Next Article in Journal
The Role of Vitamin D in Supporting Health in the COVID-19 Era
Next Article in Special Issue
Histone Deacetylase (HDAC) Inhibitors: A Promising Weapon to Tackle Therapy Resistance in Melanoma
Previous Article in Journal
Control of Bacterial Diseases of Banana Using CRISPR/Cas-Based Gene Editing
Previous Article in Special Issue
Immunotherapy for Cervical Cancer: Are We Ready for Prime Time?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exosomes in the Treatment of Pancreatic Cancer: A Moonshot to PDAC Treatment?

by
Stavros P. Papadakos
1,†,
Nikolaos Dedes
1,†,
Alexandros Pergaris
1,
Maria Gazouli
2 and
Stamatios Theocharis
1,*
1
First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(7), 3620; https://doi.org/10.3390/ijms23073620
Submission received: 27 February 2022 / Revised: 19 March 2022 / Accepted: 23 March 2022 / Published: 25 March 2022
(This article belongs to the Special Issue Molecular Advances in Cancer Therapy)

Abstract

:
Pancreatic Ductal Adenocarcinoma (PDAC) constitutes a leading cause of cancer death globally. Its mortality remains unaltered despite the considerable scientific progress made in the fields of diagnostics and treatment. Exosomes comprise of small extracellular vesicles secreted by nearly all cells; their cargo contains a vast array of biomolecules, such as proteins and microRNAs. It is currently established that their role as messengers is central to a plethora of both physiologic and pathologic processes. Accumulating data have shed light on their contributions to carcinogenesis, metastasis, and immunological response. Meanwhile, the advancement of personalized targeted therapies into everyday clinical practice necessitates the development of cost-efficient treatment approaches. The role of exosomes is currently being extensively investigated towards this direction. This review aims to summarize the current pre-clinical and clinical evidence regarding the effects of exosomal applications in the timely diagnosis, prognosis, and therapeutic management of pancreatic cancer.

1. Introduction

1.1. PDAC: Epidemiology, Pathogenesis and Current Therapeutic Management

Pancreatic cancer has long been a major public health issue and statistical figures highlight the ever-growing severity of the problem. According to GLOBOCAN 2020, 495,773 new cases and 466,003 deaths were attributed to pancreatic cancer worldwide in the year 2020 [1]. Currently, pancreatic cancer is the fourth cancer-related cause of death in the United States with the outlook being rather grim, as it is projected to be elevated to the position of second or the third place with respect to cancer mortality in Europe and North America by the end of this decade [2,3]. Incidence and mortality rates are four or five times higher in regions with a high Human Development Index such as Europe or North America when compared with countries in Sub-Saharan Africa [1]. Nonetheless, disease prognosis does not differ significantly between rich and poor countries, an observation ascribed to the lack of an effective screening process, late diagnosis, and the limited success of available treatment protocols [4]. The incidence of pancreatic cancer is slightly higher in men than in women and predominantly affects people between 60 and 80 years old with the cumulative lifetime risk of developing pancreatic cancer estimated to be around 1.5% [5,6]. It should be noted that these data are primarily derived from cases of pancreatic ductal adenocarcinoma (PDAC), which is the most common histological type (>90%) and thus is the focus of this manuscript [7]. Other less common varieties are acinar cell carcinoma or cystadenocarcinoma, intraductal papillary mucinous neoplasm with an associated invasive carcinoma intraductal tubulopapillary neoplasm with associated invasive carcinoma, mixed acinar/ductal/neuroendocrine carcinoma, pancreatoblastoma serous cystadenocarcinoma, solid-pseudopapillary neoplasm, and neuroendocrine neoplasms [8].
Regardless of histology, our understanding concerning the etiology of this disease is fundamentally lacking. As is the case with many malignancies, “modus vivendi” is a major determinant of PDAC risk. Smoking, excess body weight, and diabetes mellitus are all recognized risk factors [8,9,10,11,12,13,14,15]. Recent advances in molecular biology have served to further illuminate the mechanics PDAC. While most cases are sporadic, 5–10% are related to familial and inherited genetic disorders, such as Lynch Syndrome or Familial adenomatous polyposis, which carry a higher risk for PDAC [15,16]. Delving deeper into the genetics, mutations or deletion in genes such as PRSS1, KRAS, p16, p53, and BRCA2 are linked to an increased level of risk [17]. Of particular significance is the mutational activation of the proto-oncogene KRAS, which is harbored in up to 90% of cases [18,19]. These KRAS mutations concern mainly three hotspots, which most often are substitutions at G12 (84%). There are six possible single-base missense mutations, namely, G12D, G12V, G12C, G12A, G12S, and G12R [20]. Beyond KRAS mutations, there are a considerable number of tumor suppressors that are usually altered; some examples are CDKN2A (95%), TP53 (75%), and DPC4/SMAD4 (55%) [21,22,23,24].
Undoubtedly, the pathogenesis of PDAC remains elusive. However, a more pressing matter and a source of great concern is its insidious nature and the lack of effective treatment options. At diagnosis, PDAC is usually at least 2–4 cm in diameter and has in many occasions already infiltrated the surrounding structures and lymph nodes [8]. Unfortunately, just 10–15% of patients are candidates for surgery, which is associated with a 5-year-survival rate of around 25% [24,25,26]. The chance of survival is significantly better when surgery is combined with chemotherapy [27,28,29]. While the survival rates after surgery are not impressive, they are considerably higher than those of patients who present with metastases at diagnosis, which are no more than 2.7% [29]. For those who are not candidates for surgery and who have an Eastern Cooperative Oncology Group (ECOG) Performance Status (PS) between 0 and 1, the first treatment option is chemotherapy with leucovorin, fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX) or gemcitabine plus nab-paclitaxel. Patients with a PS of 2 generally receive gemcitabine alone, while treatment for those with a PS of 3 is generally limited to the best supportive care. Further treatment options are available for a limited number of patients with NTRK fusions, BRCA mutations, or those who are mismatch repair deficient or whose disease has high microsatellite instability [30]. Nevertheless, the available therapeutic strategies have limited efficacy and the disease usually develops resistance to even the most active agents, such as gemcitabine [28,31]. Currently, the end result is disheartening: patient median survival time is less than 6 months [32]. This state of affairs calls for a different approach to PDAC diagnosis and management, and one of the novel strategies is the manipulation of exosomes [33,34,35,36,37,38]. This review provides a precis of exosome biology and its possible applications to the therapeutics of PDAC.

1.2. Extracellular Vesicles: Characteristics and Roles in Physiology and Disease

Extracellular vesicles (EVs) are membrane-bound lipid particles secreted by all cells and function as a means of communication between the parent and the recipient cell through their biological cargo (proteins, nucleic acids, lipids, and metabolites). With regard to their classification, the International Society of Extracellular Vesicles recommends the division of EVs based on their size: medium/large EV (>150 nm) and small EV (<150 nm) [39]. Nonetheless, there is no general consensus regarding the categorization of EVs in the current literature due to the considerable overlap of vesicle sizes. However, most researchers discern three major groups of EVs: small EVs or exosomes, microvesicles, and apoptotic bodies.
Small EVs or Exosomes: Exosomes are nanoparticles with a diameter measurement in the range of 30–150 nm. The first step in the biogenesis of exosomes is the inward budding of the extracellular membrane, which forms the endosome. The endosome undergoes inward invaginations and thus encloses biological cargo, which produces the intraluminal vesicles. The total number of intraluminal vesicles and the endosome in which they are contained comprise a multivesicular body (MVB). Once formed, these MVBs can either fuse with lysosomes and have their content degraded or release their content to the extracellular matrix through the endosomal sorting complex required for the transport- (ESCRT)-dependent or the ESCRT-independent pathway. Among exosomes, there are several common features such as the tetraspanin family of surface proteins (CD9, CD63 and CD81), integrins, heat shock proteins, ESCRT, actin and flotillins, while other molecules are specific to the donor cell, such as type MHC Class I and II. The end result is the activation of a variety of signaling cascades, which are determined by the interconnection between exosome surface proteins and receptors on the recipient cells [39,40].
Microvesicles: The sizes of these vesicles range from 50 nm to 1μm and are generated by the outward budding of the cell membrane [41]. Their content is similar to that of exosomes, and they are considered to be involved on tissue homeostasis (regeneration, angiogenesis, cell cycle regulation, inflammation, etc.) [42,43].
Apoptotic Bodies: These vesicles comprise large fragments of a cell which has undergone apoptosis. They are the largest subset of the EVs with a size of up to 2 μm and therefore provide a substantial molecular pool for recipient cells [44]. Apoptotic bodies are readily cleared by macrophages, which recognize phosphatidylserine on the surface of the EV in question; the defects of this process are linked to auto-immune reactions [45]. Similarly to microvesicles, apoptotic bodies play a role in tissue homeostasis, such as bone metabolism [46].

