Skip to main content

The systematic role of pancreatic cancer exosomes: distant communication, liquid biopsy and future therapy

Abstract

Pancreatic cancer remains one of the most lethal diseases worldwide. Cancer-derived exosomes, benefiting from the protective role of the lipid membrane, exhibit remarkable stability in the circulatory system. These exosomes, released by tumor microenvironment, contain various biomolecules such as proteins, RNAs, and lipids that plays a pivotal role in mediating distant communication between the local pancreatic tumor and other organs or tissues. They facilitate the transfer of oncogenic factors to distant sites, contributing to the compromised body immune system, distant metastasis, diabetes, cachexia, and promoting a microenvironment conducive to tumor growth and metastasis in pancreatic cancer patients. Beyond their intrinsic roles, circulating exosomes in peripheral blood can be detected to facilitate accurate liquid biopsy. This approach offers a novel and promising method for the diagnosis and management of pancreatic cancer. Consequently, circulating exosomes are not only crucial mediators of systemic cell-cell communication during pancreatic cancer progression but also hold great potential as precise tools for pancreatic cancer management and treatment. Exosome-based liquid biopsy and therapy represent promising advancements in the diagnosis and treatment of pancreatic cancer. Exosomes can serve as drug delivery vehicles, enhancing the targeting and efficacy of anticancer treatments, modulating the immune system, and facilitating gene editing to suppress tumor growth. Ongoing research focuses on biomarker identification, drug delivery systems, and clinical trials to validate the safety and efficacy of exosome-based therapies, offering new possibilities for early diagnosis and precision treatment in pancreatic cancer. Leveraging the therapeutic potential of exosomes, including their ability to deliver targeted drugs and modulate immune responses, opens new avenues for innovative treatment strategies.

Background

Despite amounts of effort thrown into medical research, pancreatic ductal adenocarcinoma, generally named pancreatic cancer, is still one of the most severe malignant diseases. According to the latest GLOBOCAN report, pancreatic cancer has one of the poorest prognoses among cancers, ranking as the sixth leading cause of cancer mortality. There were an estimated 510,566 new cases and 467,005 deaths attributed to pancreatic cancer worldwide [1].

Compared with the magnificent progress in the treatments against other cancer types, the 5-year survival rate of pancreatic cancer remains about 10% [2]. Until today, radical operation provided the only way to cure pancreatic cancer. However, due to the hidden symptom in the early stage, over 80% of patients lose the opportunity to receive an operation because of distant metastasis or obvious vessel invasion [3, 4]. Therefore, systematic therapy combining surgery and chemotherapy is still the main manner to manage pancreatic cancer.

The first-line chemotherapy regimens for pancreatic cancer vary based on the treatment stage and patient condition. Neoadjuvant chemotherapy for locally advanced pancreatic cancer typically includes FOLFIRINOX or gemcitabine plus nab-paclitaxel, aiming to shrink tumors and facilitate surgical resection [5,6,7]. Adjuvant chemotherapy post-surgery often involves gemcitabine alone or combined with capecitabine, or mFOLFIRINOX for physically fit patients, to reduce recurrence risk [8]. For metastatic pancreatic cancer, palliative chemotherapy options include FOLFIRINOX or gemcitabine plus nab-paclitaxel for those in good health, and gemcitabine monotherapy for frail patients, aiming to alleviate symptoms, extend survival, and improve quality of life [9, 10]. However, they still failed to dramatically improve the prognosis of pancreatic cancer patients due to chemoresistance [11]. In addition to chemotherapy, radiotherapy, another approved method to treat a variety of solid tumors, currently serves as a palliative manner to relieve cancerous pain, but with undesirable side effects [12].

In addition to conventional chemotherapy, several novel treatment modalities are being explored for pancreatic cancer, including immunotherapy, targeted therapy, and other systemic treatments [13]. Immunotherapy has emerged as a promising approach in oncology, but its success in pancreatic cancer has been limited. Targeting focal adhesion kinase (FAK) with defactinib, combined with pembrolizumab and gemcitabine, was investigated in a phase I study for PDAC [14]. They reported an 80% disease control rate in refractory PDAC patients, yielding a median PFS of 3.6 months and OS of 7.8 months. In the maintenance cohort, the disease control rate was 70%, with a median PFS of 5.0 months and OS of 8.3 months. Another phase 1b/2 study tested the safety and antitumor activity of niraparib combined with either nivolumab or ipilimumab in advanced pancreatic cancer. The niraparib plus ipilimumab group achieved a 6-month progression-free survival (PFS) of 59.6%, while the niraparib plus nivolumab group showed a PFS of 20.6% [15]. Targeted therapies aim to exploit specific genetic mutations or pathways involved in cancer growth. The PARP inhibitor olaparib has shown promise in patients with BRCA-mutated pancreatic cancer. In the POLO trial, olaparib as maintenance therapy for germline BRCA-mutated metastatic pancreatic cancer led to a significant improvement in PFS (7.4 months with olaparib vs. 3.8 months with placebo), though the OS benefit was not statistically significant [16]. Other systemic treatments include approaches such as oncolytic virus therapy and cancer vaccines. Oncolytic viruses are designed to selectively infect and kill cancer cells, and preliminary trials have shown some promise in pancreatic cancer [17, 18]. In the phase 1 AMPLIFY-201 study, cancer vaccine ELI-002 2P showed no dose-limiting toxicities and induced mKRAS-specific T cell responses in 84% of patients, with a median relapse-free survival (RFS) of 16.33 months. The study found a significant correlation between T cell responses and tumor biomarker reduction, indicating that ELI-002 2P is safe and effective in inducing T cell responses in KRAS-mutated tumors [19].

Despite the array of novel therapeutic approaches being investigated, the prognosis for pancreatic cancer patients remains poor, with worsening social and physical quality of life [13, 20]. Therefore, effective therapy against pancreatic cancer is urgently needed to solve the current dilemma.

As the prerequisite, understanding pancreatic cancer clearly and thoroughly is the basis for developing novel therapy. Compared to massive previous studies on malignant cells and tumor microenvironment [21], local pancreatic tumor also distantly communicates with other tissue and organs, affecting the tumor-host macroenvironment [22]. Therefore, the communication among them is vital for pancreatic cancer progression [23].

Extracellular vesicles, including exosomes, microvesicles, and apoptotic bodies, differ significantly in their physical characteristics, biological functions, and origins [24,25,26] and are extensively studied for their potential diagnostic applications in pancreatic cancer [27]. Exosomes (30–150 nm) are small, spherical vesicles formed via the endosomal pathway, playing roles in intercellular communication and genetic information transfer. Microvesicles (100-1,000 nm) bud directly from the plasma membrane and are involved in cell signaling and inflammation. Apoptotic bodies (50 − 5,000 nm) arise from cells undergoing programmed cell death and contain cellular debris, aiding in the clearance of apoptotic cells. Compared to other extracellular vesicles, exosomes are the most extensively studied. They can be released by all cell types and are found in various body fluids, including serum, blood plasma, and urine. Exosomes contain proteins, metabolites, and nucleic acids, making them a vital mediator in regulating distant cell-cell communication [28] (Fig. 1).

Fig. 1
figure 1

The communication between pancreatic tumor and other organs or tissues via exosomes. Proteins and RNAs in exosomes from local pancreatic tumor could be transmitted to distant organs and tissues, promoting distant metastasis, diabetes, cachexia, and compromised immune system

Benefiting from the Protective lipid bilayer membrane with identifiable proteins (such as CD9, CD63, CD81), exosomes could mediate adjacent and long-distance cell-cell communication in both health and disease compared to other secretory molecules [29]. Originally, exosomes are derived from endosomes. After encapsulating biomolecules from cytosol through inward budding, late endosome turns to be multivesicular body (MVB) containing massive intraluminal vesicles. Then, some intracellular MVB degrade to lysosomes, while others fuse with the cell membrane and release contained exosomes to extracellular space [30]. Such biosynthesis of exosomes in cancer cells is finely regulated by many molecules under different conditions [31]. Although the precise physiology of exosomes biogenesis remains not very clear, their role in facilitating tumorigenesis is relatively widely researched.

Circulating exosomes have significant utility in cancer diagnostics [32]. They carry tumor-specific proteins, RNA, and DNA, enabling early detection and non-invasive diagnosis. As a noninvasive detection method, liquid biopsy offers notable benefits over traditional diagnostic procedures such as tissue biopsy, including enhanced cost-efficiency and convenience [33]. Exosomes can be stably present in bodily fluids, including blood, urine, and saliva, making them highly promising for liquid biopsy [34]. Exosomes can also deliver therapeutic molecules directly to cancer cells, enhancing treatment efficacy and reducing side effects. Additionally, analyzing exosomal content provides insights into tumor biology and drug resistance mechanisms, aiding in the development of personalized treatment strategies and real-time monitoring of therapeutic responses [35]. Moreover, artificially modified exosomes could also be employed as nanocarriers in anticancer therapy [36]. Given the complex tumor microenvironment and poor prognosis of pancreatic cancer, research on exosome-based liquid biopsies may offer more effective diagnostic and prognostic methods for this disease [37]. Exploring the application of exosomes in pancreatic cancer not only helps improve early detection rates but also provides new approaches for personalized treatment and improving survival rates.

In this review, we summarized current knowledge about exosomes in mediating distant cell-cell communication in pancreatic cancer. In addition, the potential clinical application and future research directions of exosomes in pancreatic cancer was also introduced.

Exosomes connecting local tumor microenvironment with other organs/tissues

Body immune system

Immune cells engage in comprehensive and dynamic crosstalk with tumor cells in the pancreatic TME via exosomes [38, 39]. For instance, LncRNA SBF2-AS1 in M2 macrophages-derived exosomes endogenously compete and suppress miR-122-5p expression in PCCs upon uptake, thus promoting XIAP protein level and accelerating PC development [40]. Similarly, M2 macrophages release miR-193b-3p [41], miR-202-5p and miR-142-5p [42] to promote the proliferation, invasion, and migration of PC. Meanwhile, PC cell-derived exosome miR-210 can promote macrophage M2 polarization via targeting and inhibiting FGFRL1, thereby activating the p-PI3K/p-AKT/p-mTOR pathway and promoting chemoresistance [43]. PC cells also generate Exosome lncRNA, such as FGD5-AS1, which induces M2 macrophage polarization and promote PC cells’ malignant behaviors [44]. Thanks to the protective effect of the lipid bilayer, exosomes secreted by PC can also act on immune cells in the circulatory system as well as in other organs/tissues (Fig. 2).

