Skip to main content

An exosomal strategy for targeting cancer-associated fibroblasts mediated tumors desmoplastic microenvironments

Abstract

Tumors desmoplastic microenvironments are characterized by abundant stromal cells and extracellular matrix (ECM) deposition. Cancer-associated fibroblasts (CAFs), as the most abundant of all stromal cells, play significant role in mediating microenvironments, which not only remodel ECM to establish unique pathological barriers to hinder drug delivery in desmoplastic tumors, but also talk with immune cells and cancer cells to promote immunosuppression and cancer stem cells-mediated drug resistance. Thus, CAFs mediated desmoplastic microenvironments will be emerging as promising strategy to treat desmoplastic tumors. However, due to the complexity of microenvironments and the heterogeneity of CAFs in such tumors, an effective deliver system should be fully considered when designing the strategy of targeting CAFs mediated microenvironments. Engineered exosomes own powerful intercellular communication, cargoes delivery, penetration and targeted property of desired sites, which endow them with powerful theranostic potential in desmoplastic tumors. Here, we illustrate the significance of CAFs in tumors desmoplastic microenvironments and the theranostic potential of engineered exosomes targeting CAFs mediated desmoplastic microenvironments in next generation personalized nano-drugs development.

Graphical Abstract

Introduction

Despite the rapid development of delivery platforms and theranostic tools, malignancies are still one of the leading causes of human death globally. Currently, most clinically targeted-therapies mainly target cancer cells, while ignoring their surrounding substances in microenvironments. The microenvironments mainly contain stromal cells (e.g., cancer-associated fibroblasts (CAFs), immune cells, pericytes and endothelial cells), cancer cells, cytokines, chemokines, extracellular matrix (ECM) and vasculature [1]. In malignant tumors, the microenvironments of desmoplastic tumors (e.g., cholangiocarcinoma, pancreatic ductal adenocarcinoma, ovarian cancer, breast cancer, colorectal cancer, prostate cancers and lung cancer) are especially complex composed of abundant stromal cells and deposited ECM, which help establish pathological barrier to hinder the effective transport of therapeutic agents to the cancer site [2,3,4,5,6,7,8,9,10,11]. Meanwhile, the barrier also can compress blood vessels to reduce blood perfusion, further decreasing the delivery and extravasation of immune cells and therapeutic agents. Thus, aiming at the treatment of such tumors, targeting the complex tumors desmoplastic microenvironments are promising therapeutic avenues.

Notably, CAFs are the most abundant of all stromal cells in desmoplastic microenvironments, which contribute to 60–90% ECM proteins and remodel ECM through exerting a physical force on cancer cells as well as depositing, crosslinking and degrading the ECM proteins [1, 12, 13]. The ECM after CAFs remodeling is rigid, which not only serves as pathological barrier to hinder the effective delivery of drug and immune cells infiltration, but also offers scaffold for the migration and invasion of tumor cells [5, 14]. Additionally, CAFs can secret abundant cytokines and ECM proteins that activate associated signaling pathways to talk with immune cells and cancer cells, promoting immune evasion of tumor cells and cancer stem cells (CSCs)-mediated resistance of therapeutic agents [1]. Therefore, CAFs play significant role in mediating the formation of complex tumor microenvironments.

Currently, the therapies of targeting CAFs mediated desmoplastic microenvironments in most of tumors are mainly performed by synthesized nanoparticles (e.g., liposomes, micelles and polymer-based synthetic nanoparticles) that can improve the solubility, efficiency, blood circulation time of chemotherapeutic agents [2]. However, desmoplastic environment presents unique and complex pathological abnormality that limit drug delivery even in nanoscale, so there is a need to a tailored targeted-delivery system for desmoplastic tumors that can penetrate pathological barrier and may be less likely to trigger an immune response. Compared with synthesized nanoparticles, exosomes originate from biological systems can inherit abundant information (e.g., nucleic acids, lipids and proteins) from the patient cells and own good biocompatibility, biodegradability, intercellular communication and low immunogenicity, which endow them with unprecedented potential as carriers for drug delivery [15,16,17]. Additionally, exosomes also exhibit excellent deep penetration due to their specific phospholipid bilayer structure and naturally small size, the mediation of transcytosis, and the carriers with ECM remodeling components, which are suitable for the treatment of desmoplastic tumors [18,19,20]. To gain multiple functional properties, engineered modifications can be flexibly exerted on outer membrane or inner cargoes of exosomes, further improving their specific targeting and accumulation of at desired sites [15, 21, 22]. Meanwhile, with the rapid development of engineered exosomes as communicator or messenger in surrounding cells and substances have endowed them with powerful potential in the theranostic of desmoplastic tumors.

In this review, we illustrate how CAFs can be significant role in tumors desmoplastic microenvironments and their detailed mechanism, while also elaborate that engineered exosomes could target CAFs mediated desmoplastic microenvironment for the effectively delivery and theranostic of such complex tumor in the future.

The significance of CAFs in tumors desmoplastic microenvironments

How CAFs constitute the complexity of desmoplastic microenvironment

Desmoplastic tumors with high-grade malignancy are characterized by fibrotic stroma and accompanied by abundant stromal cells and ECM deposition [2,3,4], including cholangiocarcinoma, pancreatic ductal adenocarcinoma, ovarian cancer, breast cancer, colorectal cancer, prostate cancers and lung cancer [5,6,7,8,9,10,11]. The dense stroma can reduce blood perfusion by compressing blood vessels, further decreasing the effective delivery and extravasation of therapeutic agents. Additionally, the surrounding substances of desmoplastic tumors establish a pathological barrier that hinders the effective transport of therapeutic agents to the cancer site, leading to the poor distribution and penetration in desmoplastic tumors [2, 3]. Thus, chemotherapy and nanomedicine therapy failed more than once to treat desmoplastic tumors even though they are sufficient to eliminate tumor cells in vitro experiments.

The complex desmoplastic microenvironments are composed of abundant stromal cells (e.g., CAFs, infiltrating immune cells, pericytes and endothelial cells), cancer cells, cytokines, chemokines, ECM and vasculature [2, 3]. Notably, CAFs, the most abundant of all stromal cells merged in tumor tissues, are activated fibroblasts embedded within ECM during cancer development, which exhibit enhanced proliferative, migratory and secretory properties compared with quiescent fibroblasts and normal activated fibroblasts [1, 12, 23]. The most prominent feature of CAFs is that they can secrete a great deal of ECM proteins and remodel ECM via exerting a physical force as well as depositing, crosslinking and degrading ECM proteins during desmoplastic tumors progression, ultimately leading to the stiffness of ECM and tumors tissue [1, 13]. Subsequently, the CAFs constructed rigid ECM not only serves as pathological barrier to hinder drugs delivery and immune cells infiltration, but also provides a scaffold for the invasion and migration of tumor cells (Fig. 1) [5, 14]. Additionally, CAFs also secrete larger numbers of cytokines and chemokines to instigate immune cells into their friends and establish an immunosuppressive microenvironment [1]. As cancer cells’ henchmen, it’s not surprising that CAFs can talk directly with cancer cells by secreting cytokines and ECM proteins. It is reported that CAFs not only can promote the dedifferentiation of cancer cells toward CSCs, but also participate in the maintenance of CSCs self-renew, further promoting resistance of therapeutic agents [24, 25]. Notably, cancer cells affected by CAFs can secrete cytokines, which further triggering the recruitment and activation of fibroblasts, leading to the accumulation of high activity of CAFs and forming a positive feedback loop [1, 14, 26]. Meanwhile, the accumulation of abundant CAFs also can compress blood vessels and form pathological barriers to impede the effective drugs delivery to the inner of desmoplastic tumors [2]. In view of mentioned above, CAFs can mediate the formation of tumors desmoplastic microenvironments. Therefore, aiming at the treatment of such tumors, targeting CAFs mediated microenvironments are promising therapeutic avenues.