1.3. Exosomes

Classically, exosomes were considered as a means for the disposal of nonessential cellular cargo. The first documentation of the term “exosome” in the literature was in a publication delineating the maturation of reticulocytes [47]. The development of more advanced cell culture techniques in conjunction with the improvement in exosomal isolation capacity and immune-electron microscopy has led to a deepening in our perception of the underlying biosynthetic and regulatory mechanisms of exosomes [48]. It is now common knowledge that they are important mediators of intercellular communication under both normal and diseased conditions [49]. In more detail, the mediation of exosomes regulates a wide variety of physiologic processes: skin regeneration [50], bone remodeling [51], placental formation [52], and organ crosstalk during exercise [53]. It also serves as a diagnostic and predictive biomarker in inflammatory diseases such as rheumatoid arthritis [54], systemic lupus erythematosus [55], and nephropathies [56].
Similarly, the participation of exosomes in cancer pathophysiology with applications in diagnosis, prognosis, and treatment has been extensively studied [57,58,59].
Focusing on PDAC, exosomes have the potential to help determine treatment decisions. One such application is the perspective given by liquid biopsy, as was hinted by Buscail et al. who demonstrated that the level Glypican-1 positive exosomes circulating tumor cells and CA19-9 are able to be used for diagnostic and prognostic purposes in patients with resectable tumors [60,61]. Moreover, Nakamura et al. found that extracted exosomes from pancreatic juice could be used to discriminate PDAC from chronic pancreatitis [62]. Furthermore, treatment adjustments could be made by monitoring exosomal miRs, known to confer chemoresistance [63].

1.4. Artificial Exosomes: Engineering and Applications

Exosomes possess abilities and characteristics that render them a highly promising drug delivery system. However, the utilization of natural exosomes comes across a variety of technical issues such as difficulties in large-scale production, isolation, drug conjugation, stability, and quality control. To compensate for these deficiencies, a considerable effort is being made that aims to develop artificial exosomes by means of nanobiotechnology. With respect to the production of these vesicles, many different approaches have been suggested, all of which can be summarized into three distinct filter categories, namely, “top-down”, “bottom-up,” and “biohybrid” technologies [64].
The “top-down” strategy aims at the creation of nanovesicles through cell manipulation. This can be achieved with multiple techniques such as forced passage through porous membranes or microfluidic devices, nitrogen cavitation, cell membrane blebbing by virtue of sulfhydryl-blocking, and exposure to alkaline solutions followed by sonication. The major advantage of the “top-down” methodology is that the produced nanovesicles are fully biological with similar features to natural exosomes. Unfortunately, their production process also involves cell destruction, which limits its production sustainability and is associated with the presence of various contaminants [65,66,67,68].
On the other hand, “bottom-up” methodologies produce exosome-mimetics by supramolecular chemistry, which combines basic components such as lipids or proteins in a progressive manner so as to form complex structures [64]. This approach offers a pure product that is eligible for mass production, while its most substantial shortcoming is attributed to the difficulties in the liposome modification necessary in order to accomplish the sufficient mimicry of natural exosomes [69,70].
Finally, biohybrids, a form of exosome-mimetic, can be created through the merging of synthetic nanoparticles with natural vesicles, which is achievable by freeze-thawing, incubation, or co-extrusion [64]. This strategy might produce a carrier with higher efficiency than liposomes and higher stability than natural exosomes. Regrettably, this methodology is the most resource-intensive [71].
In summary, the products of nanobiotechnology are expected to comprise a major drug delivery system in the near future and bring medicine closer to personalized therapy. However, this field is still in its early stages of evolution. Therefore, further research is needed to improve production protocols, devise characterization methods, and achieve a meaningful outcome. The above-mentioned techniques are presented in Figure 1.

2. PDAC Microenvironment: Role in Neoplasia

The PDAC’s microenvironment consists of a wide variety of heterogeneous cell populations, namely, endothelial cells and pericytes comprising the vascular compartment, extracellular matrix (ECM)-producing cells, neuroendocrine cells, and immune and inflammatory cells at various activation states. From a review of the preceding literature, it is apparent that their contribution to tumorigenesis has long been underappreciated. This is reflected by the utilization of nonspecific chemotherapeutic agents in PDAC’s therapy in order to achieve cell-cycle arrest. With the broadening of our understanding of PDAC’s initiation, proliferation, and metastatic potential as well as of the dynamic interconnection between tumor cells and the cells of tumor microenvironment (TME), new target-specific therapeutic approaches are under development. This complex network is interconnected with physical interactions, immune suppression, and pleiotropic crosstalk mechanisms [72]. This interconnection is mediated through exosomal intercellular communication.
As far as a tumor’s physiology is concerned, the PDAC cells are crowded in an intense stroma of fibrous tissue, which is characteristic of the disease [73]. The immune cell infiltrates constitute nearly 50% of the tumor mass [74]. A glycosaminoglycan desmoplastic reaction (analyzed below) is responsible for the notable increase in interstitial fluid pressure (IFP) [75], which constricts the vascular inflow, creating an oxygen-deprived, nutrient-poor environment [72]. Under hypoxic conditions, the pancreatic cancer cells generate and secrete exosomes carrying miR-301a-3p, which polarize the stromal macrophages into a M2 phenotype upregulating the PTEN/PI3K signaling pathway [76]. Moreover, Yang et al. demonstrated that M2 macrophages had a positive correlation with the microvessel density of human PDAC. M2 macrophage-derived exosomes were more enriched in miR-155-5p and miR-221-5p compared to M0 macrophage-derived exosomes, which promoted the angiogenic ability of endothelial cells in an E2F2-dependent manner [77]. Similarly, hypoxic PDAC cell-derived exosomal miR-30b-5p was found to promote angiogenesis through the inhibition of GJA1 in a mouse model. This observation holds the promise of becoming the basis of the development of a new diagnostic marker, as patients with PDAC had higher levels of total miR-30b-5p and exosomal miR-30b-5p in peripheral blood plasma than did healthy subjects [78]. Hypoxia is also targeted by modern therapeutics. For example, evofosfamide is reduced in hypoxic conditions, releasing its active metabolite bromo-isophosphoramide (Br-IPM). Br-IPM exerts its cytotoxic effects preferentially in the hypoxic areas of the tumor and potentiates the utilization of radiotherapy and chemotherapy as adjunct therapeutic modalities [79]. However, the TME and the disease are not static, as the transfer of miRNA in macrophage-derived exosomes induces drug resistance in PDAC [80]. Exosome-mediated intercellular communication is also central to disease progression. It has been recently found that the liver metastatic niche is prepared in advance by the PDAC-derived exosomes carrying the CD44v6/C1QBP complex, which causes hepatic fibrosis [81]. In this context, exosomal long noncoding RNA LINC01133 was demonstrated to promote the proliferation, migration, invasion, and epithelial-mesenchymal transition of pancreatic cancer cells [82].
Beyond mere description of disease pathogenesis, the exploration of exosome-mediated communication might uncover a new basis for potential treatment. Based on this thesis, Zhou et al. created an exosome-based dual delivery biosystem for enhancing PDAC immunotherapy and reversing the tumor immunosuppression of M2 macrophages through the disruption of the galectin-9/dectin 1 axis [83]. TME was also targeted in a recent clinical trial which tested the ibrutinib plus nab-paclitaxel/gemcitabine versus the placebo plus nab-paclitaxel/gemcitabine. Unfortunately, no statistically significant increase in survival was noted [84]. Alternation of the TME and immune system activation has also been attempted via the use of a modified herpes simplex virus by Wang et al., while the same aim was pursued by Jiang et al. through MEK and autophagy co-inhibition, coupled with CD40 agonism [85,86].
Taking all of this into consideration, PDAC TME is central to pathogenesis and the physical history of the disease. These processes—mediated largely by exosomes—are becoming ever more defined. Thus, the next step is to utilize the attributes of exosomes and to manipulate them in such a way that they become clinically meaningful. A brief synopsis of the above is presented in Figure 2.

3. Therapeutic Applications of Exosomes in PDAC

The urgency regarding the generation of a more personalized approach to medical diagnosis and treatment has placed exosome technology in the spotlight. The main applications that are under investigation concern the reduction of drug resistance, the utilization of exosomes as drug-carries, the therapeutic modification of the immune tumor microenvironment, and the targeting of KRAS. The contributions of exosomes in the therapy of PDAC will be presented below.