Fig. 2
figure 2

Impact of Pancreatic Cancer Exosomes on the Immune System and Tumor Microenvironment. This illustration depicts the complex interplay between pancreatic cancer cells and the body’s immune system, highlighting how pancreatic cancer exosomes weaken immune responses. On the left, the pancreatic tumor microenvironment restricts the activation and migration of lymphocytes, leading to impaired activation and proliferation of distant T and B lymphocytes (center). The diagram also shows circulating immature myeloid cells creating an immunosuppressive background, and pancreatic cancer cell (PCC) exosomes impairing humoral immunity through massive antibodies and tumor-associated antigens. On the right, circulating dendritic cells (DCs) exhibit dysfunction and humoral immune is impaired, contributing to a compromised immune system. Additionally, circulating T lymphocytes show impaired activation, exemplified by PD-1 and PD-L1 interactions. The overall effect is a compromised body immune system that struggles to effectively attack pancreatic cancer

A previous study on rats showed that murine PCCs-derived exosomes could be internalized of a variety of leukocytes located in peritoneal exudate, spleen, peripheral blood, bone marrow, and lymph nodes. Upon uptake, exosomes can hinder leukocytes activation and proliferation via down-regulating of CD44v6 and inhibiting phosphorylation of LCK, ZAP70 and ERK1/2 [45]. Also, they can occupy migration-associated ligands, such a as CD44, CD49d, CD54, and CD62L [46, 47], and significantly impair lymphocyte migration. Besides, the functions of CTLs, NKs, and tumor-lysate-loaded DCs were activated, supporting immune response induction of pancreatic cancer [45]. However, in circulating system, exosomes secreted by PCCs, especially those without SMAD4 expression, create an immunosuppressive microenvironment characterized by unbalanced immature myeloid cells by upregulating calcium fluxes and glycolysis, which potentially mediated by exo-miRNAs miR-1260a and miR-494-3p [48].

Additionally, PCCs-derived exosomes were taken up by circulating T lymphocytes, activating p38 mitogen-activated protein kinase and inducing endoplasmic reticulum stress-mediated apoptosis of recipient cells, which finally caused immunosuppression [49]. For instance, exosomal PD-L1 could inhibit circulating cytotoxic T lymphocytes and promote PC invasion [50, 51]. In addition to the cytotoxic immune response, the humoral immune response has a more large-scale battlefield involving immune tissue/organs, circulating system, and local tumor. PCCs-derived exosomes preferred to equip enriched tumor associated antigens (TAAs) on their surface and induce B cells-mediated humoral immune response [52]. However, those exosomes could absorb massive autoantibodies as a decoy and facilitate PCCs to escape from complement-mediated cytotoxicity [53].

Furthermore, PCCs-derived exosomes have been demonstrated to inhibit the immune system by deregulating DCs in PBMC. After being taken up by DCs, miR-203-containing exosomes generated by PC cells can boost intracellular miR-203 levels and suppress the production of TLR4 and downstream cytokines (TNF-α and IL-12), finally causing their dysfunction [54]. Zech et al. revealed that PCCs-derived exosomes acquired by DCs can effectively suppress IL-2/PI3K/Akt signal pathway in the lymphocytes, which activated apoptosis [45]. Also, PCCs-derived exosomes suppress RFXAP expression via miR-212-3p, resulting in decreased MHC II expression and immunological tolerance of DCs [55].

The resistance that pancreatic cancer develops to immunotherapy is partly due to the accumulation of immunosuppressive cells in the TME. Wong et al. recently discovered that myeloid-derived suppressor cells (MDSC) formation in PC could be induced by macrophage migration inhibitory factor (MIF) from the tumor-derived exosome. They also synthesized a MIF tautomerase inhibitor, which effectively inhibited exosome-induced MDSC differentiation and tumor growth in an orthotopic PC model by promoting CD8 + T cells infiltration in the TME [56].

Distant metastasis

The movement of tumor cells from vessel to tissue is important, thus the first step of distant metastasis. PCCs could transmit tissue factors to endothelial cells through circulating exosomes. With the help of factor VIIa and factor Xa, those exosomes upregulated endothelial E-selectin and IL-8 synthesis, inducing pro-adhesive and pro-inflammatory phenotype of endothelial cells and promoting metastasis [57]. Additionally, exosomes circular RNA IARS (circ-IARS) from PCCs significantly enhanced endothelial monolayer permeability and promoted blood-borne distant metastasis as well [58]. Moreover, another recent study showed that hypoxic PCC secreted exosomal miR-30b-5p targeted GJA1 in endothelial cells, promoting angiogenesis and distant metastasis [59]. However, PCC also could transmit exosomal miR-29b to endothelial cells and repressed ROBO1 and SRGAP2, which inhibited angiogenesis and metastasis [60].

Lymphatic system is an important defense system of the human body, consisting of lymphatic vessels, lymphoid tissue, and lymphoid organs [61]. Current research is being devoted to understand how PC cells transfer to the lymph nodes. A potential mediator that causes lymphatic metastasis is exosomes [62,63,64]. Exosomal lnRNA LNMAT2 was proved to facilitate lymph-angiogenesis and lymphatic metastasis in bladder cancer via VEGF-C [65]. Also, cancer-secreted exosomal miRNAs could promote lymphatic metastasis in esophageal [66, 67], cervical [68], endometrial [69], gastric [70], and breast cancer [71]. In pancreatic cancer, Zhou et al. found that PCC-derived exosomes could promote lymph-angiogenesis via downregulating ABHD11-AS1 expression and upregulating proliferation, migration, and tubes formation in lymphatic endothelial cells [72].

Fig. 3
figure 3

Mechanisms by which circulating PCC-derived exosomes act on vascular and lymphatic endothelial cells to promote metastasis

Besides the impact on vascular endothelial cells and lymphatic system (Fig. 3), the direct effects of exosomes on target organs were also significant to construct premetastatic niche. The disposition of PCC-derived exosomes in target organs, such as macrophages in lung and bone marrow, could be detected before any metastases are present [73]. The underlying molecular mechanism of exosomes in driving distant metastasis is being gradually revealed. Liver, peritoneum, and lung are common metastatic sites in pancreatic cancer patients [74]. The pattern of PCC-derived exosomes involvement in liver metastasis is illustrated in Fig. 4. Primary tumor cells could secrete exosomes to modify the extracellular matrix of other organs, rendering them to be receptive to metastatic seeding. For example, specific integrins on PCC-derived exosomes could drive different metastasis. Exosomes with integrin αvβ5 mediated liver tropism whereas α6β4 and α6β1 contributed lung tropism, which could induce pro-inflammatory pathways in recipient cells and create pre-metastatic niches in target organs [75]. Macrophage migration inhibitory factor (MIF), enriched in pancreatic tumor exosomes, could impel hepatic Kupffer cells to secrete and release TGF-β. Then, the fibronectin production of hepatic stellate cells was upregulated, which increased the recruitment of bone marrow-derived macrophages and promoted liver pre-metastatic niche formation [76]. Besides the stimulation mediated by Kupffer cells, PCC-derived exosomes containing CD44v6 and complement C1q binding protein (C1QBP) complex contributed to the phosphorylation of PI3K/AKT pathway and α-SMA expression in hepatic stellate cells directly, which enhanced liver fibrosis and liver metastasis of pancreatic cancer [77]. Additionally, another recent report suggested that PCCs-derived exosomes could reprogram the transcriptome of CD11b + cells in bone marrow, which might subsequently turn to be bone marrow-derived macrophages within tumor microenvironment and liver premetastatic niches [78]. Also, the PCCs-derived exosome tRF-GluCTC-0005 was able to recruit MDSCs and activated hepatic stellate cells in the liver, which created an immunosuppressive microenvironment and further promoted liver metastasis [79]. Moreover, PCC also transferred exosomes with metabolites, particularly palmitic acid, to Kupffer cells in liver, inducing fatty liver and systemically dysregulated metabolism in pancreatic cancer patients. As a result, the systemic immune functions were compromised, and the distant metastasis was accelerated [80].

Fig. 4
figure 4

Mechanisms by which circulating PCC-derived exosomes act on a hepatic Kupffer cells and stellate cells to promote liver metastasis

Except for promoting liver metastasis, PCCs with low protein kinase D1 (PKD1) expression preferred to secret α6β4 positive exosomes, which promoted lung metastasis of pancreatic cancer [81]. Additionally, PCCs with mutant p53 released exosomes containing certain podocalyxin levels, which could influence the motility of fibroblasts and promote the formation of pro-invasive extracellular matrix in distant organs, such as lung [82]. After injection of PC ascites-derived exosomes from PC patients in mice, the vascular permeability in the lung was promoted, leading to the extravasation and colonization of PCCs [83]. Compared with liver and lung metastasis, the underlying mechanism of possible exosome-mediated peritoneal metastasis in pancreatic cancer remains to be elucidated.

Diabetes

Extensive epidemiological evidence suggested that diabetes could serve as both a risk factor and clinical manifestation of pancreatic cancer [84, 85]. However, the underlying molecular mechanism between pancreatic cancer and diabetes remains unclear [86]. PCC-derived exosomes may cause changes in the whole pancreatic islets including intra-tumoral islets, resulting in the occurrence of pancreatic cancer-related diabetes [87, 88]. In addition, the development of diabetes is not only related to the pancreatic islets, but also to the sugar uptake and fat metabolism of peripheral tissues such as muscle tissue [89]. We have revealed the biological mechanisms of exosomes in mediating this process (Fig. 5).

Fig. 5
figure 5

Mechanisms by which circulating PCC-derived exosomes act on pancreatic β-cells, muscle cells and enteroendocrine cells to promote the development of diabetes

Tumor-derived EVs could transfer miR-122 to β-cells and target pyruvate kinase M (PKM), thus suppressing glycolysis and insulin secretion, promoting glucose tolerance, fasting hyperglycemia, eventually contributing to the impairment of whole-body glucose homeostasis [90]. In PC, adrenomedullin was up-regulated and responsible for β-cells insulin resistance [91]. PCCs-derived exosomes transmitted adrenomedullin to β-cells, which caused endoplasmic reticulum stress and β-cell dysfunction. Therefore, insulin secretion was indirectly suppressed by PCCs [92]. Also, adrenomedullin can be loaded into exosomes derived from cancer-associated fibroblasts, and then delivered to recipient adipocytes to enhance lipolysis in the adipose tissues [93], which was reckoned as a hallmark of diabetes [94]. MiR-19a contained in PCCs-derived exosomes was also reported to regulate insulin secretion. It was found that miR-19a induced β-cell dysfunction via targeting ADCY1 and EPAC2 [95], as well as inhibited insulin production via targeting Neurod1 in pancreatic β cells [96]. Additionally, another recent report showed that Vanin-1 in PCC-derived exosomes could impair adjacent islets by producing oxidative stress and β-cell dedifferentiation, leading to diabetes [97]. In addition, the secretion and function of insulin were affected by many other regulators. Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) are two important hormones contributing to glucose homeostasis through enhancing insulin secretion and sensitivity. Exosomes miRNAs (miR-6796-3p, miR-6763-5p, miR-4750-3p, and miR-197-3p) from PCCs could suppress proprotein convertase subtilisin kexin type 1/3 (PCSK1/3) in enteroendocrine cells, subsequently inhibiting GLP1 and GIP secretion [98].

Decreased peripheral glucose use also contributes to the pathogenesis of diabetes. PCCs-derived exosomes could inhibit the PI3K/AKT signaling pathway in muscle cells, resulting in compromised expression of glucose transporter 4 protein (GLUT4) and insulin resistance [99]. Besides, Wang et al. proved that PCCs-derived exosomal miR-let-7b-5p stimulated insulin resistance in skeletal muscle cells via targeting RNF20 and activating STAT3/FOXO1 pathway [100].

Cachexia

Cachexia is a prominent and frequent clinical feature of pancreatic cancer, which is mainly characterized by non-intentional weight loss, such as sarcopenia and adipopenia [101]. Importantly, cachexia is associated with poor life quality and even death in pancreatic cancer patients, attracting massive attention from clinicians and nutritionists [101]. As another systematic syndrome besides diabetes, the cachexia process in pancreatic cancer could also be partially attributed to exosomes.

