynamic interaction with the ECM not only sustains tumor progression but also enhances therapeutic resistance. Given their vital role in tumorigenesis, tumor-associated stromal cells have emerged as promising therapeutic targets, offering novel avenues to disrupt tumor development and progression while potentially overcoming resistance mechanisms.

3.1. Cancer-Associated Fibroblasts

Cancer-associated fibroblasts (CAFs) are the most abundant stromal cells within the TME, typically activated from resident fibroblasts in response to stimuli such as TGF-β [69], platelet-derived growth factor (PDGF) [70], and fibroblast growth factor-2 (FGF-2) [71,72]. Unlike normal fibroblasts, CAFs exhibit dysregulated signaling pathways and altered protein expression profiles [73], enabling them to reshape the TME architecture, hinder drug diffusion, and thereby reduce treatment efficacy [74]. Activated CAFs are defined by the upregulation of specific markers, including α-SMA [75], fibroblast-specific protein 1 (FSP1) [76], and fibroblast activation protein (FAP) [77,78]. These markers not only distinguish CAFs from normal fibroblasts but also highlight their role in driving tumor progression and resistance to therapy, making them promising targets for cancer diagnosis and treatment.
CAFs overexpressing FAP are frequently found within the tumor stroma across various cancer types [77,78]. In contrast, FAP expression is minimal in normal tissues, making it an ideal target for cancer diagnosis and therapy. Due to its enzymatic activity, FAP can cleave specific peptides, enabling the design of innovative imaging agents.
Wu et al. developed a FAP-responsive probe featuring a valine–proline dipeptide conjugated with a cyanine-derived NIR dye, exhibiting excellent FAP-targeting capability. This probe effectively captures FAP-expressing CAFs in LX-2 hepatic stellate cells—the primary source of CAFs in the liver—as well as in HCC-bearing mice. Upon FAP-mediated cleavage, the imaging probe is activated and emits strong NIR signals, demonstrating its potential for both in vitro and in vivo FAP imaging. Notably, FAP-expressing CAFs contribute to immune evasion by promoting reactive oxygen species (ROS) generation and upregulating PD-L1 expression, further underscoring their role in tumor progression and therapeutic resistance [79].
Due to the preferential expression of FAP in CAFs and their scarcity in normal tissues, radiolabeled FAP inhibitors (FAPIs) have emerged as valuable tools for cancer theranostics, particularly in tumors characterized by extensive fibrotic stroma. Several 18F- and 68Ga-labeled FAPI compounds (e.g., FAPI-04, FAPI-46, and FAPI-74) have shown promising diagnostic potential across multiple cancer types when imaging with PET scanners. In a study involving 62 patients with pancreatic adenocarcinoma, which is characterized by significant desmoplastic responses and associated hypometabolism, [18F]FAPI-04 demonstrated superior capabilities in detecting the primary tumor as well as identifying liver, peritoneal, and lymphatic metastases, compared to [18F]FDG [80]. Additionally, in a case involving a 52-year-old woman with advanced pancreatic adenocarcinoma, [68Ga]Ga-FAPI-46 exhibited a lower background signal, a stronger signal in small lesions, and a higher standard uptake value (SUV) compared to [18F]FDG [81].
In lung cancer, compared to [18F]FDG, [18F]FAPI showed a lower tumor-to-background signal in primary lesions, but high signals in lymphatic and bone metastasis [82]. These FAP-targeting probes allow better visualization of tumor boundaries, more accurate assessment of metastatic spread, and potential therapeutic response monitoring. To enhance tumor retention and tumor-to-tissue ratios, a 64Cu-labeled PET imaging probe containing four FAPI-46 motifs attached to diethylene glycol (mini-PEG) spacers was developed. Furthermore, [177Lu]Lu-4P(FAPI)4 showed enhanced tumor suppression compared to [177Lu]-FAPI-46 [83]. The glycopeptide-modified version of FAPI-42, [18F]FAPT, exhibited reduced hepatobiliary uptake and twice the tumor accumulation compared to FAPI-42 in A549-FAP tumor-bearing mice [84].
The 18F- and 68Ga-labeled FAPI probes have been used in a clinic setting, as they can be taken up against multiple cancer types [85,86]. The [68Ga]Ga-FAPI probes exhibit remarkable specificity in identifying liver nodules [87] and PDAC [88], which can be difficult to detect using alternative imaging techniques.
Recently, accumulating evidence has shown that [68Ga]Ga-FAPI may outperform [18F]FDG in revealing lung cancer [89], pancreatic cancer [90], colorectal cancer [91], and nasopharyngeal carcinoma [92]. Furthermore, the high sensitivity and accuracy of [68Ga]Ga-FAPI imaging are advantageous for staging and planning radiotherapy [93,94]. An in-depth review of 18F- and 68Ga-based FAPI PET/CT imaging is also available [95,96].
Another series of FAP-imaging probes have been developed, including PNT6555, PNT6952, and PNT6522, using the amino-terminally blocked D-Ala-boroPro as the targeting moiety. These probes were further radiolabeled with gallium-68, lutetium-177, and terbium-161, and their theranostic potential was verified in HEK-mFAP tumor-bearing mice. Notably, PNT6555, when radiolabeled with actinium-225, demonstrated significant tumor growth inhibition, highlighting the potential of FAP-targeted alpha therapy [97].
Bentivoglio et al. developed a [99mTc]Tc-FGF-2 probe that targets FGFR-2, which is overexpressed on CAFs and TAMs, and tested its efficacy in reticulum cell sarcoma-bearing mice J774A [98]. Additionally, Dai et al. designed a CAF-mimetic aggregation-induced emission (AIE) probe with a CAF-derived cell membrane coating to enhance targeting specificity. The fibronectin present on the CAF membrane enables specific binding to integrin α5 (ITGA5), which is overexpressed in cancer cells. This AIE imaging probe was applied in ovarian cancer surgery, significantly improving lesion detection compared to visual inspection alone, potentially facilitating the optical-guided surgery and increasing precision [99].
CAF-targeted imaging probes, particularly those directed at FAP, have shown remarkable potential in advancing cancer diagnosis and therapy. Radiolabeled FAPIs and FGF-2, along with innovative probes such as CAF-mimetic AIE probes, enable precise visualization of CAFs within the TME. These technologies significantly improve diagnostic accuracy, facilitate response monitoring, and enhance surgical precision by delineating tumor margins more effectively. Given the critical roles of CAFs in tumor progression, immune evasion, and treatment resistance, these advancements highlight the promise of CAF-targeted strategies in revolutionizing cancer imaging and therapeutic interventions.

3.2. Vascular and Lymphoid Networks

Blood and lymphatic vessels, formed by endothelial cells (ECs) [100], are essential for transporting blood, facilitating oxygen exchange, and recycling lymphatic fluid. In the TME, these vascular and lymphatic structures are repurposed to support rapid tumor growth and recruit immunosuppressive cells from surrounding tissues and circulation [27,101]. While these structures supply nutrients and oxygen to sustain tumor progression, they are often chaotic and dysfunctional compared to normal vessels, contributing to treatment resistance [102]. Tumor neovasculature is characterized by leaky vessel walls resulting from disrupted cell junctions and a nearly or entirely absent basement membrane. These structural abnormalities reduce the energy required for cancer cells to traverse the vessels, thereby facilitating migration and metastasis [103]. Such aberrant vascular and lymphatic changes highlight their critical roles in cancer progression, which will be discussed in detail below.

3.2.1. Vascular Vessels

To meet the heightened demand for oxygen and nutrients driven by rapid tumor growth, cancer establishes complex vascular networks through two primary mechanisms: vasculogenesis and angiogenesis [104].
Vasculogenesis involves the formation of new vessels from bone marrow-derived endothelial progenitor cells (EPCs) [105,106], which express specific markers such as CD34, VEGFR1/2, CD133, Tie-2, Nanog, and Oct-4 [27,107]. EPCs are recruited to hypoxic tumor regions in response to growth factors, cytokines, and hypoxia-induced signals. Within the TME, elevated HIF-1α drives VEGF expression, which attracts VEGFR2+ EPCs to tumor sites and promotes vasculogenesis [108]. Additionally, chemotaxis pathways, including VEGF/VEGFR, CCR2/CCL2 [109], and CCR5/CCL5, are also prominent for recruiting EPCs to the TME.
In contrast, angiogenesis involves the sprouting of new vessels from pre-existing capillaries [110], primarily driven by the VEGF/VEGFR pathway. VEGF generated by cancer cells binds to VEGFR expressed on nearby ECs, activating enzymes such as elastase, MMPs, and cathepsin G. These proteases mediate ECM remodeling, facilitating EC migration, sprouting, and proliferation, ultimately leading to the formation of new blood vessels. Intriguingly, sometimes aggressive cancer cells can also form vessel-like structures, referred to as vascular mimicry [111], which are not derived from ECs but still provide an alternative nutrient supply by mimicking blood vessels.
Tumor vasculature is usually irregular, dysfunctional, and highly permeable compared to normal vessels. This vascular leakage leads to waste accumulation, increased tumor acidity, and altered metabolism [112]. Moreover, the degree of neovascularization strongly correlates with the potential for distant metastasis. To detect tumor growth, invasion, and metastasis, imaging probes have been developed which target angiogenic factors that are overexpressed on activated ECs but are limited in normal epithelium. These targets include various VEGF isoforms (A–E) [113], VEGFRs, and integrin αvβ3 [114], offering precise tools for visualizing tumor progression.
KDR (also known as VEGFR2), which exhibits a high affinity for VEGF, is significantly upregulated in cancer cells and activated ECs within the TME, driving proangiogenic processes. Suppressing KDR expression has been shown to inhibit tumor-associated angiogenesis. For instance, strong accumulations of 188Re-labeled VEGF189 peptides were observed in KDR-overexpressing SKOV3 tumors, highlighting its potential for KDR-targeted imaging and therapy [115]. Similarly, VEGF125–136, the first identified inhibitor of VEGFA [116], was labeled with copper-64 for PET imaging, showing efficacy in melanoma-, glioblastoma-, and breast cancer-bearing murine models [117]. Furthermore, the humanized anti-VEGFR2 monoclonal antibody Ramucirumab was radiolabeled with copper-64 for PET imaging, demonstrating its specificity in non-small cell lung cancer (NSCLC) mouse models [118].
Further developments include VEGFR1- and VEGFR2-specific imaging probes, 89Zr-labeled scVR1/Zr and scVR2/Zr, respectively, which allow selective imaging of VEGFR1+ and VEGFR2+ cells at different stages of cancer progression [119]. VEGFR expression is tightly associated with the formation of pre-metastasis niches. For instance, VEGFR1+CD133+ hematopoietic progenitor cells (HPCs) have been implicated in promoting lung metastasis in subcutaneous MDA-MB-435 breast cancer-bearing mice [120]. To image the pre-metastasis niche, an 18F-labeled VEGFR1-specific single-chain VEGF mutant, [18F]AlF-NODA-scVR1 was developed to target VEGFR1+ cells. PET/CT imaging revealed increased lung signals that co-localized with IVIS/CT images. Additionally, autoradiography hot spots demonstrated high VEGFR1 intensity, corroborated by immunofluorescence staining. The study also identified the gradual recruitment of VEGFR1+CD133+ HPCs to the lungs as metastases developed in the orthotopic 4T1 mouse model [121].
Activated ECs also highly expressed integrin αvβ3, indicating that RGD peptides may be potential probes for neovascular imaging [122]. Lui et al. developed an [89Zr]Zr-DFO-heterodimeric peptide, iRGD-PEG3-(lys-[89Zr]Zr-DFO)-PEG3-DA7R, which showed specific targeting of integrin αvβ3 and VEGFR2. Both PET imaging and biodistribution data showed the probe could be retained in tumors for over 3 h [123]. Additionally, a [64Cu]Cu-DOTA-cRGD(D-BPA)K probe was designed for PET imaging and BNCT application [124]. Based on the success of FAPI probes, a dual-targeting PET probe, [68Ga]Ga-FAPI-RGD, was developed. Elevated uptake of [68Ga]Ga-FAPI-RGD was detected in both primary tumors and metastases across various cancer types in patients. Compared to 18F-FDG and [68Ga]Ga-FAPI-46, [68Ga]Ga-FAPI-RGD exhibited higher sensitivity and superior diagnostic accuracy, making it a promising tool for cancer detection and monitoring [125].
The processes of vasculogenesis, angiogenesis, and vascular mimicry in the TME play critical roles in sustaining tumor growth, promoting metastasis, and driving treatment resistance. Advances in imaging probes targeting angiogenic factors like VEGF, VEGFRs, and integrins have significantly enhanced the visualization of dynamic changes within the tumor vasculature. These innovations not only improve cancer detection and monitoring of disease progression but also hold therapeutic potential when coupled with appropriate radioisotopes, enabling cancer theranostics.