Fig. 1
figure 1

The significance of CAFs in desmoplastic tumors. CAFs secrete abundant ECM proteins and exert physical force to remodel ECM and establish pathological barrier that hinders drugs delivery and immune cells infiltration and promotes cancer cells invasion. CAFs also secret abundant cytokines and ECM proteins to induce immunosuppress and CSCs. Cancer cells affected by CAFs can secrete abundant cytokines to promote CAFs accumulation that form barrier and compress blood vessels. CSF1, colony-stimulating factor 1; FGF5, fibroblast growth factor 5; EMT, epithelial mesenchymal transition

Heterogeneity of CAFs

CAFs are heterogeneous cell populations with diverse subpopulations and functions in many desmoplastic tumor types, which might be ascribed to their various cellular origins, mainly including tissue-resident fibroblasts, mesenchymal stem cells, quiescent stellate cells, endothelial cells, epithelial cells and adipocytes (Fig. 1) [1, 27,28,29]. These precursor cells are activated, recruited, or trans-differentiated into CAFs under the stimuli of multiple factors in tumors desmoplastic microenvironments, including oxidative stress, local hypoxia, physical changes in the ECM, exosomes and DNA damage during radiation therapy, cancer-derived cytokines such as transforming growth factor-β (TGF-β), interleukin-6 (IL-6), platelet-derived growth factor (PDGF), stromal derived factor 1 (SDF1) and hepatocyte growth factor (HGF) [1, 26, 27, 30]. Activated CAFs can be identified by markers, including but not limited to fibroblast activation protein (FAP), PDGF receptors (PDGFRs) and alpha smooth muscle actin (αSMA) [1, 12, 30]. FAP is a serine protease with overexpression on CAFs in more than 90% of human cancer, which not only takes part in ECM remodeling, but also induces the immunosuppressive microenvironment [31,32,33]. PDGFRs, tyrosine kinase receptors, are mainly classified as two types-PDGFRα and PDGFRβ [34]. PDGF can recruit and activate fibroblasts into a CAF-like state to promote desmoplastic tumor growth and stimulate angiogenesis via interacting with PDGFRs [8, 34]. α-SMA, a cytoskeletal protein, is related to the secretion of TGF-β and the regulation of myofibroblast contractility, which has been served as a most frequent marker for the identification of CAFs and the prognosis factor of desmoplastic tumors [5, 35, 36]. Unfortunately, none of these markers are specifically expressed by CAFs because they are shared with at least one cell subpopulation, which also emphasizes CAFs heterogeneity that mainly exhibited by the existence of various CAF subpopulations in different desmoplastic tumor and accompanied by different markers [28].

In recent years, abundant CAF subpopulations have been identified using single-cell RNA sequencing in desmoplastic tumors and some of them exhibit different markers and biological functions (Table 1). In pancreatic ductal adenocarcinoma, researchers first identified two CAF subpopulations derived from pancreatic stellate cells, myofibroblast CAF and inflammatory CAF, which can dynamically reverse between them in vitro [37]. Among them, myofibroblast CAF expresses high level of αSMA and associated with an ECM-producing, conversely, inflammatory CAF expresses low level of αSMA, which increasingly secretes inflammatory factors (such as IL-6 IL-8, IL-11 and LIF) and characterizes by an inflammatory phenotype. Subsequently, these two CAF subpopulations also were found in breast cancer and cholangiocarcinoma patient samples [38, 39]. Four CAFs subpopulations (CAF-A to CAF-D) were identified in human pancreatic ductal adenocarcinoma with different biomarkers, in which periostin was a biomarker for subtype A and associated with tumor invasion and shorter survival, myosin-11 was biomarker for subtype B and related to lymph-node metastasis, podoplanin was biomarker for subtype C and associated with immune promotion and good prognosis, and no marker was selected in subtype D [40]. In breast cancer and ovarian cancer, four CAF subpopulations, CAF-S1 to CAF-S4, were identified with many markers, such as PDGFRβ, FAP, CD29, αSMA and S100A4 [35, 41, 42]. CAF-S1 and CAF-S4 subpopulations exhibited pro-invasive characteristics and accompanied with high expression of αSMA. Among them, CAF-S1 promotes cancer cell invasion mainly through secreting TGF-β and C-X-C chemokine ligan (CXCL) 12, while CAF-S4 through Notch pathway. Another difference is that in terms of immunosuppression, CAF-S1 can enhance the function of CD4+T and CD25+T lymphocytes and increase the capacity of Treg Cell, while CAF-S4 does not have these characteristics. In breast cancer, Brechbuhl et al. reported two CAF subtypes, CD146posCAFs and CD146negCAFs. Among them, CD146posCAFs promote oestrogen-dependent proliferation and tamoxifen sensitivity of cancer cells, whereas CD146negCAFs suppress oestrogen receptor expression and enhance tamoxifen resistance [43]. These examples, and many more (Table 1), are leading us to reveal the significant relationship between CAF subpopulations and desmoplastic tumors progression.

Table 1 CAF subpopulations and markers were identified in different desmoplastic tumors

The above studies showed that in addition to the majority of CAF subpopulations exert tumor-promoting functions, there are also a few tumor-repressing CAF subpopulations in certain cancers. Under the circumstances, nonselective targeting of CAF subpopulations may be off-target or even adverse clinical outcomes in cancer treatment. Such as, the depletion of αSMA+ myofibroblasts in pancreatic ductal adenocarcinoma can suppress immune surveillance with increased CD4+Foxp3+ Tregs, further reducing survival of patients [44]. Consequently, it is critical to identify more specific markers to distinguish tumor-promoting and tumor-repressing CAF subpopulations and design specific targeted-exosomes to treat desmoplastic tumors in the future. But here, the functions of tumor-promoting CAFs in tumors desmoplastic microenvironment were emphasized.

Briefly, CAFs paly significant role in mediating the formation of tumors desmoplastic microenvironment, resulting in an abundance of dense and fibrous tissue that acts as pathological barrier, which limit the efficacy of drugs by blocking the deep delivery to tumor sites. Clinically, desmoplastic tumors with high mortality present a great challenge for recurrence and metastasis. Thus, there is a need for a biocompatible targeted drug delivery system that can penetrate pathological barrier, further reducing mortality of desmoplastic tumors.

myCAFs myofibroblast CAFs, iCAFs inflammatory CAFs, Lox lysyl oxidase, PDPN podoplanin, TPM tropomyosins 1, POSTN periostin, MMP matrix metallopeptidase, HAS hyaluronan synthases, MHC II major histocompatibility complex class II

Functions of CAFs in desmoplastic tumors progression

In desmoplastic tumor progression, CAFs secrete abundant ECM proteins and exert physical force to remodel ECM and establish pathological barrier that hinders drugs delivery and immune cells infiltration and promotes cancer cells invasion. Furthermore, CAFs also secret abundant cytokines and ECM proteins to talk with immune cells and cancer cells, inducing immunosuppression and promoting CSCs-mediated resistance for therapeutic agents (Fig. 1).

CAFs remodel ECM

ECM, a non-cellular component, consists of macromolecules including collagen, elastin, fibrin and proteoglycan, which is a significant supporter of the tumor tissues and stromal cells in desmoplastic tumor. During desmoplasia, CAFs can secrete abundant ECM proteins and remodel ECM via exerting a physical force on cancer cells as well as continuously depositing, crosslinking and degrading the ECM proteins, resulting in the stiffness of desmoplastic tumor tissue and matricellular fibrosis that obstruct drugs delivery and immune cells infiltration and provide scaffold for the invasion and migration of tumor cells [1].

ECM remodeling also can be achieved by CAFs-mediated physical force, further promoting the invasion of tumor cells. Study reported that CAFs not only produce rich fibronectin, but also modulate fibronectin matrix by increasing contractility and traction forces that are exerted by PDGFRα and nonmuscle myosin II [50]. Then the contractile and tractive forces are transmitted to fibronectin via α5β1 integrin. Finally, CAFs organize fibronectin as parallel fibers, further promoting directional migration of prostate tumor cells. Additionally, by cell–cell contact, CAFs also exert physical forces to promote the joint invasion of CAFs and cancer cells, further remodeling ECM. The published study showed that force transmission was exerted through heterophilic adhesion between N-cadherin on CAFs and E-cadherin on tumor cells [51].