3.1. Chemoresistance in PDAC Treatment

Resistance to chemotherapy is an appreciable problem in the treatment of pancreatic cancer, and there is growing evidence that exosomes are crucially involved. The underlying mechanisms for nontargeting drugs include the upregulation of multidrug resistance proteins (MDR) which comprise several classes of drug efflux pumps such as P-glycoproteins (P-gp), multidrug-resistant protein-1 (MRP-1), and ATP-binding cassette sub-family A member 3(ABCA-3). They also include the enhancement of proliferating and anti-apoptotic pathways, the downregulation of drug targets, and the mutational alteration of drug targets. The latter also constitutes the leading cause of drug resistance in targeted therapies [87]. Exosomes contribute to drug resistance either directly (by carrying the chemotherapeutic agents outside of the cancer cells [88]) or indirectly (through their cargo-mediated effects); they derive from chemotherapy-resistant cancer cells and the cells of TME and modulate the phenotype of chemotherapy-sensitive cancer cells [87]. In recent years, preclinical data on the role of exosomes in chemotherapy resistance have increased remarkably.
Radical oxygen species (ROS) generation is a well-documented precipitant of cell death. Patel et al. documented that among the EVs, exosomes contribute the most to gemcitabine (GEM) resistance by conveying superoxide dismutase 2 (SOD2) and catalase (CAT) to detoxify ROS. In parallel, the exosomally transmitted miR-155 inhibits the expression of deoxycytidine kinase (DCK), deescalating the phosphorylation of GEM monophosphate to its active metabolite. The inhibition of mir-155 or the downregulation of DCK counteracts the exosome-induced GEM resistance [89]. Mikamori et al. shed more light on the effects of miR-155 on PDAC. They demonstrated that miR-155 enhances the secretion of exosomes, modifies their cargo, increasing its excretion, and induces GEM resistance through suppressing apoptosis by downregulating TP53INP. The interception of RAB27B’s function mitigated drug resistance by enhancing the caspase 3/7-induced apoptosis [90]. These results indicate that miR-155 in exosomes could be a potential therapeutic target. Taking a step further, survivin exerts its anti-apoptotic effects by binding and deactivating caspase-9 through the phosphorylatable Thr. Since the utilization of Survivin-T34A by adenoviral vectors showed moderate therapeutic results [91], Aspe et al. treated MIA PaCa-2 cell lines with Survivin-T34A-enriched exosomes raised by engineered melanoma cells. They reported that the combination of GEM and Survivin-T34A-enriched exosomes ameliorated nearly three times the apoptotic process compared to GEM or exosomes alone [92]. Although the role of the EPH/ephrin system on cancer has been thoroughly presented [93], we are obligated to indicate the significant role of exosomal EPHA2 in GEM resistance. A comparative analysis of the protein exosomal cargo indicated a 5-fold increase of EPHA2 in the GEM-resistant PANC-1 cell line in comparison with the more GEM-sensitive MIA PaCa-2 and BxPC-3 lines. The silencing of the EPHA2 in PANC-1 cells improved GEM resistance by approximately 25% [94]. In the complex TME, drug resistance can also be imposed by noncancerous cell populations.
Excepting the metabolic support that Cancer-associated fibroblasts (CAF) provide to the cancerous epithelial cells, Richards et al. documented that CAFs, when exposed to GEM, tend to transmit their innate resistance to GEM with exosomes highly enriched in miR-146a and Snail. Blocking the secretion of exosomes with GW4869 enhances PDAC cell killing by GEM in co-cultures and shrinks the tumor volume [95]. Analogously, miR-106b-enriched exosomes of CAF origin promote resistance upon GEM exposure, combining with TP53INP. Those actions can be alleviated by miR-106b inhibitors [96]. Tumor-associated macrophages (TAMs) and cancer stem cells are also cell populations which significantly contribute to GEM resistance. M2-derived exosomes carried circa ten times more miR-365 in comparison to M1-originated exosomes, conferring a 2.6-fold increase in the intracellular NTP pool. This stimulates the upregulation of cytidine deaminase (CDA) in the PDAC cells, which in turn deactivates GEM while the elevated dCTP concentration antagonizes dFdCTP. Genetically modified Rab27 a/b knockout mice, which are unable to secrete exosomes, metabolize GEM better than the wild-type mice, while the treatment of PDAC-carrying mice with miR-365 oppressors restored sensitivity to GEM [80]. Exosomes derived from GEM-resistant cancer stem cells are abundant in miR-MiR-210 and contribute to GEM resistance, potentiating the mTOR signaling pathway [97]. All the above are summarized in Figure 3.

3.2. Exosomes as Drug Carriers

The rationale for using exosomes as drug carriers in the treatment of pancreatic cancer stems from the similarity of their cell membranes to those of human cells. This trait is responsible for some advantageous properties, including favorable bioavailability and biocompatibility, inconsequential immunogenicity, the enhancement of drug release stability, the expansion of a drug’s half-life, and the ability to penetrate biological membranes. Τhe end result is the development of more effective drugs with improved profiles of side effects [98].
As mentioned above, the administration of drugs to cell populations of mesenchymal origin (e.g., adipose tissue, bone marrow) can trigger the generation and secretion of exosomes. This feature is exploited in order to create a model where chemotherapeutics are incorporated in mesenchymal tissues and secreted with exosomes. Another technique utilizes modern nanotechnology to load or pulse drugs directly into exosomes. Table 1 and Table 2 summarize the preclinical data regarding the effects of certain chemotherapeutics and of miRNA mimetics, respectively.

3.3. Immune Microenvironment Modifications

The discovery of exosomes and the evolution of their isolation and processing technology has transformed the logic of cancer vaccination. The initial vaccine development efforts aimed at stimulating antigen-presenting dendritic cells with cancer-specific antigens in order to create vaccines that targeted distinct cancer cell populations [112]. An increasing understanding of exosomal function improved the specificity of the vaccines and enabled the co-administration of drugs while in parallel, expanding the scope of therapeutic interventions within the immune microenvironment of the tumor. This was accomplished by the enhancement of immunotherapy; the utilization of modified exosomes as vectors in photodynamic therapy and their contribution to the reprogramming of the TME. In recent years, significant preclinical data have emerged in this direction.
Wang et al. demonstrated that under hypoxic conditions, PDAC cells are urged in an HIF-1a/2a-mediated manner to upregulate the expression of miR-301a-3p. The secretion of miR-301a-3p-enriched exosomes inhibits PTEN expression and potentiates the PI3K signaling pathway to polarize macrophages towards an M2 phenotype. The end result is a statistically significant association of miR-301a-3p with a more aggressive clinicopathological disease regarding the extent of tumor invasion, metastatic potential, perineural infiltration, and TNM staging. The silencing of miR-301a-3p reverses those effects [76] and could possibly be combined with antifibrotic treatments. The phenotype switching of macrophages may have enormous therapeutic implications. Su et al. reported that exosomes derived from miR-155 and miR-125b-2-transfected Panc-1 cells could shift macrophages from an M2 towards the immune-stimulatory M1 phenotype [113]. While Que et al. documented that PDAC cell-originated exosomes, in which the miRNA cargos were lysed, could more effectively activate dendritic cells and cytokine-induced cell killer populations [114], Xiao et al. incorporated all-trans-retinoic acid, GEM, and sunitinib into pancreatic cancer-derived exosomes and into dendritic cells in a continuum. This coalescence was able to target myeloid-derived suppressive cells (MDSCs) at a multitude of developmental stages, exhibiting prolonged survival and an improved metastatic profile [115].
These scientific efforts led to a more comprehensive theranostic approach. Yang et al. modified MIA-PaCa-2 cell-derived exosomes loading Chlorin e6 (Ce6), a potent photosensitizer. Ce6 can enhance the photoacoustic signals and trigger ROS generation upon exposure to laser irradiation. Additionally, potentiating the local antigen-presenting cells releases a cytokine reaction which generates an immune reaction by CD8 T cells. This combination of image-guided photodynamic therapy and immunotherapy showed promising results in mice with minimal side effects [116] and should enter into the stage of clinical trials. On the other hand, Zhou et al. utilized exosomes derived from bone marrow mesenchymal stem cells (BM-MSC) and, via electroporation, inserted galectin-9 siRNA and oxaliplatin. The latter exerts its action directly on pancreatic cancer cells by causing apoptosis and indirectly through causing immunogenic cell death. The enhancement of the immunological response is also achieved by the silencing of galectin-9, which polarizes macrophages towards an M1 phenotype and contributes to the aggregate cytotoxic response [83].

3.4. Targeting KRAS in PDAC

Despite the abovementioned fact that KRAS mutations drive carcinogenesis and are existent from early adenomatous lesions to late metastatic disease, there were not until recently any KRAS-targeting therapies. In fact, for decades, KRAS was considered “undruggable” based on the immense affinity of Ras for the highly abundant GDP,GTP in the cell’s cytoplasm [117]. Numerous approaches to downregulate the Ras signaling pathway were engaged. These are reviewed in depth elsewhere [118], and we intend to introduce only the fundamental pharmacologic mechanisms. Since the attempt at the allosteric inhibition of KRAS encountered difficulties, research efforts have turned towards the displacement of KRAS from the plasma membrane, the inhibition of downward signaling molecules, and the percussion of the cancer cell’s metabolic repertoire [118]. Recently, Kesler et al. discovered the druggable S I/II-pocket, a lipophilic cleft which could, upon pharmacologic inhibition, arrest the GTPase-activating protein (GAP), guanine nucleotide exchange factors (GEFs), and the downstream synergies of KRAS [119], potentially offering a pan-RAS inhibitor, while the first switch II-pocket inhibitors have gone into clinical trials [120]. In parallel, significant progress has been made in the development of exosome-based therapeutic applications.
Τhe production of cutting-edge biotechnological therapeutic products often runs into the inability to produce sufficient quantities to cover patients’ needs. Mendt et al. generated siRNA-loaded exosomes to an extensive scale. The KrasG12D-silencing RNAs were incorporated via electroporation into mesenchymal-derived exosomes inhibiting the PDAC tumor growth in mouse xenografts. After intraperitoneal administration, iExosomes were allocated preferentially in pancreatic tumor tissue in comparison to the neighboring organs with a mechanism which remains unclarified [121]. The KRAS-driven enhancement of micropinocytosis [122] might be a contributing mechanism. The prolongation of survival is documented in several PDAC mouse models [121]. In the same direction, Kamerkar et al. documented the superiority of CD47-anchored fibroblast-derived exosomes when compared with liposomes. The “don’t eat me” signal conferred by CD47 enhanced the exosome’s bioavailability, significantly limiting the side-effects profile. The incorporation of KRASG12C-silencing RNAs significantly shrank the tumor volume in the mouse models of orthotopic tumors [122]. These encouraging results led to a Phase I clinical trial. Patients with Stage IV clinical disease will be subjected to the administration of KrasG12D siRNA-carrying MSC-derived exosome at days one, four, ten, and every two weeks for three courses. The results are expected to be released soon [123].