Weight loss and malnutrition due to cachexia are often caused by muscle, fat, and even bone catabolism. Tumor-released extracellular surface heat shock protein 70 (HSP70) and Hsp90 were identified as key factors causing myotube atrophy and muscle wasting [102,103,104]. PCCs released markedly increased levels Hsp70/Hsp90-containing EVs, which targeted in myotubes and resulted in the loss of the myosin heavy chain and myotube diameter via upregulating of p38 MAPK, C/EBPβ, and LC3-II [103]. Another study by Yang et al. reported that ZIP4 could induce muscle loss and cachexia by activating CREB-regulated expression of RAB27B, a key regulator required for EVs releasing in PCCs. Those exosomes containing high levels of HSP70 and HSP90 could stimulate Toll-like receptor 4 (TLR4) on the recipient muscle cells, finally promoting p38 MAPK-mediated muscle catabolism and cachexia [105].

Additionally, the abovementioned adrenomedullin in PCCs-derived exosomes could activate ERK1/2 and p38 MAPK pathways in adipocytes, contributing to lipolysis in adipose tissue and weight loss [106]. Another study by Shibata et al. reported that intravenous injection of PCCs-derived EVs could promote lipolysis and negatively affect body weight in mice model. They also revealed that EVs in PC patients’ serum exhibit higher ITGA6 and ITGB1 expression levels, which allowed them to be preferentially distributed to the adipose tissue [107].Moreover, compared with the healthy control, the expression of genes for browning, lipolysis, fibrosis, and acute inflammation was upregulated in subcutaneous adipocytes exposed to plasma exosomes from pancreatic cancer patients [108].

A recent report also suggested that PCCs-derived exosomal miR-125a-5p could induce osteoclast differentiation and cancer-related bone loss [109], further indicating the role of exosomes in pancreatic cancer patients accompanied with cachexia.

Clinical values of exosomes in diagnosis, monitoring, and therapy

Diagnosis and monitoring

Due to non-specific symptoms at early stage, about 80% of pancreatic cancer patients are diagnosed to be the advanced stage when initially evaluated. Therefore, early detection is significant and promising for improving the dismal prognosis [110, 111]. Benefiting from the protective role of lipid bilayer membrane and CD47 transmembrane protein, exosomes carrying specific information about pancreatic cancer is relatively stable in the circulating system and less likely eliminated by monocytes [112], providing the possibility of early diagnosis via liquid biopsy (Table 1).

Table 1 Diagnosis and monitoring the status of pancreatic cancer patients via Exosomes

Studies based on proteomics suggested that protein signatures of exosomes derived from patients’ plasma could reflect the pathophysiology of pancreatic cancer [113]. Also, the phenotype signatures of plasma-derived exosomes in pancreatic cancer patients were proved to have high diagnostic accuracy and strongly correlated with tumor stages [114]. Through analyzing the levels of glypican 1 (GPC1) exosomes in serum, a study involving German patients revealed that GPC1 exosomes could perfectly distinguish pancreatic cancer from healthy donors and individuals with benign pancreatic diseases. The AUC was 1.0 (95% CI: 0.988–1.0), with 100% sensitivity and 100% specificity [115]. By performing immune lipoplex nanoparticle biochip assay, Li et al. validated that circulating exosomes GPC1 could serve as a viable biomarker for PC which was validated in clinical cohorts at multiple hospitals [116]. While GPC1 alone showed high accuracy, combining Exosomes GPC1, CD82, and serum CA199 levels could also realize better diagnostic efficiency, achieving an AUC of 0.942 (95% CI: 0.882–1.000) [117]. Similarly, the combination of exosomal markers EGFR, EpCAM, WNT2, and GPC1 demonstrated excellent diagnostic efficiency, achieving an AUC of 1.0, with a sensitivity of 82% and a specificity of 90% [118]. However, another study suggested that circulating exosomes GPC1 was not diagnostic for pancreatic cancer, but indicated high levels of miR-181a, miR-10b, miR-21, miR-30c, and low levels of miR-let7a in plasma exosomes could effectively differentiate pancreatic cancer from healthy donors and chronic pancreatitis patients [119]. The variations between different groups’ findings can be attributed to several factors, including differences in blood preservation conditions, exosome extraction methods, protein/RNA quantification techniques, and population demographics. These factors can significantly influence the results and lead to discrepancies between studies. Currently, the role of exosomes in liquid biopsy field is gaining momentum. Nakamura et al. [120]. adopted exosome-based liquid biopsy to establish a 13 miRNAs signature which not only showed high diagnostic accuracy (AUC = 0.98 for training cohort; AUC = 0.93 for validation cohort) for all stages of PDAC but also successfully identified CA19-9 negative patients (AUC = 0.96, sensitivity = 91% and specificity = 90%), demonstrating the potential superiority than CA19-9. Besides, when combined with CA19-9 levels, the diagnostic accuracy significantly improved (AUC = 0.99 compared to AUC = 0.86 for CA19-9 alone).

Other relevant studies also suggested that plasma exosomes microRNAs (miR-409, miR-191, miR-21, miR-451a, miR-19b, miR-335-5p, miR-340-5p, miR-451,miR-720), mRNAs (CK18, CD63, FGA, KRT19, HIST1H2BK, ITIH2, MARCH2, CLDN1, MAL2, TIMP1, WASF2, and ARF6), small nucleolar RNAs (SNORA74A and SNORA25), and proteins (ZIP4, EphA2, ALIX, ADAM8) could assist to pancreatic cancer diagnosis [120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135]. Besides blood, exosomes in saliva provided information about pancreatic cancer as well. Exosomes from the saliva of mice models bearing orthotopically implanted pancreatic cancer had higher mRNAs (Apbblip, Aspn, Incenp, Daf2, and Foxp1) [136]. Furthermore, Saliva-derived exosomal miR-1246 and miR-4644 fulfilled pancreatic cancer diagnosis with 83.3% sensitivity and 92.3% specificity [137], which revealed a more convenient and non-invasive method for potentially diagnosing pancreatic cancer (Table 1).

Recently, a novel method for mutational protein analysis of single extracellular vesicle has been developed, which can capture individual extracellular vesicles and analyze multiple protein markers within them based on antibody capture and fluorescence imaging [138]. Researchers found KRASmut and P53mut could serve as extracellular vesicle markers for detecting pancreatic cancer. By applying this method, PDAC with volume of merely 0.1 cubic centimeters was successfully detected, far superior to current clinical imaging detection capabilities [138].

In addition to diagnosis, accurately and dynamically monitoring the status of patients during cancer course is vital for individualized therapy and long-term survival [139] (Table 1). Lower miR-130b-5p, miR-133a-3p, miR-1273f, and higher miR-195-5p, miR-432-5p, miR-1229-3p in serum or plasma exosomes related to early recurrence after surgery in patients with pancreatic cancer [140]. Similarly, higher exosomes EphA2 in plasma also could predict early pancreatic cancer recurrence after surgery [141]. Besides predicting early recurrence, the panel containing serum exosomal miR-1299, GAPDH (mRNA), circulating mutant KRAS allele fraction, and CA199 levels could effectively predict occult distant metastasis of pancreatic cancer, which was significantly better than imaging alone [121]. Decreased exosomal EphA2 levels in plasma could reflect good/partial response to neoadjuvant therapy in pancreatic cancer patients [125]. Besides that, increased KRAS mutant allele fraction in exosomal DNA could predict disease progression during neoadjuvant treatment, which was superior to elevated CA19-9 levels [142].

However, there is no available assay approved by Food and Drug Administration for pancreatic cancer diagnosis and monitoring. Hence, the results of previous publications remain further validation in high-quality clinical trials. Additionally, the methods for isolating exosomes in clinical use should also be optimized and unified [143].

Therapy

Compared with free drug and synthetic nanoparticles such as liposomes, exosomes, which are naturally generated nanoscale EVs with innate properties well suited to lipids, shuttle proteins, and nucleic acids between cells [144], can be loaded with drugs while freely traversing across dense tumor stroma, avoiding degeneration and immune clearance [145, 146]. Additionally, PCCs-derived exosomes were preferentially taken up by tumor tissue in orthotopic mice models [147, 148]. Such enhanced uptake of exosomes might be mediated by oncogenic KRAS, which promoted macropinocytosis in pancreatic cancer [112, 149]. Therefore, exosomes provided promising novel opportunities for pancreatic cancer treatment. However, their therapeutic effectiveness is restricted due to their inability to specifically target tumor cells and a high rate of clearance by the mononuclear phagocytic system. To overcome such limitations, Creeden et al. developed novel engineered “Smart Exosomes” which increased binding ability to αvβ3 on PC cells, resulting in enhanced cellular uptake and increased chemotherapy response in both in vivo and in vitro models [150].

PCCs-derived exosomes loading with gemcitabine exhibited superior results in suppressing tumor growth in xenograft mice models than free gemcitabine. Moreover, compared with free gemcitabine administrated systemically, those autologous exosomes specifically carried gemcitabine to pancreatic cancer tissue and significantly decreased side effects [151]. Benefiting from the homing property to parent tumor cells, loading paclitaxel into the exosomes from PANC-1 cell line with surface modifications could achieve better efficiency than free paclitaxel [152]. Another recent study suggested that loading paclitaxel and gemcitabine monophosphate into the engineered exosomes might have superior anticancer effects than loading gemcitabine alone [153]. Furthermore, Zhao et al. established a novel drug delivery system based on M1 macrophage-derived exosome, which provided an effective therapeutic strategy against drug-resistant pancreatic cancer [154]. Additionally, exosomes derived from bone marrow mesenchymal stem cells (BM-MSCs) have natural anticancer properties via circ_0030167 [141]. BM-MSCs exosomes could also be loaded with miR-124, which targeted EZH2 in PCCs and sensitized pancreatic cancer to chemotherapy in vitro and in mice models [155]. Recently, Hasoglu et al. firstly reported that pancreatic islet-derived exosomes can selectively kill pancreatic cancer cells without harming healthy cells [156]. These advancements underscore the promising potential of exosome-based therapies in enhancing the efficacy and specificity of pancreatic cancer treatments.

Engineered exosomes were promising in immunotherapy as well [2]. Galectin-9 expressed in PCCs promoted M2 phenotype of macrophages and immunosuppression. In the meanwhile, some chemotherapeutic drugs such as oxaliplatin could induce immunogenic PCCs death and activate antitumor immune response. After loading oxaliplatin and antisense oligonucleotides targeting Galectin-9 into exosomes which were originated from MSCs, they exhibited superior antitumor effects than oxaliplatin and Galectin-9 inhibition alone in mice models with orthotopic tumor [157]. Another recent study showed that loading immunogenic peptides and CCL22 siRNA into exosomes could expand CD8 + T cells and repress regulatory T cells within tumor microenvironment upon intramuscular administration [158]. In addition, precise photodynamic treatment could also be realized via exosomes. Under photoacoustic imaging guidance, engineered exosomes with chlorin e6 (photosensitizer) generated high amounts of ROS within the tumor. Consequently, disrupted exosomes membrane components could stimulate the immune system as tumor antigens [159]. Given the key role of exosomes in constructing premetastatic niches, loading pirfenidone into PCC-derived exosomes could alleviate liver fibrosis and suppress liver metastasis [160].