3.2.2. Lymphatic Vessels

The lymphatic system, consisting of lymphatic vessels and lymphatic organs [126], maintains fluid balance in the body by draining excess tissue fluids and particulate matter from the bloodstream. Additionally, lymphatic vessels also help immune cells migrate to lymphoid tissues, thereby supporting immune surveillance [127,128]. A higher density of lymphatic vessels known as lymphangiogenesis develops around primary tumors, offering alternative routes for tumor dissemination [129]. This process is closely associated with metastasis and poor prognosis in cancer patients [130,131]. Within TME, lymphatic vessels are often dilated, irregular, and leaky, leading to inefficient lymphatic drainage, impaired drug delivery, and increased metastatic potential.
Radiolabeled [99mTc]Tc-sulfur colloid, [99mTc]Tc-phytate, and [99mTc]Tilmanocept (Lymphoseek®) are routinely used for lymph node scanning in the clinic. These imaging probes, which can operate on the nanoscale, are effective at entering and being retained within lymphatic vessels and provide high-resolution lymphatic imaging [132,133,134,135]. Thorek et al. developed [89Zr]Zr-ferumoxytol, a formulation of iron oxide particles modified with DFO, which enables multimodal PET and MRI of lymph nodes in prostate cancer-bearing mice [136].
Key proteins involved in lymphangiogenesis, such as prospero homeobox 1 (Prox1) [137], podoplanin (PDPN) [138], and lymphatic vessel endothelial hyaluronic acid receptor 1 (LYVE-1) [139], are essential for the formation of lymphatic networks. Prox1 upregulates the expressions of integrin α9 and VEGFR-3 on lymphatic ECs. When activated by its ligands, VEGF-C and VEGF-D, VEGFR3 promotes the maturation of lymphatic vessels [140,141,142]. In the TME, cancer cells express high levels of neuropilin-2, VEGF-A, VEGF-C, and VEGF-D, which stimulate lymphangiogenesis and increase the potential for lymphatic spread [143,144].
Imaging advances have leveraged these lymphatic markers for cancer detection. Kwon et al. injected ICG intradermally to visualize early-stage lymphatic leakage in B16F-10 tumors overexpressing VEGF-C. Their findings suggest that tumor-secreted VEGF-C destabilizes lymphatic vessels, which promotes cell extravasation and lymphatic spread [145]. In addition, another NIRF tracer, P20D680, a poly (ethylene glycol) (PEG)-based lymphatic tracer, has shown potential in evaluating lymphangiogenesis and lymphatic vessel contractility [146,147].
PDPN is essential for the formation of lymphatic vessels and has been found to be elevated in several cancers [126]. Mouse embryos lacking PDPN exhibit a phenotype characterized by the mixing of blood and lymphatic vessels, which results in blood-filled lymphatic vessels and severe edema due to the abnormal separation between blood and lymphatic vessels [148]. Given the significance of PDPN in lymphangiogenesis and tumor development, Kato et al. developed an IR700-conjugated anti-PDPN antibody that is suitable for both fluorescence imaging and near-infrared photoimmunotherapy [149].
LYVE-1, primarily found on lymphatic ECs, is involved in the transport of hyaluronan, a component abundant in the TME [150]. Elevated LYVE-1 expression is associated with lymphangiogenesis and the spread of metastasis to lymph nodes [151,152]. Anti-LYVE-1 immuno-PET, using the [125I]I-anti-LYVE-1 antibody, has shown superior performance to the conventional [18F]FDG/PET scan in detecting metastasis, as it captures lymphangiogenesis more effectively [153].
Although [125I]I-anti-VEGFR-3 antibodies accumulate specifically in lymphatic vessels, their ability to inhibit lymphangiogenesis presents challenges for imagining biological processes without interfering with them [153]. This issue could be addressed by using peptide-based probes that specifically target VEGFR-3, such as [68Ga]Ga-DOTA-TMVP1 and [68Ga]Ga-DOTA-TMVP1448. Both probes have demonstrated excellent selectivity in vivo by targeting tumor-induced metastatic lymph nodes while sparing contralateral lymph nodes. Moreover, [68Ga]Ga-DOTA-TMVP1448 also provides superior imaging resolution compared to [18F]FDG [154,155].
Lymphangiogenesis and lymphatic vessel remodeling are crucial processes that influence cancer metastasis and patient prognosis. Imaging the lymphatic system with probes targeting lymphatic markers, such as PDPN, LYVE-1, and VEGFR-3, offers non-invasive methods to assess lymphatic vessel activity and lymph node metastasis, thus offering valuable insights for predicting therapeutic response.

3.3. Immunosuppressive Cells

Interactions between immune cells and cancer cells play dual roles in oncogenesis and tumor progression [156]. Within the TME, immune cells can adopt pro-tumorigenic or anti-tumorigenic phenotypes depending on the context [157]. This section highlights recent advances in molecular imaging probes targeting various immune cells in the field of cancer research.
Immune cells within the TME can shift toward a pro-tumorigenic phenotype, fostering a favorable environment for rapid growth and potential immune evasion [157]. Myeloid cells, for instance, are recruited to the TME and may initially mediate anti-tumor inflammation, suppressing tumor growth during early tumorigenesis [158]. However, excessive cytokine release within the TME can induce immature myeloid cells to infiltrate the TME and differentiate into myeloid-derived suppressor cells (MDSCs), which suppress immune responses and promote tumor progression [159]. Hoffmann et al. radiolabeled polymorphonuclear (PMN-) and monocytic (M-) MDSCs with 64Cu-NOTA-anti-CD11b monoclonal antibodies to study their dynamics in vivo using PET imaging in breast cancer and melanoma mouse models. PET imaging revealed that both PMC- and M-MDSCs were recruited to primary tumors and lung metastases in both models, with recruitment strongly correlating with tumor progression [160].
Additionally, Zinnhardt et al. developed an [18F]F-DPA-714 TSPO probe for glioma imaging. This probe revealed the high TSPO expression in HLA-DR+ glioma-associated myeloid cells, including CD45medCD14+++ MDSCs and CD45highCD14++ TAMs. Moreover, more intense DPA-714 signals were detected in grade IV glioblastoma lesions compared to grade II oligodendroglioma, indicating the potential of this probe in staging gliomas [161]. S100A8/A9, an exosomal protein released by MDSCs, interacts with pattern recognition receptors RAGE and TLR4, initiating an autocrine loop that promotes MDSC recruitment in pre-metastatic sites. SPECT/CT imaging demonstrated elevated uptake of 111In-DTPA-αS100A9 in the lungs, spleen, and tumors of mice implanted with the lung-metastatic 4T1.2 breast cancer cells, compared to non-metastatic 67NR cells [162]. The spleen uptake of [111In]In-DTPA-αS100A9 correlated well with elevated Gr-1+CD115+CCR2highCX3CR1low monocytes, which release S100A8/A9 in response to CCL2 secreted by metastatic cancer cells. Interestingly, an increased [111In]In-DTPA-αS100A9 signal was spotted in the lungs 10 days after tumor implantation, whereas tumor signals in the lungs were only detectable on day 20. This finding underscores the potential of 111In-DTPA-αS100A9 as a potential imaging probe for detecting pre-metastatic niches.
MDSCs play a pivotal role in immune evasion by suppressing T cell and NK cell functions [163] and promoting the differentiation of regulatory T cells (Tregs) through the TGF-β and IL-10 pathways [164]. Tregs, a specialized subset of CD4+ T cells, are essential for maintaining self-tolerance and regulating immune responses [165]. Compared to natural Tregs (nTregs) originating from the thymus, induced Tregs (iTregs) are differentiated in the TME in response to high levels of TGF-β and IL-10 [166,167].
Early imaging strategies for Tregs included the transduction of murine Tregs with the human sodium/iodide symporter (NIS) and labeling with 99mTcO4 for in vivo visualization [168]. Jacob et al. differentiated Tregs from peripheral blood mononuclear cells derived from healthy volunteers and then labeled them with [89Zr]Zr-oxine. Their findings demonstrated a 4-fold higher uptake of [89Zr]Zr-oxine in Tregs cultured in Hank’s Balanced Salt Solution compared to standard X-Vivo15 medium [169]. Additionally, [89Zr]Zr-Df-Bz-NCS-anti-CD3 mAb has been used to visualize human T cell expansion in a graft-versus-host disease mouse model receiving human peripheral blood mononuclear cell transfusion. Notably, the signal correlated with T cell numbers and decreased in groups receiving syngeneic Treg transfers [170]. While these imaging probes have been validated in transplantation models, their application in tumor-bearing models remains underexplored, leaving a gap in Treg-targeted imaging in oncology.
Macrophages, differentiated from monocytes, exhibit remarkable plasticity and can be polarized into classically activated M1 macrophages (tumor killing and defense roles) or alternatively activated M2 macrophages (wound healing, tissue remodeling, and tumor progression) based on the cytokine milieu [171]. Tumor-associated macrophages (TAMs), predominantly classified under the M2 phenotype, represent the primary macrophage population in many cancers and are pivotal in tumor progression [172]. TAMs promote angiogenesis by secreting VEGF [173], induce Treg differentiation through IL-10 and TGF-β secretion [171], and are recruited to tumor sites by tumor-derived CSF-1 [174]. Their activity often compromises the efficacy of cancer therapies. Consequently, imaging TAMs within the TME could significantly aid in the development of new therapeutics.
The macrophage mannose receptor (MMR, or CD206) is a key M2 marker and is primarily expressed on MHC IIlow CD11b+ cells, especially TAMs. CD206 is a reliable target for in vivo identification of M2-like TAMs [175]. MMR-positive macrophages are immunosuppressive and angiogenic, contributing to tumor progression and correlating with poor prognosis in cancer patients [171,176]. To leverage CD206 as a molecular imaging target, an MMR-specific nanobody, [99mTc]Tc-αMMR Nb, was developed using an immunized alpaca. This probe demonstrated high specificity for MHC IIlow TAMs in wild-type 3LL-R lung carcinoma-bearing mice. Notably, tumor uptake of [99mTc]Tc-αMMR Nb was significantly reduced in CCR2-deficient mice, which exhibit impaired TAM infiltration, confirming the specificity of the probe and dependence on TAM presence [175]. However, high liver and spleen accumulation of [99mTc]Tc-αMMR Nb prompted the development of a bivalent αMMR-αMMR Nb, which is preferentially blocked in the liver and spleen. Administering a 20-fold excess of bivalent Nbs mixed with [99mTc]Tc-αMMR Nbs effectively reduced liver and spleen uptake, improving the tumor-to-tissue ratio.
To advance TAM-targeting imaging tools for clinical translation, Blykers et al. developed camelid single-domain antibody fragments (sdAbs) with dual cross-reactivity to mouse and human MMR. The anti-MMR 3.49 sdAb was radiolabeled to create both [99mTc]Tc-αMMR 3.49 and [18F]FB-αMMR 3.49 tracers, which were evaluated in wild-type C57BL/6, MMR-deficient, and CCR2-deficient mice inoculated with 3LL-R Lewis lung carcinoma cells [177]. Both nanobodies demonstrated comparable tumor-targeting efficiency, achieving approximately 2.4% injected radioactivity (IA)/g in tumor tissue. However, [99mTc]Tc-αMMR 3.49 exhibited 20-fold higher kidney accumulation compared to [18F]FB-αMMR 3.49 at the 3 h time point in 3LL-R lung carcinoma-bearing mice. This increased kidney accumulation is attributed to the lysosomal trapping of radiometal-labeled compounds in renal cells, a limitation not observed with fluorinated tracers [178]. Moreover, PET imaging with fluorine-18 offers superior sensitivity and spatial resolution compared to SPECT imaging with technetium-99m, positioning [18F]FB-αMMR 3.49 as a more suitable candidate for clinical applications. These findings highlight the potential of fluorine-based nanobody tracers for enhanced tumor imaging and improved clinical utility.
CD163 is another marker selectively found on the surface of the M2-like macrophages. A peptide-based probe, [18F]AlF-NODA-MP-C6-CTHRSSVVC, successfully targeted CD163-positive TAMs in cyclophosphamide-treated CT26 tumor-bearing mice, demonstrating its specificity [179]. Furthermore, the [64Cu]Cu-ICT-01 probe, initially developed for atherosclerosis imaging, exhibited high specificity for CD163 and holds potential for future applications in cancer imaging [180].
Inspired by the interaction of macrophages with high-density lipoprotein (HDL), two 89Zr-labeled reconstituted HDL (rHDL) nanotracers—[89Zr]Zr-apoA-I-labeled rHDL ([89Zr]Zr-AL-rHDL) and [89Zr]Zr-phospholipid-labeled rHDL ([89Zr]Zr-PL-rHDL)—were designed to image TAMs in orthotopic 4T1 breast cancer mouse models. Both tracers demonstrated similar tumor accumulation at 48 h and co-localization with Iba-1 signals at 24 h, as confirmed by ex vivo autoradiography [181]. Despite differing biodistribution profiles—[89Zr]Zr-AL-rHDL had prolonged blood circulation and higher kidney uptake, while [89Zr]Zr-PL-rHDL showed greater bone retention—flow cytometry analyses revealed that both tracers were preferentially taken up by TAMs over other immune cells such as dendritic cells, T cells, and monocytes. This underscores their potential utility in clinical TAM imaging.
Immunosuppressive cells within the TME, including MDSCs, Tregs, and TAMs, play pivotal roles in promoting immune evasion. Advances in molecular imaging probes targeting these cells have facilitated non-invasive visualization of immune cell dynamics within the TME. Probes for MDSC markers, Treg-specific molecules, and TAM markers such as CD206 and CD163 have shown encouraging results in preclinical studies. These imaging tools enhance our understanding of tumor–immune interactions and offer methods to monitor therapeutic responses. The simultaneous use of multiple probes to target diverse immune cell populations could offer a comprehensive view of the immunosuppressive landscape, paving the way for improved cancer diagnostics and precision immunotherapy.