4. Conclusions—Future Perspectives

The use of exosomes in clinical practice has the potential to become an important weapon in our therapeutic armory, capable of serving the demands imposed by personalized precision medicine. Although the consolidation of evidence concerning the importance of exosomes is relatively recent, a significant amount of preclinical data has begun to accumulate into the literature and a modest fraction of this is directed into clinical trials in order to test its diagnostic [124] or therapeutic capacity [123]. In parallel, the generation of a PDAC organoid which accurately recreates a tumor’s ecosystem has the potential to notably enrich the scientific community with data generated from the interactions among several cell populations [125]. The exosomally delivered KrasG12D siRNA constitutes the first KRAS-targeting therapy, and if it succeeds, it could transform the architecture of PDAC’s treatment algorithms, while the favorable usage profile of exosomes with an extensive distribution in body fluids and a minor triggering of immune responses [126] could render them ideal carriers of the established chemotherapeutics paclitaxel [99,102], GEM [100,101], and repurposed natural extracts such as curcumin [103]. Scientific advancements have given us the means to engineer exosomes providing theranostics technological applications [83,116] which combine diagnostic and radicalized treatment avenues such as the modification of a tumor’s immune ecosystem in conjunction with photodynamic therapy (PDT) [116]. The continual delineating of drug-resistance mechanisms elucidates the principal role of exosomes in the process and could ideally offer new treatment-enhancing targets. Finally, given the implications exosomes hold in the generation of the pre-metastatic niche [127,128], the introduction of exosome inhibitors might revolutionize therapy for metastatic disease, which is the main cause of death in cancer patients.