CAFs can deposit ECM proteins, such as I, III, IV and V types of collagens, fibronectins, hyaluronan, laminins, glycoproteins and proteoglycans, which contribute to the stiffness of ECM and go on as pathological barrier that hinders drugs delivery and the infiltration of immune cells [1, 52]. Pietilä et al. demonstrated that increased ECM stiffness can protect ovarian cancer cells from cisplatin mediated apoptosis via focal adhesion kinases and yes-associated protein signaling pathway [7]. The process of deposition often matches with matrix crosslinking enzymes. For instance, lysyl oxidase-like 2 is responsible for crosslinking collagen and elastin leading to the stiffness of ECM, which can be used as a biomarker of poor prognosis in cholangiocarcinoma and pancreatic ductal adenocarcinoma [5, 53]. Intriguingly, instead of isolating cancer cells, the barrier for ECM stiffening facilitates their invasion and spread [7, 54]. The overexpression of FAP in pancreatic cancer CAF can remodel ECM through modulating fibronectin levels and increasing the organization of collagen fiber, further forming a hardened and parallel fiber that increases the directionality of tumor cell invasion [31]. Simultaneously, in the process of desmoplasia, CAFs produce ECM-degrading proteases, such as MMPs, which can remodel ECM and promote the metastasis and invasion of tumor cells. The overexpressed MMPs (such as MMP-2, MMP7, MMP-9, MMP-11 and MMP-14) are frequently induced by TGF-α/β, NF-κB and WNT, which are related to poor prognosis for cholangiocarcinoma and breast cancer [5, 55, 56]. Therefore, desmoplasia is a dynamic stromal alteration process via CAFs-mediated local remodeling of ECM.

CAFs induce immunosuppression

Except for the pathological barrier caused by CAFs and ECM to hinder immune cells infiltration, CAFs can secrete larger numbers of cytokines and chemokines to recruit immunosuppressive cells or reduce the activities of immune effector cells, promoting the immune escape of cancer cells [1, 27, 57].

In desmoplastic tumors progression, the most common immunosuppressive cells, such as tumor associated macrophages (TAM) 2 and myeloid-derived suppressor cells (MDSCs), are related to poor clinical prognosis of patients [58, 59]. TAMs are conventionally divided into two subpopulations according to the differentiation degree and function, TAM1 and TAM2, in which TAM1 primarily plays an antitumor role with proinflammatory properties, whereas TAM2 exhibits tumor-promoting activity [59, 60]. CAFs-derived colony-stimulating factor 1 can recruit monocyte and trans-differentiation toward the TAM2 via the interaction of colony-stimulating factor 1 and its receptor to promote cancer progression [61]. IL-8 secreted from CAFs can attract monocytes and subsequently facilitate the polarization of macrophages into TAM2, which inhibits the functioning of natural killer cells in colorectal cancer [62]. Reciprocally, TAM2 recruited by CAFs can also further stimulate CAFs activation and progression. For instance, CAFs-derived SDF1 and CXCL14 can promote monocyte recruitment and trans-differentiation toward the TAM2, in turn, TAM2 can secrete SDF-1 and IL-6 to activate CAFs, further making up a positive loop that facilitates immunosuppression and prostate cancers progression [63, 64]. FAP+CAFs-derived C–C chemokine ligand (CCL) 2 can mediate the recruitment of MDSCs through the FAP-STAT3-CCL2 pathway, further establishing an immunosuppressive environment [32]. Subsequently, a similar act of CAF-derived CCL2 on MDSCs was discovered in lung cancer, and the accumulation of MDSCs suppressed CD8 + T cell function [65]. Additionally, CAFs also secrete SDF-1 recruit monocytes and trigger their differentiation into MDSCs via IL-6/STAT3, further inhibiting T cell proliferation [66].

Cytotoxic T lymphocytes (CTLs, also called CD8+T) mainly induce cytotoxic activities to promote the apoptosis of cancer cells, which are taken as significant component of antitumor effects [59, 67]. However, the existence of CAFs could inhibit the growth, infiltration and antitumor immunity of CD8+T cells [68]. CXCL12 is upregulated in activated pancreatic stellate cells, which can decrease the migration of CD8+T cells to cancer islets in pancreatic ductal adenocarcinoma [69]. CAFs can also secrete IL-6, βig-h3 (also called TGFβi) and TGFβ to restrict the activity of CD8+T cell [66, 70, 71]. Additionally, CAFs also can suppress CD8+T cells by the activation of immune checkpoint. Lakins et al. [72] reported that CAFs participate in sampling, process, cross-present antigen and induce CD8+T cells death via coincidental upregulation of FAS and PD-1 on T cells and FASL and PD-L2 on CAFs.

CAFs promote CSCs

CSCs are special stem-like subpopulation in tumors that can self-renew and differentiate into tumor cells, leading to resistance of therapeutic agents [73, 74]. In desmoplastic tumors progression, CAFs-secreted cytokines and ECM proteins not only can promote the dedifferentiation of cancer cells toward CSCs‐like phenotype, but also participate in the maintenance of CSCs self-renewal, further promoting the resistance of therapeutic agents [24, 25].

CAFs can transform cancer cells into CSCs. Chen et al. demonstrated that differentiated lung cancer cells can dedifferentiate into CSCs when co-culturing with CAFs [75]. Under the stimulation of cancer-released hedgehog ligands, CAFs can secret fibroblast growth factor 5 to facilitate the dedifferentiation of breast cancer cells into CSCs [76]. Breast CAF-secreted TGF-β can increase ZEB1 transcription and induce the dedifferentiation of tumor cells into CSCs state [77]. Colorectal CAF-secreted HGF can promote the transformation of tumor cells into CSCs through Wnt/β-catenin signaling pathway [78]. Prostate cancer-derived IL-6 can activate fibroblast, and then activated prostate CAFs can secrete MMPs and trigger epithelial mesenchymal transition in cancer cells to facilitate the transformation into CSCs‐like phenotype [79]. In ovarian cancer, CAFs-secret IL-8 also can activate Notch signaling pathway to promote CSCs [80].

Additionally, CAFs play significant role in the maintenance of CSCs. CAFs-derived insulin-like growth factor-II (IGF-II) can activate IGF-II/IGF1 receptor/Nanog signaling to maintain CSCs, which is related to poor prognosis in lung cancer patients [75]. In breast cancer, CAFs-secreted CCL2 can induce CSCs self-renewal via Notch1 signaling [81]. Under hypoxia tumor microenvironment, colorectal CAFs-secreted TGF-β can promote CSCs maintenance and reduce sensitivity to 5-fluorouracil and oxaliplatin by hypoxia/TGF-β/GLI2 signature [82]. CD10+GPR77+ CAFs-derived IL-6 and IL-8 can induce CSCs self-renewal and enrichment, further protecting breast and lung cancer cells from neo-adjuvant chemotherapy-induced cell death [45]. In colorectal cancer, under the stimulation of clinically relevant chemotherapy agents (5-fluorouracil, oxaliplatin and leucovorin), CAF-secreted IL-17A can maintain CSCs self-renewal [83]. Furthermore, research showed that stimulated CAFs can promote the formation of fibrillar collagen that provides a supportive niche for CSCs self-renewal, further reducing the sensitivity of docetaxel chemotherapy [76].

Engineered exosomes serve as promising drug delivery platform for targeting CAFs mediated desmoplastic microenvironments

CAFs mediated desmoplastic microenvironments have become an appealing target for treating desmoplastic tumors. Due to the existence of pathological barrier, targeting cancer cells remains challenging for clinicians. The synthesized nanoparticles (e.g., liposomes, micelles and polymer-based synthetic nanoparticles) are currently the most commonly used carriers for drug delivery, for which their features (e.g., diameter, surface charge, hydrophobicity/hydrophilicity and receptor-mediated endocytosis) can be easily adjusted or modified. Thus, synthesized nanoparticles can significantly improve the residence time, cellular internalization and release of chemotherapeutic agents [16]. However, due to the existence of unique and complex pathological barrier of desmoplastic tumors (e.g., CAFs, ECM and compressed blood vessels), which prevent some synthesized nanoparticles from going further into tumor sites, and most of which are not biodegradable leading to immunogenicity or accumulative toxicity. Compared with synthesized nanoparticles, the inherent properties of exosomes, including biocompatibility, biodegradability, intercellular communication and low immunogenicity, endow them with unprecedented potential as carriers for drug delivery [2, 15, 84]. Additionally, exosomes also exhibit excellent deep penetration due to their specific phospholipid bilayer structure and naturally small size, the mediation of transcytosis, or the carriers of ECM remodeling components, which are easier to cross ECM and, most importantly, easier fusion with receptor cells to release drugs into them [18,19,20]. Exosomes are also highly engineerable. To improve the targeted delivery of natural exosomes, further engineered modifications are needed to optimize them more specific targeting property. Based on the above advantages, engineered exosomes are suitable for targeting CAFs mediated tumors desmoplastic microenvironments, which could be taken as a promising drug delivery platform.