4. Conclusions

The tumor microenvironment (TME) represents a complex and dynamic ecosystem that plays a crucial role in cancer progression, metastasis, and resistance to therapy. Comprised of cancer cells and diverse stromal components—including the extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), vascular and lymphatic networks, and immunosuppressive immune cells—the TME orchestrates a supportive niche for tumor development.
Recent advancements in molecular imaging of the TME have greatly enhanced our understanding of tumor biology (Figure 2). These imaging techniques allow for non-invasive assessments of ECM remodeling, stromal cell activity, vascular and lymphatic dynamics, and interactions among immune cells. In addition to imaging, radiopharmaceutical therapies using isotopes such as Lutetium-177, copper-67, lead-212, bismuth-212, actinium-225 are emerging as promising strategies to target the TME and cancer cells with high specificity to eradicate cancer cells and prevent metastasis potentially. These therapies specifically deliver radiation doses to tumors while minimizing off-target effects [182,183,184]. By combining imaging and therapeutic capabilities, these isotopes enable theranostic approaches that integrate diagnosis, treatment, and monitoring into a single platform.
However, the clinical translation of novel imaging probes faces several regulatory challenges. These include stringent requirements for demonstrating safety, efficacy, and reproducibility in human studies, as mandated by regulatory agencies such as the FDA and EMA. The high costs and lengthy timelines of preclinical and clinical trials complicate drug development. A key challenge is the need for standardized probe production and quality control, especially for probes targeting the complex and often poorly understood components of the tumor TME. Additionally, integrating these probes into clinical workflows may require updated imaging hardware, software, and personnel training, posing logistical challenges for widespread adoption.
Despite these hurdles, integrating artificial intelligence (AI) may further increase detection specificity and sensitivity, reducing the time for imaging processing and filing reports, and increasing the throughput. Additionally, more information can be extracted from the obtained images using radiomics, a technique that quantifies imaging features to reveal patterns not visible to the human eye. This method correlates features with multi-omics data.
Radiomics provides a non-invasive way to assess tumor heterogeneity, offering biomarkers for disease characterization, prognosis prediction, and treatment response evaluation. When combined with advanced molecular imaging, these technologies can provide unprecedented insights into tumor progression and response to therapy. The continuous refinement and clinical application of imaging probes can transform cancer management by enhancing diagnostic accuracy and allowing for better monitoring of treatment responses. Additionally, AI techniques provide priceless support in expediting regulatory approval by improving the interpretability of imaging data and aiding in validating robust biomarkers.
Future efforts should address regulatory, technical, and accessibility barriers through interdisciplinary collaboration and public–private partnerships. Clearer regulatory and approval processes, along with global standardization, are essential for fully realizing the clinical potential of novel imaging probes. These developments enhance diagnostic accuracy and allow for more effective monitoring of treatment responses, ultimately leading to improved patient outcomes.

