Background

CCA remains a highly lethal malignancy of the biliary system. It can be classified into three major groups based on their lesion locations: intrahepatic, perihilar, and distal CCA. Globally, the proportion of CCA is second only to hepatocellular carcinoma (HCC) among all primary liver tumors, accounting for roughly 15%, and accounts for 3% of all gastrointestinal malignant tumors [1, 2]. Despite advancements advances in CCA cognition, diagnosis, and treatment for the past few years, due to its high malignant degree, strong invasiveness, and the occult in initial, most patients are first diagnosed already in the late-stage that severely constrains therapeutic options. Although curative surgery is a preferred selection for some early-stage patients, the fact of tumor recurrence or metastasis after resection remains frustrating. Meanwhile, on account of the high heterogeneity of CCAs, the systemic therapeutics bring little effect for advanced patients who are not possible for radical surgery, leading to a very poor prognosis that only 7%-20% of patients can reach the five-year survival [3, 4]. Nevertheless, with the advancements in genetic profiling of CCAs, emerging treatments such as targeted or immunological therapeutics may be able to assist patients with this deadly cancer to get better outcomes. Considering the current situation of CCAs, exploring new diagnostic and therapeutic strategies remains a matter of priority.

EVs are enveloped by a lipid bilayer that can be discharged by numerous cell sorts. According to the size and formation pathway of vesicles, it can be classified into two major subsets approximately, called ectosomes and exosomes [5]. The former are vesicles with a diameter of 50 nm ~ 1 μm via plasma membrane budding outward directly, while exosomes are EVs ranging from 40 ~ 150 nm in diameter generated in the opposite way, which involves plasma membrane invagination and endosomal formation [5]. Exosomes contain multiple substances and are broadly distributed in different body fluids like plasma, urine, bile, and cerebrospinal fluid (CSF), which play important roles in a variety of normal or abnormal biological behaviors [6,7,8]. Recently, researches about cancer exosomes have received tremendous attention. Intercellular communication in the microenvironment plays a significant role in regulating tumor development, where exosomes are key messengers that mediate this cell-to-cell communication [5, 9]. Previous researches have illustrated that exosomes participate in tumorigenesis or metastasis in multiple ways, their potential usages in cancer diagnosis and prognosis have also been deeply explored [9]. Although certain studies have reviewed the roles of EVs in the progression of CCA [10], a more comprehensive summarization of exosomes in CCA remains insufficient up to now.

In this article, we systematically summarized the research status of exosomes in the tumor fields. Based on the existing researches of exosomes in CCA, we specifically emphasized their significant roles in regulating tumor development and potential values in diagnosis and treatment.

Research status of exosomes

Biogenesis, secretion and internalization

As a type of EVs, the synthesis progress of exosomes involves three major phases: 1) plasma membrane invagination and early endosomes formation. 2) intraluminal vesicles (ILVs) and intracellular multivesicular bodies (MVBs) generation. 3) the fusion of MVBs and plasma membrane leads to exosomes secretion [9]. Generally, the biogenesis of MVBs mainly depends on the following two pathways: endosomal sorting complexes required for transport (ESCRT)-dependent or ESCRT-independent mechanisms, and the former is the most classic pathway [6]. Once mature, MVBs can integrate with autophagosomes then degrade through the lysosomal pathway or secrete into extracellular space as exosomes by fusing with the plasma membrane [11]. In this biogenesis and secretion process, other components such as tumor susceptibility gene 101 (TSG101), Rab family of GTPases (like Rab27A and Rab27B), soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, apoptosis-linked gene 2-interacting protein X (Alix), ceramide, tetraspanins (CD63, CD9, CD81), and phospholipids are also getting involved [5, 12, 13].