Besides chemotherapy and immunotherapy, targeted therapy against oncogenic KRAS, the most important driver mutation in pancreatic cancer, has attracting amounts attention from oncologists and pharmacologists for over three decades. Despite the breakthrough of KRAS G12C inhibitors in treating non-small-cell lung cancer, other KRAS-targeted therapy still has poor clinical efficacy [161, 162]. Specifically delivering antisense oligonucleotides or CRISPR/Cas9 via exosomes to target mutant KRAS in PCCs may also be a novel way to treat pancreatic cancer [163]. In orthotopic mice models, exosomes derived from MSCs carrying short interfering RNA effectively suppressed tumor growth through targeting KRAS G12D, the most common mutation subtype in pancreatic cancer [112, 164]. Moreover, a relevant phase I clinical trial is currently activating (NCT03608631). Additionally, pancreatic cancer progression could also be restrained by other types of engineered exosomes, such as PCCs-derived Exosomes loading with short interfering RNA against P21-activated kinase 4 (PAK4) [165], and BM-MSCs loading with miR-1231 [166]. Exosome-based treatments for pancreatic cancer were summarised in Table 2.

Table 2 Overview of exosome-based treatments for pancreatic cancer

However, there are many challenges of exosome-based therapy at present. Besides therapeutic RNAs and proteins, engineered exosomes also carried other molecules. Therefore, prion particles, oncogenes, and viral miRNAs might be co-delivered to recipient cells, producing side effects. Additionally, the normal intercellular communication mediated by endogenous exosomes might also be interfered by exogenous exosomes [146]. In general, exosomes-based therapy is promising but remains further study.

Exosome characterization techniques

Extracellular vehicles, including exosomes, can be characterized using a variety of advanced techniques [167]. High-resolution imaging methods like scanning electron microscopy (SEM), transmission electron microscopy (TEM), cryo-electron microscopy (Cryo-EM), and atomic force microscopy (AFM) are used for morphological characterization to visualize EVs’ structural features [168,169,170]. Proteomic analysis through mass spectrometry, Western blot, and enzyme-linked immunosorbent assay (ELISA) identifies key proteins such as CD9, CD63, and CD81 [171], ensuring purity and origin assessment. Particle size analysis of exosomes is mainly performed by dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), and flow cytometry, these techniques measure the size distribution and concentration of exosomes, thus further characterizing the physical properties of exosomes [172, 173]. The lipidomics characterization of exosomes is conducted using chromatography techniques combined with mass spectrometry (LC-MS/MS). This approach enables the analysis of the lipid composition of exosomes, revealing their role in the structure and function of cell membranes [174, 175]. Emerging techniques, such as single-exosome detection methods, are also being developed to enhance the precision and detail of exosome analysis [174, 176]. Notably, innovative isolation techniques such as microfluidic technology, immunoaffinity capture, and resistive pulse sensing have been highlighted [177, 178]. These methods offer higher purity and specificity in isolating extracellular vesicles, thereby improving the reliability of subsequent analyses.

When applying these characterization techniques to different tissue types, the extraction methods may need to be adjusted based on the tissue’s specific properties. For example, the dense nature of pancreatic cancer tissue requires modifications to the standard protocols to effectively isolate exosomes. It is recommended to moderately increase the concentration of collagenase, the shaking speed, and the incubation time when extracting exosomes from pancreatic cancer tissue compared to other reported tissues such as gliomas and melanomas [76, 179, 180].

Discussion

Tumor microenvironment is a highly heterozygous organization. Multiple cells and ECM coordinate with each other, synergistically promoting pancreatic cancer progression. Exosomes are vital mediators to realize rapid and abundant communication among different PCCs, CAFs, and immune cells within tumor microenvironment. Recently, a novel method showed that pure exosomes could be effectively isolated from tissues directly [181, 182]. Therefore, the role of exosomes in the tumor microenvironment are ready to be clear in the future study. Besides that, liquid biopsy via circulating exosomes is significant in diagnosis and stratifying patients to achieve individualized treatment. However, there are still some barriers restraining its clinical application. For example, exosomes could be extracted from blood or other body fluid through a variety of methods, such as ultracentrifugation and size exclusion chromatography, which may contribute to unrepeatable results in different clinical studies. Therefore, clinical standard in isolating exosomes from body fluid remains to be worked out.

The development of standardized protocols and methodologies is paramount to advancing EV research in pancreatic cancer. Consistent and reproducible results across different studies are essential to validate the potential of EVs as reliable biomarkers and therapeutic agents [144, 183]. Current research suffers from variability in isolation techniques, characterization methods, and analytical approaches [183]. Establishing consensus guidelines for these processes will enhance the comparability of data and facilitate the translation of EV-based diagnostics and therapies into clinical practice.

To validate the diagnostic potential of EV liquid biopsies, multicenter studies with large sample sizes are crucial [184, 185]. Such studies can provide robust evidence of the sensitivity and specificity of EV-derived biomarkers in detecting pancreatic cancer at various stages. There is also an urgent need to expedite Phase I and Phase II clinical trials to assess the safety, efficacy, and optimal delivery methods of engineered exosome-based therapy [186, 187]. Early-phase clinical trials will provide critical insights into the therapeutic potential of exosome and identify any potential challenges that need to be addressed before large-scale implementation [188].

In addition to exosomes, circulating DNA (cfDNA), metabolites, and circulating RNA (cfRNA) offer complementary insights into the tumor microenvironment and tumor burden [189, 190]. Combining these biomarkers in a multi-omics approach can enhance the sensitivity and specificity of liquid biopsies. Utilizing integrated multi-omics data models can provide a comprehensive view of the molecular landscape of pancreatic cancer, improving diagnostic accuracy and enabling precise patient stratification for personalized treatment [191].

Conclusion

Exosomes could carry a variety of cargos including proteins, RNAs, metabolites, mediating the communication among heterogeneous cellular components within tumor microenvironment or even between different organs. The true potential utility of extracellular vesicles, particularly exosomes, lies in their ability to serve as both diagnostic biomarkers and therapeutic agents. Engineered exosomes are promising to implement targeted therapy, and circulating exosomes in plasma could be detected as liquid biopsy to accurately diagnose or monitor pancreatic cancer patients. However, for this promise to become a reality, additional work is needed. Standardization of isolation and characterization techniques, large-scale multicenter validation, and early-phase clinical trials are essential steps. Combining exosomal biomarkers with other biological materials like circulating DNA (cfDNA), metabolites, and circulating RNA (cfRNA) in a multi-omics approach can further enhance their clinical utility. These efforts will pave the way for innovative biomarkers discovery and the development of effective therapeutic strategies, ultimately improving patient outcomes in pancreatic cancer.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

PDAC:

Pancreatic Ductal Adenocarcinoma

TME:

Tumor Microenvironment

EVs:

Extracellular Vesicles

MVB:

Multivesicular Body

PCC:

Pancreatic Cancer Cells

MDSC:

Myeloid-Derived Suppressor Cells

DC:

Dendritic Cells

NK:

Natural Killer cells

miRNA:

MicroRNA

HSP:

Heat Shock Protein

GPC1:

Glypican 1

EpCAM:

Epithelial Cell Adhesion Molecule

KRAS:

Kirsten Rat Sarcoma Viral Oncogene Homolog

cfDNA:

Cell-Free DNA

cfRNA:

Cell-Free RNA

GLUT4:

Glucose Transporter Type 4

TLR4:

Toll-Like Receptor 4

CREB:

cAMP Response Element-Binding Protein

IL-8:

Interleukin 8

VEGF-C:

Vascular Endothelial Growth Factor C

TNF-α:

Tumor Necrosis Factor Alpha

IL-12:

Interleukin 12

EMT:

Epithelial-Mesenchymal Transition

CD:

Cluster of Differentiation

LncRNA:

Long Non-Coding RNA

RNA:

Ribonucleic Acid

CD63:

Cluster of Differentiation 63

CD81:

Cluster of Differentiation 81

FGFRL1:

Fibroblast Growth Factor Receptor Like 1

PI3K:

Phosphoinositide 3-Kinase

AKT:

Protein Kinase B

mTOR:

Mechanistic Target of Rapamycin

XIAP:

X-Linked Inhibitor of Apoptosis Protein

circRNA:

Circular RNA

ROBO1:

Roundabout Guidance Receptor 1

SRGAP2:

SLIT-ROBO Rho GTPase Activating Protein 2

LN:

Lymph Node

cfMiRNA:

Cell-Free MicroRNA

SNORA:

Small Nucleolar RNA

ADAM8:

A Disintegrin and Metalloprotease Domain 8

ZIP4:

Zinc Transporter 4

PCSK1/3:

Proprotein Convertase Subtilisin/Kexin Type 1/3

RNF20:

Ring Finger Protein 20

STAT3:

Signal Transducer and Activator of Transcription 3

FOXO1:

Forkhead Box O1

CRISPR/Cas9:

Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR Associated Protein 9

MSC:

Mesenchymal Stem Cells

PAK4:

p21-Activated Kinase 4

BM-MSCs:

Bone Marrow Mesenchymal Stem Cells

NTA:

Nanoparticle Tracking Analysis

DLS:

Dynamic Light Scattering

SEM:

Scanning Electron Microscopy

TEM:

Transmission Electron Microscopy

Cryo-EM:

Cryo-Electron Microscopy

AFM:

Atomic Force Microscopy

LC-MS/MS:

Liquid Chromatography–Mass Spectrometry/Mass Spectrometry

PBMC:

Peripheral Blood Mononuclear Cells

FOLFIRINOX:

A combination chemotherapy regimen including Folinic Acid (Leucovorin), Fluorouracil (5-FU), Irinotecan, and Oxaliplatin

GIP:

Glucose-Dependent Insulinotropic Peptide

GLP-1:

Glucagon-Like Peptide-1

ADCY1:

Adenylate Cyclase 1

EPAC2:

Exchange Protein Directly Activated by cAMP 2

ER:

Endoplasmic Reticulum

ECM:

Extracellular Matrix

References

  1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–63.

    Article  PubMed  Google Scholar 

  2. Zhang M, Hu S, Liu L, Dang P, Liu Y, Sun Z, et al. Engineered exosomes from different sources for cancer-targeted therapy. Signal Transduct Target Ther. 2023;8(1):124.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Huang L, Jansen L, Balavarca Y, Molina-Montes E, Babaei M, van der Geest L, et al. Resection of pancreatic cancer in Europe and USA: an international large-scale study highlighting large variations. Gut. 2019;68(1):130–9.

    Article  PubMed  Google Scholar 

  4. Wu W, Miao Y, Yang Y, Lou W, Zhao Y, on behalf of Chinese Pancreatic Surgery Association CSoS, Chinese Medical Association. Real-world study of surgical treatment of pancreatic cancer in China: annual report of China Pancreas Data Center (2016–2020). Journal of Pancreatology. 2022;5(1):1–9.

  5. Alva-Ruiz R, Yohanathan L, Yonkus JA, Abdelrahman AM, Gregory LA, Halfdanarson TR, et al. Neoadjuvant Chemotherapy Switch in Borderline Resectable/Locally Advanced Pancreatic Cancer. Ann Surg Oncol. 2022;29(3):1579–91.

    Article  PubMed  Google Scholar 

  6. Perri G, Prakash L, Qiao W, Varadhachary GR, Wolff R, Fogelman D, et al. Response and Survival Associated with First-line FOLFIRINOX vs Gemcitabine and nab-Paclitaxel Chemotherapy for localized pancreatic ductal adenocarcinoma. JAMA Surg. 2020;155(9):832–9.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, et al. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol. 2023;20(5):318–37.

