International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
The incidence and mortality rates of pancreatic cancer (PC) have been steadily increasing; there were 441,000 cases of PC worldwide in 2017, compared with only 196,000 cases in 1990 (1). According to epidemiological data, PC has the lowest 5-year relative survival rate among all solid tumors, estimated at ~13% (2). Surgical resection combined with adjuvant chemotherapy represents the current standard of care and the most widely adopted clinical strategy for the treatment of PC (3). However, due to the asymptomatic nature of early-stage PC, the majority of patients are diagnosed at an advanced stage. Consequently, only a small subset of patients remains eligible for surgical intervention (4). Furthermore, the development of chemotherapy resistance, often resulting from prolonged treatment, notably impacts both the survival and quality of life of patients with PC. The pathogenesis of PC remains incompletely elucidated; however, in addition to modifiable risk factors such as smoking, alcohol consumption, obesity and diabetes, genetic mutations, familial inheritance and a complex tumor microenvironment (TME) are also recognized as critical contributing factors (1,5,6). The TME of PC is a complex ecosystem composed of cancer cells, stromal cells, immune cells and various extracellular components. PC cells actively drive genetic mutations; stromal cells produce abundant collagen and extracellular matrix leading to tissue fibrosis and highly expressed inflammatory factors create a pro-inflammatory environment. This unique TME establishes a robust foundation for the growth and metastasis of PC (7). Therefore, deepening the understanding of the molecular mechanisms underlying PC, developing more effective therapeutic strategies and overcoming chemotherapy resistance represent critical research directions that require further exploration.
Mesenchymal stem cells (MSCs) are multipotent stem cells characterized by high differentiation potential and strong self-renewal capacity, which originate from the mesoderm and ectoderm during early development (8). Studies have indicated that MSCs actively participate in the progression of PC, including tumor growth and metastasis of cancer cells, as well as the modulation of TME (9–11). Studies have reported that MSCs can promote the progression of PC by secreting pro-tumorigenic factors (12) and differentiating into cancer-associated fibroblasts (CAFs) (13). However, other evidence suggests that MSCs can also suppress the development of PC. For example, Mohr et al (14) demonstrated that systemic MSC-mediated delivery of soluble tumor necrosis factor-related apoptosis inducing ligand, combined with X-linked inhibitor of apoptosis protein inhibition, could inhibit PC growth and metastasis. Additionally, IL-10-modified human MSCs were shown to inhibit PC progression by suppressing the secretion of pro-inflammatory cytokines IL-6 and TNF-α and inhibiting tumor angiogenesis (15). The role of MSCs in PC is closely associated with their derived EXOs (16).
EXOs are extracellular vesicles (EVs) with a diameter of 30–150 nm that are capable of transmitting complex information between cells (17), across distant tissues (18) and between tumor and stromal compartments (19). They originate from a wide variety of sources and are present in nearly all bodily fluids, and EXOs from different origins exhibit distinct functions (20). MSC-derived EXOs (MSC-EXOs) share numerous functional similarities with MSCs. However, compared with MSCs, MSC-EXOs demonstrate enhanced safety, superior penetrability, improved compatibility and higher stability when interacting with tumor cells (21,22). In recent years, the role of MSC-EXOs in the treatment of PC has been extensively investigated. For instance, it has been reported that human umbilical cord MSC-EXOs (hUC-MSC-EXOs) can promote the growth of pancreatic ductal adenocarcinoma by transferring microRNA (miR/miRNA)-100-5p into the PC tumor model (23). By contrast, Xie et al (24) reported opposing findings, demonstrating that hsa-miRNA-128-3p carried by hUC-MSC-EXOs suppressed the proliferation, invasion and migration of pancreatic ductal adenocarcinoma cells by targeting galectin-3. These findings indicate that MSC-EXOs serve a dual role in PC. The present article systematically elaborates on the dual tumor-promoting and tumor-suppressing mechanisms of MSC-EXOs in PC. It also clarifies the potential factors influencing this duality and provides direction for their application in targeted PC therapy.
The International Society for Cell Therapy has established the minimal defining criteria for MSCs: i) They must exhibit plastic-adherence under standard culture conditions; ii) they must express specific surface markers such as CD73, CD90 and CD105; iii) they must possess the capacity to differentiate into osteoblasts, chondrocytes and adipocytes in vitro; and iv) they must lack expression of CD14, CD34, CD45, CD11b, CD79a, CD19 and human leukocyte antigen-DR (25,26). MSCs can be isolated from a wide range of biological tissues, including bone marrow, adipose tissue, umbilical cord, placenta and peripheral blood (27–29). In previous years, MSCs have demonstrated notable potential in the treatment of PC; however, several limitations have also been identified. For example, studies have indicated that MSCs may increase the risk of tumorigenicity and cell death (30,31). To address these concerns, researchers have proposed using MSC-EXOs as an alternative therapeutic approach for PC. These EXOs exhibit a number of functions similar to those of MSCs, offer improved safety and stability profiles and can serve as excellent carriers for delivering antitumor drugs (32).
EXOs are nanoscale EVs enclosed by a double-layer lipid membrane (33), which are widely derived from various cell types, such as cancer cells, immune cells, stem cells and even food or plant cells. The biogenesis and synthesis of EXOs involve a complex process (Fig. 1). Initially, their formation begins with the invagination of the cell membrane, where ubiquitination of surface receptors initiates the endocytic process, leading to the formation of early endosomes. Over time, early endosomes mature into late endosomes, within which multiple intraluminal vesicles (ILVs) are formed through inward budding; the entire structure is referred to as a multivesicular body (MVB) (34,35). MVBs primarily face two fates: One is fusion with lysosomes resulting in degradation of their contents, and the other is fusion with the plasma membrane, through which ILVs are released into the extracellular environment as EXOs via exocytosis (36). This process involves various key molecules, including Rab GTPase proteins; endosomal sorting complexes required for transport, tetraspanins and ceramide (37–40). EXOs carry a diverse range of bioactive molecules. Their protein cargo includes heat shock proteins (HSP70, HSP90), GTPases, tetraspanins (CD63, CD81, CD9, CD82), as well as various transport, fusion and adhesion molecules (41–43). Additionally, EXOs contain lipids such as cholesterol, sphingomyelin, glycosphingolipids, phosphatidylcholine, phosphatidylserine and ceramides (44), along with various nucleic acids such as mRNA, miRNA, circular (circ)RNA, long non-coding (lnc)RNA and small nuclear (sn)RNA (45).