Considering the difference in the origin and microenvironment, exosomes have a strong heterogeneity, which is mainly reflected in the regulation of the target cell functions [5]. Once exosomes are secreted by the host cells, they can be absorbed by target cells through various approaches like endocytosis, plasma membrane integration, and specific protein interactions [14]. Among these internalization ways, endocytosis is the most widely studied pattern. According to the characteristics of components involved in endocytosis, several subtypes are broadly divided up, including phagocytosis, macro-pinocytosis, clathrin-mediated endocytosis (CME), and caveolin-dependent endocytosis (CDE), as well as lipid raft-mediated internalization [15]. Moreover, several proteins such as tetraspanins, integrins, proteoglycans, and lectins, also participate in the internalization of exosomes by the unique ligand-receptor interactions[9]. However, on account of the heterogeneity of exosomes, whether or not exosomes uptake is specific remains controversial(15). Therefore, it is essential to further investigate the detailed routes of exosomes uptake (Fig. 1).

Fig. 1
figure 1

Biogenesis, secretion, and internalization of exosomes. The formation of exosomes initially depends on the invagination of the plasma membrane, followed by the generation of ILVs and MVBs. Once mature, MVBs can fuse with lysosomes and be degraded, or integrate with the plasma membrane and finally get released, i.e., exosomes. During this process of synthesis and secretion, ESCRT-dependent and ESCRT-independent mechanisms are two common approaches, other components like the Rab family of GTPases, SNARE, ceramide, and tetraspanins are also involved. Exosomes can be uptake by receptor cells to perform specific functions through various mechanisms, such as phagocytosis, macro-pinocytosis, ligand-receptor interaction, CME, and CDE. As plasma membrane-derived vesicles with lipid bilayer structure, exosomes carry a variety of components, including RNAs (mRNA, MiRNA, LncRNA, and CircRNA), proteins (TSG101, Alix, HSP, CD9, CD63, and CD81) and metabolites, etc.

Isolation and identification

Currently, frequently-used isolation strategies include centrifugation (differential or density gradient centrifugation), particle size separation, size-exclusion chromatography, microfluidic technique, and immunoaffinity capture [16]. Until now, the most common method is still differential centrifugation due to its high exosome yields and relatively cheap price. However, it also has some deficiencies like complicated procedures, low separation efficiency, and susceptibility to contamination by soluble substances in cell culture medium or other body fluids [17]. Other isolation methods like size-exclusion chromatography, with a relatively high yield but difficult to achieve mass production, and immunoaffinity capture, advanced in specific separation yet costly with low yields [16, 17]. So far there is not a standardized method that can achieve both economic and high purity at the same time. Therefore, the exploration of better purification methods remains a major challenge in the exosome-related fields.

In terms of identification, the International Society of Extracellular Vesicles proposed to identify exosomes mainly from the following three aspects: 1) Exosomal morphology identification, 2) Exosomal size detection, 3) Exosomal biomarkers identification [18, 19]. Among them, transmission electron microscopy (TEM), cryo-electron microscopy (Cryo-EM), and atomic force microscopy (AFM) are the most direct methods for visual observation of exosomes [20]. Real-time nanoparticle tracking technology based on the principle of Brownian motion can be used to obtain the size distribution of exosomes [21]. In addition, enzyme-linked immunosorbent assay (ELISA), flow cytometry (FCM), and western blotting (WB) are available means to detect the specific proteins or other markers expressed on exosomes [20, 22]. Reportedly, several transmembrane proteins like CD9, CD63, and CD81 are considered to be representative hallmarks, however, a recent study suggested that compared with other tetraspanins, CD63 is the unique biomarker, while CD9 and CD81 are not specific for exosomes [23]. Moreover, other components related to the formation of exosomes such as Alix, TSG101, and Heat shock proteins (HSP) can also serve as classical hallmarks [5].

The roles of exosomes in malignancies

Since exosomes have played an essential role in multiple pathological changes through mediating intercellular communication, it has also received enormous concerns in the cancer area over the past few years [9]. Related studies have pointed out that cancer-cell-derived exosomes can modulate tumor progression through a variety of mechanisms [9, 24]. Besides, as mentioned above, exosomes contain complex cargoes that are widespread in various body fluids, which also partly represent the heterogeneity of their parental cells, making them available for cancer diagnosis and prognostic by serving as novel biomarkers [25]. Moreover, recent studies have focused more on the tumor microenvironment (TME), where the signal interaction mediated by exosomes also makes a difference in tumor development [5, 26].