    Article  PubMed  Google Scholar 

  8. Conroy T, Lambert A, Ducreux M. Adjuvant and neoadjuvant approaches in pancreatic cancer. Curr Opin Oncol. 2023;35(4):326–33.

    Article  CAS  PubMed  Google Scholar 

  9. Sohal DPS, Kennedy EB, Khorana A, Copur MS, Crane CH, Garrido-Laguna I, et al. Metastatic pancreatic Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol. 2018;36(24):2545–56.

    Article  PubMed  Google Scholar 

  10. Klein-Brill A, Amar-Farkash S, Lawrence G, Collisson EA, Aran D. Comparison of FOLFIRINOX vs Gemcitabine Plus Nab-Paclitaxel as First-Line chemotherapy for metastatic pancreatic ductal adenocarcinoma. JAMA Netw Open. 2022;5(6):e2216199.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Zeng S, Pottler M, Lan B, Grutzmann R, Pilarsky C, Yang H. Chemoresistance in Pancreatic Cancer. Int J Mol Sci. 2019;20(18).

  12. Buwenge M, Macchia G, Arcelli A, Frakulli R, Fuccio L, Guerri S, et al. Stereotactic radiotherapy of pancreatic cancer: a systematic review on pain relief. J Pain Res. 2018;11:2169–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hu ZI, O’Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2024;21(1).

  14. Wang-Gillam A, Lim K-H, McWilliams R, Suresh R, Lockhart AC, Brown A, et al. Defactinib, Pembrolizumab, and Gemcitabine in patients with Advanced Treatment Refractory Pancreatic Cancer: a phase I dose escalation and expansion study. Clin Cancer Res. 2022;28(24):5254–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Reiss KA, Mick R, Teitelbaum U, O’Hara M, Schneider C, Massa R, et al. Niraparib plus Nivolumab or Niraparib plus Ipilimumab in patients with platinum-sensitive advanced pancreatic cancer: a randomised, phase 1b/2 trial. Lancet Oncol. 2022;23(8):1009–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance olaparib for germline BRCA-Mutated metastatic pancreatic Cancer. N Engl J Med. 2019;381(4):317–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bazan-Peregrino M, Garcia-Carbonero R, Laquente B, Álvarez R, Mato-Berciano A, Gimenez-Alejandre M et al. VCN-01 disrupts pancreatic cancer stroma and exerts antitumor effects. J Immunother Cancer. 2021;9(11).

  18. Musher BL, Rowinsky EK, Smaglo BG, Abidi W, Othman M, Patel K, et al. LOAd703, an oncolytic virus-based immunostimulatory gene therapy, combined with chemotherapy for unresectable or metastatic pancreatic cancer (LOKON001): results from arm 1 of a non-randomised, single-centre, phase 1/2 study. Lancet Oncol. 2024;25(4):488–500.

    Article  CAS  PubMed  Google Scholar 

  19. Pant S, Wainberg ZA, Weekes CD, Furqan M, Kasi PM, Devoe CE, et al. Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: the phase 1 AMPLIFY-201 trial. Nat Med. 2024;30(2):531–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bauer MR, Bright EE, MacDonald JJ, Cleary EH, Hines OJ, Stanton AL. Quality of life in patients with pancreatic Cancer and their caregivers: a systematic review. Pancreas. 2018;47(4):368–75.

    Article  PubMed  Google Scholar 

  21. Thomas D, Radhakrishnan P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol Cancer. 2019;18(1):14.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Flint TR, Janowitz T, Connell CM, Roberts EW, Denton AE, Coll AP, et al. Tumor-Induced IL-6 reprograms host metabolism to suppress anti-tumor immunity. Cell Metab. 2016;24(5):672–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Asao T, Tobias GC, Lucotti S, Jones DR, Matei I, Lyden D. Extracellular vesicles and particles as mediators of long-range communication in cancer: connecting biological function to clinical applications. Extracell Vesicles Circulating Nucleic Acids. 2023;4(3):461–85.

    Article  CAS  Google Scholar 

  24. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.

    Article  CAS  PubMed  Google Scholar 

  25. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–28.

    Article  PubMed  Google Scholar 

  26. Raposo G, Stahl PD. Extracellular vesicles - on the cusp of a new language in the biological sciences. Extracell Vesicles Circulating Nucleic Acids. 2023;4(2):240–54.

    Article  CAS  Google Scholar 

  27. Couto N, Elzanowska J, Maia J, Batista S, Pereira CE, Beck HC, et al. IgG + extracellular vesicles measure therapeutic response in Advanced Pancreatic Cancer. Cells. 2022;11:18.

    Article  Google Scholar 

  28. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021;20(1):131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478).

  30. Farahmand Y, Tehrany PM, Nazari A, Nava ZH, Alsaffar MF, Yazdani O, et al. A comprehensive survey into the role of exosomes in pancreatic cancer; from the origin of cancer to the progress and possibility of diagnosis and treatment. Pathol Res Pract. 2023;245:154465.

    Article  CAS  PubMed  Google Scholar 

  31. McAndrews KM, Kalluri R. Mechanisms associated with biogenesis of exosomes in cancer. Mol Cancer. 2019;18(1):52.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kok VC, Yu C-C. Cancer-Derived exosomes: their role in Cancer Biology and Biomarker Development. Int J Nanomed. 2020;15:8019–36.

    Article  CAS  Google Scholar 

  33. Wang K, Wang X, Pan Q, Zhao B. Liquid biopsy techniques and pancreatic cancer: diagnosis, monitoring, and evaluation. Mol Cancer. 2023;22(1):167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, et al. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21(1):56.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Liang G, Zhu Y, Ali DJ, Tian T, Xu H, Si K, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnol. 2020;18(1):10.

    Article  CAS  Google Scholar 

  36. Raguraman R, Bhavsar D, Kim D, Ren X, Sikavitsas V, Munshi A, et al. Tumor-targeted exosomes for delivery of anticancer drugs. Cancer Lett. 2023;558:216093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lan B, Zeng S, Grutzmann R, Pilarsky C. The role of exosomes in Pancreatic Cancer. Int J Mol Sci. 2019;20(18).

  38. Liu X, Xu J, Zhang B, Liu J, Liang C, Meng Q, et al. The reciprocal regulation between host tissue and immune cells in pancreatic ductal adenocarcinoma: new insights and therapeutic implications. Mol Cancer. 2019;18(1):184.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Rubin SJS, Sojwal RS, Gubatan J, Rogalla S. The Tumor Immune Microenvironment in Pancreatic Ductal Adenocarcinoma: neither Hot nor Cold. Cancers (Basel). 2022;14(17).

  40. Yin Z, Zhou Y, Ma T, Chen S, Shi N, Zou Y, et al. Down-regulated lncRNA SBF2-AS1 in M2 macrophage-derived exosomes elevates mir-122-5p to restrict XIAP, thereby limiting pancreatic cancer development. J Cell Mol Med. 2020;24(9):5028–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang K, Li Y-J, Peng L-J, Gao H-F, Liu L-M, Chen H. M2 macrophage-derived exosomal miR-193b-3p promotes progression and glutamine uptake of pancreatic cancer by targeting TRIM62. Biol Direct. 2023;18(1):1.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Chen Y, Lei Y, Li J, Wang X, Li G. Macrophage-derived exosomal microRNAs promote metastasis in pancreatic ductal adenocarcinoma. Int Immunopharmacol. 2024;129:111590.

    Article  CAS  PubMed  Google Scholar 

  43. Guo Y, Cui J, Liang X, Chen T, Lu C, Peng T. Pancreatic cancer stem cell-derived exosomal miR-210 mediates macrophage M2 polarization and promotes gemcitabine resistance by targeting FGFRL1. Int Immunopharmacol. 2024;127:111407.

    Article  CAS  PubMed  Google Scholar 

  44. He Z, Wang J, Zhu C, Xu J, Chen P, Jiang X, et al. Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated pancreatic cancer cell proliferation and metastasis. Cancer Lett. 2022;548:215751.

    Article  CAS  PubMed  Google Scholar 

  45. Zech D, Rana S, Büchler MW, Zöller M. Tumor-exosomes and leukocyte activation: an ambivalent crosstalk. Cell Commun Signal. 2012;10(1):37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Steeber DA, Venturi GM, Tedder TF. A new twist to the leukocyte adhesion cascade: intimate cooperation is key. Trends Immunol. 2005;26(1).

  47. Rose DM, Alon R, Ginsberg MH. Integrin modulation and signaling in leukocyte adhesion and migration. Immunol Rev. 2007;218:126–34.

    Article  CAS  PubMed  Google Scholar 

  48. Basso D, Gnatta E, Padoan A, Fogar P, Furlanello S, Aita A, et al. PDAC-derived exosomes enrich the microenvironment in MDSCs in a SMAD4-dependent manner through a new calcium related axis. Oncotarget. 2017;8(49):84928–44.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Shen T, Huang Z, Shi C, Pu X, Xu X, Wu Z, et al. Pancreatic cancer-derived exosomes induce apoptosis of T lymphocytes through the p38 MAPK-mediated endoplasmic reticulum stress. FASEB J. 2020;34(6):8442–58.

    Article  CAS  PubMed  Google Scholar 

  50. Ye L, Zhu Z, Chen X, Zhang H, Huang J, Gu S et al. The importance of exosomal PD-L1 in Cancer Progression and its potential as a therapeutic target. Cells. 2021;10(11).

  51. Xiang J, Zheng B, Zhao L, He Y, Lou F, Li R et al. Exo70 promotes the Invasion of Pancreatic Cancer cells via the regulation of Exosomes. Cancers (Basel). 2024;16(2).

  52. Hu S, Ma J, Su C, Chen Y, Shu Y, Qi Z, et al. Engineered exosome-like nanovesicles suppress tumor growth by reprogramming tumor microenvironment and promoting tumor ferroptosis. Acta Biomater. 2021;135:567–81.

    Article  CAS  PubMed  Google Scholar 

  53. Capello M, Vykoukal JV, Katayama H, Bantis LE, Wang H, Kundnani DL, et al. Exosomes harbor B cell targets in pancreatic adenocarcinoma and exert decoy function against complement-mediated cytotoxicity. Nat Commun. 2019;10(1):254.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Zhou M, Chen J, Zhou L, Chen W, Ding G, Cao L. Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell Immunol. 2014;292(1–2):65–9.

    Article  CAS  PubMed  Google Scholar 

  55. Ding G, Zhou L, Qian Y, Fu M, Chen J, Chen J, et al. Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget. 2015;6(30):29877–88.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Jia X, Xi J, Tian B, Zhang Y, Wang Z, Wang F, et al. The tautomerase activity of Tumor Exosomal MIF promotes pancreatic Cancer Progression by modulating MDSC differentiation. Cancer Immunol Res. 2024;12(1):72–90.

    Article  CAS  PubMed  Google Scholar 

  57. Che SPY, Park JY, Stokol T. Tissue factor-expressing tumor-derived extracellular vesicles activate quiescent endothelial cells via protease-activated Receptor-1. Front Oncol. 2017;7:261.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, et al. Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 2018;37(1):177.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Chen K, Wang Q, Liu X, Wang F, Yang Y, Tian X. Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. Int J Biol Sci. 2022;18(3):1220–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wang L, Yang L, Zhuang T, Shi X. Tumor-derived exosomal miR-29b reduces angiogenesis in pancreatic Cancer by silencing ROBO1 and SRGAP2. J Immunol Res. 2022;2022:4769385.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Xu J-Q, Liu Q-Q, Huang S-Y, Duan C-Y, Lu H-B, Cao Y, et al. The lymphatic system: a therapeutic target for central nervous system disorders. Neural Regen Res. 2023;18(6):1249–56.