Exosomes mediate intercellular communication

Exosomes, with small diameter of 30–200 nm and lipid bilayer membrane, are extracellular vesicles secreted by cells, which originate from membrane invaginations, and then multivesicular bodies are formed through inward budding of the endosomal membrane, finally, exosomes can be released by fusing with plasma membrane [85, 86]. Intraluminal vesicles (future exosomes) are generated by the inward budding of membrane and accumulate in multivesicular bodies, which involves cargo sorting machinery, mainly including the endosomal sorting complex required for transport (ESCRT)-dependent or ESCRT-independent [86]. The ESCRT machinery contains five subcomplexes, in which ESCRT-0 and ESCRT-l take charge of ubiquitinated proteins cluster, ESCRT-I and ESCRT-II induce bud formation, ESCRT-III drives vesicle cleavage and releases intraluminal vesicles into the endosome to form multivesicular bodies, ATPase vacuolar protein sorting gene 4 depolymerizes the polymerized ESCRT-III that allow recycling of ESCRT [87]. Additionally, endosomal sorting also can be accomplished by ESCRT-independent machinery. The published studies showed that ceramide and tetraspanin CD63 are involved in this progress [88, 89]. Finally, the multivesicular bodies with various intraluminal vesicles can either be degraded by fusing with lysosomes or be released by fusing with plasma membrane [17].

Released exosomes from donor cells express various surface molecules, including tetraspanins, lipids, lectins and integrins, which enable them to target recipient cells and deliver contents (nucleic acids, lipids and proteins) to regulate the biochemical composition and behavior of recipient cells [17, 90]. Once exosomes attach to the plasma membrane of target cells, then can activate the intracellular signaling pathway via receptor binding to elicit functional responses [17]. Exosomes also can be internalized by clathrin, caveolae and lipid rafts-mediated endocytosis or phagocytosis/macropinocytosis and then reach multivesicular bodies, which could either be degraded via lysosomes or release contents into the cytoplasm of target cell by back fusion with the membrane of multivesicular bodies [17, 21]. Additionally, exosome contents can be directly released into the cytoplasm via membrane fusion of exosomes and recipient cell [17, 91]. Thus, in the body, exosomes can be released by almost all cells and communicate with their donor cells or neighboring/distant recipient cells to regulate various physiological and pathological processes, including blood coagulation, inflammation, immune response and tumor progression, among others [92,93,94].

Engineered exosomes serve as multifunctional carriers for drug delivery

Synthesized nanoparticles are currently the most commonly used carrier for drug delivery, which can improve the residence time and selectivity of chemotherapeutic agents [15]. But the clinical application of these nanoparticles has some difficulties, such as toxicity, low bioavailability of drugs and the trigger of immune response [2]. Additionally, synthesized nanoparticles rely heavily on the enhanced permeability and retention for passive targeting and accumulation, which limit drug delivery in desmoplastic tumor due to the existence of unique and complex pathological barrier [2]. Notably, the following advantages of exosomes offer the maximum possibility for solving these problems. First, exosomes originate from biological systems and could be acquired from the patient’s cells, which may be less likely to trigger an immune response than synthetic carrier [17, 94]. Additionally, CD47 on exosomal surface can protect them from macrophages-mediated phagocytosis via binding with signal regulatory protein alpha resulting in a don’t eat me signal, which prolongs circulation time of exosomes and improves therapeutic efficacy [95]. Second, exosomes have lipid bilayer membranes that can directly fuse with the membranes of target cell to improve the efficiency of drug internalization [15, 96]. Additionally, this membrane also can protect the loaded therapeutic agents from degradation, such as natural products, small interfering RNA (siRNA), microRNA (miRNA) and proteins [16]. Third, exosomes have superb deep penetration due to their specific phospholipid bilayer structure and naturally small size, the mediation of transcytosis, and the carriers with ECM remodeling components, which are easier to cross ECM barriers [18,19,20]. Existing reports showed that exosomes loaded with cargo can penetrate pathological barrier and improve the efficacy of therapeutic agents compared with free drug, and it has been recently revealed that the transcytosis may be associated with crossing this barrier [18, 97]. For instance, exosome carried with neuropilin-1 can penetrate blood–brain barrier and pancreatic cancer tissues through receptor-mediated transcytosis [98, 99]. Furthermore, exosomes carried ECM remodeling components can achieve deep penetration and tumor accumulation in desmoplastic tumor via degrading dense ECM. Feng et al. and Wei et al. reported that exosomes loaded with hyaluronidase PH20 and MMPs (e.g., MMP-9 and MMP-14) can penetrate dense ECM barrier via ablating them, further enhancing drug delivery and immune cells infiltration [19, 100]. Therefore, based on above advantages, exosomes as a carrier for drug delivery can carry therapeutic agents for long-time stable circulation in the body and penetrate the pathological barrier to reach the desired sites and then exert anti-tumor effects.

Engineered exosomes can target CAFs mediated desmoplastic microenvironments

Compared with traditional nanomedicine, the homing and penetration property of exosomes endow them with the advantage of targeting recipient cells in dense desmoplastic tumors. Nevertheless, in order to increase the capacity of targeting desired sites for exosomes, further engineered modifications are necessary. The precise targeting of recipient cells is achieved mainly through the high affinity between the receptor and ligand. Thus, appropriately modified exosomes can easily target the specific sites of CAFs mediated tumors desmoplastic microenvironments, such as cancer cells, CAFs, ECM and immune cells. The following are engineering modification strategies of exosomes.

Genetic engineering is the most widely used modification strategy, which aims to fuse ligands with targeted function to transmembrane proteins (Lamp2b, CD9, CD63, CD81, glycosyl phosphatidylinositol, PDGFR, C1C2 region of lactadherin) on the exosomal surface [22, 101, 102]. The targeting peptides, such as iRGD and tLyP-1, can be fused to Lamp2b on exosomes to target cancer cells by binding to the αv integrins and neuropilin-1/2 receptors on their membranes, respectively [96, 103]. In addition to the targeting peptide, transmembrane proteins also can be fused with antibody fragments. For instance, PDGFR transmembrane region can be fused with antibodies, such as anti-human CD3 and HER2, which dually target CD3 receptor of T-cell and HER2 receptors of breast cancer [104]. The CD63/CD9/CD81, C1C2 domain of lactadherin and glycosyl phosphatidylinositol-anchored proteins on exosomes also can be engineered with different ligands to target desired cells by binding to their affinity receptors [105,106,107]. Additionally, parental cells of exosomes can be modified by using plasmids or viruses that encode fusion ligands on cells, and then acquire abundant exosomes with the expression of gene-associated ligands [101]. Hu et al. showed that membrane-anchored FAP can be modified on the cancer cells by a lentiviral vector [33]. Then obtained cancer-derived exosomes with the expression of FAP antigen that can target CAFs to exert anti-tumor effects. Hong et al. designed that glycosylphosphatidylinositol-anchored hyaluronidase can be modified on HEK293T cells by plasmid encoding full-length PH20, and then isolated exosomes can target CAFs-mediated overly accumulated ECM [108]. Thus, genetic engineering can select the expression of proteins on the exosome and endow them with the capacity of targeting CAFs mediated tumors desmoplastic microenvironments.