The tumor-promoting role of MSC-EXOs in PC has been demonstrated in multiple studies (23,46,47). For instance, B-MSC-EXOs, AD-MSC-EXOs and UC-MSC-EXOs can jointly exert cancer-promoting effects through different mechanisms of action (Fig. 2). The current section will focus on elucidating the specific molecular mechanisms and signaling pathways underlying their pro-tumorigenic effects, including promoting tumor cell proliferation, shaping an immunosuppressive microenvironment and mediating chemotherapy resistance (Table I). By systematically elaborating these mechanisms, the present study aimed to provide researchers with a clear understanding of the tumor-promoting functions of MSC-EXOs in PC and establish a theoretical foundation for developing future exosome-targeted therapeutic strategies.
MSC-EXOs can directly promote the proliferation of PC cells, thereby exerting tumor-promoting effects. In the study by Ding et al (23), hUC-MSC-EXOs carrying miR-100-5p were demonstrated to promote the proliferation of PC cells PANC1 and BxPC3 in both in vivo and in vitro studies, consequently accelerating the growth of pancreatic ductal adenocarcinoma. This finding not only reveals the key mechanism by which hUC-MSC-EXOs contribute to the development and progression of pancreatic ductal adenocarcinoma through the transfer of miR-100-5p, but also provides new insights for targeted intervention. Inhibiting miR-100-5p may represent a potential therapeutic strategy to block EXO-mediated pro-tumorigenic effects, thereby opening new avenues for precision therapy in pancreatic ductal adenocarcinoma.
MSC-EXOs can participate in shaping the immunosuppressive microenvironment of PC through complex mechanisms, thereby providing favorable conditions for tumor initiation and progression. For example, Gao et al (46) found that bone marrow MSC-EXOs (B-MSC-EXOs) carrying hsa_circ_0006790 can recruit chromobox protein homolog 7, which subsequently recruits DNA methyltransferases, leading to hypermethylation of the promoter region of the downstream target gene S100A11 and resulting in its downregulation. As a key molecule involved in immunoregulation, the decreased expression of S100A11 ultimately induces immune escape in PC cells. This study by Gao et al (46) not only reveals the specific mechanism by which B-MSC-EXOs contribute to the formation of the immunosuppressive microenvironment in PC but also provides a potential target for developing novel immunotherapy strategies targeting the exosomal signaling axis.
MSC-EXOs can participate in mediating chemotherapy resistance in PC. For example, a study showed that human adipose MSC-EXOs (hAD-MSC-EXOs) are enriched with the lncRNA NEAT1, which can competitively bind to miR-491-5p, thereby relieving its inhibition of the transcription factor Snail. This subsequently leads to downregulation of suppressor of cytokine signaling 3 (SOCS3) expression, ultimately inducing resistance to gemcitabine in PC cells (47). This mechanism highlights the critical role of exosome-carried lncRNA in mediating chemotherapy resistance, not only providing a new molecular perspective for understanding drug resistance in PC but also offering potential therapeutic strategies for reversing resistance, such as targeting the NEAT1/miR-491-5p/Snail/SOCS3 signaling axis to reduce chemoresistance.
In conclusion, although MSC-EXOs derived from different tissues such as umbilical cord, bone marrow and adipose can promote the progression of PC through multiple mechanisms, related research is still relatively limited, and more experimental evidence is needed to verify the specific cancer-promoting mechanisms. In addition, apart from gemcitabine, there are currently numerous drugs used to inhibit the development of PC. Therefore, whether hAD-MSC-EXOs still exhibit cancer-promoting effects when used in conjuction with other anticancer drugs remains to be elucidated. In addition, whether MSC-EXOs derived from the same tissue only serve a promoting role in PC also remains to be elucidated. Through in-depth sorting and analysis, a more complex fact has been demonstrated: Even MSC-EXOs derived from the same tissue exhibit a dual role in PC and may either promote tumor development or inhibit its progression.
Although MSC-EXOs can promote the initiation and progression of PC through various mechanisms, current research focuses more on their tumor-suppressive roles. For instance, UC-MSC-EXOs, B-MSC-EXOs, hA-MSC-EXOs, hAF-MSC-EXOs and Dental-MSC-EXOs can jointly exert anti-cancer effects through different mechanisms of action (Fig. 3). In fact, owing to their notable immunomodulatory capabilities and tumor-homing properties, MSC-EXOs exhibit multifaceted functions in tumor suppression: On the one hand, they can directly inhibit the proliferation of PC cells and induce apoptosis; on the other hand, they can indirectly exert antitumor effects by modulating immune responses. Furthermore, due to their high biocompatibility, low immunogenicity and efficient targeted delivery capacity, MSC-EXOs serve as a highly promising drug delivery system and are widely used for loading antitumor agents, thereby further enhancing their value in suppressing PC (Table II) (48,49). Therefore, despite reports indicating that MSC-EXOs may promote the growth and development of PC, their functions in tumor suppression remain more comprehensive and hold greater translational potential.