Author Contributions

Conceptualization, S.P.P., N.D. and S.T.; Writing—original draft preparation, S.P.P. and N.D.; writing—review and editing, A.P.; visualization, S.P.P. and N.D.; supervision, M.G. and S.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Ferlay, J.; Partensky, C.; Bray, F. More deaths from pancreatic cancer than breast cancer in the EU by 2017. Acta Oncol. 2016, 55, 1158–1160. [Google Scholar] [CrossRef] [PubMed]
  3. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting Cancer Incidence and Deaths to 2030: The Unexpected Burden of Thyroid, Liver, and Pancreas Cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Karim-Kos, H.E.; de Vries, E.; Soerjomataram, I.; Lemmens, V.; Siesling, S.; Coebergh, J.W.W. Recent trends of cancer in Europe: A combined approach of incidence, survival and mortality for 17 cancer sites since the 1990s. Eur. J. Cancer 2008, 44, 1345–1389. [Google Scholar] [CrossRef]
  5. Becker, A.E.; Hernandez, Y.G.; Frucht, H.; Lucas, A.L. Pancreatic ductal adenocarcinoma: Risk factors, screening, and early detection. World J. Gastroenterol. 2014, 20, 11182–11198. [Google Scholar] [CrossRef]
  6. Ilic, M.; Ilic, I. Epidemiology of pancreatic cancer. World J. Gastroenterol. 2016, 22, 9694–9705. [Google Scholar] [CrossRef]
  7. Chen, R.; Lai, L.A.; Sullivan, Y.; Wong, M.; Wang, L.; Riddell, J.; Jung, L.; Pillarisetty, V.G.; Brentnall, T.A.; Pan, S. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer. Sci. Rep. 2017, 7, 7950. [Google Scholar] [CrossRef] [Green Version]
  8. Haeberle, L.; Esposito, I. Pathology of pancreatic cancer. Transl. Gastroenterol. Hepatol. 2019, 4, 1–12. [Google Scholar] [CrossRef] [PubMed]
  9. Vrieling, A.; Bueno-de-Mesquita, H.B.; Boshuizen, H.C.; Michaud, D.S.; Severinsen, M.T.; Overvad, K.; Olsen, A.; Tjønneland, A.; Clavel-Chapelon, F.; Boutron-Ruault, M.C.; et al. Cigarette smoking, environmental tobacco smoke exposure and pancreatic cancer risk in the European Prospective Investigation into Cancer and Nutrition. Int. J. Cancer 2010, 126, 2394–2403. [Google Scholar] [CrossRef]
  10. Iodice, S.; Gandini, S.; Maisonneuve, P.; Lowenfels, A.B. Tobacco and the risk of pancreatic cancer: A review and meta-analysis. Langenbecks Arch. Surg. 2008, 393, 535–545. [Google Scholar] [CrossRef]
  11. Rastogi, T.; Devesa, S.; Mangtani, P.; Mathew, A.; Cooper, N.; Kao, R.; Sinha, R. Cancer incidence rates among South Asians in four geographic regions: India, Singapore, UK and US. Int. J. Epidemiol. 2007, 37, 147–160. [Google Scholar] [CrossRef] [PubMed]
  12. Maisonneuve, P.; Lowenfels, A.B. Risk factors for pancreatic cancer: A summary review of meta-analytical studies. Int. J. Epidemiol. 2015, 44, 186–198. [Google Scholar] [CrossRef] [PubMed]
  13. Bosetti, C.; Rosato, V.; Li, D.; Silverman, D.; Petersen, G.M.; Bracci, P.M.; Neale, R.E.; Muscat, J.; Anderson, K.; Gallinger, S.; et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: An analysis from the International Pancreatic Cancer Case-Control Consortium. Ann. Oncol. 2014, 25, 2065–2072. [Google Scholar] [CrossRef] [PubMed]
  14. Rosato, V.; Polesel, J.; Bosetti, C.; Serraino, D.; Negri, E.; Vecchia, C. La Population attributable risk for pancreatic cancer in northern Italy. Pancreas 2015, 44, 216–220. [Google Scholar] [CrossRef] [PubMed]
  15. Jacobs, E.J.; Chanock, S.J.; Fuchs, C.S.; LaCroix, A.; McWilliams, R.R.; Steplowski, E.; Stolzenberg-Solomon, R.Z.; Arslan, A.A.; Bas Bueno-de-Mesquita, H.; Gross, M.; et al. Family history of cancer and risk of pancreatic cancer: A pooled analysis from the Pancreatic Cancer Cohort Consortium (PanScan). Int. J. Cancer 2010, 127, 1421–1428. [Google Scholar] [CrossRef] [PubMed]
  16. Greer, J.B.; Whitcomb, D.C.; Brand, R.E. Genetic predisposition to pancreatic cancer: A brief review. Am. J. Gastroenterol. 2007, 102, 2564–2569. [Google Scholar] [CrossRef] [PubMed]
  17. Slebos, R.J.C.; Hoppin, J.A.; Tolbert, P.E.; Holly, E.A.; Brock, J.W.; Zhang, R.H.; Bracci, P.M.; Foley, J.; Stockton, P.; McGregor, L.M.; et al. K-ras and p53 in pancreatic cancer: Association with medical history, histopathology, and environmental exposures in a population-based study. Cancer Epidemiol. Prev. Biomark. 2000, 9, 1223–1232. [Google Scholar]
  18. Hashimoto, D.; Arima, K.; Yokoyama, N.; Chikamoto, A.; Taki, K.; Inoue, R.; Kaida, T.; Higashi, T.; Nitta, H.; Ohmuraya, M.; et al. Heterogeneity of KRAS Mutations in Pancreatic Ductal Adenocarcinoma. Pancreas 2016, 45, 1111–1114. [Google Scholar] [CrossRef]
  19. Singh, H.; Longo, D.L.; Chabner, B.A. Improving prospects for targeting RAS. J. Clin. Oncol. 2015, 33, 3650–3659. [Google Scholar] [CrossRef]
  20. Hobbs, G.A.; Baker, N.M.; Miermont, A.M.; Thurman, R.D.; Tran, T.H.; Anderson, A.O.; Waters, A.M.; Nathaniel, J.; Papke, B.; Hodge, R.G.; et al. Atypical KRASG12R Mutant Is Impaired in PI3K Signaling and Macropinocytosis in Pancreatic Cancer. Cancer Discov. 2020, 10, 104–123. [Google Scholar] [CrossRef]
  21. Waddell, N.; Pajic, M.; Patch, A.M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hingorani, S.R.; Wang, L.; Multani, A.S.; Combs, C.; Deramaudt, T.B.; Hruban, R.H.; Rustgi, A.K.; Chang, S.; Tuveson, D.A. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005, 7, 469–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Martinez-Useros, J.; Garcia-Foncillas, J. Can molecular biomarkers change the paradigm of pancreatic cancer prognosis? BioMed Res. Int. 2016, 2016, 4873089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Longo, D.L.; Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic Adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1088. [Google Scholar] [CrossRef]
  25. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2015. CA Cancer J. Clin. 2015, 65, 5–29. [Google Scholar] [CrossRef] [PubMed]
  26. Stathis, A.; Moore, M.J. Advanced pancreatic carcinoma: Current treatment and future challenges. Nat. Rev. Clin. Oncol. 2010, 7, 163–172. [Google Scholar] [CrossRef] [PubMed]
  27. Dhir, M.; Malhotra, G.K.; Sohal, D.P.S.; Hein, N.A.; Smith, L.M.; O’Reilly, E.M.; Bahary, N.; Are, C. Neoadjuvant treatment of pancreatic adenocarcinoma: A systematic review and meta-analysis of 5520 patients. World J. Surg. Oncol. 2017, 15, 183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.-L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  29. Zhao, J.; Schlößer, H.A.; Wang, Z.; Qin, J.; Li, J.; Popp, F.; Popp, M.C.; Alakus, H.; Chon, S.H.; Hansen, H.P.; et al. Tumor-derived extracellular vesicles inhibit natural killer cell function in pancreatic cancer. Cancers 2019, 11, 874. [Google Scholar] [CrossRef] [Green Version]
  30. Sohal, D.P.S.; Kennedy, E.B.; Cinar, P.; Conroy, T.; Copur, M.S.; Crane, C.H.; Garrido-Laguna, I.; Lau, M.W.; Johnson, T.; Krishnamurthi, S.; et al. Metastatic pancreatic cancer: ASCO guideline update. J. Clin. Oncol. 2020, 38, 3217–3230. [Google Scholar] [CrossRef]
  31. Middleton, G.; Palmer, D.H.; Greenhalf, W.; Ghaneh, P.; Jackson, R.; Cox, T.; Evans, A.; Shaw, V.E.; Wadsley, J.; Valle, J.W.; et al. Vandetanib plus gemcitabine versus placebo plus gemcitabine in locally advanced or metastatic pancreatic carcinoma (ViP): A prospective, randomised, double-blind, multicentre phase 2 trial. Lancet Oncol. 2017, 18, 486–499. [Google Scholar] [CrossRef]
  32. Kimura, H.; Yamamoto, H.; Harada, T.; Fumoto, K.; Osugi, Y.; Sada, R.; Maehara, N.; Hikita, H.; Mori, S.; Eguchi, H.; et al. CKAP4, a DKK1 receptor, is a biomarker in exosomes derived from pancreatic cancer and a molecular target for therapy. Clin. Cancer Res. 2019, 25, 1936–1947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Chung, I.M.; Rajakumar, G.; Venkidasamy, B.; Subramanian, U.; Thiruvengadam, M. Exosomes: Current use and future applications. Clin. Chim. Acta 2020, 500, 226–232. [Google Scholar] [CrossRef] [PubMed]
  34. Jiang, L.; Gu, Y.; Du, Y.; Liu, J. Exosomes: Diagnostic Biomarkers and Therapeutic Delivery Vehicles for Cancer. Mol. Pharm. 2019, 16, 3333–3349. [Google Scholar] [CrossRef]
  35. Huang, T.; Deng, C.X. Current Progresses of Exosomes as Cancer Diagnostic and Prognostic Biomarkers. Int. J. Biol. Sci. 2019, 15, 1–11. [Google Scholar] [CrossRef]
  36. Liu, Q.; Li, S.; Dupuy, A.; Le Mai, H.; Sailliet, N.; Logé, C.; Robert, J.M.H.; Brouard, S. Exosomes as new biomarkers and drug delivery tools for the prevention and treatment of various diseases: Current perspectives. Int. J. Mol. Sci. 2021, 22, 7763. [Google Scholar] [CrossRef] [PubMed]
  37. Kok, V.C.; Yu, C.C. Cancer-derived exosomes: Their role in cancer biology and biomarker development. Int. J. Nanomed. 2020, 15, 8019–8036. [Google Scholar] [CrossRef] [PubMed]
  38. Moris, D.; Beal, E.W.; Chakedis, J.; Burkhart, R.A.; Schmidt, C.; Dillhoff, M.; Zhang, X.; Theocharis, S.; Pawlik, T.M. Role of exosomes in treatment of hepatocellular carcinoma. Surg. Oncol. 2017, 26, 219–228. [Google Scholar] [CrossRef]
  39. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  40. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef]