    Article  CAS  PubMed  Google Scholar 

  62. Weidle UH, Birzele F, Kollmorgen G, Rüger R. The multiple roles of exosomes in Metastasis. Cancer Genomics Proteom. 2017;14(1).

  63. Liu S-L, Sun P, Li Y, Liu S-S, Lu Y. Exosomes as critical mediators of cell-to-cell communication in cancer pathogenesis and their potential clinical application. Transl Cancer Res. 2019;8(1):298–311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang L, Li L, Zhu G. Role of Extracellular vesicles on Cancer Lymphangiogenesis and Lymph Node Metastasis. Front Oncol. 2021;11:721785.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chen C, Luo Y, He W, Zhao Y, Kong Y, Liu H, et al. Exosomal long noncoding RNA LNMAT2 promotes lymphatic metastasis in bladder cancer. J Clin Invest. 2020;130(1):404–21.

    Article  CAS  PubMed  Google Scholar 

  66. Liu T, Li P, Li J, Qi Q, Sun Z, Shi S, et al. Exosomal and intracellular miR-320b promotes lymphatic metastasis in esophageal squamous cell carcinoma. Mol Ther Oncolytics. 2021;23:163–80.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Xiao Z, Feng X, Zhou Y, Li P, Luo J, Zhang W et al. Exosomal mir-10527-5p inhibits Migration, Invasion, Lymphangiogenesis and Lymphatic Metastasis by affecting Wnt/β-Catenin signaling via Rab10 in esophageal squamous cell carcinoma. Int J Nanomed. 2023;18.

  68. Zhou C-F, Ma J, Huang L, Yi H-Y, Zhang Y-M, Wu X-G, et al. Cervical squamous cell carcinoma-secreted exosomal mir-221-3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene. 2019;38(8):1256–68.

    Article  CAS  PubMed  Google Scholar 

  69. Wang J, Gong X, Yang L, Li L, Gao X, Ni T, et al. Loss of exosomal miR-26a-5p contributes to endometrial cancer lymphangiogenesis and lymphatic metastasis. Clin Transl Med. 2022;12(5):e846.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. An HJ, Lee JS, Yang JW, Kim MH, Na JM, Song DH. RAB27A and RAB27B expression may predict Lymph Node Metastasis and Survival in patients with gastric Cancer. Cancer Genomics Proteom. 2022;19(5):606–13.

    Article  CAS  Google Scholar 

  71. Ding J, Xu Z, Zhang Y, Tan C, Hu W, Wang M, et al. Exosome-mediated miR-222 transferring: an insight into NF-κB-mediated breast cancer metastasis. Exp Cell Res. 2018;369(1):129–38.

    Article  CAS  PubMed  Google Scholar 

  72. Zhou X, Zhong F, Yan Y, Wu S, Wang H, Liu J et al. Pancreatic Cancer cell-derived exosomes promote lymphangiogenesis by downregulating ABHD11-AS1 expression. Cancers (Basel). 2022;14(19).

  73. Satake T, Suetsugu A, Nakamura M, Kunisada T, Saji S, Moriwaki H, et al. Color-coded imaging of the Fate of Cancer-cell-derived Exosomes during Pancreatic Cancer metastases in a nude-mouse model. Anticancer Res. 2019;39(8):4055–60.

    Article  CAS  PubMed  Google Scholar 

  74. Houg DS, Bijlsma MF. The hepatic pre-metastatic niche in pancreatic ductal adenocarcinoma. Mol Cancer. 2018;17(1):95.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Xie Z, Gao Y, Ho C, Li L, Jin C, Wang X et al. Exosome-delivered CD44v6/C1QBP complex drives pancreatic cancer liver metastasis by promoting fibrotic liver microenvironment. Gut. 2021.

  78. Maia J, Otake AH, Pocas J, Carvalho AS, Beck HC, Magalhaes A, et al. Transcriptome reprogramming of CD11b(+) bone marrow cells by pancreatic Cancer Extracellular vesicles. Front Cell Dev Biol. 2020;8:592518.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Chen W, Peng W, Wang R, Bai S, Cao M, Xiong S, et al. Exosome-derived tRNA fragments tRF-GluCTC-0005 promotes pancreatic cancer liver metastasis by activating hepatic stellate cells. Cell Death Dis. 2024;15(1):102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wang G, Li J, Bojmar L, Chen H, Li Z, Tobias GC, et al. Tumour extracellular vesicles and particles induce liver metabolic dysfunction. Nature. 2023;618(7964):374–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Armacki M, Polaschek S, Waldenmaier M, Morawe M, Ruhland C, Schmid R, et al. Protein kinase D1, reduced in human pancreatic tumors, increases secretion of small extracellular vesicles from Cancer cells that promote metastasis to lung in mice. Gastroenterology. 2020;159(3):1019–e3522.

    Article  CAS  PubMed  Google Scholar 

  82. Novo D, Heath N, Mitchell L, Caligiuri G, MacFarlane A, Reijmer D, et al. Mutant p53s generate pro-invasive niches by influencing exosome podocalyxin levels. Nat Commun. 2018;9(1):5069.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Kimoto A, Kadoi Y, Tsuruda T, Kim Y-S, Miyoshi M, Nomoto Y, et al. Exosomes in ascites from patients with human pancreatic cancer enhance remote metastasis partially through endothelial-mesenchymal transition. Pancreatology. 2023;23(4):377–88.

    Article  CAS  PubMed  Google Scholar 

  84. Roy A, Sahoo J, Kamalanathan S, Naik D, Mohan P, Kalayarasan R. Diabetes and pancreatic cancer: exploring the two-way traffic. World J Gastroenterol. 2021;27(30):4939–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Ruze R, Chen Y, Xu R, Song J, Yin X, Wang C, et al. Obesity, diabetes mellitus, and pancreatic carcinogenesis: correlations, prevention, and diagnostic implications. Biochim Biophys Acta Rev Cancer. 2023;1878(1):188844.

    Article  CAS  PubMed  Google Scholar 

  86. Paternoster S, Falasca M. The intricate relationship between diabetes, obesity and pancreatic cancer. Biochim Biophys Acta Rev Cancer. 2020;1873(1):188326.

    Article  CAS  PubMed  Google Scholar 

  87. Xiong L, Chen L, Wu L, He W, Chen D, Peng Z, et al. Lipotoxicity-induced circGlis3 impairs beta cell function and is transmitted by exosomes to promote islet endothelial cell dysfunction. Diabetologia. 2022;65(1):188–205.

    Article  CAS  PubMed  Google Scholar 

  88. Mattke J, Vasu S, Darden CM, Kumano K, Lawrence MC, Naziruddin B. Role of exosomes in Islet Transplantation. Front Endocrinol (Lausanne). 2021;12:681600.

    Article  PubMed  Google Scholar 

  89. Lega IC, Lipscombe LL, Review. Diabetes, obesity, and Cancer-Pathophysiology and Clinical implications. Endocr Rev. 2020;41(1).

  90. Cao M, Isaac R, Yan W, Ruan X, Jiang L, Wan Y, et al. Cancer-cell-secreted extracellular vesicles suppress insulin secretion through miR-122 to impair systemic glucose homeostasis and contribute to tumour growth. Nat Cell Biol. 2022;24(6):954–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Aggarwal G, Ramachandran V, Javeed N, Arumugam T, Dutta S, Klee GG et al. Adrenomedullin is up-regulated in patients with pancreatic cancer and causes insulin resistance in β cells and mice. Gastroenterology. 2012;143(6).

  92. Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, et al. Pancreatic Cancer-derived exosomes cause paraneoplastic beta-cell dysfunction. Clin Cancer Res. 2015;21(7):1722–33.

    Article  CAS  PubMed  Google Scholar 

  93. Kong F, Li L, Du Y, Zhu H, Li Z, Kong X. Exosomal adrenomedullin derived from cancer-associated fibroblasts promotes lipolysis in adipose tissue. Gut. 2018;67(12):2226–7.

    Article  PubMed  Google Scholar 

  94. Sancar G, Liu S, Gasser E, Alvarez JG, Moutos C, Kim K et al. FGF1 and insulin control lipolysis by convergent pathways. Cell Metab. 2022;34(1).

  95. Pang W, Yao W, Dai X, Zhang A, Hou L, Wang L, et al. Pancreatic cancer-derived exosomal microRNA-19a induces β-cell dysfunction by targeting ADCY1 and EPAC2. Int J Biol Sci. 2021;17(13):3622–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Su J, Pang W, Zhang A, Li L, Yao W, Dai X. Exosomal miR-19a decreases insulin production by targeting Neurod1 in pancreatic cancer associated diabetes. Mol Biol Rep. 2021.

  97. Qin W, Kang M, Li C, Zheng W, Guo Q. VNN1 overexpression in pancreatic cancer cells inhibits paraneoplastic islet function by increasing oxidative stress and inducing β–cell dedifferentiation. Oncol Rep. 2023;49(6).

  98. Zhang Y, Huang S, Li P, Chen Q, Li Y, Zhou Y, et al. Pancreatic cancer-derived exosomes suppress the production of GIP and GLP-1 from STC-1cells in vitro by down-regulating the PCSK1/3. Cancer Lett. 2018;431:190–200.

    Article  CAS  PubMed  Google Scholar 

  99. Wang L, Zhang B, Zheng W, Kang M, Chen Q, Qin W, et al. Exosomes derived from pancreatic cancer cells induce insulin resistance in C2C12 myotube cells through the PI3K/Akt/FoxO1 pathway. Sci Rep. 2017;7(1):5384.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Wang L, Li X, Wu J, Tang Q. Pancreatic Cancer-derived exosomal mir-Let-7b-5p stimulates insulin resistance in skeletal muscle cells through RNF20/STAT3/FOXO1 Axis Regulation. Diabetes Metab Syndr Obes. 2023;16:3133–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Poulia KA, Sarantis P, Antoniadou D, Koustas E, Papadimitropoulou A, Papavassiliou AG et al. Pancreatic Cancer and Cachexia-metabolic mechanisms and Novel insights. Nutrients. 2020;12(6).

  102. Zhang G, Liu Z, Ding H, Zhou Y, Doan HA, Sin KWT, et al. Tumor induces muscle wasting in mice through releasing extracellular Hsp70 and Hsp90. Nat Commun. 2017;8(1):589.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Wu HY, Trevino JG, Fang BL, Riner AN, Vudatha V, Zhang GH et al. Patient-derived pancreatic Cancer cells induce C2C12 myotube atrophy by releasing Hsp70 and Hsp90. Cells. 2022;11(17).

  104. Niu M, Song S, Su Z, Wei L, Li L, Pu W, et al. Inhibition of heat shock protein (HSP) 90 reverses signal transducer and activator of transcription (STAT) 3-mediated muscle wasting in cancer cachexia mice. Br J Pharmacol. 2021;178(22):4485–500.