Author Contributions

H.-W.C. wrote the manuscript draft and drew the figures; D.-Y.K. acquired the grant and revised the manuscript; P.-W.S. revised the manuscript; H.-Y.C. proposed the idea of the manuscript, acquired the grants, and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grant No. 112DN19 from the Far Eastern Memorial Hospital-National Yang-Ming University Joint Research Program (D.-Y. Kuo and H.-Y. Chuang). This study was supported by grant NSTC112-2314-B-A498-066 from the National Science and Technology Council, TAIWAN (H.-Y. Chuang).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef] [PubMed]
  2. Langley, R.R.; Fidler, I.J. The seed and soil hypothesis revisited--the role of tumor-stroma interactions in metastasis to different organs. Int. J. Cancer 2011, 128, 2527–2535. [Google Scholar] [CrossRef]
  3. Fidler, I.J.; Poste, G. The “seed and soil” hypothesis revisited. Lancet Oncol. 2008, 9, 808. [Google Scholar] [CrossRef]
  4. Neophytou, C.M.; Panagi, M.; Stylianopoulos, T.; Papageorgis, P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers 2021, 13, 2053. [Google Scholar] [CrossRef]
  5. Xiao, Y.; Yu, D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol. Ther. 2021, 221, 107753. [Google Scholar] [CrossRef] [PubMed]
  6. Du, W.; Xia, X.; Hu, F.; Yu, J. Extracellular matrix remodeling in the tumor immunity. Front. Immunol. 2023, 14, 1340634. [Google Scholar] [CrossRef] [PubMed]
  7. Reese, K.L.; Pantel, K.; Smit, D.J. Multibiomarker panels in liquid biopsy for early detection of pancreatic cancer—A comprehensive review. J. Exp. Clin. Cancer Res. 2024, 43, 250. [Google Scholar] [CrossRef]
  8. Yao, S.; Han, Y.; Yang, M.; Jin, K.; Lan, H. Integration of liquid biopsy and immunotherapy: Opening a new era in colorectal cancer treatment. Front. Immunol. 2023, 14, 1292861. [Google Scholar] [CrossRef]
  9. Zhang, K.; Xu, H.; Li, K. Molecular Imaging for Early-Stage Disease Diagnosis. Adv. Exp. Med. Biol. 2023, 1199, 39–58. [Google Scholar] [CrossRef]
  10. Bai, J.W.; Qiu, S.Q.; Zhang, G.J. Molecular and functional imaging in cancer-targeted therapy: Current applications and future directions. Signal Transduct. Target. Ther. 2023, 8, 89. [Google Scholar] [CrossRef]
  11. Lilburn, D.M.L.; Groves, A.M. The role of PET in imaging of the tumour microenvironment and response to immunotherapy. Clin. Radiol. 2021, 76, 784.e1–784.e15. [Google Scholar] [CrossRef] [PubMed]
  12. Kiraga, L.; Kucharzewska, P.; Paisey, S.; Cheda, L.; Domanska, A.; Rogulski, Z.; Rygiel, T.P.; Boffi, A.; Król, M. Nuclear imaging for immune cell tracking. Comparison of various cell labeling methods and their application. Coordin. Chem. Rev. 2021, 445, 214008. [Google Scholar] [CrossRef]
  13. Hoffmann, E.; Masthoff, M.; Kunz, W.G.; Seidensticker, M.; Bobe, S.; Gerwing, M.; Berdel, W.E.; Schliemann, C.; Faber, C.; Wildgruber, M. Multiparametric MRI for characterization of the tumour microenvironment. Nat. Rev. Clin. Oncol. 2024, 21, 428–448. [Google Scholar] [CrossRef]
  14. Smith, B.L.; Hunt, K.K.; Carr, D.; Blumencranz, P.W.; Hwang, E.S.; Gadd, M.A.; Stone, K.; Dyess, D.L.; Dodge, D.; Valente, S.; et al. Intraoperative Fluorescence Guidance for Breast Cancer Lumpectomy Surgery. NEJM Evid. 2023, 2, EVIDoa2200333. [Google Scholar] [CrossRef]
  15. Huynh, A.S.; Cohen, A.S.; Doligalski, M.; Casagni, T.J.; Moberg, V.E.; Huang, X.; Morse, J.; Abrahams, D.; Lloyd, M.C.; Centeno, B.A.; et al. Intraoperative Guidance of Pancreatic Cancer Resection Using a Toll-like Receptor 2-Targeted Fluorescence Molecular Imaging Agent. Cancer Res. Commun. 2024, 4, 2877–2887. [Google Scholar] [CrossRef]
  16. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef] [PubMed]
  17. Muiznieks, L.D.; Keeley, F.W. Molecular assembly and mechanical properties of the extracellular matrix: A fibrous protein perspective. Biochim. Biophys. Acta 2013, 1832, 866–875. [Google Scholar] [CrossRef]
  18. Fattahi, R.; Mohebichamkhorami, F.; Taghipour, N.; Keshel, S.H. The effect of extracellular matrix remodeling on material-based strategies for bone regeneration: Review article. Tissue Cell 2022, 76, 101748. [Google Scholar] [CrossRef] [PubMed]
  19. Gattazzo, F.; Urciuolo, A.; Bonaldo, P. Extracellular matrix: A dynamic microenvironment for stem cell niche. Biochim. Biophys. Acta 2014, 1840, 2506–2519. [Google Scholar] [CrossRef]
  20. Yuan, Z.; Li, Y.; Zhang, S.; Wang, X.; Dou, H.; Yu, X.; Zhang, Z.; Yang, S.; Xiao, M. Extracellular matrix remodeling in tumor progression and immune escape: From mechanisms to treatments. Mol. Cancer 2023, 22, 48. [Google Scholar] [CrossRef]
  21. Hastings, J.F.; Skhinas, J.N.; Fey, D.; Croucher, D.R.; Cox, T.R. The extracellular matrix as a key regulator of intracellular signalling networks. Br. J. Pharmacol. 2019, 176, 82–92. [Google Scholar] [CrossRef] [PubMed]
  22. Tanaka, H.Y.; Nakazawa, T.; Enomoto, A.; Masamune, A.; Kano, M.R. Therapeutic Strategies to Overcome Fibrotic Barriers to Nanomedicine in the Pancreatic Tumor Microenvironment. Cancers 2023, 15, 724. [Google Scholar] [CrossRef] [PubMed]
  23. Belhabib, I.; Zaghdoudi, S.; Lac, C.; Bousquet, C.; Jean, C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers 2021, 13, 3466. [Google Scholar] [CrossRef] [PubMed]
  24. Mai, Z.; Lin, Y.; Lin, P.; Zhao, X.; Cui, L. Modulating extracellular matrix stiffness: A strategic approach to boost cancer immunotherapy. Cell Death Dis. 2024, 15, 307. [Google Scholar] [CrossRef]
  25. Szulczewski, J.M.; Inman, D.R.; Proestaki, M.; Notbohm, J.; Burkel, B.M.; Ponik, S.M. Directional cues in the tumor microenvironment due to cell contraction against aligned collagen fibers. Acta Biomater. 2021, 129, 96–109. [Google Scholar] [CrossRef]
  26. Zhang, X.; Zhao, Y.; Chen, X. Collagen extracellular matrix promotes gastric cancer immune evasion by activating IL4I1-AHR signaling. Transl. Oncol. 2024, 49, 102113. [Google Scholar] [CrossRef]
  27. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef]
  28. Kai, F.; Drain, A.P.; Weaver, V.M. The Extracellular Matrix Modulates the Metastatic Journey. Dev. Cell 2019, 49, 332–346. [Google Scholar] [CrossRef]
  29. Sethi, T.; Rintoul, R.C.; Moore, S.M.; MacKinnon, A.C.; Salter, D.; Choo, C.; Chilvers, E.R.; Dransfield, I.; Donnelly, S.C.; Strieter, R.; et al. Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: A mechanism for small cell lung cancer growth and drug resistance in vivo. Nat. Med. 1999, 5, 662–668. [Google Scholar] [CrossRef]
  30. Sherman-Baust, C.A.; Weeraratna, A.T.; Rangel, L.B.; Pizer, E.S.; Cho, K.R.; Schwartz, D.R.; Shock, T.; Morin, P.J. Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells. Cancer Cell 2003, 3, 377–386. [Google Scholar] [CrossRef]
  31. Frantz, C.; Stewart, K.M.; Weaver, V.M. The extracellular matrix at a glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef] [PubMed]
  32. Ding, D.Y.; Jiang, S.Y.; Zu, Y.X.; Yang, Y.; Gan, X.J.; Yuan, S.X.; Zhou, W.P. Collagen in hepatocellular carcinoma: A novel biomarker and therapeutic target. Hepatol. Commun. 2024, 8, e0489. [Google Scholar] [CrossRef] [PubMed]
  33. De Martino, D.; Bravo-Cordero, J.J. Collagens in Cancer: Structural Regulators and Guardians of Cancer Progression. Cancer Res. 2023, 83, 1386–1392. [Google Scholar] [CrossRef] [PubMed]
  34. Desogere, P.; Tapias, L.F.; Rietz, T.A.; Rotile, N.; Blasi, F.; Day, H.; Elliott, J.; Fuchs, B.C.; Lanuti, M.; Caravan, P. Optimization of a Collagen-Targeted PET Probe for Molecular Imaging of Pulmonary Fibrosis. J. Nucl. Med. 2017, 58, 1991–1996. [Google Scholar] [CrossRef] [PubMed]
  35. Desogere, P.; Tapias, L.F.; Hariri, L.P.; Rotile, N.J.; Rietz, T.A.; Probst, C.K.; Blasi, F.; Day, H.; Mino-Kenudson, M.; Weinreb, P.; et al. Type I collagen-targeted PET probe for pulmonary fibrosis detection and staging in preclinical models. Sci. Transl. Med. 2017, 9, eaaf4696. [Google Scholar] [CrossRef]
  36. Esfahani, S.A.; Ma, H.; Krishna, S.; Shuvaev, S.; Sabbagh, M.; Deffler, C.; Rotile, N.; Weigand-Whittier, J.; Zhou, I.Y.; Catana, C.; et al. Collagen type I PET/MRI enables evaluation of treatment response in pancreatic cancer in pre-clinical and first-in-human translational studies. Theranostics 2024, 14, 5745–5761. [Google Scholar] [CrossRef]
  37. Caravan, P.; Das, B.; Dumas, S.; Epstein, F.H.; Helm, P.A.; Jacques, V.; Koerner, S.; Kolodziej, A.; Shen, L.; Sun, W.C.; et al. Collagen-targeted MRI contrast agent for molecular imaging of fibrosis. Angew. Chem. Int. Ed. Engl. 2007, 46, 8171–8173. [Google Scholar] [CrossRef]
  38. Kader, A.; Kaufmann, J.O.; Mangarova, D.B.; Moeckel, J.; Adams, L.C.; Brangsch, J.; Heyl, J.L.; Zhao, J.; Verlemann, C.; Karst, U.; et al. Collagen-Specific Molecular Magnetic Resonance Imaging of Prostate Cancer. Int. J. Mol. Sci. 2022, 24, 711. [Google Scholar] [CrossRef]
  39. Guo, P.; Qi, A.; Shang, W.; Cai, Z.; Hu, S.; Dai, P.; Chen, Z.; Sun, M.; Wang, Z.; Tong, Z.; et al. Targeting tumour surface collage with hydrogel probe: A new strategy to enhance intraoperative imaging sensitivity and stability of bladder cancer. Eur. J. Nucl. Med. Mol. Imaging 2024, 51, 4165–4176. [Google Scholar] [CrossRef]
  40. Xu, J.; Liu, Y.; Hsu, S.H. Hydrogels Based on Schiff Base Linkages for Biomedical Applications. Molecules 2019, 24, 3005. [Google Scholar] [CrossRef]
  41. Trebacz, H.; Barzycka, A. Mechanical Properties and Functions of Elastin: An Overview. Biomolecules 2023, 13, 574. [Google Scholar] [CrossRef] [PubMed]
  42. Yasui, Y.; Abe, T.; Kurosaki, M.; Higuchi, M.; Komiyama, Y.; Yoshida, T.; Hayashi, T.; Kuwabara, K.; Takaura, K.; Nakakuki, N.; et al. Elastin Fiber Accumulation in Liver Correlates with the Development of Hepatocellular Carcinoma. PLoS ONE 2016, 11, e0154558. [Google Scholar] [CrossRef]
  43. Fang, T.; Zhang, L.; Yin, X.; Wang, Y.; Zhang, X.; Bian, X.; Jiang, X.; Yang, S.; Xue, Y. The prognostic marker elastin correlates with epithelial-mesenchymal transition and vimentin-positive fibroblasts in gastric cancer. J. Pathol. Clin. Res. 2023, 9, 56–72. [Google Scholar] [CrossRef]