Table II.A summary of research on the therapeutic potential of MSCs-EXOs of different sources in PC. |
MSC-EXOs exert notable tumor-suppressive effects by inhibiting tumor cell growth and inducing apoptosis. Multiple studies support this conclusion through various mechanisms. For instance, hsa-miRNA-128-3p in hUC-MSC-EXOs can effectively inhibit the proliferation, invasion and migration of PC PANC1 cells by targeting galectin-3 (24). Meanwhile, circ_0030167 from human (h)B-MSC-EXOs suppresses the proliferation, invasion and metastasis of PC cells by sponging miR-338-5p and regulating the Wnt inhibitory factor 1/Wnt8/β-catenin axis (50). Additionally, miRNA-1231 from B-MSC-EXOs negatively regulates the proliferation, migration, invasion and matrix adhesion of both BxPC-3 and PANC1 cells (51). Xu et al (52) demonstrated that B-MSC-EXOs carrying miR-124 inhibit proliferation, invasion and metastasis, while promoting apoptosis in AsPC1 and PANC1 cells by modulating enhancer of zeste homolog 2 expression. Another study indicated that miR-484, carried by hB-MSC-EXOs, suppresses proliferation and migration and induces apoptosis in PC cells via regulation of the Wnt/MAPK axis (53). Furthermore, human amniotic MSC-EXOs inhibit the proliferation of PANC1 cells by downregulating the expression of Sugen kinase 269, E-cadherin, vimentin and the Snail transcription factor (54), a finding further supported by studies from Safari and Dadvar (55) and Safari et al (56). Collectively, these findings demonstrate that MSC-EXOs inhibit PC cell proliferation and promote apoptosis through multiple molecular mechanisms, serving a crucial role in suppressing PC progression.
MSC-EXOs can exert antitumor effects by modulating immune responses. For example, a study by Chen et al (9) demonstrated that human amniotic fluid MSC-EXOs (hAF-MSC-EXOs) markedly upregulate the expression of pro-apoptotic factors caspase-3, caspase-8 and Bax, thereby inhibiting the invasion and metastatic capabilities of PANC1 cells. This finding suggests that hAF-MSC-EXOs may enhance immune effector mechanisms by upregulating pro-apoptotic factors, consequently suppressing the progression of PC.
MSC-EXOs are regarded as a highly promising drug delivery vehicle for anti-PC therapy due to their excellent biocompatibility, low immunogenicity and inherent targeting capability. A study has shown that B-MSC-EXOs can serve as carriers for a triple-drug combination [galectin-9 small interfering (si)RNA/dodecyloxybenzyl gemcitabine/indocyanine green], demonstrating favorable tumor-targeting ability and pH-responsive release characteristics both in vivo and in vitro, notably enhancing the anti-PC efficacy (48). Furthermore, B-MSC-EXOs, as delivery vehicles for paclitaxel and gemcitabine, effectively overcome chemotherapy resistance in PC owing to their superior homing and penetration capabilities during delivery (49). Zhou et al (57) developed a dual-delivery biosystem using B-MSC-EXOs capable of co-loading oxaliplatin and siRNA. This system not only elicits an antitumor immune response by inhibiting tumor-associated macrophage polarization, promoting T lymphocyte recruitment and downregulating regulatory T cell activity, but also exhibits higher stability and lower side effects compared with conventional synthetic carriers. An in vitro study by Klimova et al (58) found that EXOs derived from human dental pulp MSCs can act as carriers for gemcitabine, significantly inhibiting the growth of PC cells. In summary, the multiple advantages demonstrated by MSC-EXOs in drug delivery indicate their potential as an efficient and safe nanoscale platform for the treatment of PC.
Over time, genetic engineering has evolved into a pivotal approach for treating various diseases, including hematological disorders, genetic conditions, and cancers (59–61). Particularly in the field of oncology, continuous technological advancements and expanding applications have notably enhanced the efficacy and safety of genetic engineering-based therapies, demonstrating broad prospects for clinical application. MSC-EXOs have garnered considerable attention in the treatment of PC due to their unique biocompatibility, low immunogenicity and targeted delivery capabilities. However, natural EXOs face limitations such as insufficient targeting specificity, limited efficacy of their cargo and rapid clearance, which substantially restrict their clinical translation and therapeutic effectiveness. Consequently, combining them with genetic engineering strategies can markedly enhance their tumor-targeting ability, treatment specificity and immunomodulatory functions in PC.
PC CAFs serve a critical role in the initiation and progression of PC. Targeted reprogramming of CAFs may represent a promising therapeutic strategy for PC. Zhou et al (62) employed a genetic engineering approach to endogenously modify B-MSC-EXOs, enabling surface display of integrin α5 targeting peptides and concomitant overexpression of miR-148a-3p, thereby achieving precise reprogramming of CAFs and providing new insights for clinical translation of this strategy. On the other hand, Buocikova et al (63) genetically engineered EXOs from placental MSCs to express the yeast cytosine deaminase::uracil phosphoribosyltransferase fusion enzyme. In a model of pancreatic ductal adenocarcinoma, this approach demonstrated potent cytotoxicity-reducing effects. Therefore, it offers a promising cell-free therapeutic strategy for PC. Although genetically engineered MSC-EXOs are considered a promising strategy for cancer treatment, their current application in PC remains in its early stages and requires further in-depth research to advance their development. The development of more efficient and safer engineered exosome-based therapeutic strategies remains a notable challenge in the current treatment of PC.