Exosomes induce or accelerate tumorigenesis. Exosomes secreted by HCC cancer cells promoted tumorigenesis through the Hedgehog pathway [27]. Mirna-224-5p-enriched exosomes secreted by non-small cell lung cancer (NSCLC) cells accelerated neoplasia by directly binding with the androgen receptor (AR) [28]. On the contrary, exosomes distributed in the plasma of patients with medulloblastoma inhibited tumorigenesis by targeting FOXP4 (forkhead box protein 4) and EZH2 (enhancer of zeste 2 polycomb repressive complex 2 subunit) directly through their miRNA cargoes [

Availability of data and materials

Not applicable.

Abbreviations

CCA:

Cholangiocarcinoma

EVs:

Extracellular vesicles

HCC:

Hepatocellular carcinoma

CSF:

Cerebrospinal fluid

ILVs:

Intraluminal vesicles

MVBs:

Multivesicular bodies

ESCRT:

Endosomal sorting complexes required for transport

TSG101:

Tumor susceptibility gene 101

SNARE:

Soluble N-ethylmaleimide-sensitive factor attachment protein receptor

Alix:

Apoptosis-linked gene 2-interacting protein X

CME:

Clathrin-mediated endocytosis

CDE:

Caveolin-dependent endocytosis

TEM:

Transmission electron microscopy

Cryo-EM:

Cryo-electron microscopy

AFM:

Atomic force microscopy

ELISA:

Enzyme-linked immunosorbent assay

FCM:

Flow cytometry

WB:

Western blotting

HSP:

Heat shock protein

MiRNA:

MicroRNA

LncRNA:

Long noncoding RNA

CircRNA:

Circular RNA

NSCLC:

Non-small cell lung cancer

AR:

Androgen receptor

FOXP4:

Forkhead box protein 4

EZH2:

Enhancer of zeste 2 polycomb repressive complex 2 subunit

PTEN:

Phosphatase and tensin homolog

CRC:

Colorectal cancer

KLF2:

Kruppel like factor 2

KLF4:

Kruppel like factor 4

sE-cad:

Soluble E-cadherin

TME:

Tumor microenvironment

CAFs:

Cancer-associated fibroblasts

EOC:

Epithelial ovarian cancer

EMT:

Epithelial-mesenchymal transition

GSK-3β:

Glycogen synthase kinase 3 beta

OSCC:

Oral squamous cell carcinoma

MMP9:

Matrix metalloproteinase 9

CSF-1:

Colony stimulating factor 1

MCP-1:

Monocyte chemoattractant protein-1

TGFβ:

Transforming growth factor beta

UHRF1:

Ubiquitin-like with PHD and ring finger domain 1

NKs:

Natural killer cells

TIM-3:

T cell immunoglobulin domain and mucin domain 3

DCs:

Dendritic cells

AFP:

α-Fetoprotein

PD-L1:

Programmed death 1 ligand

PD-1:

Programmed death 1

SNHG7:

Small nucleolar RNA host gene 7

LUAD:

Lung adenocarcinoma

TAM:

Tumor-associated macrophage

ATG5:

Autophagy related 5

GPC1:

Glypican-1

KEGG:

Kyoto encyclopedia of genes and genomes

BMSCs:

Bone marrow mesenchymal stem cells

α-SMA:

Alpha-smooth muscle actin

FAP:

Fibroblast activation protein alpha

IL-6:

Interleukin- 6

STAT3:

Signal transducer and activator of transcription 3

LY6E:

Lymphocyte antigen 6 family member E

CCAC1:

Cholangiocarcinoma-associated circular RNA 1

YY1:

Yin Yang 1

CAMLG:

Calcium modulating ligand

FZD 10:

Frizzled class receptor 10

CIKs:

Cytokine-induced killer cells

TNF-α:

Tumor necrosis factor-α

PSC:

Primary sclerosing cholangitis

UC:

Ulcerative colitis

CMIP:

C-Maf inducing protein

GAD1:

Glutamate decarboxylase 1

NDKP1:

Nucleoside diphosphate kinase 1

CDS1:

CDP-diacylglycerol synthase 1

CKS1B:

Cyclin-dependent kinase regulatory subunit 1

UBE2C:

Ubiquitin-conjugating enzyme E2C

SERPINB1:

Serine protease inhibitor B1

MALAT1:

Metastasis associated lung adenocarcinoma transcript 1

Cripto-1:

Teratocarcinoma-derived growth factor 1 (TDGF-1)

PHCCA:

Perihilar cholangiocarcinoma

AMPN:

Aminopeptidase N

VNN1:

Pantetheinase

PIGR:

Polymeric immunoglobulin receptor

FIBG:

Fibrinogen gamma chain

A1AG1:

Alpha1-acid glycoprotein 1

S100A8:

S100 calcium binding protein A8

iCCA:

Intrahepatic cholangiocarcinoma

pCCA:

Perihilar cholangiocarcinoma

dCCA:

Distal cholangiocarcinoma

IDH1/2:

Isocitrate dehydrogenase1/2

KRAS:

Kirsten ratsarcoma viral oncogene homolog

BAP1:

BRCA1 associated protein 1

TP53:

Tumor protein p53

FGFR:

Fibroblast growth factor receptor

PRKACA:

Protein kinase cAMP-activated catalytic subunit alpha

PRKACB:

Protein kinase cAMP-activated catalytic subunit beta

ELF3:

E74 like ETS transcription factor 3

AMSCs:

Adipose tissue-derived mesenchymal stem cells

mTOR:

Mechanistic target of rapamycin kinase

References

  1. Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, et al. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol. 2016;13(5):261–80.

    Article  PubMed  Google Scholar 

  2. Banales JM, Marin JJG, Lamarca A, Rodrigues PM, Khan SA, Roberts LR, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol. 2020;17(9):557–88.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Cambridge WA, Fairfield C, Powell JJ, Harrison EM, Soreide K, Wigmore SJ, et al. Meta-analysis and meta-regression of survival after liver transplantation for unresectable perihilar cholangiocarcinoma. Ann Surg. 2021;273(2):240–50.

    Article  PubMed  Google Scholar 

  4. Bertuccio P, Malvezzi M, Carioli G, Hashim D, Boffetta P, El-Serag HB, et al. Global trends in mortality from intrahepatic and extrahepatic cholangiocarcinoma. J Hepatol. 2019;71(1):104–14.

    Article  PubMed  Google Scholar 

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

  6. Guay C, Regazzi R. Exosomes as new players in metabolic organ cross-talk. Diabetes Obes Metab. 2017;19(Suppl 1):137–46.

    Article  PubMed  Google Scholar 

  7. Budnik V, Ruiz-Canada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17(3):160–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14(3):195–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu J, Ren L, Li S, Li W, Zheng X, Yang Y, et al. The biology, function, and applications of exosomes in cancer. Acta Pharm Sin B. 2021;11(9):2783–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bai M, Fu W, Su G, Cao J, Gao L, Huang C, et al. The role of extracellular vesicles in cholangiocarcinoma. Cancer Cell Int. 2020;20(1):435.

    Article  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  12. Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 2019;18(1):75.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Mathieu M, Martin-Jaular L, Lavieu G, Thery C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17.

    Article  CAS  PubMed  Google Scholar 

  14. Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3.

  16. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in Exosome Isolation Techniques. Theranostics. 2017;7(3):789–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):152.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Witwer KW, Soekmadji C, Hill AF, Wauben MH, Buzas EI, Di Vizio D, et al. Updating the MISEV minimal requirements for extracellular vesicle studies: building bridges to reproducibility. J Extracell Vesicles. 2017;6(1):1396823.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913.

    Article  PubMed  Google Scholar 

  20. Szatanek R, Baj-Krzyworzeka M, Zimoch J, Lekka M, Siedlar M, Baran J. The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int J Mol Sci. 2017;18(6).

  21. Saveyn H, De Baets B, Thas O, Hole P, Smith J, Van der Meeren P. Accurate particle size distribution determination by nanoparticle tracking analysis based on 2-D Brownian dynamics simulation. J Colloid Interface Sci. 2010;352(2):593–600.