  44. Keller, S.; Borde, T.; Brangsch, J.; Reimann, C.; Kader, A.; Schulze, D.; Buchholz, R.; Kaufmann, J.O.; Karst, U.; Schellenberger, E.; et al. Assessment of the hepatic tumor extracellular matrix using elastin-specific molecular magnetic resonance imaging in an experimental rabbit cancer model. Sci. Rep. 2020, 10, 20785. [Google Scholar] [CrossRef]
  45. Collettini, F.; Reimann, C.; Brangsch, J.; Chapiro, J.; Savic, L.J.; Onthank, D.C.; Robinson, S.P.; Karst, U.; Buchholz, R.; Keller, S.; et al. Elastin-specific MRI of extracellular matrix-remodelling following hepatic radiofrequency-ablation in a VX2 liver tumor model. Sci. Rep. 2021, 11, 6814. [Google Scholar] [CrossRef]
  46. Kader, A.; Brangsch, J.; Reimann, C.; Kaufmann, J.O.; Mangarova, D.B.; Moeckel, J.; Adams, L.C.; Zhao, J.; Saatz, J.; Traub, H.; et al. Visualization and Quantification of the Extracellular Matrix in Prostate Cancer Using an Elastin Specific Molecular Probe. Biology 2021, 10, 1217. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, X.; Shen, Y.; Zhang, X.; Lin, R.; Jia, Q.; Chang, Y.; Liu, W.; Liu, W. Brachytherapy Using Elastin-Like Polypeptides with (131)I Inhibit Tumor Growth in Rabbits with VX2 Liver Tumor. Dig. Dis. Sci. 2016, 61, 2921–2927. [Google Scholar] [CrossRef]
  48. To, W.S.; Midwood, K.S. Plasma and cellular fibronectin: Distinct and independent functions during tissue repair. Fibrogenesis Tissue Repair 2011, 4, 21. [Google Scholar] [CrossRef]
  49. Wang, Y.J.; Lin, J.F.; Cheng, L.H.; Chang, W.T.; Kao, Y.H.; Chang, M.M.; Wang, B.J.; Cheng, H.C. Pterostilbene prevents AKT-ERK axis-mediated polymerization of surface fibronectin on suspended lung cancer cells independently of apoptosis and suppresses metastasis. J. Hematol. Oncol. 2017, 10, 72. [Google Scholar] [CrossRef]
  50. Lin, T.C.; Yang, C.H.; Cheng, L.H.; Chang, W.T.; Lin, Y.R.; Cheng, H.C. Fibronectin in Cancer: Friend or Foe. Cells 2019, 9, 27. [Google Scholar] [CrossRef]
  51. Patras, L.; Paul, D.; Matei, I.R. Weaving the nest: Extracellular matrix roles in pre-metastatic niche formation. Front. Oncol. 2023, 13, 1163786. [Google Scholar] [CrossRef]
  52. Zhou, F.; Sun, J.; Ye, L.; Jiang, T.; Li, W.; Su, C.; Ren, S.; Wu, F.; Zhou, C.; Gao, G. Fibronectin promotes tumor angiogenesis and progression of non-small-cell lung cancer by elevating WISP3 expression via FAK/MAPK/HIF-1alpha axis and activating wnt signaling pathway. Exp. Hematol. Oncol. 2023, 12, 61. [Google Scholar] [CrossRef] [PubMed]
  53. Cheng, Z.; Jin, Y.; Li, J.; Shi, G.; Yu, L.; Shao, B.; Tian, J.; Du, Y.; Yuan, Z. Fibronectin-targeting and metalloproteinase-activatable smart imaging probe for fluorescence imaging and image-guided surgery of breast cancer. J. Nanobiotechnol. 2023, 21, 112. [Google Scholar] [CrossRef] [PubMed]
  54. Zhou, Z.; Wu, X.; Kresak, A.; Griswold, M.; Lu, Z.R. Peptide targeted tripod macrocyclic Gd(III) chelates for cancer molecular MRI. Biomaterials 2013, 34, 7683–7693. [Google Scholar] [CrossRef] [PubMed]
  55. Zhou, Z.; Qutaish, M.; Han, Z.; Schur, R.M.; Liu, Y.; Wilson, D.L.; Lu, Z.R. MRI detection of breast cancer micrometastases with a fibronectin-targeting contrast agent. Nat. Commun. 2015, 6, 7984. [Google Scholar] [CrossRef]
  56. Yang, N.; Huang, Y.; Wang, X.; Wang, D.; Yao, D.; Ren, G. Fibronectin-Targeting Dual-Modal MR/NIRF Imaging Contrast Agents for Diagnosis of Gastric Cancer and Peritoneal Metastasis. Bioconjug Chem. 2024, 35, 843–854. [Google Scholar] [CrossRef]
  57. Zollinger, A.J.; Smith, M.L. Fibronectin, the extracellular glue. Matrix Biol. 2017, 60–61, 27–37. [Google Scholar] [CrossRef]
  58. Zardi, L.; Carnemolla, B.; Siri, A.; Petersen, T.E.; Paolella, G.; Sebastio, G.; Baralle, F.E. Transformed human cells produce a new fibronectin isoform by preferential alternative splicing of a previously unobserved exon. EMBO J. 1987, 6, 2337–2342. [Google Scholar] [CrossRef]
  59. Han, Z.; Zhou, Z.; Shi, X.; Wang, J.; Wu, X.; Sun, D.; Chen, Y.; Zhu, H.; Magi-Galluzzi, C.; Lu, Z.R. EDB Fibronectin Specific Peptide for Prostate Cancer Targeting. Bioconjug Chem. 2015, 26, 830–838. [Google Scholar] [CrossRef]
  60. Ye, X.X.; Zhao, Y.Y.; Wang, Q.; Xiao, W.; Zhao, J.; Peng, Y.J.; Cao, D.H.; Lin, W.J.; Si-Tu, M.Y.; Li, M.Z.; et al. EDB Fibronectin-Specific SPECT Probe (99m)Tc-HYNIC-ZD2 for Breast Cancer Detection. ACS Omega 2017, 2, 2459–2468. [Google Scholar] [CrossRef]
  61. Santimaria, M.; Moscatelli, G.; Viale, G.L.; Giovannoni, L.; Neri, G.; Viti, F.; Leprini, A.; Borsi, L.; Castellani, P.; Zardi, L.; et al. Immunoscintigraphic detection of the ED-B domain of fibronectin, a marker of angiogenesis, in patients with cancer. Clin. Cancer Res. 2003, 9, 571–579. [Google Scholar] [PubMed]
  62. Poli, G.L.; Bianchi, C.; Virotta, G.; Bettini, A.; Moretti, R.; Trachsel, E.; Elia, G.; Giovannoni, L.; Neri, D.; Bruno, A. Radretumab radioimmunotherapy in patients with brain metastasis: A 124I-L19SIP dosimetric PET study. Cancer Immunol. Res. 2013, 1, 134–143. [Google Scholar] [CrossRef] [PubMed]
  63. Tijink, B.M.; Neri, D.; Leemans, C.R.; Budde, M.; Dinkelborg, L.M.; Stigter-van Walsum, M.; Zardi, L.; van Dongen, G.A. Radioimmunotherapy of head and neck cancer xenografts using 131I-labeled antibody L19-SIP for selective targeting of tumor vasculature. J. Nucl. Med. 2006, 47, 1127–1135. [Google Scholar]
  64. Rossin, R.; Berndorff, D.; Friebe, M.; Dinkelborg, L.M.; Welch, M.J. Small-animal PET of tumor angiogenesis using a (76)Br-labeled human recombinant antibody fragment to the ED-B domain of fibronectin. J. Nucl. Med. 2007, 48, 1172–1179. [Google Scholar] [CrossRef]
  65. Zhao, Y.; Shen, M.; Wu, L.; Yang, H.; Yao, Y.; Yang, Q.; Du, J.; Liu, L.; Li, Y.; Bai, Y. Stromal cells in the tumor microenvironment: Accomplices of tumor progression? Cell Death Dis. 2023, 14, 587. [Google Scholar] [CrossRef]
  66. Guo, S.; Deng, C.X. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int. J. Biol. Sci. 2018, 14, 2083–2093. [Google Scholar] [CrossRef] [PubMed]
  67. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef]
  68. Yang, D.; Liu, J.; Qian, H.; Zhuang, Q. Cancer-associated fibroblasts: From basic science to anticancer therapy. Exp. Mol. Med. 2023, 55, 1322–1332. [Google Scholar] [CrossRef]
  69. Shi, X.; Young, C.D.; Zhou, H.; Wang, X. Transforming Growth Factor-beta Signaling in Fibrotic Diseases and Cancer-Associated Fibroblasts. Biomolecules 2020, 10, 1666. [Google Scholar] [CrossRef]
  70. Ren, X.; Li, L.; Wu, J.; Lin, K.; He, Y.; Bian, L. PDGF-BB regulates the transformation of fibroblasts into cancer-associated fibroblasts via the lncRNA LURAP1L-AS1/LURAP1L/IKK/IkappaB/NF-kappaB signaling pathway. Oncol. Lett. 2021, 22, 537. [Google Scholar] [CrossRef]
  71. Wu, H.; Ma, S.; Xiang, M.; Tong, S. HTRA1 promotes transdifferentiation of normal fibroblasts to cancer-associated fibroblasts through activation of the NF-kappaB/bFGF signaling pathway in gastric cancer. Biochem. Biophys. Res. Commun. 2019, 514, 933–939. [Google Scholar] [CrossRef] [PubMed]
  72. Suh, J.; Kim, D.H.; Lee, Y.H.; Jang, J.H.; Surh, Y.J. Fibroblast growth factor-2, derived from cancer-associated fibroblasts, stimulates growth and progression of human breast cancer cells via FGFR1 signaling. Mol. Carcinog. 2020, 59, 1028–1040. [Google Scholar] [CrossRef]
  73. Gascard, P.; Tlsty, T.D. Carcinoma-associated fibroblasts: Orchestrating the composition of malignancy. Genes Dev. 2016, 30, 1002–1019. [Google Scholar] [CrossRef]
  74. Zhao, Z.; Li, T.; Sun, L.; Yuan, Y.; Zhu, Y. Potential mechanisms of cancer-associated fibroblasts in therapeutic resistance. Biomed. Pharmacother. 2023, 166, 115425. [Google Scholar] [CrossRef] [PubMed]
  75. Muchlinska, A.; Nagel, A.; Popeda, M.; Szade, J.; Niemira, M.; Zielinski, J.; Skokowski, J.; Bednarz-Knoll, N.; Zaczek, A.J. Alpha-smooth muscle actin-positive cancer-associated fibroblasts secreting osteopontin promote growth of luminal breast cancer. Cell. Mol. Biol. Lett. 2022, 27, 45. [Google Scholar] [CrossRef]
  76. Zhao, Z.; Li, T.; Yuan, Y.; Zhu, Y. What is new in cancer-associated fibroblast biomarkers? Cell Commun. Signal. 2023, 21, 96. [Google Scholar] [CrossRef] [PubMed]
  77. Fitzgerald, A.A.; Weiner, L.M. The role of fibroblast activation protein in health and malignancy. Cancer Metastasis Rev. 2020, 39, 783–803. [Google Scholar] [CrossRef]
  78. Xin, L.; Gao, J.; Zheng, Z.; Chen, Y.; Lv, S.; Zhao, Z.; Yu, C.; Yang, X.; Zhang, R. Fibroblast Activation Protein-alpha as a Target in the Bench-to-Bedside Diagnosis and Treatment of Tumors: A Narrative Review. Front. Oncol. 2021, 11, 648187. [Google Scholar] [CrossRef]
  79. Wu, C.C.; Mao, Y.T.; Zhang, F.H.; Wang, X.; Fan, N.N.; Zhang, W.; Zhang, W.; Li, P.; Tang, B. Uncovering the mechanism of cancer-associated fibroblasts induced immune evasion of hepatocellular carcinoma cells via in situ fluorescence imaging. Sensor Actuat B-Chem 2023, 389, 133891. [Google Scholar] [CrossRef]
  80. Li, X.; Lu, N.; Lin, L.; Chen, Y.; Yang, S.; Wang, H.; Liu, X.; Wu, C.; Xue, X.; Su, X.; et al. (18)F-FAPI-04 Outperforms (18)F-FDG PET/CT in Clinical Assessments of Patients with Pancreatic Adenocarcinoma. J. Nucl. Med. 2024, 65, 206–212. [Google Scholar] [CrossRef]
  81. 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] [PubMed]
  82. Wei, Y.; Ma, L.; Li, P.; Lu, J.; Ren, J.; Yan, S.; Wu, H.; Yuan, S.; Fu, Z.; Yu, J. FAPI Compared with FDG PET/CT for Diagnosis of Primary and Metastatic Lung Cancer. Radiology 2023, 308, e222785. [Google Scholar] [CrossRef] [PubMed]
  83. Pang, Y.; Zhao, L.; Fang, J.; Chen, J.; Meng, L.; Sun, L.; Wu, H.; Guo, Z.; Lin, Q.; Chen, H. Development of FAPI Tetramers to Improve Tumor Uptake and Efficacy of FAPI Radioligand Therapy. J. Nucl. Med. 2023, 64, 1449–1455. [Google Scholar] [CrossRef] [PubMed]
  84. Huang, J.; Fu, L.; Zhang, X.; Huang, S.; Dong, Y.; Hu, K.; Han, Y.; Zhou, K.; Min, C.; Huang, Y.; et al. Noninvasive imaging of FAP expression using positron emission tomography: A comparative evaluation of a [(18)F]-labeled glycopeptide-containing FAPI with [(18)F]FAPI-42. Eur. J. Nucl. Med. Mol. Imaging 2023, 50, 3363–3374. [Google Scholar] [CrossRef]