MSC-EXOs exhibit a dual role in PC, a phenomenon that warrants in-depth consideration. The underlying mechanisms are likely closely associated with the following factors. Firstly, the functional properties of EXOs are notably influenced by their cellular origin. Although EXOs derived from different sources of MSCs share common characteristics, such as high self-renewal capacity, multipotent differentiation potential, immunomodulatory activity, pro-angiogenic effects and tumor-homing ability (64–74) (Fig. 4), the specific molecules present on their surface and their cargo compositions may vary considerably, potentially leading to distinct functional orientations (75–79) (Table III). MSC-EXOs from different tissue sources have demonstrated a dual role in PC, which has been discussed in the present review. On the other hand, the stage of PC progression may also affect the functional outcomes of EXOs. For instance, early-stage PC may respond more favorably to therapy with MSC-EXOs compared with advanced-stage disease; however, this hypothesis still lacks robust experimental evidence and thus represents a critical question that urgently requires validation in future research. In addition, the differences in experimental design within this research field are another reason for the contradictions among different research results. Specifically, the differences in the extraction and identification methods of EXOs, cell co-culture systems and animal models (such as mouse strains, quantities and tumor-bearing sites) may all introduce notable heterogeneity, thereby affecting the comparability and repeatability of research conclusions.
In summary, MSC-EXOs exhibit a notable dual role in PC. The present review has systematically discussed their specific molecular mechanisms and functional characteristics in PC, and summarized the latest research advances in engineered modification of MSC-EXOs. In-depth elucidation of the key molecules, proteins and signaling pathways involved will provide new research directions and intervention strategies for the treatment of PC.
Despite the considerable tumor-suppressive potential of MSC-EXOs in PC, several unresolved issues and challenges remain. First, the techniques for isolating and extracting MSC-EXOs are still inadequate. Although multiple methods have been developed for exosome isolation and extraction, such as ultracentrifugation (80,81), ultrafiltration (82–84), flushing separation (85), precipitation (86–88), immunoaffinity capture (89–91), microfluidics (92–95) and mass spectrometry (96), each of these approaches has its own limitations. There is currently a lack of a simple, efficient, cost-effective and standardized extraction protocol suitable for clinical application. Second, research on and preparation of MSC-EXOs lack unified standards. No authoritative institutions have yet established clear guidelines for their isolation, preparation, characterization and functional evaluation. Third, large-scale production, storage and transportation systems remain underdeveloped. Systematic research and solutions are still needed regarding large-scale preparation, long-term stable storage methods and potential loss of activity and integrity during transportation. Fourth, evidence for clinical translation is insufficient. Most current studies are limited to animal experiments; future research should focus on clinical trials to validate the safety and efficacy of these EXOs in human applications. Fifth, the immunogenicity risks brought about by the engineering modification of MSC-EXOs need to be systematically evaluated. For example, whether the natural low immunogenicity advantage of MSC-EXOs will be altered after engineering. Or, whether it may trigger an immune response in the body, especially in the context of repeated administration for PC, which would be a core issue regarding treatment safety. Sixth, the dense fibrotic matrix of PC may severely impede the penetration ability of EXOs, and their pharmacokinetic performance in this environment requires further research and confirmation. Finally, there is a lack of horizontal efficacy comparisons among various treatment methods for PC. For instance, compared with traditional nanoparticles (such as liposomes) that have entered clinical application, MSC-EXOs may have inherent advantages in active targeting and biocompatibility, but they are slightly inferior in drug loading capacity and the maturity of production processes (97). However, future research should prospectively explore strategies for combining the two to achieve synergistic therapeutic effects.
Although the present article systematically expounds the research progress of MSC-EXOs in PC, there are still certain limitations. Firstly, due to the rapid development of research in this field, this article may not be able to cover all the latest released research achievements. Secondly, the discussion on the mechanism of MSC-EXOs in promoting cancer still needs to be further clarified. In addition, the analysis of the potential and challenges of EXOs derived from engineered MSCs in this field needs to be strengthened. Looking to the future, first of all, researchers should focus on developing a simple, efficient and low-cost standardized extraction scheme for the separation and extraction of EXOs. Secondly, more in-depth exploration and research should be performed on the role of MSC-EXOs in PC to address the potential loss issues that may arise during their large-scale production, storage and transportation. In addition, a systematic safety assessment of the immunogenicity risk and in vivo pharmacokinetic behavior of engineered modified EXOs is necessary, which is a prerequisite for their clinical transformation. Another important task lies in performing a horizontal efficacy comparison between exosome therapy and other treatment methods, with the aim of providing the optimal treatment strategy for patients with PC. Finally, accelerating the transformation of MSC-EXOs from basic research to clinical practice makes it possible for them to become an important component of the comprehensive treatment system for PC.
MSC-EXOs serve as highly plastic messengers in PC, where their role in promoting or suppressing tumor progression is not fixed but rather determined by their specific surface molecules and cargo contents, which are closely associated with the diverse origins of MSCs. Future research should focus on transforming MSC-EXOs from a ‘double-edged sword’ into a precise weapon against PC. The primary task is to deeply explore its specific active ingredients and their precise regulatory mechanisms in PC and, on this basis, strategies for engineering and modifying MSC-EXOs should be established. At the same time, establishing standardized, repeatable, low-cost yet high-purity separation and purification technologies is the core step for it to move towards clinical application. In addition, it is important to actively explore the combined application of EXOs therapy and existing treatment methods. Only through in-depth exploration at multiple levels can this promising treatment method be advanced from the laboratory to clinical practice, bringing new treatment options for patients with PC.
Not applicable.
Funding: No funding was received.
Not applicable.