    Article  CAS  PubMed  Google Scholar 

  22. Chiriaco MS, Bianco M, Nigro A, Primiceri E, Ferrara F, Romano A, et al. Lab-on-Chip for Exosomes and Microvesicles Detection and Characterization. Sensors (Basel). 2018;18(10).

  23. Mathieu M, Nevo N, Jouve M, Valenzuela JI, Maurin M, Verweij FJ, et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat Commun. 2021;12(1):4389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen H, Chengalvala V, Hu H, Sun D. Tumor-derived exosomes: Nanovesicles made by cancer cells to promote cancer metastasis. Acta Pharm Sin B. 2021;11(8):2136–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhou B, Xu K, Zheng X, Chen T, Wang J, Song Y, et al. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther. 2020;5(1):144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020;5(1):145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li L, Zhao J, Zhang Q, Tao Y, Shen C, Li R, et al. Cancer Cell-Derived Exosomes Promote HCC Tumorigenesis Through Hedgehog Pathway. Front Oncol. 2021;11:756205.

  28. Zhou J, Wang H, Sun Q, Liu X, Wu Z, Wang X, et al. miR-224-5p-enriched exosomes promote tumorigenesis by directly targeting androgen receptor in non-small cell lung cancer. Mol Ther Nucleic Acids. 2021;23:1217–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Xue P, Huang S, Han X, Zhang C, Yang L, **ao W, et al. Exosomal miR-101–3p and miR-423–5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ. 2021.

  30. He L, Zhu W, Chen Q, Yuan Y, Wang Y, Wang J, et al. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics. 2019;9(26):8206–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zeng Z, Li Y, Pan Y, Lan X, Song F, Sun J, et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018;9(1):5395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tang MKS, Yue PYK, Ip PP, Huang RL, Lai HC, Cheung ANY, et al. Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat Commun. 2018;9(1):2270.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9(1):191.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Chen X, Zhou J, Li X, Wang X, Lin Y, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018;435:80–91.

    Article  CAS  PubMed  Google Scholar 

  35. Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, et al. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine. 2018;36:209–20.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Wu J, Gao W, Tang Q, Yu Y, You W, Wu Z, et al. M2 Macrophage-Derived Exosomes Facilitate HCC Metastasis by Transferring alphaM beta2 Integrin to Tumor Cells. Hepatology. 2021;73(4):1365–80.

    Article  CAS  PubMed  Google Scholar 

  37. Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, et al. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019;38(26):5158–73.

    Article  CAS  PubMed  Google Scholar 

  38. Zhang PF, Gao C, Huang XY, Lu JC, Guo XJ, Shi GM, et al. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol Cancer. 2020;19(1):110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lu Z, Zuo B, **g R, Gao X, Rao Q, Liu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.

    Article  CAS  PubMed  Google Scholar 

  40. **e F, Xu M, Lu J, Mao L, Wang S. The role of exosomal PD-L1 in tumor progression and immunotherapy. Mol Cancer. 2019;18(1):146.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity. 2018;48(3):434–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yin Z, Yu M, Ma T, Zhang C, Huang S, Karimzadeh MR, et al. Mechanisms underlying low-clinical responses to PD-1/PD-L1 blocking antibodies in immunotherapy of cancer: a key role of exosomal PD-L1. J Immunother Cancer. 2021;9(1).

  43. Zhang K, Chen J, Li C, Yuan Y, Fang S, Liu W, et al. Exosome-mediated transfer of SNHG7 enhances docetaxel resistance in lung adenocarcinoma. Cancer Lett. 2021.

  44. Zheng P, Chen L, Yuan X, Luo Q, Liu Y, **e G, et al. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 2017;36(1):53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. **n L, Zhou LQ, Liu C, Zeng F, Yuan YW, Zhou Q, et al. Transfer of LncRNA CRNDE in TAM-derived exosomes is linked with cisplatin resistance in gastric cancer. EMBO Rep. 2021:e52124.

  46. Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, et al. Transfer of miRNA in Macrophage-Derived Exosomes Induces Drug Resistance in Pancreatic Adenocarcinoma. Cancer Res. 2018;78(18):5287–99.