  85. Kratochwil, C.; Flechsig, P.; Lindner, T.; Abderrahim, L.; Altmann, A.; Mier, W.; Adeberg, S.; Rathke, H.; Röhrich, M.; Winter, H.; et al. (68)Ga-FAPI PET/CT: Tracer Uptake in 28 Different Kinds of Cancer. J. Nucl. Med. 2019, 60, 801–805. [Google Scholar] [CrossRef]
  86. Tian, X.; Wang, Y.; Zhang, Y.; Teng, Y.; Cui, Z.; Liu, J. Evaluating the diagnostic value of 18F-FAPI-04 PET/CT in various malignant tumors: A head-to-head comparison with 18F-FDG PET/CT. Jpn. J. Radiol. 2024. [Google Scholar] [CrossRef]
  87. Shi, X.; Xing, H.; Yang, X.; Li, F.; Yao, S.; Zhang, H.; Zhao, H.; Hacker, M.; Huo, L.; Li, X. Fibroblast imaging of hepatic carcinoma with 68Ga-FAPI-04 PET/CT: A pilot study in patients with suspected hepatic nodules. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 196–203. [Google Scholar] [CrossRef]
  88. 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 <sup>68</sup>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]
  89. Wang, L.; Tang, G.; Hu, K.; Liu, X.; Zhou, W.; Li, H.; Huang, S.; Han, Y.; Chen, L.; Zhong, J.; et al. Comparison of 68Ga-FAPI and 18F-FDG PET/CT in the Evaluation of Advanced Lung Cancer. Radiology 2022, 303, 191–199. [Google Scholar] [CrossRef]
  90. Liu, Q.; Shi, S.; Liu, S.; Xu, X.; Hu, S.; Zhang, J.; Wang, C.; Yu, X.; Song, S. The added value of [68Ga]Ga-DOTA-FAPI-04 PET/CT in pancreatic cancer: A comparison to [18F]F-FDG. Eur. Radiol. 2023, 33, 5007–5016. [Google Scholar] [CrossRef]
  91. Kömek, H.; Can, C.; Kaplan, İ.; Gündoğan, C.; Kepenek, F.; Karaoglan, H.; Demirkıran, A.; Ebinç, S.; Güzel, Y.; Gündeş, E. Comparison of [68 Ga]Ga-DOTA-FAPI-04 PET/CT and [18F]FDG PET/CT in colorectal cancer. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 3898–3909. [Google Scholar] [CrossRef] [PubMed]
  92. Qin, C.; Liu, F.; Huang, J.; Ruan, W.; Liu, Q.; Gai, Y.; Hu, F.; Jiang, D.; Hu, Y.; Yang, K.; et al. A head-to-head comparison of 68Ga-DOTA-FAPI-04 and 18F-FDG PET/MR in patients with nasopharyngeal carcinoma: A prospective study. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 3228–3237. [Google Scholar] [CrossRef] [PubMed]
  93. Giesel, F.L.; Adeberg, S.; Syed, M.; Lindner, T.; Jiménez-Franco, L.D.; Mavriopoulou, E.; Staudinger, F.; Tonndorf-Martini, E.; Regnery, S.; Rieken, S.; et al. FAPI-74 PET/CT Using Either (18)F-AlF or Cold-Kit (68)Ga Labeling: Biodistribution, Radiation Dosimetry, and Tumor Delineation in Lung Cancer Patients. J. Nucl. Med. 2021, 62, 201–207. [Google Scholar] [CrossRef] [PubMed]
  94. Röhrich, M.; Syed, M.; Liew, D.P.; Giesel, F.L.; Liermann, J.; Choyke, P.L.; Wefers, A.K.; Ritz, T.; Szymbara, M.; Schillings, L.; et al. 68Ga-FAPI-PET/CT improves diagnostic staging and radiotherapy planning of adenoid cystic carcinomas—Imaging analysis and histological validation. Radiother. Oncol. 2021, 160, 192–201. [Google Scholar] [CrossRef] [PubMed]
  95. Mori, Y.; Haberkorn, U.; Giesel, F.L. 68Ga- or 18F-FAPI PET/CT—What it can and cannot. Eur. Radiol. 2023, 33, 7877–7878. [Google Scholar] [CrossRef]
  96. Chandekar, K.R.; Prashanth, A.; Vinjamuri, S.; Kumar, R. FAPI PET/CT Imaging—An Updated Review. Diagnostics 2023, 13, 2018. [Google Scholar] [CrossRef]
  97. Poplawski, S.E.; Hallett, R.M.; Dornan, M.H.; Novakowski, K.E.; Pan, S.; Belanger, A.P.; Nguyen, Q.D.; Wu, W.; Felten, A.E.; Liu, Y.; et al. Preclinical Development of PNT6555, a Boronic Acid-Based, Fibroblast Activation Protein-alpha (FAP)-Targeted Radiotheranostic for Imaging and Treatment of FAP-Positive Tumors. J. Nucl. Med. 2024, 65, 100–108. [Google Scholar] [CrossRef]
  98. Bentivoglio, V.; Galli, F.; Varani, M.; Ranieri, D.; Nayak, P.; D’Elia, A.; Soluri, A.; Massari, R.; Lauri, C.; Signore, A. Radiolabelled FGF-2 for Imaging Activated Fibroblasts in the Tumor Micro-Environment. Biomolecules 2024, 14, 491. [Google Scholar] [CrossRef]
  99. Dai, J.; Ouyang, H.; Wei, S.; Chen, B.; Dong, X.; Hu, J.J.; Wu, M.; Wang, S.; Xia, F.; Lou, X. Cancer-Associated Fibroblast Mimetic AIE Probe for Precision Imaging-Guided Full-Cycle Management of Ovarian Cancer Surgery. Anal. Chem. 2023, 95, 15068–15077. [Google Scholar] [CrossRef]
  100. Bautch, V.L.; Caron, K.M. Blood and lymphatic vessel formation. Cold Spring Harb. Perspect. Biol. 2015, 7, a008268. [Google Scholar] [CrossRef]
  101. Kataru, R.P.; Ly, C.L.; Shin, J.; Park, H.J.; Baik, J.E.; Rehal, S.; Ortega, S.; Lyden, D.; Mehrara, B.J. Tumor Lymphatic Function Regulates Tumor Inflammatory and Immunosuppressive Microenvironments. Cancer Immunol. Res. 2019, 7, 1345–1358. [Google Scholar] [CrossRef] [PubMed]
  102. Paduch, R. The role of lymphangiogenesis and angiogenesis in tumor metastasis. Cell. Oncol. 2016, 39, 397–410. [Google Scholar] [CrossRef] [PubMed]
  103. Pflicke, H.; Sixt, M. Preformed portals facilitate dendritic cell entry into afferent lymphatic vessels. J. Exp. Med. 2009, 206, 2925–2935. [Google Scholar] [CrossRef]
  104. Patan, S. Vasculogenesis and angiogenesis. Cancer Treat. Res. 2004, 117, 3–32. [Google Scholar] [CrossRef]
  105. Marcola, M.; Rodrigues, C.E. Endothelial progenitor cells in tumor angiogenesis: Another brick in the wall. Stem Cells Int. 2015, 2015, 832649. [Google Scholar] [CrossRef] [PubMed]
  106. Nolan, D.J.; Ciarrocchi, A.; Mellick, A.S.; Jaggi, J.S.; Bambino, K.; Gupta, S.; Heikamp, E.; McDevitt, M.R.; Scheinberg, D.A.; Benezra, R.; et al. Bone marrow-derived endothelial progenitor cells are a major determinant of nascent tumor neovascularization. Genes Dev. 2007, 21, 1546–1558. [Google Scholar] [CrossRef]
  107. Romagnani, P.; Annunziato, F.; Liotta, F.; Lazzeri, E.; Mazzinghi, B.; Frosali, F.; Cosmi, L.; Maggi, L.; Lasagni, L.; Scheffold, A.; et al. CD14+CD34low cells with stem cell phenotypic and functional features are the major source of circulating endothelial progenitors. Circ. Res. 2005, 97, 314–322. [Google Scholar] [CrossRef]
  108. de la Puente, P.; Muz, B.; Azab, F.; Azab, A.K. Cell trafficking of endothelial progenitor cells in tumor progression. Clin. Cancer Res. 2013, 19, 3360–3368. [Google Scholar] [CrossRef]
  109. Deng, T.; Zhao, J.; Tong, Y.; Chen, Z.; He, B.; Li, J.; Chen, B.; Li, R.; Deng, L.; Yu, H.; et al. Crosstalk between endothelial progenitor cells and HCC through periostin/CCL2/CD36 supports formation of the pro-metastatic microenvironment in HCC. Oncogene 2024, 43, 944–961. [Google Scholar] [CrossRef]
  110. Liu, Z.L.; Chen, H.H.; Zheng, L.L.; Sun, L.P.; Shi, L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct. Target. Ther. 2023, 8, 198. [Google Scholar] [CrossRef]
  111. Luo, Q.; Wang, J.; Zhao, W.; Peng, Z.; Liu, X.; Li, B.; Zhang, H.; Shan, B.; Zhang, C.; Duan, C. Vasculogenic mimicry in carcinogenesis and clinical applications. J. Hematol. Oncol. 2020, 13, 19. [Google Scholar] [CrossRef] [PubMed]
  112. Kozlova, N.; Grossman, J.E.; Iwanicki, M.P.; Muranen, T. The Interplay of the Extracellular Matrix and Stromal Cells as a Drug Target in Stroma-Rich Cancers. Trends Pharmacol. Sci. 2020, 41, 183–198. [Google Scholar] [CrossRef] [PubMed]
  113. Shibuya, M. Vascular endothelial growth factor and its receptor system: Physiological functions in angiogenesis and pathological roles in various diseases. J. Biochem. 2013, 153, 13–19. [Google Scholar] [CrossRef]
  114. Brooks, P.C.; Clark, R.A.; Cheresh, D.A. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 1994, 264, 569–571. [Google Scholar] [CrossRef]
  115. Qin, Z.X.; Li, Q.W.; Liu, G.Y.; Luo, C.X.; Xie, G.F.; Zheng, L.; Huang, D.D. Imaging targeted at tumor with (188)Re-labeled VEGF(189) exon 6-encoded peptide and effects of the transfecting truncated KDR gene in tumor-bearing nude mice. Nucl. Med. Biol. 2009, 36, 535–543. [Google Scholar] [CrossRef]
  116. Jia, H.; Jezequel, S.; Lohr, M.; Shaikh, S.; Davis, D.; Soker, S.; Selwood, D.; Zachary, I. Peptides encoded by exon 6 of VEGF inhibit endothelial cell biological responses and angiogenesis induced by VEGF. Biochem. Biophys. Res. Commun. 2001, 283, 164–173. [Google Scholar] [CrossRef]
  117. Hu, K.; Shang, J.; Xie, L.; Hanyu, M.; Zhang, Y.; Yang, Z.; Xu, H.; Wang, L.; Zhang, M.R. PET Imaging of VEGFR with a Novel (64)Cu-Labeled Peptide. ACS Omega 2020, 5, 8508–8514. [Google Scholar] [CrossRef] [PubMed]
  118. Luo, H.; England, C.G.; Graves, S.A.; Sun, H.; Liu, G.; Nickles, R.J.; Cai, W. PET Imaging of VEGFR-2 Expression in Lung Cancer with 64Cu-Labeled Ramucirumab. J. Nucl. Med. 2016, 57, 285–290. [Google Scholar] [CrossRef] [PubMed]
  119. Meyer, J.P.; Edwards, K.J.; Kozlowski, P.; Backer, M.V.; Backer, J.M.; Lewis, J.S. Selective Imaging of VEGFR-1 and VEGFR-2 Using 89Zr-Labeled Single-Chain VEGF Mutants. J. Nucl. Med. 2016, 57, 1811–1816. [Google Scholar] [CrossRef]
  120. Meng, D.; Meng, M.; Luo, A.; Jing, X.; Wang, G.; Huang, S.; Luo, M.; Shao, S.; Zhao, X.; Liu, R. Effects of VEGFR1(+) hematopoietic progenitor cells on pre-metastatic niche formation and in vivo metastasis of breast cancer cells. J. Cancer Res. Clin. Oncol. 2019, 145, 411–427. [Google Scholar] [CrossRef]
  121. Mason, C.A.; Carter, L.M.; Mandleywala, K.; de Souza Franca, P.D.; Meyer, J.P.; Mamun, T.; Backer, J.M.; Backer, M.V.; Reiner, T.; Lewis, J.S. Imaging Early-Stage Metastases Using an (18)F-Labeled VEGFR-1-Specific Single Chain VEGF Mutant. Mol. Imaging Biol. 2021, 23, 340–349. [Google Scholar] [CrossRef] [PubMed]
  122. Danhier, F.; Le Breton, A.; Preat, V. RGD-based strategies to target alpha(v) beta(3) integrin in cancer therapy and diagnosis. Mol. Pharm. 2012, 9, 2961–2973. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, W.; Ma, H.; Li, F.; Cai, H.; Liang, R.; Chen, X.; Lan, T.; Yang, J.; Liao, J.; Yang, Y.; et al. PET imaging of VEGFR and integrins in glioma tumor xenografts using (89)Zr labelled heterodimeric peptide. Bioorg. Med. Chem. 2022, 59, 116677. [Google Scholar] [CrossRef]
  124. Kim, S.; Mushtaq, S.; Lee, K.C.; Park, J.A.; Kim, J.Y. (64)Cu-Labeled Boron-Containing Cyclic RGD Peptides for BNCT and PET Imaging. ACS Med. Chem. Lett. 2024, 15, 344–348. [Google Scholar] [CrossRef]
  125. Zhao, L.; Wen, X.; Xu, W.; Pang, Y.; Sun, L.; Wu, X.; Xu, P.; Zhang, J.; Guo, Z.; Lin, Q.; et al. Clinical Evaluation of (68)Ga-FAPI-RGD for Imaging of Fibroblast Activation Protein and Integrin alpha(v)beta(3) in Various Cancer Types. J. Nucl. Med. 2023, 64, 1210–1217. [Google Scholar] [CrossRef] [PubMed]