HZ contributed to the conception and overall design of the study. HZ drafted the manuscript and prepared the figures and tables. XS reviewed and revised the manuscript. All authors read and approved the final version of the manuscript. Data authentication was not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Klein AP: Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 18:493–502. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024.PubMed/NCBI | |
|
Park W, Chawla A and O'Reilly EM: Pancreatic cancer: A review. JAMA. 326:851–862. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH and Neoptolemos JP: Pancreatic cancer. Nat Rev Dis Primers. 2:160222016. View Article : Google Scholar : PubMed/NCBI | |
|
Halbrook CJ, Lyssiotis CA, Pasca di Magliano M and Maitra A: Pancreatic cancer: Advances and challenges. Cell. 186:1729–1754. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Klatte DCF, Wallace MB, Löhr M, Bruno MJ and van Leerdam ME: Hereditary pancreatic cancer. Best Pract Res Clin Gastroenterol. 58–59. 1017832022.PubMed/NCBI | |
|
Wood LD, Canto MI, Jaffee EM and Simeone DM: Pancreatic cancer: Pathogenesis, screening, diagnosis, and treatment. Gastroenterology. 163:386–402.e1. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Tsai CC, Su PF, Huang YF, Yew TL and Hung SC: Oct4 and Nanog directly regulate Dnmt1 to maintain self-renewal and undifferentiated state in mesenchymal stem cells. Mol Cell. 47:169–182. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Chen YC, Lan YW, Huang SM, Yen CC, Chen W, Wu WJ, Staniczek T, Chong KY and Chen CM: Human amniotic fluid mesenchymal stem cells attenuate pancreatic cancer cell proliferation and tumor growth in an orthotopic xenograft mouse model. Stem Cell Res Ther. 13:2352022. View Article : Google Scholar : PubMed/NCBI | |
|
Kabashima-Niibe A, Higuchi H, Takaishi H, Masugi Y, Matsuzaki Y, Mabuchi Y, Funakoshi S, Adachi M, Hamamoto Y, Kawachi S, et al: Mesenchymal stem cells regulate epithelial-mesenchymal transition and tumor progression of pancreatic cancer cells. Cancer Sci. 104:157–164. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Beckermann BM, Kallifatidis G, Groth A, Frommhold D, Apel A, Mattern J, Salnikov AV, Moldenhauer G, Wagner W, Diehlmann A, et al: VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br J Cancer. 99:622–631. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Saito K, Sakaguchi M, Maruyama S, Iioka H, Putranto EW, Sumardika IW, Tomonobu N, Kawasaki T, Homma K and Kondo E: Stromal mesenchymal stem cells facilitate pancreatic cancer progression by regulating specific secretory molecules through mutual cellular interaction. J Cancer. 9:2916–2929. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Miyazaki Y, Oda T, Inagaki Y, Kushige H, Saito Y, Mori N, Takayama Y, Kumagai Y, Mitsuyama T and Kida YS: Adipose-derived mesenchymal stem cells differentiate into heterogeneous cancer-associated fibroblasts in a stroma-rich xenograft model. Sci Rep. 11:46902021. View Article : Google Scholar : PubMed/NCBI | |
|
Mohr A, Albarenque SM, Deedigan L, Yu R, Reidy M, Fulda S and Zwacka RM: Targeting of XIAP combined with systemic mesenchymal stem cell-mediated delivery of sTRAIL ligand inhibits metastatic growth of pancreatic carcinoma cells. Stem Cells. 28:2109–2120. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao C, Pu Y, Zhang H, Hu X, Zhang R, He S, Zhao Q and Mu B: IL10-modified human mesenchymal stem cells inhibit pancreatic cancer growth through angiogenesis inhibition. J Cancer. 11:5345–5352. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Keshtkar S, Azarpira N and Ghahremani MH: Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Res Ther. 9:632018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou J, Tan X, Tan Y, Li Q, Ma J and Wang G: Mesenchymal stem cell derived exosomes in cancer progression, metastasis and drug delivery: A comprehensive review. J Cancer. 9:3129–3137. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, et al: Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 17:816–826. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Han S, Gonzalo DH, Feely M, Rinaldi C, Belsare S, Zhai H, Kalra K, Gerber MH, Forsmark CE and Hughes SJ: Stroma-derived extracellular vesicles deliver tumor-suppressive miRNAs to pancreatic cancer cells. Oncotarget. 9:5764–5777. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, Mark MT, Molina H, Martin AB, Bojmar L, et al: Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 20:332–343. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zagrean AM, Hermann DM, Opris I, Zagrean L and Popa-Wagner A: Multicellular crosstalk between exosomes and the neurovascular unit after cerebral ischemia. therapeutic implications. Front Neurosci. 12:8112018. View Article : Google Scholar : PubMed/NCBI | |
|
Lee JR, Park BW, Kim J, Choo YW, Kim HY, Yoon JK, Kim H, Hwang JW, Kang M, Kwon SP, et al: Nanovesicles derived from iron oxide nanoparticles-incorporated mesenchymal stem cells for cardiac repair. Sci Adv. 6:eaaz09522020. View Article : Google Scholar : PubMed/NCBI | |
|
Ding Y, Mei W, Zheng Z, Cao F, Liang K, Jia Y, Wang Y, Liu D, Li J and Li F: Exosomes secreted from human umbilical cord mesenchymal stem cells promote pancreatic ductal adenocarcinoma growth by transferring miR-100-5p. Tissue Cell. 73:1016232021. View Article : Google Scholar : PubMed/NCBI | |
|