  41. Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: Artefacts no more. Trends Cell Biol. 2009, 19, 43–51. [Google Scholar] [CrossRef] [PubMed]
  42. Ratajczak, M.Z.; Ratajczak, J. Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia 2020, 34, 3126–3135. [Google Scholar] [CrossRef] [PubMed]
  43. Dini, L.; Tacconi, S.; Carata, E.; Tata, A.M.; Vergallo, C.; Panzarini, E. Microvesicles and exosomes in metabolic diseases and inflammation. Cytokine Growth Factor Rev. 2020, 51, 27–39. [Google Scholar] [CrossRef] [PubMed]
  44. Caruso, S.; Poon, I.K.H. Apoptotic cell-derived extracellular vesicles: More than just debris. Front. Immunol. 2018, 9, 1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Green, D.R. Introduction: Apoptosis in the development and function of the immune system. Nat. Immunol. 2003, 15, 121–123. [Google Scholar] [CrossRef]
  46. Ma, Q.; Liang, M.; Wu, Y.; Ding, N.; Duan, L.; Yu, T.; Bai, Y.; Kang, F.; Dong, S.; Xu, J.; et al. Mature osteoclast- derived apoptotic bodies promote osteogenic differentiation via RANKL-mediated reverse signaling. J. Biol. Chem. 2019, 294, 11240–11247. [Google Scholar] [CrossRef]
  47. Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [CrossRef]
  48. Théry, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and Characterization of Exosomes from Cell Culture Supernatants. Curr. Protoc. Cell Biol. 2006, 30, 3–22. [Google Scholar] [CrossRef] [PubMed]
  49. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  50. Marofi, F.; Alexandrovna, K.I.; Margiana, R.; Bahramali, M.; Suksatan, W.; Abdelbasset, W.K.; Chupradit, S.; Nasimi, M.; Maashi, M.S. MSCs and their exosomes: A rapidly evolving approach in the context of cutaneous wounds therapy. Stem Cell Res. Ther. 2021, 12, 597. [Google Scholar] [CrossRef]
  51. Masaoutis, C.; Theocharis, S. The Role of Exosomes in Bone Remodeling: Implications for Bone Physiology and Disease. Dis. Markers 2019, 2019, 9417914. [Google Scholar] [CrossRef] [PubMed]
  52. Weingrill, R.B.; Paladino, S.L.; Souza, M.L.R.; Pereira, E.M.; Marques, A.L.X.; Silva, E.C.O.; da Silva Fonseca, E.J.; Ursulino, J.S.; Aquino, T.M.; Bevilacqua, E.; et al. Exosome-Enriched Plasma Analysis as a Tool for the Early Detection of Hypertensive Gestations. Front. Physiol. 2021, 12, 767112. [Google Scholar] [CrossRef] [PubMed]
  53. Sabaratnam, R.; Wojtaszewski, J.F.P.; Højlund, K. Factors Mediating Exercise-induced Organ Crosstalk. Acta Physiol. 2022, 234, e13766. [Google Scholar] [CrossRef] [PubMed]
  54. Shin, M.J.; Park, J.Y.; Lee, D.H.; Khang, D. Stem Cell Mimicking Nanoencapsulation for Targeting Arthritis. Int. J. Nanomed. 2021, 16, 8485–8507. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, W.; Yue, C.; Gao, S.; Li, S.; Zhou, J.; Chen, J.; Fu, J.; Sun, W.; Hua, C. Promising Roles of Exosomal microRNAs in Systemic Lupus Erythematosus. Front. Immunol. 2021, 12, 757096. [Google Scholar] [CrossRef] [PubMed]
  56. Masaoutis, C.; Al Besher, S.; Koutroulis, I.; Theocharis, S. Exosomes in Nephropathies: A Rich Source of Novel Biomarkers. Dis. Markers 2020, 2020, 8897833. [Google Scholar] [CrossRef]
  57. Hadjimichael, A.C.; Pergaris, A.; Kaspiris, A.; Foukas, A.F.; Theocharis, S.E. Liquid Biopsy: A New Translational Diagnostic and Monitoring Tool for Musculoskeletal Tumors. Int. J. Mol. Sci. 2021, 22, 11526. [Google Scholar] [CrossRef]
  58. Masaoutis, C.; Mihailidou, C.; Tsourouflis, G.; Theocharis, S. Exosomes in lung cancer diagnosis and treatment. From the translating research into future clinical practice. Biochimie 2018, 151, 27–36. [Google Scholar] [CrossRef]
  59. Georgantzoglou, N.; Pergaris, A.; Masaoutis, C.; Theocharis, S.; Mione, M.C. Extracellular Vesicles as Biomarkers Carriers in Bladder Cancer: Diagnosis, Surveillance, and Treatment. Int. J. Mol. Sci. 2021, 22, 2744. [Google Scholar] [CrossRef]
  60. Buscail, E.; Chauvet, A.; Quincy, P.; Degrandi, O.; Buscail, C.; Lamrissi, I.; Moranvillier, I.; Caumont, C.; Verdon, S.; Brisson, A.; et al. CD63-GPC1-Positive Exosomes Coupled with CA19-9 Offer Good Diagnostic Potential for Resectable Pancreatic Ductal Adenocarcinoma. Transl. Oncol. 2019, 12, 1395–1403. [Google Scholar] [CrossRef]
  61. Buscail, E.; Alix-Panabières, C.; Quincy, P.; Cauvin, T.; Chauvet, A.; Degrandi, O.; Caumont, C.; Verdon, S.; Lamrissi, I.; Moranvillier, I.; et al. High clinical value of liquid biopsy to detect circulating tumor cells and tumor exosomes in pancreatic ductal adenocarcinoma patients eligible for up-front surgery. Cancers 2019, 11, 1656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Nakamura, S.; Sadakari, Y.; Ohtsuka, T.; Okayama, T.; Nakashima, Y.; Gotoh, Y.; Saeki, K.; Mori, Y.; Nakata, K.; Miyasaka, Y.; et al. Pancreatic Juice Exosomal MicroRNAs as Biomarkers for Detection of Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2019, 26, 2104–2111. [Google Scholar] [CrossRef] [PubMed]
  63. Comandatore, A.; Immordino, B.; Balsano, R.; Capula, M.; Garajovà, I.; Ciccolini, J.; Giovannetti, E.; Morelli, L. Potential Role of Exosomes in the Chemoresistance to Gemcitabine and Nab-Paclitaxel in Pancreatic Cancer. Diagnostics 2022, 12, 286. [Google Scholar] [CrossRef] [PubMed]
  64. Li, Y.J.; Wu, J.Y.; Liu, J.; Xu, W.; Qiu, X.; Huang, S.; Hu, X.B.; Xiang, D.X. Artificial exosomes for translational nanomedicine. J. Nanobiotechnol. 2021, 19, 242. [Google Scholar] [CrossRef] [PubMed]
  65. Patty, P.J.; Frisken, B.J. The pressure-dependence of the size of extruded vesicles. Biophys. J. 2003, 85, 996–1004. [Google Scholar] [CrossRef] [Green Version]
  66. Liu, D.; Zhang, H.; Fontana, F.; Hirvonen, J.T.; Santos, H.A. Current developments and applications of microfluidic technology toward clinical translation of nanomedicines. Adv. Drug Deliv. Rev. 2018, 128, 54–83. [Google Scholar] [CrossRef] [Green Version]
  67. Go, G.; Lee, J.; Choi, D.S.; Kim, S.S.; Gho, Y.S. Extracellular Vesicle–Mimetic Ghost Nanovesicles for Delivering Anti-Inflammatory Drugs to Mitigate Gram-Negative Bacterial Outer Membrane Vesicle–Induced Systemic Inflammatory Response Syndrome. Adv. Healthc. Mater. 2019, 8, 1801082. [Google Scholar] [CrossRef]
  68. Sezgin, E.; Kaiser, H.J.; Baumgart, T.; Schwille, P.; Simons, K.; Levental, I. Elucidating membrane structure and protein behavior using giant plasma membrane vesicles. Nat. Protoc. 2012, 7, 1042–1051. [Google Scholar] [CrossRef]
  69. De La Peña, H.; Madrigal, J.A.; Rusakiewicz, S.; Bencsik, M.; Cave, G.W.V.; Selman, A.; Rees, R.C.; Travers, P.J.; Dodi, I.A. Artificial exosomes as tools for basic and clinical immunology. J. Immunol. Methods 2009, 344, 121–132. [Google Scholar] [CrossRef]
  70. Li, K.; Chang, S.; Wang, Z.; Zhao, X.; Chen, D. A novel micro-emulsion and micelle assembling method to prepare DEC205 monoclonal antibody coupled cationic nanoliposomes for simulating exosomes to target dendritic cells. Int. J. Pharm. 2015, 491, 105–112. [Google Scholar] [CrossRef]
  71. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef] [PubMed]
  72. Halbrook, C.J.; Lyssiotis, C.A. Employing Metabolism to Improve the Diagnosis and Treatment of Pancreatic Cancer. Cancer Cell 2017, 31, 5–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chu, G.C.; Kimmelman, A.C.; Hezel, A.F.; DePinho, R.A. Stromal biology of pancreatic cancer. J. Cell. Biochem. 2007, 101, 887–907. [Google Scholar] [CrossRef] [PubMed]
  74. Feig, C.; Gopinathan, A.; Neesse, A.; Chan, D.S.; Cook, N.; Tuveson, D.A. The pancreas cancer microenvironment. Clin. Cancer Res. 2012, 18, 4266–4276. [Google Scholar] [CrossRef] [Green Version]
  75. Provenzano, P.P.; Hingorani, S.R. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br. J. Cancer 2013, 108, 1–8. [Google Scholar] [CrossRef] [Green Version]
  76. Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kg to promote pancreatic cancer metastasis. Cancer Res. 2018, 78, 4586–4598. [Google Scholar] [CrossRef] [Green Version]
  77. Yang, Y.; Guo, Z.; Chen, W.; Wang, X.; Cao, M.; Han, X.; Zhang, K.; Teng, B.; Cao, J.; Wu, W.; et al. M2 Macrophage-Derived Exosomes Promote Angiogenesis and Growth of Pancreatic Ductal Adenocarcinoma by Targeting E2F2. Mol. Ther. 2021, 29, 1226–1238. [Google Scholar] [CrossRef]
  78. 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, 1220–1237. [Google Scholar] [CrossRef]
  79. Kishimoto, S.; Brender, J.R.; Chandramouli, G.V.R.; Saida, Y.; Yamamoto, K.; Mitchell, J.B.; Krishna, M.C. Hypoxia-Activated Prodrug Evofosfamide Treatment in Pancreatic Ductal Adenocarcinoma Xenografts Alters the Tumor Redox Status to Potentiate Radiotherapy. Antioxid. Redox Signal. 2020, 35, 904–915. [Google Scholar] [CrossRef]
  80. Binenbaum, Y.; Fridman, E.; Yaari, Z.; Milman, N.; Schroeder, A.; David, G. Ben; Shlomi, T.; Gil, Z. Transfer of miRNA in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Cancer Res. 2018, 78, 5287–5299. [Google Scholar] [CrossRef] [Green Version]
  81. Xie, Z.; Gao, Y.; Ho, C.; Li, L.; Jin, C.; Wang, X.; Zou, C.; Mao, Y.; Wang, X.; Li, Q.; et al. Exosome-delivered CD44v6/C1QBP complex drives pancreatic cancer liver metastasis by promoting fibrotic liver microenvironment. Gut 2022, 71, 568–579. [Google Scholar] [CrossRef] [PubMed]