  126. Hu, Z.; Zhao, X.; Wu, Z.; Qu, B.; Yuan, M.; Xing, Y.; Song, Y.; Wang, Z. Lymphatic vessel: Origin, heterogeneity, biological functions, and therapeutic targets. Signal Transduct. Target. Ther. 2024, 9, 9. [Google Scholar] [CrossRef]
  127. Hampton, H.R.; Chtanova, T. Lymphatic Migration of Immune Cells. Front. Immunol. 2019, 10, 1168. [Google Scholar] [CrossRef]
  128. Steele, M.M.; Lund, A.W. Afferent Lymphatic Transport and Peripheral Tissue Immunity. J. Immunol. 2021, 206, 264–272. [Google Scholar] [CrossRef]
  129. Zhou, H.; Lei, P.J.; Padera, T.P. Progression of Metastasis through Lymphatic System. Cells 2021, 10, 627. [Google Scholar] [CrossRef]
  130. Naxerova, K. Defining the role of lymph node metastasis in systemic breast cancer evolution. EBioMedicine 2020, 57, 102852. [Google Scholar] [CrossRef]
  131. Sha, M.; Jeong, S.; Wang, X.; Tong, Y.; Cao, J.; Sun, H.Y.; Xia, L.; Xu, N.; Xi, Z.F.; Zhang, J.J.; et al. Tumor-associated lymphangiogenesis predicts unfavorable prognosis of intrahepatic cholangiocarcinoma. BMC Cancer 2019, 19, 208. [Google Scholar] [CrossRef] [PubMed]
  132. Eshima, D.; Eshima, L.A.; Gotti, N.M.; Herda, S.C.; Algozine, C.A.; Burris, T.G.; Vansant, J.P.; Alazraki, N.P.; Taylor, A.T. Technetium-99m-sulfur colloid for lymphoscintigraphy: Effects of preparation parameters. J. Nucl. Med. 1996, 37, 1575–1578. [Google Scholar] [PubMed]
  133. Alavi, A.; Staum, M.M.; Shesol, B.F.; Bloch, P.H. Technetium-99m stannous phytate as an imaging agent for lymph nodes. J. Nucl. Med. 1978, 19, 422–426. [Google Scholar] [PubMed]
  134. Leong, S.P.; Kim, J.; Ross, M.; Faries, M.; Scoggins, C.R.; Metz, W.L.; Cope, F.O.; Orahood, R.C. A phase 2 study of (99m)Tc-tilmanocept in the detection of sentinel lymph nodes in melanoma and breast cancer. Ann. Surg. Oncol. 2011, 18, 961–969. [Google Scholar] [CrossRef] [PubMed]
  135. Leong, S.P. Detection of melanoma, breast cancer and head and neck squamous cell cancer sentinel lymph nodes by Tc-99m Tilmanocept (Lymphoseek(R)). Clin. Exp. Metastasis 2022, 39, 39–50. [Google Scholar] [CrossRef]
  136. Thorek, D.L.; Ulmert, D.; Diop, N.F.; Lupu, M.E.; Doran, M.G.; Huang, R.; Abou, D.S.; Larson, S.M.; Grimm, J. Non-invasive mapping of deep-tissue lymph nodes in live animals using a multimodal PET/MRI nanoparticle. Nat. Commun. 2014, 5, 3097. [Google Scholar] [CrossRef]
  137. Liu, P.; Ding, P.; Sun, C.; Chen, S.; Lowe, S.; Meng, L.; Zhao, Q. Lymphangiogenesis in gastric cancer: Function and mechanism. Eur. J. Med. Res. 2023, 28, 405. [Google Scholar] [CrossRef]
  138. Bieniasz-Krzywiec, P.; Martin-Perez, R.; Ehling, M.; Garcia-Caballero, M.; Pinioti, S.; Pretto, S.; Kroes, R.; Aldeni, C.; Di Matteo, M.; Prenen, H.; et al. Podoplanin-Expressing Macrophages Promote Lymphangiogenesis and Lymphoinvasion in Breast Cancer. Cell Metab. 2019, 30, 917–936.e910. [Google Scholar] [CrossRef]
  139. Jackson, D.G.; Prevo, R.; Clasper, S.; Banerji, S. LYVE-1, the lymphatic system and tumor lymphangiogenesis. Trends Immunol. 2001, 22, 317–321. [Google Scholar] [CrossRef]
  140. Cueni, L.N.; Detmar, M. New insights into the molecular control of the lymphatic vascular system and its role in disease. J. Investig. Dermatol. 2006, 126, 2167–2177. [Google Scholar] [CrossRef]
  141. Christiansen, A.; Detmar, M. Lymphangiogenesis and cancer. Genes Cancer 2011, 2, 1146–1158. [Google Scholar] [CrossRef] [PubMed]
  142. Dellinger, M.T.; Witte, M.H. Lymphangiogenesis, lymphatic systemomics, and cancer: Context, advances and unanswered questions. Clin. Exp. Metastasis 2018, 35, 419–424. [Google Scholar] [CrossRef] [PubMed]
  143. Su, J.L.; Yen, C.J.; Chen, P.S.; Chuang, S.E.; Hong, C.C.; Kuo, I.H.; Chen, H.Y.; Hung, M.C.; Kuo, M.L. The role of the VEGF-C/VEGFR-3 axis in cancer progression. Br. J. Cancer 2007, 96, 541–545. [Google Scholar] [CrossRef] [PubMed]
  144. Tacconi, C.; Ungaro, F.; Correale, C.; Arena, V.; Massimino, L.; Detmar, M.; Spinelli, A.; Carvello, M.; Mazzone, M.; Oliveira, A.I.; et al. Activation of the VEGFC/VEGFR3 Pathway Induces Tumor Immune Escape in Colorectal Cancer. Cancer Res. 2019, 79, 4196–4210. [Google Scholar] [CrossRef]
  145. Kwon, S.; Velasquez, F.C.; Sevick-Muraca, E.M. Near-infrared fluorescence lymphatic imaging in vascular endothelial growth factor-C overexpressing murine melanoma. Biomed. Opt. Express 2018, 9, 4631–4637. [Google Scholar] [CrossRef]
  146. Proulx, S.T.; Luciani, P.; Christiansen, A.; Karaman, S.; Blum, K.S.; Rinderknecht, M.; Leroux, J.C.; Detmar, M. Use of a PEG-conjugated bright near-infrared dye for functional imaging of rerouting of tumor lymphatic drainage after sentinel lymph node metastasis. Biomaterials 2013, 34, 5128–5137. [Google Scholar] [CrossRef]
  147. Chong, C.; Scholkmann, F.; Bachmann, S.B.; Luciani, P.; Leroux, J.C.; Detmar, M.; Proulx, S.T. In vivo visualization and quantification of collecting lymphatic vessel contractility using near-infrared imaging. Sci. Rep. 2016, 6, 22930. [Google Scholar] [CrossRef]
  148. Schacht, V.; Ramirez, M.I.; Hong, Y.K.; Hirakawa, S.; Feng, D.; Harvey, N.; Williams, M.; Dvorak, A.M.; Dvorak, H.F.; Oliver, G.; et al. T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J. 2003, 22, 3546–3556. [Google Scholar] [CrossRef]
  149. Kato, T.; Furusawa, A.; Okada, R.; Inagaki, F.; Wakiyama, H.; Furumoto, H.; Fukushima, H.; Okuyama, S.; Choyke, P.L.; Kobayashi, H. Near-Infrared Photoimmunotherapy Targeting Podoplanin-Expressing Cancer Cells and Cancer-Associated Fibroblasts. Mol. Cancer Ther. 2023, 22, 75–88. [Google Scholar] [CrossRef]
  150. Banerji, S.; Lawrance, W.; Metcalfe, C.; Briggs, D.C.; Yamauchi, A.; Dushek, O.; van der Merwe, P.A.; Day, A.J.; Jackson, D.G. Homodimerization of the Lymph Vessel Endothelial Receptor LYVE-1 through a Redox-labile Disulfide Is Critical for Hyaluronan Binding in Lymphatic Endothelium. J. Biol. Chem. 2016, 291, 25004–25018. [Google Scholar] [CrossRef]
  151. Karinen, S.; Juurikka, K.; Hujanen, R.; Wahbi, W.; Hadler-Olsen, E.; Svineng, G.; Eklund, K.K.; Salo, T.; Astrom, P.; Salem, A. Tumour cells express functional lymphatic endothelium-specific hyaluronan receptor in vitro and in vivo: Lymphatic mimicry promotes oral oncogenesis? Oncogenesis 2021, 10, 23. [Google Scholar] [CrossRef] [PubMed]
  152. Wang, Q.S.; He, R.; Yang, F.; Kang, L.J.; Li, X.Q.; Fu, L.; Sun, B.; Feng, Y.M. FOXF2 deficiency permits basal-like breast cancer cells to form lymphangiogenic mimicry by enhancing the response of VEGF-C/VEGFR3 signaling pathway. Cancer Lett. 2018, 420, 116–126. [Google Scholar] [CrossRef]
  153. Mumprecht, V.; Honer, M.; Vigl, B.; Proulx, S.T.; Trachsel, E.; Kaspar, M.; Banziger-Tobler, N.E.; Schibli, R.; Neri, D.; Detmar, M. In vivo imaging of inflammation- and tumor-induced lymph node lymphangiogenesis by immuno-positron emission tomography. Cancer Res. 2010, 70, 8842–8851. [Google Scholar] [CrossRef]
  154. Chen, X.; Zhang, Z.; Wang, L.; Zhang, J.; Zhao, T.; Cai, J.; Dang, Y.; Guo, R.; Liu, R.; Zhou, Y.; et al. Homodimeric peptide radiotracer [(68)Ga]Ga-NOTA-(TMVP1)(2) for VEGFR-3 imaging of cervical cancer patients. Eur. J. Nucl Med. Mol. Imaging 2024, 51, 2338–2352. [Google Scholar] [CrossRef] [PubMed]
  155. Yuan, Y.; Dong, X.; Chen, Y.; Xi, L.; Ma, D.; Dai, J.; Li, F. TMVP1448, a novel peptide improves detection of primary tumors and metastases by specifically targeting VEGFR-3. Biomed. Pharmacother. 2024, 177, 116980. [Google Scholar] [CrossRef]
  156. Wang, J.; Li, D.; Cang, H.; Guo, B. Crosstalk between cancer and immune cells: Role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019, 8, 4709–4721. [Google Scholar] [CrossRef] [PubMed]
  157. Pinard, C.J.; International Immuno-Oncology Biomarker Working, G.; Lagree, A.; Lu, F.I.; Klein, J.; Oblak, M.L.; Salgado, R.; Cardenas, J.C.P.; Brunetti, B.; Muscatello, L.V.; et al. Comparative Evaluation of Tumor-Infiltrating Lymphocytes in Companion Animals: Immuno-Oncology as a Relevant Translational Model for Cancer Therapy. Cancers 2022, 14, 5008. [Google Scholar] [CrossRef] [PubMed]
  158. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
  159. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef]
  160. Hoffmann, S.H.L.; Reck, D.I.; Maurer, A.; Fehrenbacher, B.; Sceneay, J.E.; Poxleitner, M.; Oz, H.H.; Ehrlichmann, W.; Reischl, G.; Fuchs, K.; et al. Visualization and quantification of in vivo homing kinetics of myeloid-derived suppressor cells in primary and metastatic cancer. Theranostics 2019, 9, 5869–5885. [Google Scholar] [CrossRef]
  161. Zinnhardt, B.; Muther, M.; Roll, W.; Backhaus, P.; Jeibmann, A.; Foray, C.; Barca, C.; Doring, C.; Tavitian, B.; Dolle, F.; et al. TSPO imaging-guided characterization of the immunosuppressive myeloid tumor microenvironment in patients with malignant glioma. Neuro-Oncology 2020, 22, 1030–1043. [Google Scholar] [CrossRef] [PubMed]
  162. Eisenblaetter, M.; Flores-Borja, F.; Lee, J.J.; Wefers, C.; Smith, H.; Hueting, R.; Cooper, M.S.; Blower, P.J.; Patel, D.; Rodriguez-Justo, M.; et al. Visualization of Tumor-Immune Interaction—Target-Specific Imaging of S100A8/A9 Reveals Pre-Metastatic Niche Establishment. Theranostics 2017, 7, 2392–2401. [Google Scholar] [CrossRef] [PubMed]
  163. Lindau, D.; Gielen, P.; Kroesen, M.; Wesseling, P.; Adema, G.J. The immunosuppressive tumour network: Myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2013, 138, 105–115. [Google Scholar] [CrossRef] [PubMed]
  164. Haist, M.; Stege, H.; Grabbe, S.; Bros, M. The Functional Crosstalk between Myeloid-Derived Suppressor Cells and Regulatory T Cells within the Immunosuppressive Tumor Microenvironment. Cancers 2021, 13, 210. [Google Scholar] [CrossRef]
  165. Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T cells and immune tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef]