Xie X, Ji J, Chen X, Xu W, Chen H, Zhu S, Wu J, Wu Y, Sun Y, Sai W, et al: Human umbilical cord mesenchymal stem cell-derived exosomes carrying hsa-miRNA-128-3p suppress pancreatic ductal cell carcinoma by inhibiting Galectin-3. Clin Transl Oncol. 24:517–531. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj and Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 8:315–317. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Lv FJ, Tuan RS, Cheung KM and Leung VY: Concise review: The surface markers and identity of human mesenchymal stem cells. Stem Cells. 32:1408–1419. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Teixeira FG, Panchalingam KM, Anjo SI, Manadas B, Pereira R, Sousa N, Salgado AJ and Behie LA: Do hypoxia/normoxia culturing conditions change the neuroregulatory profile of Wharton Jelly mesenchymal stem cell secretome? Stem Cell Res Ther. 6:1332015. View Article : Google Scholar : PubMed/NCBI | |
|
Kisiel AH, McDuffee LA, Masaoud E, Bailey TR, Esparza Gonzalez BP and Nino-Fong R: Isolation, characterization, and in vitro proliferation of canine mesenchymal stem cells derived from bone marrow, adipose tissue, muscle, and periosteum. Am J Vet Res. 73:1305–1317. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy MB, Moncivais K and Caplan AI: Mesenchymal stem cells: Environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 45:e542013. View Article : Google Scholar : PubMed/NCBI | |
|
Trounson A and McDonald C: Stem cell therapies in clinical trials: Progress and challenges. Cell Stem Cell. 17:11–22. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
He Z, Wang J, Zhu C, Xu J, Chen P, Jiang X, Chen Y, Jiang J and Sun C: Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated pancreatic cancer cell proliferation and metastasis. Cancer Lett. 548:2157512022. View Article : Google Scholar : PubMed/NCBI | |
|
Cai J, Wu J, Wang J, Li Y, Hu X, Luo S and Xiang D: Extracellular vesicles derived from different sources of mesenchymal stem cells: Therapeutic effects and translational potential. Cell Biosci. 10:692020. View Article : Google Scholar : PubMed/NCBI | |
|
Singh S, Dansby C, Agarwal D, Bhat PD, Dubey PK and Krishnamurthy P: Exosomes: Methods for isolation and characterization in biological samples. Methods Mol Biol. 2835:181–213. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Henne WM, Buchkovich NJ and Emr SD: The ESCRT pathway. Dev Cell. 21:77–91. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, Sibiak R, Piotrowska-Kempisty H, Iżycki D, Bukowska D, et al: Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J Clin Med. 9:4362020. View Article : Google Scholar : PubMed/NCBI | |
|
Gurung S, Perocheau D, Touramanidou L and Baruteau J: The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun Signal. 19:472021. View Article : Google Scholar : PubMed/NCBI | |
|
Joo HS, Suh JH, Lee HJ, Bang ES and Lee JM: Current knowledge and future perspectives on mesenchymal stem cell-derived exosomes as a new therapeutic agent. Int J Mol Sci. 21:7272020. View Article : Google Scholar : PubMed/NCBI | |
|
Kowal J, Tkach M and Théry C: Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 29:116–125. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wortzel I, Dror S, Kenific CM and Lyden D: Exosome-mediated metastasis: Communication from a distance. Dev Cell. 49:347–360. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Doyle LM and Wang MZ: Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 8:7272019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Hong Y, Cho E, Kim GB and Kim IS: Extracellular vesicles as a platform for membrane-associated therapeutic protein delivery. J Extracell Vesicles. 7:14401312018. View Article : Google Scholar : PubMed/NCBI | |
|
Kugeratski FG, Hodge K, Lilla S, McAndrews KM, Zhou X, Hwang RF, Zanivan S and Kalluri R: Quantitative proteomics identifies the core proteome of exosomes with syntenin-1 as the highest abundant protein and a putative universal biomarker. Nat Cell Biol. 23:631–641. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hánělová K, Raudenská M, Masařík M and Balvan J: Protein cargo in extracellular vesicles as the key mediator in the progression of cancer. Cell Commun Signal. 22:252024. View Article : Google Scholar : PubMed/NCBI | |
|
Choi DS, Kim DK, Kim YK and Gho YS: Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 13:1554–1571. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Gao G, Wang L and Li C: Circ_0006790 carried by bone marrow mesenchymal stem cell-derived exosomes regulates S100A11 DNA methylation through binding to CBX7 in pancreatic ductal adenocarcinoma. Am J Cancer Res. 12:1934–1959. 2022.PubMed/NCBI | |
|
Wu R, Su Z, Zhao L, Pei R, Ding Y, Li D, Zhu S, Xu L, Zhao W and Zhou W: Extracellular vesicle-loaded oncogenic lncRNA NEAT1 from adipose-derived mesenchymal stem cells confers gemcitabine resistance in pancreatic cancer via miR-491-5p/Snail/SOCS3 axis. Stem Cells Int. 2023:65105712023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang R, Zhang Y, Hao F, Su Z, Duan X and Song X: Exosome-mediated triple drug delivery enhances apoptosis in pancreatic cancer cells. Apoptosis. 30:1893–1911. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Zhou W, Chen X, Wang Q, Li C, Chen Q, Zhang Y, Lu Y, Ding X and Jiang C: Bone marrow mesenchymal stem cells-derived exosomes for penetrating and targeted chemotherapy of pancreatic cancer. Acta Pharm Sin B. 10:1563–1575. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yao X, Mao Y, Wu D, Zhu Y, Lu J, Huang Y, Guo Y, Wang Z, Zhu S, Li X and Lu Y: Exosomal circ_0030167 derived from BM-MSCs inhibits the invasion, migration, proliferation and stemness of pancreatic cancer cells by sponging miR-338-5p and targeting the Wif1/Wnt8/β-catenin axis. Cancer Lett. 512:38–50. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shang S, Wang J, Chen S, Tian R, Zeng H, Wang L, Xia M, Zhu H and Zuo C: Exosomal miRNA-1231 derived from bone marrow mesenchymal stem cells inhibits the activity of pancreatic cancer. Cancer Med. 8:7728–7740. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Liu N, Wei Y, Zhou D, Lin R, Wang X and Shi B: 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 (Albany NY). 12:19660–19676. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin T, Pu X, Zhou S, Huang Z, Chen Q, Zhang Y, Mao Q, Liang Y and Ding G: Identification of exosomal miR-484 role in reprogramming mitochondrial metabolism in pancreatic cancer through Wnt/MAPK axis control. Pharmacol Res. 197:1069802023. View Article : Google Scholar : PubMed/NCBI | |