Although less explored, exosomes could also be engineered by chemical methods. Some targeted peptides are conjugated to the membrane of exosomes by the cycloaddition reaction and bio-orthogonal click reaction to target CAFs mediated desmoplastic microenvironments [101, 102]. The study reported that RGE peptide can be conjugated with the membrane of loaded-curcumin exosome by click chemistry, which can easily reach glioma by binding to neuropilin-1 to exert the imaging and therapeutic functions [99]. The CREKA peptide can target to CAF based on the high affinity between the peptide and overexpressed fibronectin [109]. Additionally, the amphipathic molecules of distearoyl phosphoethanolamine-polyethylene glycol (DSPE-PEG) can be inserted into exosomes membrane, which is another chemical method. Subsequently, some targeted molecules are conjugated to DSPE-PEG for targeting cancer or CAF sites. It is reported that RGD and folate can be conjugated to DSPE-PEG and inserted into exosomal membrane, endowing them with the ability to target cancer sites [102, 110]. Macrophage-derived exosomes with aminoethyl anisamide (AA)-DSPE-PEG can target overexpressed sigma receptor on non-small cell lung cancer [111]. Previous study revealed that sigma receptor also is overexpressed on activated CAFs and AA-DSPE-PEG can target CAFs [112]. FH peptide (FHKHKSPALSPVGGG) can be conjugated to DSPE-PEG to target overexpressed tenascin C on CAFs [113]. Chemical modification has the advantages of short time consuming, high efficiency and mild reactions.

Theranostic potential of engineered exosomes for targeting CAFs mediated desmoplastic microenvironments

Desmoplastic tumors are characterized by abundant accumulation of CAFs and ECM deposition, which surround and infiltrate the tumor cells and block the infiltration of immune cells [2,3,4]. Thus, targeting the main ingredients (such as CAFs, ECM, immune cells and cancer cells) in CAFs mediated desmoplastic microenvironments and influencing their behavior, might solve the current clinical and therapeutic stalemate in desmoplastic tumors. The rapid development of engineered exosomes as cargo delivery carriers have endowed them with power potential in the treatment of desmoplastic tumors (Fig. 2, Table 2). Immune cells and cancer cells-derived exosomes may serve as vehicles to trigger anti-tumor immune responses through antigen presentation. Therefore, exosomes modified appropriately with antigens may target CAFs mediated desmoplastic microenvironments to and exert anti-tumor immune effects. Except for antigenic modifications on the membrane, engineered exosomes could load therapeutic agents into their lumen and membrane, including natural products, siRNA, miRNA and proteins, which can be effectively released into CAFs mediated desmoplastic microenvironments to treat desmoplastic tumors mainly through removing ECM, inactivating CAFs and inhibiting the signal transduction of CAF-tumor cells and CAF-immune cells crosstalk. Additionally, engineered exosomes carrying imaging agents have been applied in vivo imaging to precisely track the biodistribution of therapeutics-loaded exosomes and accumulation of targeted sites and obtain real-time tumor delineation, further helping physicians to diagnose and monitor treatment response of desmoplastic tumors.

Fig. 2
figure 2

Schematic illustration of engineered exosomes targeting CAFs mediated desmoplastic microenvironments to exert theranostic potential on desmoplastic tumors models. After injection, antigen and theranostic agents-loaded exosomes can target CAFs mediated desmoplastic microenvironments to exert theranostic potential via stimulating the activation of CD8+T cell, removing ECM, inactivating CAFs, inhibiting the signal transduction of CAF-tumor cells and CAF-immune cells crosstalk, and imaging in vivo

Table 2 Theranostic potential of engineered exosomes for targeting CAFs mediated desmoplastic microenvironments

Therapeutic potential of antigens-modified exosomes

Immune cells and cancer cells-derived exosomes can inherit amount antigens or proteins from their donor cells, which can participate in antigen presentation and trigger CTLs to exert specific anti-tumor immune responses [126]. Therefore, exosomes themselves or exosomes modified with appropriate antigens can target CAFs mediated desmoplastic microenvironments to exert anti-tumor immune effects, further treating desmoplastic tumors.

Dendritic cells and B lymphoma cells-derived exosomes contain MHC I/II, CD86 and heat shock proteins, which can facilitate antigen presentation to prime CD4+T cell and stimulate the activation of CD8+T cell, further exerting efficient antitumor effects [126, 127]. Tumor cell-derived exosomes inherit MHC II and amount tumor-specific antigens, which can present tumor antigens to CTLs and trigger efficient antitumor effects [22, 114]. Additionally, exosomes modified with appropriate antigens also can target CAFs to reprogram CAF-mediated immunosuppressive microenvironment. Hu et al. performed specifically antigenically modified on tumor cell-derived exosomes to achieve dual targeting of cancer cells and CAF [33]. For instance, in breast, lung and colon cancer models, the FAP gene-engineered tumor-derived exosomes not only retain tumor antigen but also FAP antigen, which could induce CTLs against cancer cells and FAP+CAFs and reprogram immunosuppressive microenvironment by transforming TAM2 into TAM1 and reducing the infiltration of MDSCs. Furthermore, the specific anti-tumor immune responses also can facilitate cancer ferroptosis through the CTLs-derived interferon-gamma from and the depletion of FAP+CAFs.

Therapeutic potential of therapeutic agents-loaded exosomes

Engineered exosomes exhibit higher biocompatibility, targeted delivery, penetration and drug protection for long-time stable circulation, which improve the effectiveness of natural products and protecting nucleic acids and proteins from degradation [15, 16]. Finally, the loaded contents can effectively exert anti-tumor effects mainly through removing ECM, inactivating CAFs and inhibiting the signal transduction of CAF-tumor cells and CAF-immune cells crosstalk.

Hyaluronan and collagen are main components of ECM secreted by CAFs, which can help to establish a pathological barrier and hinder drug delivery [1, 2, 128]. Exosomes carried with protease and ECM-associated miRNAs can penetrate dense ECM barrier via ablating them and then enhance drug delivery and immune cells infiltration. Feng et al. reported that PH20 hyaluronidase and folic acid-modified exosomes can enhance doxorubicin delivery by degrading ECM and inhibit the metastasis of breast cancer caused by hyaluronidase treatment [19]. Hong et al. designed a PH20-modified exosomes that can target CAF-mediated overly accumulated ECM and ablate them to increase immune cells infiltration and the penetration of doxorubicin in prostate cancers and re-expand the tumor blood vessel [108]. Hong et al. also reported that PH20-modified exosomes not only deeply penetrate breast cancer tissues via hyaluronan degradation, but also activate the maturation of CD103+ dendritic cells in vivo and CD8+T cells activation [115]. Al Faruque et al. designed the extracellular vesicles modified with RGD and magnetic nanoparticles and loaded with paclitaxel, which exhibit two key characteristics: first, the modified vehicles can effectively penetrate the fibrotic barrier via spontaneously removing CAFs-mediated αSMA and type 1 collagen, second, extracellular vesicles loaded with paclitaxel can be effectively internalized into pancreatic cancer cells and eventually regressing the tumors [116]. Furthermore, metalloproteinase (MMPs) was also widely reported for degrading collagen of ECM. Macrophage-derived exosomes overexpressed MMP9 and MMP14 could degrade tumor collagens, further enhancing CD8+ T cells infiltration and the deep delivery of anti-PD-1 antibody to exert anti-tumor effects [100]. Besides protease, ECM-associated miRNAs also could degrade ECM. Rana et al. reported that exosomes carried miRNA-494 and miRNA-542-3p could be delivered to neighboring stromal cells, further activating MMPs and degrading ECM in pancreatic cancer [117]. SiRNA can trigger the post-transcriptional gene silencing and have the potential for cancer treatment [129]. Engineered exosomes with gene silencing can inactivate CAFs and inhibit the signal transduction of CAF-tumor cells to exert anti-tumor effects. Li et al. reported that exosomes with silenced dicer can significantly decrease FAP and α-SMA level of CAFs in ovarian carcinoma [118]. The upregulation of S100A4 in CAFs is associated with tumor metastasis [1]. Zhao et al. designed the cationic bovine serum albumin-siS100A4-loaded exosomes that can trigger the post-transcriptional gene silencing and further inhibit the CAF-mediated lung metastasis of breast cancer [119]. Additionally, engineered exosomes also can inhibit the signal transduction of CAFs-immune cells. The main source of CXCL12 is FAP+CAFs that can induce immunosuppression through CXCL12/CXCR4 axis [130]. In breast cancer, Yue et al. designed the miR-5100-load exosomes that can inhibit the CXCL12/CXCR4 axis, further suppressing the invasion of tumor cells [120].