    Article  CAS  PubMed  Google Scholar 

  105. Yang J, Zhang Z, Zhang Y, Ni X, Zhang G, Cui X, et al. ZIP4 promotes muscle wasting and Cachexia in mice with Orthotopic pancreatic tumors by stimulating RAB27B-Regulated release of Extracellular vesicles from Cancer cells. Gastroenterology. 2019;156(3):722–34. e6.

    Article  CAS  PubMed  Google Scholar 

  106. Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut. 2016;65(7):1165–74.

    Article  CAS  PubMed  Google Scholar 

  107. Shibata C, Otsuka M, Seimiya T, Kishikawa T, Ishigaki K, Fujishiro M. Lipolysis by pancreatic cancer-derived extracellular vesicles in cancer-associated cachexia via specific integrins. Clin Transl Med. 2022;12(11):e1089.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Sah RP, Sharma A, Nagpal S, Patlolla SH, Sharma A, Kandlakunta H, et al. Phases of metabolic and soft tissue changes in months preceding a diagnosis of pancreatic ductal adenocarcinoma. Gastroenterology. 2019;156(6):1742–52.

    Article  PubMed  Google Scholar 

  109. Zhou Y, Zhu Y, Dong X, Cao G, Li Y, Fan Y, et al. Exosomes Derived from Pancreatic Cancer cells induce osteoclast differentiation through the miR125a-5p/TNFRSF1B pathway. Onco Targets Ther. 2021;14:2727–39.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Zhou B, Xu JW, Cheng YG, Gao JY, Hu SY, Wang L, et al. Early detection of pancreatic cancer: where are we now and where are we going? Int J Cancer. 2017;141(2):231–41.

    Article  CAS  PubMed  Google Scholar 

  111. Søreide K, Ismail W, Roalsø M, Ghotbi J, Zaharia C. Early diagnosis of pancreatic Cancer: clinical premonitions, timely Precursor Detection and increased curative-intent surgery. Cancer Control. 2023;30:10732748231154711.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Fahrmann JF, Mao X, Irajizad E, Katayama H, Capello M, Tanaka I et al. Plasma-derived extracellular vesicles convey protein signatures that reflect pathophysiology in lung and pancreatic adenocarcinomas. Cancers (Basel). 2020;12(5).

  114. Zhang W, Wang L, Li D, Campbell DH, Walsh BJ, Packer NH, et al. Phenotypic profiling of pancreatic ductal adenocarcinoma plasma-derived small extracellular vesicles for cancer diagnosis and cancer stage prediction: a proof-of-concept study. Anal Methods. 2022;14(23):2255–65.

    Article  CAS  PubMed  Google Scholar 

  115. Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Li H, Chiang C-L, Kwak KJ, Wang X, Doddi S, Ramanathan LV et al. Extracellular vesicular analysis of glypican 1 mRNA and protein for pancreatic Cancer diagnosis and prognosis. Adv Sci (Weinh). 2024:e2306373.

  117. Xiao D, Dong Z, Zhen L, Xia G, Huang X, Wang T, et al. Combined exosomal GPC1, CD82, and serum CA19-9 as multiplex targets: a specific, sensitive, and Reproducible Detection Panel for the diagnosis of pancreatic Cancer. Mol Cancer Res. 2020;18(2):300–10.

    Article  CAS  PubMed  Google Scholar 

  118. Yang KS, Im H, Hong S, Pergolini I, Del Castillo AF, Wang R et al. Multiparametric plasma EV profiling facilitates diagnosis of pancreatic malignancy. Sci Transl Med. 2017;9(391).

  119. Lai X, Wang M, McElyea SD, Sherman S, House M, Korc M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 2017;393:86–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Nakamura K, Zhu Z, Roy S, Jun E, Han H, Munoz RM et al. An exosome-based Transcriptomic Signature for Noninvasive, early detection of patients with pancreatic ductal adenocarcinoma: a Multicenter Cohort Study. Gastroenterology. 2022;163(5).

  121. Yang Z, LaRiviere MJ, Ko J, Till JE, Christensen T, Yee SS, et al. A Multianalyte Panel Consisting of Extracellular Vesicle miRNAs and mRNAs, cfDNA, and CA19-9 shows utility for diagnosis and staging of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2020;26(13):3248–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Yu S, Li Y, Liao Z, Wang Z, Wang Z, Li Y, et al. Plasma extracellular vesicle long RNA profiling identifies a diagnostic signature for the detection of pancreatic ductal adenocarcinoma. Gut. 2020;69(3):540–50.

    Article  CAS  PubMed  Google Scholar 

  123. Goto T, Fujiya M, Konishi H, Sasajima J, Fujibayashi S, Hayashi A, et al. An elevated expression of serum exosomal microRNA-191, – 21, -451a of pancreatic neoplasm is considered to be efficient diagnostic marker. BMC Cancer. 2018;18(1):116.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Jin H, Liu P, Wu Y, Meng X, Wu M, Han J, et al. Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 2018;109(9):2946–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Liang K, Liu F, Fan J, Sun D, Liu C, Lyon CJ et al. Nanoplasmonic quantification of Tumor-derived Extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat Biomed Eng. 2017;1.

  126. Kitagawa T, Taniuchi K, Tsuboi M, Sakaguchi M, Kohsaki T, Okabayashi T, et al. Circulating pancreatic cancer exosomal RNAs for detection of pancreatic cancer. Mol Oncol. 2019;13(2):212–27.

    Article  CAS  PubMed  Google Scholar 

  127. Yang J, Zhang Y, Gao X, Yuan Y, Zhao J, Zhou S, et al. Plasma-derived Exosomal ALIX as a Novel Biomarker for diagnosis and classification of pancreatic Cancer. Front Oncol. 2021;11:628346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Chen J, Yao D, Chen W, Li Z, Guo Y, Zhu F et al. Serum exosomal miR-451a acts as a candidate marker for pancreatic cancer. Int J Biol Markers. 2022:17246008211070018.

  129. Wang L, Wu J, Ye N, Li F, Zhan H, Chen S, et al. Plasma-derived Exosome MiR-19b acts as a diagnostic marker for pancreatic Cancer. Front Oncol. 2021;11:739111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Guo S, Qin H, Liu K, Wang H, Bai S, Liu S, et al. Blood small extracellular vesicles derived miRNAs to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Clin Transl Med. 2021;11(9):e520.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Verel-Yilmaz Y, Fernandez JP, Schafer A, Nevermann S, Cook L, Gercke N, et al. Extracellular vesicle-based detection of pancreatic Cancer. Front Cell Dev Biol. 2021;9:697939.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Yang G, Qiu J, Xu J, Xiong G, Zhao F, Cao Z et al. Using a microRNA panel of circulating exosomes for diagnosis of pancreatic cancer: multicentre case-control study. Br J Surg. 2022.

  133. Marin AM, Mattar SB, Amatuzzi RF, Chammas R, Uno M, Zanette DL et al. Plasma exosome-derived microRNAs as potential diagnostic and prognostic biomarkers in Brazilian pancreatic Cancer patients. Biomolecules. 2022;12(6).

  134. Hong L, Xu L, Jin L, Xu K, Tang W, Zhu Y, et al. Exosomal circular RNA hsa_circ_0006220, and hsa_circ_0001666 as biomarkers in the diagnosis of pancreatic cancer. J Clin Lab Anal. 2022;36(6):e24447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Makler A, Asghar W. Exosomal miRNA Biomarker Panel for pancreatic ductal adenocarcinoma detection in patient plasma: a pilot study. Int J Mol Sci. 2023;24(6).

  136. Lau C, Kim Y, Chia D, Spielmann N, Eibl G, Elashoff D, et al. Role of pancreatic cancer-derived exosomes in salivary biomarker development. J Biol Chem. 2013;288(37):26888–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Machida T, Tomofuji T, Maruyama T, Yoneda T, Ekuni D, Azuma T, et al. miR1246 and miR4644 in salivary exosome as potential biomarkers for pancreatobiliary tract cancer. Oncol Rep. 2016;36(4):2375–81.

    Article  CAS  PubMed  Google Scholar 

  138. Ferguson S, Yang KS, Zelga P, Liss AS, Carlson JCT, Del Castillo CF, et al. Single-EV analysis (sEVA) of mutated proteins allows detection of stage 1 pancreatic cancer. Sci Adv. 2022;8(16):eabm3453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Ye Q, Ling S, Zheng S, Xu X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer. 2019;18(1):114.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Nishiwada S, Cui Y, Sho M, Jun E, Akahori T, Nakamura K et al. Transcriptomic Profiling Identifies an Exosomal microRNA Signature for Predicting Recurrence Following Surgery in Patients with Pancreatic Ductal Adenocarcinoma. Ann Surg. 2021.

  141. Wei Q, Li Z, Feng H, Ren L. Serum exosomal EphA2 is a prognostic biomarker in patients with pancreatic Cancer. Cancer Manag Res. 2021;13:3675–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Bernard V, Kim DU, San Lucas FA, Castillo J, Allenson K, Mulu FC, et al. Circulating nucleic acids are Associated with outcomes of patients with pancreatic Cancer. Gastroenterology. 2019;156(1):108–18. e4.

    Article  CAS  PubMed  Google Scholar 

  143. Yee NS, Zhang S, He HZ, Zheng SY. Extracellular vesicles as potential biomarkers for early detection and diagnosis of pancreatic Cancer. Biomedicines. 2020;8(12).

  144. Aafreen S, Feng J, Wang W, Liu G. Theranostic extracellular vesicles: a concise review of current imaging technologies and labeling strategies. Extracell Vesicles Circulating Nucleic Acids. 2023;4(1):107–32.

    Article  CAS  Google Scholar 

  145. Witwer KW, Wolfram J. Extracellular vesicles versus synthetic nanoparticles for drug delivery. Nat Reviews Mater. 2021;6(2):103–6.

    Article  CAS  Google Scholar 

  146. Armstrong JPK, Stevens MM. Strategic design of extracellular vesicle drug delivery systems. Adv Drug Deliv Rev. 2018;130:12–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Erb U, Zhao K, Wang Z, Xiao L, Zoller M. Murine and human pancreatic tumor exosome recovery in mouse serum: diagnostic and prognostic potential and target cell delivery. Cancer Lett. 2017;403:1–12.

    Article  CAS  PubMed  Google Scholar 

  148. Xu L, Faruqu FN, Liam-Or R, Abu Abed O, Li D, Venner K, et al. Design of experiment (DoE)-driven in vitro and in vivo uptake studies of exosomes for pancreatic cancer delivery enabled by copper-free click chemistry-based labelling. J Extracell Vesicles. 2020;9(1):1779458.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ, Hackett S, et al. Macropinocytosis of protein is an amino acid supply route in ras-transformed cells. Nature. 2013;497(7451):633–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Creeden JF, Sevier J, Zhang J-T, Lapitsky Y, Brunicardi FC, Jin G, et al. Smart exosomes enhance PDAC targeted therapy. J Control Release. 2024;368:413–29.

    Article  CAS  PubMed  Google Scholar 

  151. Li Y-J, Wu J-Y, Wang J-M, Hu X-B, Cai J-X, Xiang D-X. Gemcitabine loaded autologous exosomes for effective and safe chemotherapy of pancreatic cancer. Acta Biomater. 2020;101:519–30.

    Article  CAS  PubMed  Google Scholar 

  152. Al Faruque H, Choi E-S, Kim J-H, Kim E. Enhanced effect of autologous EVs delivering paclitaxel in pancreatic cancer. J Control Release. 2022;347:330–46.