|
Alidoust Saharkhiz Lahiji M and Safari F: Potential therapeutic effects of hAMSCs secretome on Panc1 pancreatic cancer cells through downregulation of SgK269, E-cadherin, vimentin, and snail expression. Biologicals. 76:24–30. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Safari F and Dadvar F: In vitro evaluation of autophagy and cell death induction in Panc1 pancreatic cancer by secretome of hAMSCs through downregulation of p-AKT/p-mTOR and upregulation of p-AMPK/ULK1 signal transduction pathways. Tissue Cell. 84:1021602023. View Article : Google Scholar : PubMed/NCBI | |
|
Safari F, Shafiee Nejad N and Aghaei Nejad A: The inhibition of Panc1 cancer cells invasion by hAMSCs secretome through suppression of tyrosine phosphorylation of SGK223 (at Y411 site), c-Src (at Y416, Y530 sites), AKT activity, and JAK1/Stat3 signaling. Med Oncol. 39:282022. View Article : Google Scholar : PubMed/NCBI | |
|
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. 268:1205462021. View Article : Google Scholar : PubMed/NCBI | |
|
Klimova D, Jakubechova J, Altanerova U, Nicodemou A, Styk J, Szemes T, Repiska V and Altaner C: Extracellular vesicles derived from dental mesenchymal stem/stromal cells with gemcitabine as a cargo have an inhibitory effect on the growth of pancreatic carcinoma cell lines in vitro. Mol Cell Probes. 67:1018942023. View Article : Google Scholar : PubMed/NCBI | |
|
Hu X, Zhou W, Pi R, Zhao X and Wang W: Genetically modified cancer vaccines: Current status and future prospects. Med Res Rev. 42:1492–1517. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K and Sadelain M: Gene therapy comes of age. Science. 359:eaan46722018. View Article : Google Scholar : PubMed/NCBI | |
|
Doering CB, Archer D and Spencer HT: Delivery of nucleic acid therapeutics by genetically engineered hematopoietic stem cells. Adv Drug Deliv Rev. 62:1204–1212. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou P, Ding X, Du X, Wang L and Zhang Y: Targeting reprogrammed cancer-associated fibroblasts with engineered mesenchymal stem cell extracellular vesicles for pancreatic cancer treatment. Biomater Res. 28:00502024. View Article : Google Scholar : PubMed/NCBI | |
|
Buocikova V, Altanerova U, Soltysova A, Andrezal M, Vanova D, Jakubechova J, Cihova M, Burikova M, Urbanova M, Juhasikova L, et al: Placental mesenchymal stem cells: A promising platform for advancing gene therapy in pancreatic ductal adenocarcinoma. Biomed Pharmacother. 190:1184282025. View Article : Google Scholar : PubMed/NCBI | |
|
Amati E, Perbellini O, Rotta G, Bernardi M, Chieregato K, Sella S, Rodeghiero F, Ruggeri M and Astori G: High-throughput immunophenotypic characterization of bone marrow- and cord blood-derived mesenchymal stromal cells reveals common and differentially expressed markers: Identification of angiotensin-converting enzyme (CD143) as a marker differentially expressed between adult and perinatal tissue sources. Stem Cell Res Ther. 9:102018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Liu Y, Yin W and Hu X: Adipose-derived stromal cells in regulation of hematopoiesis. Cell Mol Biol Lett. 25:162020. View Article : Google Scholar : PubMed/NCBI | |
|
Xu T, Yu X, Yang Q, Liu X, Fang J and Dai X: Autologous micro-fragmented adipose tissue as stem cell-based natural scaffold for cartilage defect repair. Cell Transplant. 28:1709–1720. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Schäffler A and Büchler C: Concise review: Adipose tissue-derived stromal cells-basic and clinical implications for novel cell-based therapies. Stem Cells. 25:818–827. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Kozlowska U, Krawczenko A, Futoma K, Jurek T, Rorat M, Patrzalek D and Klimczak A: Similarities and differences between mesenchymal stem/progenitor cells derived from various human tissues. World J Stem Cells. 11:347–374. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC and Deng WP: Revisiting the advances in isolation, characterization and secretome of adipose-derived stromal/stem cells. Int J Mol Sci. 19:22002018. View Article : Google Scholar : PubMed/NCBI | |
|
Fukuchi Y, Nakajima H, Sugiyama D, Hirose I, Kitamura T and Tsuji K: Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells. 22:649–658. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Oliveira MS and Barreto-Filho JB: Placental-derived stem cells: Culture, differentiation and challenges. World J Stem Cells. 7:769–775. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ulrich C, Abruzzese T, Maerz JK, Ruh M, Amend B, Benz K, Rolauffs B, Abele H, Hart ML and Aicher WK: Human placenta-derived CD146-positive mesenchymal stromal cells display a distinct osteogenic differentiation potential. Stem Cells Dev. 24:1558–1569. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Huang KJ, Wu JC, Hu MS, Sanyal M, Hu M, Longaker MT and Lorenz HP: Peripheral blood-derived mesenchymal stem cells: Candidate cells responsible for healing critical-sized calvarial bone defects. Stem Cells Transl Med. 4:359–368. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Cao C and Dong Y and Dong Y: Study on culture and in vitro osteogenesis of blood-derived human mesenchymal stem cells. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 19:642–647. 2005.(In Chinese). PubMed/NCBI | |