Theranostic potential of imaging agents-labeled exosomes

Exosomes carried with imaging agents can be applied to non-invasive theranostic of desmoplastic tumors. According to the information presented by vivo imaging, such as the biodistribution and accumulation of therapeutics-loaded exosomes and real-time tumor delineation, physicians are able to easily diagnose and monitor the treatment response of desmoplastic tumors. Fluorescent dyes, fluorescent proteins, metal nanoparticles and radioactive isotopes are common imaging agents carried by exosomes.

PKH26, DiR and cyanine are the most commonly used fluorescent dyes for labeling engineered exosomes [22, 101, 121]. Jing et al. reported that cancer-derived exosomes labeled with cyanine 7 can be used as near-infrared fuorescence imaging of colon cancer to provide a real-time tumor delineation [122]. Genetically encoded reporters of fluorescent proteins (e.g., red/green fluorescent protein and antares2) are commonly attached to transmembrane proteins (e.g., CD63, CD9 and CD81) of engineered exosomes [22, 121, 123]. For instance, prostate cancer-derived exosomes loaded with antares2-CD63 demonstrated their homing property for targeting desired organs and tissues [123]. Additionally, metal nanoparticles (such as gold-iron oxide nanoparticles, superparamagnetic iron oxide nanoparticles and gadopentetate dimeglumine) as contrast agents also have been used as magnetic resonance imaging in vivo [121, 124, 131]. Bose et al. developed exosomes that carry both therapeutic agent (anti-miR-21) and imaging agent (gold-iron oxide nanoparticles) for better breast cancer theranostics [124]. Among them, anti-miR-21 exert anti-tumor effect by reducing doxorubicin resistance, gold-iron oxide nanoparticles exhibit excellent magnetic resonance imaging that achieves the precisely track the biodistribution of anti-miR-21-loaded exosomes and tumor-specific accumulation. In addition to imaging, gold-iron oxide nanoparticles can also selectively damage tumor cells via thermal ablation [125]. The study exhibits that gold-iron oxide nanoparticles have an efficient photothermal effect in 4T1 cells. Furthermore, vivo imaging of exosomes also can be achieved by radioactive isotopes, such as 99mTc, 131I and 111In-oxine [101, 121]. Generally, radiolabeling exosomes are more sensitive and give more precise information than the mentioned above imaging methods. Jing et al. showed that cancer-derived exosomes labeled with 99mTc can be used as nanoprobes in single-photon emission computed tomography of colon cancer to help physicians precisely obtain the biodistribution of exosomes [122].

Conclusions and perspectives

CAFs, as the most abundant of all stromal cells merged in tumor tissues, play critical role in mediating desmoplastic tumors progression, which can not only remodel ECM to establish pathological barriers that hinder drug delivery and immune cells infiltration, but also secret abundant cytokines and ECM proteins to promote immunosuppression and CSCs-mediated drugs resistance. In this review, aiming at the significance of CAFs in tumors desmoplastic microenvironments and the complexity of microenvironment, we elaborate that engineered exosomes as promising delivery platform or directly applied for theranostic purpose can target CAFs mediated microenvironments and overcome pathological barrier to exert precise and efficient therapy of desmoplastic tumors.

Although exosomes have been applied in clinical trials, there are still some challenges need to be overcome. The first challenge for clinical use is the isolation of large amount of high purity exosomes. Traditional ultracentrifugation is unmatched with massive production of exosomes due to the low purity and time consuming [15, 22]. Some isolation methods for massive production, such as size-exclusion chromatography and polymer precipitation have been developed, but they have some disadvantages, such as the expensive equipment and low purity, respectively [132, 133]. Thus, the selection of the most suitable isolation methods will be crucial for cancer treatment. The second challenge is the high loading efficiency of exosomes. Theranostic agents could be loaded into exosomes either before or after exosome isolation. Cell transfection of peptides, proteins and RNA, is most commonly used method for the pre-isolation cargo loading of exosomes, but has the disadvantage of cytotoxicity and difficult purification [15, 16, 90]. Post-isolation loading methods of exosomes mainly contain co-incubation, electroporation and extrusion. Among them, electroporation and extrusion have a high loading efficiency, but they have the disadvantages of cargo aggregation and the probable damage of exosomal membrane, respectively. Thus, more suitable approach should be charily selected based on physicochemical properties of cargo to achieve maximum theranostic efficiency in the future.

Finally, it cannot be ignored that except for targeted delivery vehicles for desmoplastic tumors treatment, exosomes contents have been considered as potential biomarkers for early cancer theranostic prediction, for that they can inherent various information from donor cells and be non-invasively obtained from biological fluids, including blood, urine, saliva and tears [15, 21, 91, 134]. Therefore, a combination of in vitro and vivo applications of exosomes may be likely to burst out power potential in clinical theranostic, which not only can be applied for early diagnosis of cancers, but also can target CAFs mediated desmoplastic microenvironments to treat cancers and real-time monitor the treatment response, providing the next generation personalized nano-drugs for desmoplastic tumors therapy.

Availability of data and materials

Not applicable.

Abbreviations

CAFs:

Cancer-associated fibroblasts

ECM:

Extracellular matrix

CSCs:

Cancer stem cells

CSF1:

Colony-stimulating factor 1

FGF5:

Fibroblast growth factor 5

EMT:

Epithelial mesenchymal transition

TGFβ:

Transforming growth factor-β

IL-6:

Interleukin-6

PDGFR:

Platelet-derived growth factor receptor

SDF1:

Stromal derived factor 1

HGF:

Hepatocyte growth factor

FAP:

Fibroblast activation protein

PDGFRs:

Platelet-derived growth factor receptors

α-SMA:

Alpha smooth muscle actin

CXCL:

C-X-C chemokine ligand

myCAFs:

Myofbroblastic CAFs

iCAFs:

Infammatory CAFs

Lox:

Lysyl oxidase

PDPN:

Podoplanin

TPM:

Tropomyosins 1

POSTN:

Periostin

MMP:

Matrix metallopeptidase

HAS:

Hyaluronan synthases

MHC II:

Major histocompatibility complex class II

TAM:

Tumor associated macrophage

MDSCs:

Myeloid-derived suppressor cells

CCL2:

C–C chemokine ligand 2

CTLs:

Cytotoxic T lymphocytes

IGF-II:

Insulin-like growth factor-II

ESCRT:

Endosomal sorting complex required for transport

siRNA:

Small interfering RNA

miRNA:

MicroRNA

DSPE-PEG:

Distearoyl phosphoethanolamine-polyethylene glycol

References

  1. Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18:99–115.

    Article  CAS  PubMed  Google Scholar 

  2. Han X, Xu Y, Geranpayehvaghei M, Anderson GJ, Li Y, Nie G. Emerging nanomedicines for anti-stromal therapy against desmoplastic tumors. Biomaterials. 2020;232: 119745.

    Article  CAS  PubMed  Google Scholar 

  3. Chen R, Huang L, Hu K. Natural products remodel cancer-associated fibroblasts in desmoplastic tumors. Acta Pharm Sin B. 2020;10:2140–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gonzalez-Molina J, Moyano-Galceran L, Single A, Gultekin O, Alsalhi S, Lehti K. Chemotherapy as a regulator of extracellular matrix-cell communication: implications in therapy resistance. Semin Cancer Biol. 2022;86:224–36.

    Article  CAS  PubMed  Google Scholar 

  5. Hogdall D, Lewinska M, Andersen JB. Desmoplastic tumor microenvironment and immunotherapy in cholangiocarcinoma. Trends Cancer. 2018;4:239–55.