  82. Liu, Y.; Tang, T.; Yang, X.; Qin, P.; Wang, P.; Zhang, H.; Bai, M.; Wu, R.; Li, F. Tumor-derived exosomal long noncoding RNA LINC01133, regulated by Periostin, contributes to pancreatic ductal adenocarcinoma epithelial-mesenchymal transition through the Wnt/β-catenin pathway by silencing AXIN2. Oncogene 2021, 40, 3164–3179. [Google Scholar] [CrossRef] [PubMed]
  83. Zhou, W.; Zhou, Y.; Chen, X.; Ning, T.; Chen, H.; Guo, Q.; Zhang, Y.; Liu, P.; Zhang, Y.; Li, C.; et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials 2021, 268, 120546. [Google Scholar] [CrossRef] [PubMed]
  84. Tempero, M.; Oh, D.Y.; Tabernero, J.; Reni, M.; Van Cutsem, E.; Hendifar, A.; Waldschmidt, D.T.; Starling, N.; Bachet, J.B.; Chang, H.M.; et al. Ibrutinib in combination with nab-paclitaxel and gemcitabine for first-line treatment of patients with metastatic pancreatic adenocarcinoma: Phase III RESOLVE study. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2021, 32, 600–608. [Google Scholar] [CrossRef] [PubMed]
  85. Jiang, H.; Courau, T.; Borison, J.; Ritchie, A.J.; Mayer, A.T.; Krummel, M.F.; Collisson, E.A. Activating Immune Recognition in Pancreatic Ductal Adenocarcinoma via Autophagy Inhibition, MEK Blockade, and CD40 Agonism. Gastroenterology 2022, 162, 590–603. [Google Scholar] [CrossRef]
  86. Wang, R.; Chen, J.; Wang, W.; Zhao, Z.; Wang, H.; Liu, S.; Li, F.; Wan, Y.; Yin, J.; Wang, R.; et al. CD40L-armed oncolytic herpes simplex virus suppresses pancreatic ductal adenocarcinoma by facilitating the tumor microenvironment favorable to cytotoxic T cell response in the syngeneic mouse model. J. Immunother. Cancer 2022, 10, e003809. [Google Scholar] [CrossRef]
  87. Sharma, A. Chemoresistance in cancer cells: Exosomes as potential regulators of therapeutic tumor heterogeneity. Nanomedicine 2017, 12, 2137–2148. [Google Scholar] [CrossRef]
  88. Koch, R.; Aung, T.; Vogel, D.; Chapuy, B.; Wenzel, D.; Becker, S.; Sinzig, U.; Venkataramani, V.; Von Mach, T.; Jacob, R.; et al. Cancer Therapy: Preclinical Nuclear Trapping through Inhibition of Exosomal Export by Indomethacin Increases Cytostatic Efficacy of Doxorubicin and Pixantrone. Clin. Cancer Res. 2016, 22, 395–404. [Google Scholar] [CrossRef] [Green Version]
  89. Patel, G.K.; Khan, M.A.; Bhardwaj, A.; Srivastava, S.K.; Zubair, H.; Patton, M.C.; Singh, S.; Khushman, M.; Singh, A.P. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br. J. Cancer 2017, 116, 609–619. [Google Scholar] [CrossRef] [Green Version]
  90. Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K.; et al. MicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma OPEN. Sci. Rep. 2017, 7, 42339. [Google Scholar] [CrossRef]
  91. Mesri, M.; Wall, N.R.; Li, J.; Kim, R.W.; Altieri, D.C. Cancer gene therapy using survivin mutant adenovirus. J. Clin. Investig. 2001, 108, 981–990. [Google Scholar] [CrossRef] [PubMed]
  92. Aspe, J.R.; Osterman, C.J.D.; Jutzy, J.M.S.; Deshields, S.; Whang, S.; Wall, N.R. Enhancement of Gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome-mediated delivery of the Survivin-T34A mutant. J. Extracell. Vesicles 2014, 3, 23244. [Google Scholar] [CrossRef] [PubMed]
  93. Pergaris, A.; Danas, E.; Goutas, D.; Sykaras, A.G.; Soranidis, A.; Theocharis, S. Molecular Sciences The Clinical Impact of the EPH/Ephrin System in Cancer: Unwinding the Thread. Int. J. Mol. Sci. 2021, 22, 8412. [Google Scholar] [CrossRef]
  94. Fan, J.; Wei, Q.; Koay, E.J.; Liu, Y.; Ning, B.; Bernard, P.W.; Zhang, N.; Han, H.; Katz, M.H.; Zhao, Z.; et al. Chemoresistance Transmission via Exosome-Mediated EphA2 Transfer in Pancreatic Cancer. Theranostics 2018, 8, 21. [Google Scholar] [CrossRef] [PubMed]
  95. Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Physiol. Behav. 2017, 176, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Fang, Y.; Zhou, W.; Rong, Y.; Kuang, T.; Xu, X.; Wu, W.; Wang, D.; Lou, W. Exosomal miRNA-106b from cancer-associated fibroblast promotes gemcitabine resistance in pancreatic cancer. Exp. Cell Res. 2019, 383, 111543. [Google Scholar] [CrossRef] [PubMed]
  97. Yang, Z.; Zhao, N.; Cui, J.; Wu, H.; Xiong, J.; Peng, T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell. Oncol. 2020, 43, 123–136. [Google Scholar] [CrossRef] [Green Version]
  98. Yang, D.-H.; John, S.; Xu, Y.; Yang, J.; Li, L.; Wang, J.; Song, H.; Liu, B.; Dong, B.; Xu, J.; et al. Exosome-Based Delivery of Natural Products in Cancer Therapy. Front. Cell Dev. Biol. 2021, 9, 650426. [Google Scholar] [CrossRef]
  99. Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Extracell. Vesicles 2015, 192, 99–107. [Google Scholar] [CrossRef]
  100. Bonomi, A.; Sordi, V.; Dugnani, E.; Ceserani, V.; Dossena, M.; Coccè, V.; Cavicchini, L.; Ciusani, E.; Bondiolotti, G.; Piovani, G.; et al. Gemcitabine-releasing mesenchymal stromal cells inhibit in vitro proliferation of human pancreatic carcinoma cells. Cytotherapy 2015, 17, 1687–1695. [Google Scholar] [CrossRef] [Green Version]
  101. 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, 519–530. [Google Scholar] [CrossRef] [PubMed]
  102. Zhou, Y.; Zhou, W.; Chen, X.; Wang, Q.; Li, C.; Chen, Q.; Zhang, Y.; Lu, Y.; Ding, X.; Jiang, C. Bone marrow mesenchymal stem cells-derived exosomes for penetrating and targeted chemotherapy of pancreatic cancer. Acta Pharm. Sin. B 2020, 10, 1563–1575. [Google Scholar] [CrossRef] [PubMed]
  103. Osterman, C.J.D.; Lynch, J.C.; Leaf, P.; Gonda, A.; Bennit, H.R.F.; Griffiths, D.; Wall, N.R. Curcumin modulates pancreatic adenocarcinoma cell-derived exosomal function. PLoS ONE 2015, 10, e0132845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Shang, S.; Wang, J.; Chen, S.; Tian, R.; Zeng, H.; Wang, L.; Xia, M.; Zhu, H.; Zuo, C. Exosomal miRNA-1231 derived from bone marrow mesenchymal stem cells inhibits the activity of pancreatic cancer. Cancer Med. 2019, 8, 7728–7740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wu, D.M.; Wen, X.; Han, X.R.; Wang, S.; Wang, Y.J.; Shen, M.; Fan, S.H.; Zhang, Z.F.; Shan, Q.; Li, M.Q.; et al. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal MicroRNA-126-3p Inhibits Pancreatic Cancer Development by Targeting ADAM9. Mol. Ther.-Nucleic Acids 2019, 16, 229–245. [Google Scholar] [CrossRef] [Green Version]
  106. Ding, Y.; Cao, F.; Sun, H.; Wang, Y.; Liu, S.; Wu, Y.; Cui, Q.; Mei, W.T.; Li, F. Exosomes derived from human umbilical cord mesenchymal stromal cells deliver exogenous miR-145-5p to inhibit pancreatic ductal adenocarcinoma progression. Cancer Lett. 2019, 442, 351–361. [Google Scholar] [CrossRef] [PubMed]
  107. Shang, D.; Xie, C.; Hu, J.; Tan, J.; Yuan, Y.; Liu, Z.; Yang, Z. Pancreatic cancer cell–derived exosomal microRNA-27a promotes angiogenesis of human microvascular endothelial cells in pancreatic cancer via BTG2. J. Cell. Mol. Med. 2020, 24, 588–604. [Google Scholar] [CrossRef] [Green Version]
  108. Takikawa, T.; Masamune, A.; Yoshida, N.; Hamada, S.; Kogure, T.; Shimosegawa, T. Exosomes derived from pancreatic stellate cells. Pancreas 2017, 46, 19–27. [Google Scholar] [CrossRef]
  109. Wang, Z.; Sun, H.; Provaznik, J.; Hackert, T.; Zöller, M. Pancreatic cancer-initiating cell exosome message transfer into noncancer-initiating cells: The importance of CD44v6 in reprogramming. J. Exp. Clin. Cancer Res. 2019, 38, 132. [Google Scholar] [CrossRef] [Green Version]
  110. Yin, Z.; Ma, T.; Huang, B.; Lin, L.; Zhou, Y.; Yan, J.; Zou, Y.; Chen, S. Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. J. Exp. Clin. Cancer Res. 2019, 38, 310. [Google Scholar] [CrossRef] [Green Version]
  111. Wu, M.; Tan, X.; Liu, P.; Yang, Y.; Huang, Y.; Liu, X.; Meng, X.; Yu, B.; Wu, Y.; Jin, H. Role of exosomal microRNA-125b-5p in conferring the metastatic phenotype among pancreatic cancer cells with different potential of metastasis. Life Sci. 2020, 255, 117857. [Google Scholar] [CrossRef] [PubMed]
  112. Harzstark, A.L.; Small, E.J. Immunotherapy for prostate cancer using antigen-loaded antigen-presenting cells: APC8015(Provenge®). Expert Opin. Biol. Ther. 2007, 7, 1275–1280. [Google Scholar] [CrossRef] [PubMed]
  113. Su, M.J.; Aldawsari, H.; Amiji, M. Pancreatic cancer cell exosome-mediated macrophage reprogramming and the role of MicroRNAs 155 and 125b2 transfection using nanoparticle delivery systems. Sci. Rep. 2016, 6, 30110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Que, R.S.; Lin, C.; Ding, G.P.; Wu, Z.R.; Cao, L.P. ping Increasing the immune activity of exosomes: The effect of miRNA-depleted exosome proteins on activating dendritic cell/cytokine-induced killer cells against pancreatic cancer. J. Zhejiang Univ. Sci. B 2016, 17, 352–360. [Google Scholar] [CrossRef] [Green Version]
  115. Xiao, L.; Erb, U.; Zhao, K.; Hackert, T.; Zöller, M. Efficacy of vaccination with tumor-exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology 2017, 6, e1319044. [Google Scholar] [CrossRef] [Green Version]