    Article  CAS  PubMed  Google Scholar 

  47. Zhu X, Shen H, Yin X, Yang M, Wei H, Chen Q, et al. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J Exp Clin Cancer Res. 2019;38(1):81.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 2020;19(1):43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Han M, Hu J, Lu P, Cao H, Yu C, Li X, et al. Exosome-transmitted miR-567 reverses trastuzumab resistance by inhibiting ATG5 in breast cancer. Cell Death Dis. 2020;11(1):43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Salehi M, Sharifi M. Exosomal miRNAs as novel cancer biomarkers: Challenges and opportunities. J Cell Physiol. 2018;233(9):6370–80.

    Article  CAS  PubMed  Google Scholar 

  51. Li W, Li C, Zhou T, Liu X, Liu X, Li X, et al. Role of exosomal proteins in cancer diagnosis. Mol Cancer. 2017;16(1):145.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Zhou CY, Dong YP, Sun X, Sui X, Zhu H, Zhao YQ, et al. High levels of serum glypican-1 indicate poor prognosis in pancreatic ductal adenocarcinoma. Cancer Med. 2018;7(11):5525–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Conde-Vancells J, Rodriguez-Suarez E, Gonzalez E, Berisa A, Gil D, Embade N, et al. Candidate biomarkers in exosome-like vesicles purified from rat and mouse urine samples. Proteomics Clin Appl. 2010;4(4):416–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kitdumrongthum S, Metheetrairut C, Charoensawan V, Ounjai P, Janpipatkul K, Panvongsa W, et al. Dysregulated microRNA expression profiles in cholangiocarcinoma cell-derived exosomes. Life Sci. 2018;210:65–75.

    Article  CAS  PubMed  Google Scholar 

  55. Haga H, Yan IK, Takahashi K, Wood J, Zubair A, Patel T. Tumour cell-derived extracellular vesicles interact with mesenchymal stem cells to modulate the microenvironment and enhance cholangiocarcinoma growth. J Extracell Vesicles. 2015;4:24900.

    Article  PubMed  Google Scholar 

  56. Qin X, Lu M, Li G, Zhou Y, Liu Z. Downregulation of tumor-derived exosomal miR-34c induces cancer-associated fibroblast activation to promote cholangiocarcinoma progress. Cancer Cell Int. 2021;21(1):373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Li L, Piontek K, Ishida M, Fausther M, Dranoff JA, Fu R, et al. Extracellular vesicles carry microRNA-195 to intrahepatic cholangiocarcinoma and improve survival in a rat model. Hepatology. 2017;65(2):501–14.

    Article  CAS  PubMed  Google Scholar 

  58. Ota Y, Takahashi K, Otake S, Tamaki Y, Okada M, Aso K, et al. Extracellular vesicle-encapsulated miR-30e suppresses cholangiocarcinoma cell invasion and migration via inhibiting epithelial-mesenchymal transition. Oncotarget. 2018;9(23):16400–17.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Wang S, Hu Y, Lv X, Li B, Gu D, Li Y, et al. Circ-0000284 arouses malignant phenotype of cholangiocarcinoma cells and regulates the biological functions of peripheral cells through cellular communication. Clin Sci (Lond). 2019;133(18):1935–53.

    Article  CAS  Google Scholar 

  60. Xu Y, Leng K, Yao Y, Kang P, Liao G, Han Y, et al. A Circular RNA, Cholangiocarcinoma-Associated Circular RNA 1, Contributes to Cholangiocarcinoma Progression, Induces Angiogenesis, and Disrupts Vascular Endothelial Barriers. Hepatology. 2021;73(4):1419–35.

    Article  CAS  PubMed  Google Scholar 

  61. Scavo MP, Depalo N, Rizzi F, Ingrosso C, Fanizza E, Chieti A, et al. FZD10 Carried by Exosomes Sustains Cancer Cell Proliferation. Cells. 2019;8(8).