    Article  PubMed  Google Scholar 

  6. Liu L, Kshirsagar PG, Gautam SK, Gulati M, Wafa EI, Christiansen JC, et al. Nanocarriers for pancreatic cancer imaging, treatments, and immunotherapies. Theranostics. 2022;12:1030–60.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Pietilä EA, Gonzalez-Molina J, Moyano-Galceran L, Jamalzadeh S, Zhang K, Lehtinen L, et al. Co-evolution of matrisome and adaptive adhesion dynamics drives ovarian cancer chemoresistance. Nat Commun. 2021;12.

  8. Primac I, Maquoi E, Blacher S, Heljasvaara R, Van Deun J, Smeland HYH, et al. Stromal integrin α11 regulates PDGFRβ signaling and promotes breast cancer progression. J Clin Invest. 2019;129:4609–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ragusa S, Prat-Luri B, Gonzalez-Loyola A, Nassiri S, Squadrito ML, Guichard A, et al. Antiangiogenic immunotherapy suppresses desmoplastic and chemoresistant intestinal tumors in mice. J Clin Invest. 2020;130:1199–216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Loessner D, Holzapfel BM, Clements JA. Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses. Adv Drug Deliv Rev. 2014;79–80:193–213.

    Article  PubMed  Google Scholar 

  11. Altorki NK, Markowitz GJ, Gao D, Port JL, Saxena A, Stiles B, et al. The lung microenvironment: an important regulator of tumour growth and metastasis. Nat Rev Cancer. 2019;19:9–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16:582–98.

    Article  CAS  PubMed  Google Scholar 

  13. Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G, De Maria R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer. 2019;18:70.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Biffi G, Tuveson DA. Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 2020;

  15. Tenchov R, Sasso JM, Wang X, Liaw WS, Chen CA, Zhou QA. Exosomes—nature’s lipid nanoparticles, a rising star in drug delivery and diagnostics. ACS Nano. 2022;16:17802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bie N, Yong T, Wei Z, Gan L, Yang X. Extracellular vesicles for improved tumor accumulation and penetration. Adv Drug Deliv Rev. 2022;188: 114450.

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  18. Morad G, Carman CV, Hagedorn EJ, Perlin JR, Zon LI, Mustafaoglu N, et al. Tumor-derived extracellular vesicles breach the intact blood-brain barrier via transcytosis. ACS Nano. 2019;13:13853–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Feng C, Xiong Z, Wang C, Xiao W, Xiao H, Xie K, et al. Folic acid-modified Exosome-PH20 enhances the efficiency of therapy via modulation of the tumor microenvironment and directly inhibits tumor cell metastasis. Bioact Mater. 2021;6:963–74.

    CAS  PubMed  Google Scholar 

  20. 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;10:1563–75.

    Article  CAS  PubMed  Google Scholar 

  21. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367.

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

    Article  PubMed  PubMed Central  Google Scholar 

  23. Hu D, Li Z, Zheng B, Lin X, Pan Y, Gong P, et al. Cancer-associated fibroblasts in breast cancer: challenges and opportunities. Cancer Commun (London). 2022;42:401–34.

    Article  Google Scholar 

  24. Najafi M, Farhood B, Mortezaee K. Cancer stem cells (CSCs) in cancer progression and therapy. J Cell Physiol. 2019;234:8381–95.

    Article  CAS  PubMed  Google Scholar 

  25. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 2020;5:8.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20:174–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Rimal R, Desai P, Daware R, Hosseinnejad A, Prakash J, Lammers T, et al. Cancer-associated fibroblasts: Origin, function, imaging, and therapeutic targeting. Adv Drug Deliv Rev. 2022;189: 114504.

    Article  CAS  PubMed  Google Scholar 

  29. Arina A, Idel C, Hyjek EM, Alegre ML, Wang Y, Bindokas VP, et al. Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci U S A. 2016;113:7551–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18:792–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee H-O, Mullins SR, Franco-Barraza J, Valianou M, Cukierman E, Cheng JD. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer. 2011;11.

  32. Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, et al. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3–CCL2 signaling. Cancer Res. 2016;76:4124–35.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  34. Kristian P, Jessica P, Gabriele B, Douglas H. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med. 2008;5:e19.

    Article  Google Scholar 

  35. Pelon F, Bourachot B, Kieffer Y, Magagna I, Mermet-Meillon F, Bonnet I, et al. Cancer-associated fibroblast heterogeneity in axillary lymph nodes drives metastases in breast cancer through complementary mechanisms. Nat Commun. 2020;11.

  36. Han C, Liu T, Yin R. Biomarkers for cancer-associated fibroblasts. Biomark Res. 2020;8:64.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017;214:579–96.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Wu SZ, Roden DL, Wang C, Holliday H, Harvey K, Cazet AS, et al. Stromal cell diversity associated with immune evasion in human triple‐negative breast cancer. The EMBO J. 2020;39.

  39. Affo S, Nair A, Brundu F, Ravichandra A, Bhattacharjee S, Matsuda M, et al. Promotion of cholangiocarcinoma growth by diverse cancer-associated fibroblast subpopulations. Cancer Cell. 2021;39:866-882.e11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Neuzillet C, Tijeras-Raballand A, Ragulan C, Cros J, Patil Y, Martinet M, et al. Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma. J Pathol. 2019;248:51–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33:463-479.e10.

    Article  CAS  PubMed  Google Scholar 

  42. Givel A-M, Kieffer Y, Scholer-Dahirel A, Sirven P, Cardon M, Pelon F, et al. miR200-regulated CXCL12β promotes fibroblast heterogeneity and immunosuppression in ovarian cancers. Nat Commun. 2018;9.

  43. Brechbuhl HM, Finlay-Schultz J, Yamamoto TM, Gillen AE, Cittelly DM, Tan AC, et al. Fibroblast subtypes regulate responsiveness of luminal breast cancer to estrogen. Clin Cancer Res. 2017;23:1710–21.

    Article  CAS  PubMed  Google Scholar 

  44. Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Su S, Chen J, Yao H, Liu J, Yu S, Lao L, et al. CD10(+)GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell. 2018;172(841–856): e16.

    Google Scholar 

  46. Lambrechts D, Wauters E, Boeckx B, Aibar S, Nittner D, Burton O, et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 2018;24:1277–89.

    Article  CAS  PubMed  Google Scholar 

  47. Bernard V, Semaan A, Huang J, San Lucas FA, Mulu FC, Stephens BM, et al. Single-cell transcriptomics of pancreatic cancer precursors demonstrates epithelial and microenvironmental heterogeneity as an early event in neoplastic progression. Clin Cancer Res. 2019;25:2194–205.

    Article  CAS  PubMed  Google Scholar 

  48. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, et al. IL1-induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 2019;9:282–301.

    Article  PubMed  Google Scholar 

  49. Li H, Courtois ET, Sengupta D, Tan Y, Chen KH, Goh JJL, et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat Genet. 2017;49:708–18.

    Article  CAS  PubMed  Google Scholar 

  50. Erdogan B, Ao M, White LM, Means AL, Brewer BM, Yang L, et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 2017;216:3799–816.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Labernadie A, Kato T, Brugués A, Serra-Picamal X, Derzsi S, Arwert E, et al. A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat Cell Biol. 2017;19:224–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Vaish U, Jain T, Are AC, Dudeja V. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma: an update on heterogeneity and therapeutic targeting. Int J Mol Sci. 2021;22.

  53. Alonso-Nocelo M, Ruiz-Canas L, Sancho P, Gorgulu K, Alcala S, Pedrero C, et al. Macrophages direct cancer cells through a LOXL2-mediated metastatic cascade in pancreatic ductal adenocarcinoma. Gut. 2023;72:345–59.

    Article  CAS  PubMed  Google Scholar 

  54. Calvo F, Ege N, Grande-Garcia A, Hooper S, Jenkins RP, Chaudhry SI, et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat Cell Biol. 2013;15:637–46.

    Article  CAS  PubMed  Google Scholar 

  55. Têtu B, Brisson J, Wang CS, Lapointe H, Beaudry G, Blanchette C, et al. The influence of MMP-14, TIMP-2 and MMP-2 expression on breast cancer prognosis. Breast Cancer Res. 2006;8.

  56. Stuelten CH, Byfield SD, Arany PR, Karpova TS, Stetler-Stevenson WG, Roberts AB. Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of TNF-α and TGF-β. J Cell Sci. 2005;118:2143–53.