  116. Jang, Y.; Kim, H.; Yoon, S.; Lee, H.; Hwang, J.; Jung, J.; Chang, J.H.; Choi, J.; Kim, H. Exosome-based photoacoustic imaging guided photodynamic and immunotherapy for the treatment of pancreatic cancer. J. Control. Release 2021, 330, 293–304. [Google Scholar] [CrossRef]
  117. Cox, A.D.; Fesik, S.W.; Kimmelman, A.C.; Luo, J.; Der, C.J. Drugging the undruggable RAS: Mission Possible? Nat. Rev. Drug Discov. 2014, 13, 828–851. [Google Scholar] [CrossRef] [Green Version]
  118. Gorfe, A.A.; Cho, K.-J. Approaches to inhibiting oncogenic K-Ras. Small GTPases 2021, 12, 96–105. [Google Scholar] [CrossRef]
  119. Kessler, D.; Gmachl, M.; Mantoulidis, A.; Martin, L.J.; Zoephel, A.; Mayer, M.; Gollner, A.; Covini, D.; Fischer, S.; Gerstberger, T.; et al. Drugging an undruggable pocket on KRAS. Proc. Natl. Acad. Sci. USA 2019, 116, 15823–15829. [Google Scholar] [CrossRef] [Green Version]
  120. Kessler, D.; Bergner, A.; Böttcher, J.; Fischer, G.; Döbel, S.; Hinkel, M.; Müllauer, B.; Weiss-Puxbaum, A.; McConnell, D.B. Drugging all RAS isoforms with one pocket. Future Med. Chem. 2020, 12, 1911–1923. [Google Scholar] [CrossRef]
  121. Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.C.; Gagea, M.; Yang, S.; Blanko, E.V.R.; Peng, Q.; Ma, X.; et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 2018, 3, e99263. [Google Scholar] [CrossRef] [PubMed]
  122. Kamerkar, S.; Lebleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes Facilitate Therapeutic Targeting of Oncogenic Kras in Pancreatic Cancer Sushrut. Nature 2017, 176, 139–148. [Google Scholar] [CrossRef]
  123. iExosomes in Treating Participants with Metastatic Pancreas Cancer with KrasG12D Mutation—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03608631 (accessed on 29 September 2021).
  124. Circulating Extracellular Exosomal Small RNA as Potential Biomarker for Human Pancreatic Cancer—Full Text View—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04636788?term=exosomes&cond=Pancreas+Cancer&draw=2&rank=8 (accessed on 5 October 2021).
  125. Below, C.R.; Kelly, J.; Brown, A.; Humphries, J.D.; Hutton, C.; Xu, J.; Lee, B.Y.; Cintas, C.; Zhang, X.; Hernandez-Gordillo, V.; et al. A microenvironment-inspired synthetic three-dimensional model for pancreatic ductal adenocarcinoma organoids. Nat. Mater. 2022, 21, 110–119. [Google Scholar] [CrossRef]
  126. Johnsen, K.B.; Gudbergsson, J.M.; Skov, M.N.; Pilgaard, L.; Moos, T.; Duroux, M. A comprehensive overview of exosomes as drug delivery vehicles—Endogenous nanocarriers for targeted cancer therapy. Biochim. Biophys. Acta—Rev. Cancer 2014, 1846, 75–87. [Google Scholar] [CrossRef]
  127. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef] [PubMed]
  128. Yu, Z.; Zhao, S.; Ren, L.; Wang, L.; Chen, Z.; Hoffman, R.M.; Zhou, J. Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget 2017, 8, 63461–63483. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The construction of artificial exosomes (A,B), the “top-down” strategy: forced passage through porous membranes or microfluidic devices, nitrogen cavitation; cell membrane blebbing. (C) The supramolecular construction of artificial exosomes with biohybrid technologies (D), the “bottom-up” methodology: the supramolecular construction of complex structures from basic components. Created with BioRender.com.
Figure 1. The construction of artificial exosomes (A,B), the “top-down” strategy: forced passage through porous membranes or microfluidic devices, nitrogen cavitation; cell membrane blebbing. (C) The supramolecular construction of artificial exosomes with biohybrid technologies (D), the “bottom-up” methodology: the supramolecular construction of complex structures from basic components. Created with BioRender.com.
Ijms 23 03620 g001
Figure 2. The PDAC microenvironment. Created with BioRender.com.
Figure 2. The PDAC microenvironment. Created with BioRender.com.
Ijms 23 03620 g002
Figure 3. The contribution of the PDAC microenvironment to the generation of drug resistance. (A) GEM-treated PDAC cells secrete miR-155 through exosomes enhancing the antioxidant capacity of cancer cells (through the downregulation of SOD2 and CAT), disrupting the activation of GEM (through the downregulation of DCK) and cellular stress-sensing systems (by upregulating TP53INP1). (B) GEM-treated CAFs exosomally secreted miR-106b, miR-146a, and Snail, all of which mediate their intrinsic resistance to GEM. (C) The M2-derived exosomally excreted miR-365 increased the intracellular NTP pool and the CDA. (D) GEM-resistant cancer stem cells secreted through exosomes miR-210 induced drug resistance through mTOR signaling potentiation. (E) The knockdown of EPHA2 in drug-resistant cells decreased their efficiency in transmitting GEM resistance. Created with BioRender.com.
Figure 3. The contribution of the PDAC microenvironment to the generation of drug resistance. (A) GEM-treated PDAC cells secrete miR-155 through exosomes enhancing the antioxidant capacity of cancer cells (through the downregulation of SOD2 and CAT), disrupting the activation of GEM (through the downregulation of DCK) and cellular stress-sensing systems (by upregulating TP53INP1). (B) GEM-treated CAFs exosomally secreted miR-106b, miR-146a, and Snail, all of which mediate their intrinsic resistance to GEM. (C) The M2-derived exosomally excreted miR-365 increased the intracellular NTP pool and the CDA. (D) GEM-resistant cancer stem cells secreted through exosomes miR-210 induced drug resistance through mTOR signaling potentiation. (E) The knockdown of EPHA2 in drug-resistant cells decreased their efficiency in transmitting GEM resistance. Created with BioRender.com.
Ijms 23 03620 g003
Table 1. The effects of chemotherapeutics utilized against PDAC in in vivo and in vitro experiments.
Table 1. The effects of chemotherapeutics utilized against PDAC in in vivo and in vitro experiments.
DrugOrigin of ExosomeCell Line/AnimalResultsReferences
PaclitaxelMesenchymalCFPAC-1 cellsLess than 10% greater IC50 of PTX-MV vs. PTX[99]
GEMMesenchymalCFPAC-1 cellsBM-MSCsGCB-CM has 40% less V50 than pMSCsGCB-CM[100]
GEMAutologousPanc-1 cells/BALB/c nude miceAt day 16 (after treatment): ExoGEM 10 mg/kg-50% lower tumor volume[101]
At day 16 (after treatment): GEM 50 mg/kg-2 fold increase in tumor volume
GEM and PaclitaxelBone Marrow-MSCMiaPaca-2 cells/nude miceSurvival:
Exo-GEMP-PTX group: 88 days
Exo group: 42 days
GEM group: 60 days
Exo-GEM group: 63 days
GEMnab-PTX group: 73 days
[102]
CurcuminAutologousPANC-1 and MIA PaCa-2 cell linesPANC-1 cells at 72 h:
curcumin-positive exosomes 50% cell killing vs. curcumin-negative exosomes 0% cell killing
[103]
MIA PaCa-2 at 72 h: curcumin-positive exosomes 60% cell killing vs. curcumin-negative exosomes 0% cell killing
Table 2. The preclinical data regarding the impact of certain exosomal miRNAs in PDAC.
Table 2. The preclinical data regarding the impact of certain exosomal miRNAs in PDAC.
DrugExosome OriginCell Line/AnimalPathway/FunctionOutcomesReferences
miR-1231 mimicsBM-MSCsBxPC-3 and PANC-1/female BALB/C nude miceEGFR, Cyclin EDownregulation[104]
Wound healingDeterioration
InvasionDeterioration
Tumor volumeDeterioration
miR-126-3p mimicsBM-MSCsPANC-1 cellsProliferation, migration, invasionDeterioration[105]
ApoptosisIncrease
ADAM9Downregulation
Growth rhythm/Tumor volumeDecrease
miR-145-5p mimicsUmbilical cord-MSCsCapan-1, CFPAC-1, BxPC3 and Panc-1 cell lines/nude miceSMAD3Suppression[106]
Tumor proliferationDecrease
miR-27a inhibitorsPC cell-derivedPDAC cell lines: H6c7, SW1990, Capan-1, BxPc-3 and PANC-1 and microvascular endothelial cell line: HMEC-1/nude miceBTG2Upregulation[107]
proliferation, migration, invasion, angiogenesisSuppression
ApoptosisIncrease
GW4869Pancreatic stellate cellPANC-1 and Suit-2 cell linesProliferation, migrationIncrease[108]
CXCL-1,-2Increase
RTK inhibitorsCancer-initiating cell (CIC)Capan-1, A818.4 cell linesCD44v6kd non-cic, Tspan8kd non-CICAlterations[109]
SurvivalIncrease
Tumor cell invasionSuppression
miR-501-3p antagomiRM2 macrophagesPANC-1, BxPC-3/male BALB/c nude miceTumor volumeSuppression[110]
Metastatic burdenSuppression
TGFBR3Increase
miR-125b-5p inhibitorPC-1.0 derived (greatly invasive)PC-1, PC-1.0 cellsSTARD13Downregulation[111]
Migration, invasionSuppression
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Papadakos, S.P.; Dedes, N.; Pergaris, A.; Gazouli, M.; Theocharis, S. Exosomes in the Treatment of Pancreatic Cancer: A Moonshot to PDAC Treatment? Int. J. Mol. Sci. 2022, 23, 3620. https://doi.org/10.3390/ijms23073620

AMA Style

Papadakos SP, Dedes N, Pergaris A, Gazouli M, Theocharis S. Exosomes in the Treatment of Pancreatic Cancer: A Moonshot to PDAC Treatment? International Journal of Molecular Sciences. 2022; 23(7):3620. https://doi.org/10.3390/ijms23073620

Chicago/Turabian Style

Papadakos, Stavros P., Nikolaos Dedes, Alexandros Pergaris, Maria Gazouli, and Stamatios Theocharis. 2022. "Exosomes in the Treatment of Pancreatic Cancer: A Moonshot to PDAC Treatment?" International Journal of Molecular Sciences 23, no. 7: 3620. https://doi.org/10.3390/ijms23073620

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

Article Metrics

Back to TopTop