  62. Dutta S, Reamtong O, Panvongsa W, Kitdumrongthum S, Janpipatkul K, Sangvanich P, et al. Proteomics profiling of cholangiocarcinoma exosomes: A potential role of oncogenic protein transferring in cancer progression. Biochim Biophys Acta. 2015;1852(9):1989–99.

    Article  CAS  PubMed  Google Scholar 

  63. Chen JH, **ang JY, Ding GP, Cao LP. Cholangiocarcinoma-derived exosomes inhibit the antitumor activity of cytokine-induced killer cells by down-regulating the secretion of tumor necrosis factor-alpha and perforin. J Zhejiang Univ Sci B. 2016;17(7):537–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lapitz A, Arbelaiz A, O'Rourke CJ, Lavin JL, Casta A, Ibarra C, et al. Patients with Cholangiocarcinoma Present Specific RNA Profiles in Serum and Urine Extracellular Vesicles Mirroring the Tumor Expression: Novel Liquid Biopsy Biomarkers for Disease Diagnosis. Cells. 2020;9(3).

  65. Shen L, Chen G, **a Q, Shao S, Fang H. Exosomal miR-200 family as serum biomarkers for early detection and prognostic prediction of cholangiocarcinoma. Int J Clin Exp Pathol. 2019;12(10):3870–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Hu C, Zhang Y, Zhang M, Li T, Zheng X, Guo Q, et al. Exosomal Cripto-1 Serves as a Potential Biomarker for Perihilar Cholangiocarcinoma. Front Oncol. 2021;11:730615.

  67. Ge X, Wang Y, Nie J, Li Q, Tang L, Deng X, et al. The diagnostic/prognostic potential and molecular functions of long non-coding RNAs in the exosomes derived from the bile of human cholangiocarcinoma. Oncotarget. 2017;8(41):69995–70005.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Xue XY, Liu YX, Wang C, Gu XJ, Xue ZQ, Zang XL, et al. Identification of exosomal miRNAs as diagnostic biomarkers for cholangiocarcinoma and gallbladder carcinoma. Signal Transduct Target Ther. 2020;5(1):77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Weeraphan C, Phongdara A, Chaiyawat P, Diskul-Na-Ayudthaya P, Chokchaichamnankit D, Verathamjamras C, et al. Phosphoproteome Profiling of Isogenic Cancer Cell-Derived Exosome Reveals HSP90 as a Potential Marker for Human Cholangiocarcinoma. Proteomics. 2019;19(12):e1800159.

  70. Ikeda C, Haga H, Makino N, Inuzuka T, Kurimoto A, Ueda T, et al. Utility of Claudin-3 in extracellular vesicles from human bile as biomarkers of cholangiocarcinoma. Sci Rep. 2021;11(1):1195.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Arbelaiz A, Azkargorta M, Krawczyk M, Santos-Laso A, Lapitz A, Perugorria MJ, et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology. 2017;66(4):1125–43.

    Article  CAS  PubMed  Google Scholar 

  72. Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362(14):1273–81.

    Article  CAS  PubMed  Google Scholar 

  73. Shroff RT, Javle MM, **ao L, Kaseb AO, Varadhachary GR, Wolff RA, et al. Gemcitabine, Cisplatin, and nab-Paclitaxel for the Treatment of Advanced Biliary Tract Cancers: A Phase 2 Clinical Trial. JAMA Oncol. 2019;5(6):824–30.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Wang J, Ilyas S. Targeting the tumor microenvironment in cholangiocarcinoma: implications for therapy. Expert Opin Investig Drugs. 2021;30(4):429–38.

    Article  CAS  PubMed  Google Scholar 

  75. Rizvi S, Khan SA, Hallemeier CL, Kelley RK, Gores GJ. Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol. 2018;15(2):95–111.

    Article  CAS  PubMed  Google Scholar 

  76. Farshidfar F, Zheng S, Gingras MC, Newton Y, Shih J, Robertson AG, et al. Integrative Genomic Analysis of Cholangiocarcinoma Identifies Distinct IDH-Mutant Molecular Profiles. Cell Rep. 2017;18(11):2780–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015;47(9):1003–10.