  166. Whiteside, T.L. The role of regulatory T cells in cancer immunology. Immunotargets Ther. 2015, 4, 159–171. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, Y.A.; Li, X.L.; Mo, Y.Z.; Fan, C.M.; Tang, L.; Xiong, F.; Guo, C.; Xiang, B.; Zhou, M.; Ma, J.; et al. Effects of tumor metabolic microenvironment on regulatory T cells. Mol. Cancer 2018, 17, 168. [Google Scholar] [CrossRef]
  168. Sharif-Paghaleh, E.; Sunassee, K.; Tavare, R.; Ratnasothy, K.; Koers, A.; Ali, N.; Alhabbab, R.; Blower, P.J.; Lechler, R.I.; Smyth, L.A.; et al. In vivo SPECT reporter gene imaging of regulatory T cells. PLoS ONE 2011, 6, e25857. [Google Scholar] [CrossRef]
  169. Jacob, J.; Volpe, A.; Peng, Q.; Lechler, R.I.; Smyth, L.A.; Lombardi, G.; Fruhwirth, G.O. Radiolabelling of Polyclonally Expanded Human Regulatory T Cells (Treg) with (89)Zr-oxine for Medium-Term In Vivo Cell Tracking. Molecules 2023, 28, 1482. [Google Scholar] [CrossRef]
  170. Pektor, S.; Schloder, J.; Klasen, B.; Bausbacher, N.; Wagner, D.C.; Schreckenberger, M.; Grabbe, S.; Jonuleit, H.; Miederer, M. Using immuno-PET imaging to monitor kinetics of T cell-mediated inflammation and treatment efficiency in a humanized mouse model for GvHD. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1314–1325. [Google Scholar] [CrossRef]
  171. Chen, Y.; Song, Y.; Du, W.; Gong, L.; Chang, H.; Zou, Z. Tumor-associated macrophages: An accomplice in solid tumor progression. J. Biomed. Sci. 2019, 26, 78. [Google Scholar] [CrossRef] [PubMed]
  172. Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef] [PubMed]
  173. Hwang, I.; Kim, J.W.; Ylaya, K.; Chung, E.J.; Kitano, H.; Perry, C.; Hanaoka, J.; Fukuoka, J.; Chung, J.Y.; Hewitt, S.M. Tumor-associated macrophage, angiogenesis and lymphangiogenesis markers predict prognosis of non-small cell lung cancer patients. J. Transl. Med. 2020, 18, 443. [Google Scholar] [CrossRef] [PubMed]
  174. Maldonado, M.D.M.; Schlom, J.; Hamilton, D.H. Blockade of tumor-derived colony-stimulating factor 1 (CSF1) promotes an immune-permissive tumor microenvironment. Cancer Immunol. Immunother. 2023, 72, 3349–3362. [Google Scholar] [CrossRef]
  175. Movahedi, K.; Schoonooghe, S.; Laoui, D.; Houbracken, I.; Waelput, W.; Breckpot, K.; Bouwens, L.; Lahoutte, T.; De Baetselier, P.; Raes, G.; et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res. 2012, 72, 4165–4177. [Google Scholar] [CrossRef]
  176. Zhang, B.; Yao, G.; Zhang, Y.; Gao, J.; Yang, B.; Rao, Z.; Gao, J. M2-polarized tumor-associated macrophages are associated with poor prognoses resulting from accelerated lymphangiogenesis in lung adenocarcinoma. Clinics 2011, 66, 1879–1886. [Google Scholar] [CrossRef]
  177. Blykers, A.; Schoonooghe, S.; Xavier, C.; D’Hoe, K.; Laoui, D.; D’Huyvetter, M.; Vaneycken, I.; Cleeren, F.; Bormans, G.; Heemskerk, J.; et al. PET Imaging of Macrophage Mannose Receptor-Expressing Macrophages in Tumor Stroma Using 18F-Radiolabeled Camelid Single-Domain Antibody Fragments. J. Nucl. Med. 2015, 56, 1265–1271. [Google Scholar] [CrossRef]
  178. Wu, A.M. Engineered antibodies for molecular imaging of cancer. Methods 2014, 65, 139–147. [Google Scholar] [CrossRef]
  179. Fernandes, B.; Antunes, I.F.; Prasad, K.; Vazquez-Matias, D.A.; De Mattos, E.P.; Szymanski, W.; Jeckel, C.M.M.; de Vries, E.F.J.; Elsinga, P.H. Synthesis and preclinical evaluation of [(18)F]AlF-NODA-MP-C6-CTHRSSVVC as a PET tracer for CD163-positive tumor-infiltrating macrophages. Nucl. Med. Biol. 2024, 138–139, 108946. [Google Scholar] [CrossRef]
  180. Zhang, X.; Heo, G.S.; Li, A.; Lahad, D.; Detering, L.; Tao, J.; Gao, X.; Zhang, X.; Luehmann, H.; Sultan, D.; et al. Development of a CD163-Targeted PET Radiotracer That Images Resident Macrophages in Atherosclerosis. J. Nucl. Med. 2024, 65, 775–780. [Google Scholar] [CrossRef]
  181. Perez-Medina, C.; Tang, J.; Abdel-Atti, D.; Hogstad, B.; Merad, M.; Fisher, E.A.; Fayad, Z.A.; Lewis, J.S.; Mulder, W.J.; Reiner, T. PET Imaging of Tumor-Associated Macrophages with 89Zr-Labeled High-Density Lipoprotein Nanoparticles. J. Nucl. Med. 2015, 56, 1272–1277. [Google Scholar] [CrossRef] [PubMed]
  182. Nelson, B.J.B.; Andersson, J.D.; Wuest, F. Targeted Alpha Therapy: Progress in Radionuclide Production, Radiochemistry, and Applications. Pharmaceutics 2021, 13, 49. [Google Scholar] [CrossRef] [PubMed]
  183. Nelson, B.J.B.; Wilson, J.; Andersson, J.D.; Wuest, F. Theranostic Imaging Surrogates for Targeted Alpha Therapy: Progress in Production, Purification, and Applications. Pharmaceuticals 2023, 16, 1622. [Google Scholar] [CrossRef]
  184. Sharma, S.; Pandey, M.K. Radiometals in Imaging and Therapy: Highlighting Two Decades of Research. Pharmaceuticals 2023, 16, 1460. [Google Scholar] [CrossRef]
Figure 1. Composition of the Tumor Microenvironment. The tumor microenvironment (TME) comprises cancer cells, the extracellular matrix (ECM), stromal cells, and diverse immune cell populations, all of which collectively influence tumor progression. The ECM provides structural support and regulates cell behavior, with abnormal remodeling promoting stiffness, invasion, and metastasis. Stromal cells, including cancer-associated fibroblasts (CAFs), modulate ECM structure, secrete growth factors, and contribute to immunosuppression. Immune cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and regulatory T cells (Tregs) foster immune suppression and support tumor growth. Together, these components interact dynamically, reshaping the mechanical structure, altering cytokine and chemokine profiles, and facilitating immune evasion to drive tumor progression and therapeutic resistance.
Figure 1. Composition of the Tumor Microenvironment. The tumor microenvironment (TME) comprises cancer cells, the extracellular matrix (ECM), stromal cells, and diverse immune cell populations, all of which collectively influence tumor progression. The ECM provides structural support and regulates cell behavior, with abnormal remodeling promoting stiffness, invasion, and metastasis. Stromal cells, including cancer-associated fibroblasts (CAFs), modulate ECM structure, secrete growth factors, and contribute to immunosuppression. Immune cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and regulatory T cells (Tregs) foster immune suppression and support tumor growth. Together, these components interact dynamically, reshaping the mechanical structure, altering cytokine and chemokine profiles, and facilitating immune evasion to drive tumor progression and therapeutic resistance.
Pharmaceuticals 17 01663 g001
Figure 2. Imaging probes targeting various components of the tumor microenvironment. These imaging tools enable a comprehensive understanding of the TME, supporting diagnostic and therapeutic advancements by targeting ECM, stromal cells, vascular and lymphatic networks, and immune cell populations. Probes for ECM visualization focus on collagen, elastin, and fibronectin. CAF probes target markers such as FAP and FGF-2 to assess stromal cell activity. Vascular and lymphatic network imaging utilizes probes targeting angiogenesis and lymphangiogenesis markers, including VEGF, VEGFRs, podoplanin, and LYVE-1. Immune-targeted probes, specific to MDSCs, TAMs, and Tregs, provide insights into the immunosuppressive cell dynamics within the TME.
Figure 2. Imaging probes targeting various components of the tumor microenvironment. These imaging tools enable a comprehensive understanding of the TME, supporting diagnostic and therapeutic advancements by targeting ECM, stromal cells, vascular and lymphatic networks, and immune cell populations. Probes for ECM visualization focus on collagen, elastin, and fibronectin. CAF probes target markers such as FAP and FGF-2 to assess stromal cell activity. Vascular and lymphatic network imaging utilizes probes targeting angiogenesis and lymphangiogenesis markers, including VEGF, VEGFRs, podoplanin, and LYVE-1. Immune-targeted probes, specific to MDSCs, TAMs, and Tregs, provide insights into the immunosuppressive cell dynamics within the TME.
Pharmaceuticals 17 01663 g002
Table 1. Imaging techniques mentioned in the present article.
Table 1. Imaging techniques mentioned in the present article.
Imaging ModalityAdvantagesLimitations
Positron Emission Tomography (PET)
High sensitivity
Quantifiable molecular data
Real-time functional imaging
Lack of anatomical information
Radiation exposure
High cost of radiotracers
Single Photon
Emission Computed Tomography (SPECT)
High sensitivity
Wider availability
Relatively cost-effective compared to PET
Lack of anatomical information
Radiation exposure
Lower resolution than PET
Magnetic Resonance Imaging (MRI)
Non-ionizing radiation
High soft-tissue contrast
Functional and molecular information
High cost
Long acquisition time
Not suitable for patients who have metal-based implants
Computed
Tomography (CT)
Excellent anatomical information
Short imaging acquisition time
Widely available
Radiation exposure
Poor soft-tissue contrast
Potential nephrotoxicity may occur due to the use of contrast agents.
Ultrasound
Non-radiation
Real-time imaging
Portable and cost-effective
Operator dependency
Limited penetration depth
Poor resolution and scatter due to the presence of air in tissues
Fluorescence
Imaging
Non-radiation
High molecule specificity when combined with targeted probes
Real-time surgical guidance
Insufficient tissue penetration
High autofluorescence background within visible and Near-Infrared-I (NIR-I) window
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

Chan, H.-W.; Kuo, D.-Y.; Shueng, P.-W.; Chuang, H.-Y. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals 2024, 17, 1663. https://doi.org/10.3390/ph17121663

AMA Style

Chan H-W, Kuo D-Y, Shueng P-W, Chuang H-Y. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals. 2024; 17(12):1663. https://doi.org/10.3390/ph17121663

Chicago/Turabian Style

Chan, Hui-Wen, Deng-Yu Kuo, Pei-Wei Shueng, and Hui-Yen Chuang. 2024. "Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells" Pharmaceuticals 17, no. 12: 1663. https://doi.org/10.3390/ph17121663

APA Style

Chan, H. -W., Kuo, D. -Y., Shueng, P. -W., & Chuang, H. -Y. (2024). Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals, 17(12), 1663. https://doi.org/10.3390/ph17121663

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