|
Tracy SA, Ahmed A, Tigges JC, Ericsson M, Pal AK, Zurakowski D and Fauza DO: A comparison of clinically relevant sources of mesenchymal stem cell-derived exosomes: Bone marrow and amniotic fluid. J Pediatr Surg. 54:86–90. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, He Z, Liang S, Yang Q, Cheng P and Chen A: Comprehensive proteomic analysis of exosomes derived from human bone marrow, adipose tissue, and umbilical cord mesenchymal stem cells. Stem Cell Res Ther. 11:5112020. View Article : Google Scholar : PubMed/NCBI | |
|
Ji L, Bao L, Gu Z, Zhou Q, Liang Y, Zheng Y, Xu Y, Zhang X and Feng X: Comparison of immunomodulatory properties of exosomes derived from bone marrow mesenchymal stem cells and dental pulp stem cells. Immunol Res. 67:432–442. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Katsuda T, Tsuchiya R, Kosaka N, Yoshioka Y, Takagaki K, Oki K, Takeshita F, Sakai Y, Kuroda M and Ochiya T: Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep. 3:11972013. View Article : Google Scholar : PubMed/NCBI | |
|
Pomatto M, Gai C, Negro F, Cedrino M, Grange C, Ceccotti E, Togliatto G, Collino F, Tapparo M, Figliolini F, et al: Differential therapeutic effect of extracellular vesicles derived by bone marrow and adipose mesenchymal stem cells on wound healing of diabetic ulcers and correlation to their cargoes. Int J Mol Sci. 22:38512021. View Article : Google Scholar : PubMed/NCBI | |
|
Langevin SM, Kuhnell D, Orr-Asman MA, Biesiada J, Zhang X, Medvedovic M and Thomas HE: Balancing yield, purity and practicality: A modified differential ultracentrifugation protocol for efficient isolation of small extracellular vesicles from human serum. RNA Biol. 16:5–12. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gardiner C, Di Vizio D, Sahoo S, Théry C, Witwer KW, Wauben M and Hill AF: Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J Extracell Vesicles. 5:329452016. View Article : Google Scholar : PubMed/NCBI | |
|
Van Deun J, Mestdagh P, Sormunen R, Cocquyt V, Vermaelen K, Vandesompele J, Bracke M, De Wever O and Hendrix A: The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J Extracell Vesicles. 3:2014. View Article : Google Scholar : PubMed/NCBI | |
|
Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A and Möller A: Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 4:270312015. View Article : Google Scholar : PubMed/NCBI | |
|
Bhattacharjee C and Singh M: Studies on the applicability of artificial neural network (ANN) in continuous stirred ultrafiltration. Chem Eng Technol. 25:1187–1192. 2002. View Article : Google Scholar | |
|
Cheng H, Fang H, Xu RD, Fu MQ, Chen L, Song XY, Qian JY, Zou YZ, Ma JY and Ge JB: Development of a rinsing separation method for exosome isolation and comparison to conventional methods. Eur Rev Med Pharmacol Sci. 23:5074–5083. 2019.PubMed/NCBI | |
|
Soares Martins T, Catita J, Martins Rosa I, AB da Cruz E, Silva O and Henriques AG: Exosome isolation from distinct biofluids using precipitation and column-based approaches. PLoS One. 13:e01988202018. View Article : Google Scholar : PubMed/NCBI | |
|
Buschmann D, Kirchner B, Hermann S, Märte M, Wurmser C, Brandes F, Kotschote S, Bonin M, Steinlein OK, Pfaffl MW, et al: Evaluation of serum extracellular vesicle isolation methods for profiling miRNAs by next-generation sequencing. J Extracell Vesicles. 7:14813212018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Yu J, Wang J, Li H, Che J and Cao B: Isolation and identification of miRNAs in exosomes derived from serum of colon cancer patients. J Cancer. 8:1145–1152. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Popovic M, Mazzega E, Toffoletto B and De Marco A: Isolation of anti-extra-cellular vesicle single-domain antibodies by direct panning on vesicle-enriched fractions. Microb Cell Fact. 17:62018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, Hu BY, Qin W, Zou TT, Fu Y, et al: Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 13:1522020. View Article : Google Scholar : PubMed/NCBI | |
|
Pei S, Sun W, Han Q, Wang H and Liang Q: Bifunctional immunoaffinity magnetic nanoparticles for high-efficiency separation of exosomes based on host-guest interaction. Talanta. 272:1257902024. View Article : Google Scholar : PubMed/NCBI | |
|
Iliescu FS, Vrtačnik D, Neuzil P and Iliescu C: Microfluidic technology for clinical applications of exosomes. Micromachines (Basel). 10:3922019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang P, Zhou X, He M, Shang Y, Tetlow AL, Godwin AK and Zeng Y: Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat Biomed Eng. 3:438–451. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hua X, Zhu Q, Liu Y, Zhou S, Huang P, Li Q and Liu S: A double tangential flow filtration-based microfluidic device for highly efficient separation and enrichment of exosomes. Anal Chim Acta. 1258:3411602023. View Article : Google Scholar : PubMed/NCBI | |
|
Ding L, Liu X, Zhang Z, Liu LE, He S, Wu Y, Effah CY, Yang R, Zhang A, Chen W, et al: Magnetic-nanowaxberry-based microfluidic ExoSIC for affinity and continuous separation of circulating exosomes towards cancer diagnosis. Lab Chip. 23:1694–1702. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Abramowicz A, Marczak L, Wojakowska A, Zapotoczny S, Whiteside TL, Widlak P and Pietrowska M: Harmonization of exosome isolation from culture supernatants for optimized proteomics analysis. PLoS One. 13:e02054962018. View Article : Google Scholar : PubMed/NCBI | |
|
Iqbal Z, Rehman K, Mahmood A, Shabbir M, Liang Y, Duan L and Zeng H: Exosome for mRNA delivery: Strategies and therapeutic applications. J Nanobiotechnology. 22:3952024. View Article : Google Scholar : PubMed/NCBI |