    Article  CAS  PubMed  Google Scholar 

  78. Tabernero J, Bahleda R, Dienstmann R, Infante JR, Mita A, Italiano A, et al. Phase I Dose-Escalation Study of JNJ-42756493, an Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients With Advanced Solid Tumors. J Clin Oncol. 2015;33(30):3401–8.

    Article  CAS  PubMed  Google Scholar 

  79. Javle M, Roychowdhury S, Kelley RK, Sadeghi S, Macarulla T, Weiss KH, et al. Infigratinib (BGJ398) in previously treated patients with advanced or metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements: mature results from a multicentre, open-label, single-arm, phase 2 study. Lancet Gastroenterol Hepatol. 2021;6(10):803–15.

    Article  PubMed  Google Scholar 

  80. Homet Moreno B, Ribas A. Anti-programmed cell death protein-1/ligand-1 therapy in different cancers. Br J Cancer. 2015;112(9):1421–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Piha-Paul SA, Oh DY, Ueno M, Malka D, Chung HC, Nagrial A, et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: Results from the KEYNOTE-158 and KEYNOTE-028 studies. Int J Cancer. 2020;147(8):2190–8.

    Article  CAS  PubMed  Google Scholar 

  82. Gilligan KE, Dwyer RM. Engineering Exosomes for Cancer Therapy. Int J Mol Sci. 2017;18(6).

  83. Bandiera S, Pfeffer S, Baumert TF, Zeisel MB. miR-122–a key factor and therapeutic target in liver disease. J Hepatol. 2015;62(2):448–57.

    Article  CAS  PubMed  Google Scholar 

  84. Lou G, Song X, Yang F, Wu S, Wang J, Chen Z, et al. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 2015;8:122.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Lou G, Chen L, **a C, Wang W, Qi J, Li A, et al. MiR-199a-modified exosomes from adipose tissue-derived mesenchymal stem cells improve hepatocellular carcinoma chemosensitivity through mTOR pathway. J Exp Clin Cancer Res. 2020;39(1):4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  87. Chaiyadet S, Sotillo J, Krueajampa W, Thongsen S, Brindley PJ, Sripa B, et al. Vaccination of hamsters with Opisthorchis viverrini extracellular vesicles and vesicle-derived recombinant tetraspanins induces antibodies that block vesicle uptake by cholangiocytes and reduce parasite burden after challenge infection. PLoS Negl Trop Dis. 2019;13(5):e0007450.

  88. Gao Y, Zhang H, Zhou N, Xu P, Wang J, Gao Y, et al. Methotrexate-loaded tumour-cell-derived microvesicles can relieve biliary obstruction in patients with extrahepatic cholangiocarcinoma. Nat Biomed Eng. 2020;4(7):743–53.

    Article  CAS  PubMed  Google Scholar 

  89. Luo C, **n H, Zhou Z, Hu Z, Sun R, Yao N, et al. Tumor-Derived Exosomes Induce Immunosuppressive Macrophages to Foster Intrahepatic Cholangiocarcinoma Progression. Hepatology. 2022.

  90. Chen S, Chen Z, Li Z, Li S, Wen Z, Cao L, et al. Tumor-associated macrophages promote cholangiocarcinoma progression via exosomal Circ_0020256. Cell Death Dis. 2022;13(1):94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Thanks for the support from National Natural Science Foundation of China (No.81874062, 82072730), and Youth Program of National Natural Science Foundation of China (81902439). And thanks for two correspondence authors' wholehearted guidance and other author's cooperation.

Funding

The work was supported by National Natural Science Foundation of China (No. 81874062, 82072730), and Youth Program of National Natural Science Foundation of China (81902439).

Author information

Authors and Affiliations

Authors

Contributions

KZ and WY designed the review. KZ drafted the manuscript. WY and J-MW participated in the critical revision of the manuscript. X-YL, Y-XS, YL, PQ, Z-DD participated in the search for the articles cited. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Wei Yao or Jianming Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, K., Li, X., Shi, Y. et al. Exosomes in the tumor microenvironment of cholangiocarcinoma: current status and future perspectives. J Transl Med 20, 117 (2022). https://doi.org/10.1186/s12967-022-03294-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-022-03294-x

Keywords