    Article  CAS  PubMed  Google Scholar 

  153. Zhou Y, Zhou W, Chen X, Wang Q, Li C, Chen Q, et al. Bone marrow mesenchymal stem cells-derived exosomes for penetrating and targeted chemotherapy of pancreatic cancer. Acta Pharm Sin B. 2020;10(8):1563–75.

    Article  CAS  PubMed  Google Scholar 

  154. Zhao Y, Zheng Y, Zhu Y, Zhang Y, Zhu H, Liu T. M1 macrophage-derived exosomes loaded with Gemcitabine and Deferasirox against Chemoresistant Pancreatic Cancer. Pharmaceutics. 2021;13(9).

  155. Xu Y, Liu N, Wei Y, Zhou D, Lin R, Wang X, et al. Anticancer effects of miR-124 delivered by BM-MSC derived exosomes on cell proliferation, epithelial mesenchymal transition, and chemotherapy sensitivity of pancreatic cancer cells. Aging. 2020;12(19):19660–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Hasoglu I, Karatug Kacar A. The therapeutic effects of exosomes the first time isolated from pancreatic islet-derived progenitor cells in the treatment of pancreatic cancer. Protoplasma. 2024;261(2):281–91.

    Article  CAS  PubMed  Google Scholar 

  157. Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546.

    Article  CAS  PubMed  Google Scholar 

  158. Zhou W, Chen X, Zhou Y, Shi S, Liang C, Yu X, et al. Exosomes derived from immunogenically dying tumor cells as a versatile tool for vaccination against pancreatic cancer. Biomaterials. 2022;280:121306.

    Article  CAS  PubMed  Google Scholar 

  159. Jang Y, Kim H, Yoon S, Lee H, Hwang J, Jung J, et al. Exosome-based photoacoustic imaging guided photodynamic and immunotherapy for the treatment of pancreatic cancer. J Control Release. 2021;330:293–304.

    Article  CAS  PubMed  Google Scholar 

  160. Zhao J, Zhu Y, Li Z, Liang J, Zhang Y, Zhou S, et al. Pirfenidone-loaded exosomes derived from pancreatic ductal adenocarcinoma cells alleviate fibrosis of premetastatic niches to inhibit liver metastasis. Biomater Sci. 2022;10(22):6614–26.

    Article  CAS  PubMed  Google Scholar 

  161. Uprety D, Adjei AA. KRAS: from undruggable to a druggable Cancer target. Cancer Treat Rev. 2020;89:102070.

    Article  CAS  PubMed  Google Scholar 

  162. Moore AR, Rosenberg SC, McCormick F, Malek S. RAS-targeted therapies: is the undruggable drugged? Nat Rev Drug Discov. 2020;19(8):533–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. McAndrews KM, Xiao F, Chronopoulos A, LeBleu VS, Kugeratski FG, Kalluri R. Exosome-mediated delivery of CRISPR/Cas9 for targeting of oncogenic KrasG12D in pancreatic cancer. Life Sci Alliance. 2021;4(9).

  164. Mendt M, Kamerkar S, Sugimoto H, McAndrews KM, Wu CC, Gagea M et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight. 2018;3(8).

  165. Xu L, Faruqu FN, Lim YM, Lim KY, Liam-Or R, Walters AA, et al. Exosome-mediated RNAi of PAK4 prolongs survival of pancreatic cancer mouse model after loco-regional treatment. Biomaterials. 2021;264:120369.

    Article  CAS  PubMed  Google Scholar 

  166. Shang S, Wang J, Chen S, Tian R, Zeng H, Wang L, et al. Exosomal miRNA-1231 derived from bone marrow mesenchymal stem cells inhibits the activity of pancreatic cancer. Cancer Med. 2019;8(18):7728–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Welsh JA, Goberdhan DCI, O’Driscoll L, Buzas EI, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024;13(2):e12404.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Weick E-M, Puno MR, Januszyk K, Zinder JC, DiMattia MA, Lima CD. Helicase-dependent RNA decay illuminated by a Cryo-EM structure of a Human Nuclear RNA Exosome-MTR4 complex. Cell. 2018;173(7).

  169. Saumell-Esnaola M, Delgado D, García Del Caño G, Beitia M, Sallés J, González-Burguera I et al. Isolation of platelet-derived exosomes from human platelet-rich plasma: biochemical and morphological characterization. Int J Mol Sci. 2022;23(5).

  170. Yan W, Li T, Yin T, Hou Z, Qu K, Wang N, et al. M2 macrophage-derived exosomes promote the c-KIT phenotype of vascular smooth muscle cells during vascular tissue repair after intravascular stent implantation. Theranostics. 2020;10(23):10712–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Gandhi J, Sushma MV, Rengan AK, Naik MN, Mishra DK, Boyinpally SR, et al. Proteomic profiling of exosomes in a mouse model of Candida albicans endophthalmitis. Exp Cell Res. 2022;417(2):113222.

    Article  CAS  PubMed  Google Scholar 

  172. van der Pol E, Coumans FAW, Grootemaat AE, Gardiner C, Sargent IL, Harrison P, et al. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J Thromb Haemost. 2014;12(7):1182–92.

    Article  PubMed  Google Scholar 

  173. Tian Y, Gong M, Hu Y, Liu H, Zhang W, Zhang M, et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles. 2020;9(1):1697028.

    Article  CAS  PubMed  Google Scholar 

  174. Mallick MS, Misbah I, Ohannesian N, Shih W-C. Single-exosome counting and 3D, Subdiffraction Limit localization using dynamic Plasmonic nanoaperture label-free imaging. Adv Nanobiomed Res. 2023;3(9).

  175. Mo Y, Leung LL, Mak CSL, Wang X, Chan W-S, Hui LMN, et al. Tumor-secreted exosomal miR-141 activates tumor-stroma interactions and controls premetastatic niche formation in ovarian cancer metastasis. Mol Cancer. 2023;22(1):4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Liu C, Xu X, Li B, Situ B, Pan W, Hu Y, et al. Single-exosome-counting immunoassays for Cancer Diagnostics. Nano Lett. 2018;18(7):4226–32.

    Article  CAS  PubMed  Google Scholar 

  177. Zhang L, Wang H, Zhao G, Li N, Wang X, Li Y, et al. Anti-Tim4 grafting strongly Hydrophilic Metal-Organic frameworks Immunoaffinity Flake for High-Efficiency capture and separation of Exosomes. Anal Chem. 2021;93(16):6534–43.

    Article  CAS  PubMed  Google Scholar 

  178. Kim JS, Kwon SY, Lee JY, Kim SD, Kim DY, Kim H, et al. High-throughput multi-gate microfluidic resistive pulse sensing for biological nanoparticle detection. Lab Chip. 2023;23(7):1945–53.

    Article  CAS  PubMed  Google Scholar 

  179. Xu J, Zhang J, Zhang Z, Gao Z, Qi Y, Qiu W, et al. Hypoxic glioma-derived exosomes promote M2-like macrophage polarization by enhancing autophagy induction. Cell Death Dis. 2021;12(4):373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Leary N, Walser S, He Y, Cousin N, Pereira P, Gallo A, et al. Melanoma-derived extracellular vesicles mediate lymphatic remodelling and impair tumour immunity in draining lymph nodes. J Extracell Vesicles. 2022;11(2):e12197.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Crescitelli R, Lasser C, Lotvall J. Isolation and characterization of extracellular vesicle subpopulations from tissues. Nat Protoc. 2021;16(3):1548–80.

    Article  CAS  PubMed  Google Scholar 

  182. Inoue H, Eguchi A, Kobayashi Y, Usugi E, Yamada R, Tsuboi J et al. Extracellular vesicles from pancreatic ductal adenocarcinoma endoscopic ultrasound-fine needle aspiration samples contain a protein barcode. J Hepatobiliary Pancreat Sci. 2021.

  183. Cheng L, Hill AF. Therapeutically harnessing extracellular vesicles. Nat Rev Drug Discov. 2022;21(5):379–99.

    Article  CAS  PubMed  Google Scholar 

  184. Johnson J, Law SQK, Shojaee M, Hall AS, Bhuiyan S, Lim MBL, et al. First-in-human clinical trial of allogeneic, platelet-derived extracellular vesicles as a potential therapeutic for delayed wound healing. J Extracell Vesicles. 2023;12(7):e12332.

    Article  PubMed  Google Scholar 

  185. Shuen TWH, Alunni-Fabbroni M, Öcal E, Malfertheiner P, Wildgruber M, Schinner R, et al. Extracellular vesicles may predict response to Radioembolization and Sorafenib Treatment in Advanced Hepatocellular Carcinoma: an exploratory analysis from the SORAMIC Trial. Clin Cancer Res. 2022;28(17):3890–901.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Wang C, Li N, Li Y, Hou S, Zhang W, Meng Z, et al. Engineering a HEK-293T exosome-based delivery platform for efficient tumor-targeting chemotherapy/internal irradiation combination therapy. J Nanobiotechnol. 2022;20(1):247.

    Article  Google Scholar 

  187. Huang X, Wu W, Jing D, Yang L, Guo H, Wang L, et al. Engineered exosome as targeted lncRNA MEG3 delivery vehicles for osteosarcoma therapy. J Control Release. 2022;343:107–17.

    Article  CAS  PubMed  Google Scholar 

  188. Sadeghi S, Tehrani FR, Tahmasebi S, Shafiee A, Hashemi SM. Exosome engineering in cell therapy and drug delivery. Inflammopharmacology. 2023;31(1):145–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Gao Q, Lin YP, Li BS, Wang GQ, Dong LQ, Shen BY, et al. Unintrusive multi-cancer detection by circulating cell-free DNA methylation sequencing (THUNDER): development and independent validation studies. Ann Oncol. 2023;34(5):486–95.

    Article  CAS  PubMed  Google Scholar 

  190. Chen S, Jin Y, Wang S, Xing S, Wu Y, Tao Y et al. Cancer type classification using plasma cell-free RNAs derived from human and microbes. Elife. 2022;11.

  191. Li Y, Jiang G, Wu W, Yang H, Jin Y, Wu M, et al. Multi-omics integrated circulating cell-free DNA genomic signatures enhanced the diagnostic performance of early-stage lung cancer and postoperative minimal residual disease. EBioMedicine. 2023;91:104553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Weibin Wang received the support from Beijing Natural Science Foundation (No. 7232127), the National Natural Science Foundation of China (No. 82173074), the National High Level Hospital Clinical Research Funding (No. 2022-PUMCH-B-004), the National High Level Hospital Clinical Research Funding (No. 2022-PUMCH-D-001), the CAMS Innovation Fund for Medical Sciences (CIFMS) (No. 2021-I2M-1-002), Capital’s Funds for Health Improvement and Research (2024-2-4017) and the Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences (2018PT32014). Cheng Qin received the support from Postdoctoral Fellowship Program of CPSF (GZC20240146).

Author information

Authors and Affiliations

Authors

Contributions

WWB directed and guided this study. QC, LTY, LC collected related literature and drafted this manuscript. ZBB, LZR, ZYT made critical revisions to this manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Weibin Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The manuscript is approved by all authors for publication.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qin, C., Li, T., Lin, C. et al. The systematic role of pancreatic cancer exosomes: distant communication, liquid biopsy and future therapy. Cancer Cell Int 24, 264 (2024). https://doi.org/10.1186/s12935-024-03456-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03456-5

Keywords