    Article  CAS  PubMed  Google Scholar 

  57. Barrett RL, Pure E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. Elife. 2020;9.

  58. Fridman WH, Zitvogel L, Sautes-Fridman C, Kroemer G. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017;14:717–34.

    Article  CAS  PubMed  Google Scholar 

  59. Bruni D, Angell HK, Galon J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020;20:662–80.

    Article  CAS  PubMed  Google Scholar 

  60. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677–86.

    Article  CAS  PubMed  Google Scholar 

  61. Buechler MB, Fu W, Turley SJ. Fibroblast-macrophage reciprocal interactions in health, fibrosis, and cancer. Immunity. 2021;54:903–15.

    Article  CAS  PubMed  Google Scholar 

  62. Zhang R, Qi F, Zhao F, Li G, Shao S, Zhang X, et al. Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis. 2019;10.

  63. Comito G, Giannoni E, Segura CP, Barcellos-de-Souza P, Raspollini MR, Baroni G, et al. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 2013;33:2423–31.

    Article  PubMed  Google Scholar 

  64. Augsten M, Sjoberg E, Frings O, Vorrink SU, Frijhoff J, Olsson E, et al. Cancer-associated fibroblasts expressing CXCL14 rely upon NOS1-derived nitric oxide signaling for their tumor-supporting properties. Cancer Res. 2014;74:2999–3010.

    Article  CAS  PubMed  Google Scholar 

  65. Xiang H, Ramil CP, Hai J, Zhang C, Wang H, Watkins AA, et al. Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res. 2020;8:436–50.

    Article  CAS  PubMed  Google Scholar 

  66. Deng Y, Cheng J, Fu B, Liu W, Chen G, Zhang Q, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2016;36:1090–101.

    Article  PubMed  Google Scholar 

  67. Farhood B, Najafi M, Mortezaee K. CD8+ cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2018;234:8509–21.

    Article  PubMed  Google Scholar 

  68. Freeman P, Mielgo A. Cancer-associated fibroblast mediated inhibition of CD8+ cytotoxic T cell accumulation in tumours: mechanisms and therapeutic opportunities. Cancers. 2020;12:2687.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013;145:1121–32.

    Article  CAS  PubMed  Google Scholar 

  70. Goehrig D, Nigri J, Samain R, Wu Z, Cappello P, Gabiane G, et al. Stromal protein βig-h3 reprogrammes tumour microenvironment in pancreatic cancer. Gut. 2019;68:693–707.

    Article  CAS  PubMed  Google Scholar 

  71. Thomas DA, Massague J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8:369–80.

    Article  CAS  PubMed  Google Scholar 

  72. Lakins MA, Ghorani E, Munir H, Martins CP, Shields JD. Cancer-associated fibroblasts induce antigen-specific deletion of CD8+T Cells to protect tumour cells. Nat Commun. 2018;9.

  73. Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011;17:313–9.

    Article  CAS  PubMed  Google Scholar 

  74. Zhou B-BS, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discovery. 2009;8:806–23.

    Article  CAS  PubMed  Google Scholar 

  75. Chen W-J, Ho C-C, Chang Y-L, Chen H-Y, Lin C-A, Ling T-Y, et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat Commun. 2014;5.

  76. Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan C-L, et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun. 2018;9.

  77. Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt F, et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 2013;154:61–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de Jong JH, Borovski T, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010;12:468–76.

    Article  CAS  PubMed  Google Scholar 

  79. Giannoni E, Bianchini F, Masieri L, Serni S, Torre E, Calorini L, et al. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010;70:6945–56.

    Article  CAS  PubMed  Google Scholar 

  80. Ji Z, Tian W, Gao W, Zang R, Wang H, Yang G. Cancer-associated fibroblast-derived interleukin-8 promotes ovarian cancer cell stemness and malignancy through the notch3-mediated signaling. Front Cell Dev Biol. 2021;9.

  81. Tsuyada A, Chow A, Wu J, Somlo G, Chu P, Loera S, et al. CCL2 mediates cross-talk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells. Cancer Res. 2012;72:2768–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Tang Y-A, Chen Y, Bao Y, Mahara S, Yatim SMJM, Oguz G, et al. Hypoxic tumor microenvironment activates GLI2 via HIF-1α and TGF-β2 to promote chemoresistance in colorectal cancer. Proc Natl Acad Sci. 2018;115.

  83. Lotti F, Jarrar AM, Pai RK, Hitomi M, Lathia J, Mace A, et al. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A. J Exp Med. 2013;210:2851–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Garcia-Fernandez J, Fuente FM. Exosome-like systems: Nanotechnology to overcome challenges for targeted cancer therapies. Cancer Lett. 2023;561: 216151.

    Article  CAS  PubMed  Google Scholar 

  85. Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem. 2019;88:487–514.

    Article  CAS  PubMed  Google Scholar 

  86. Luo X, Li Y, Hua Z, Xue X, Wang X, Pang M, et al. Exosomes-mediated tumor metastasis through reshaping tumor microenvironment and distant niche. J Control Release. 2023;353:327–36.

    Article  CAS  PubMed  Google Scholar 

  87. 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;21:9–17.

    Article  CAS  PubMed  Google Scholar 

  88. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319:1244.

    Article  CAS  PubMed  Google Scholar 

  89. van Niel G, Charrin S, Simoes S, Romao M, Rochin L, Saftig P, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21:708–21.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  92. Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164:1226–32.

    Article  CAS  PubMed  Google Scholar 

  93. Matsumoto A, Takahashi Y, Nishikawa M, Sano K, Morishita M, Charoenviriyakul C, et al. Accelerated growth of B16BL6 tumor in mice through efficient uptake of their own exosomes by B16BL6 cells. Cancer Sci. 2017;108:1803–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. El Andaloussi S, Lakhal S, Mager I, Wood MJ. Exosomes for targeted siRNA delivery across biological barriers. Adv Drug Deliv Rev. 2013;65:391–7.

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35:2383–90.

    Article  CAS  PubMed  Google Scholar 

  97. Qu M, Lin Q, Huang L, Fu Y, Wang L, He S, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J Control Release. 2018;287:156–66.

    Article  CAS  PubMed  Google Scholar 

  98. Liu X, Lin P, Perrett I, Lin J, Liao Y-P, Chang CH, et al. Tumor-penetrating peptide enhances transcytosis of silicasome-based chemotherapy for pancreatic cancer. J Clin Invest. 2017;127:2007–18.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Jia G, Han Y, An Y, Ding Y, He C, Wang X, et al. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials. 2018;178:302–16.

    Article  CAS  PubMed  Google Scholar 

  100. Wei Z, Zhang X, Yong T, Bie N, Zhan G, Li X, et al. Boosting anti-PD-1 therapy with metformin-loaded macrophage-derived microparticles. Nat Commun. 2021;12:440.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Salunkhe S, Dheeraj, Basak M, Chitkara D, Mittal A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: strategies and significance. J Control Release. 2020;326:599–614.

    Article  CAS  PubMed  Google Scholar 

  102. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics. 2021;11:3183–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Bai J, Duan J, Liu R, Du Y, Luo Q, Cui Y, et al. Engineered targeting tLyp-1 exosomes as gene therapy vectors for efficient delivery of siRNA into lung cancer cells. Asian J Pharm Sci. 2020;15:461–71.

    Article  PubMed  Google Scholar 

  104. Shi X, Cheng Q, Hou T, Han M, Smbatyan G, Lang JE, et al. Genetically engineered cell-derived nanoparticles for targeted breast cancer immunotherapy. Mol Ther. 2020;28:536–47.

    Article  CAS  PubMed  Google Scholar 

  105. Liang G, Kan S, Zhu Y, Feng S, Feng W, Gao S. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int J Nanomed. 2018;13:585–99.

    Article  CAS  Google Scholar 

  106. Rountree RB, Mandl SJ, Nachtwey JM, Dalpozzo K, Do L, Lombardo JR, et al. Exosome targeting of tumor antigens expressed by cancer vaccines can improve antigen immunogenicity and therapeutic efficacy. Cancer Res. 2011;71:5235–44.

    Article  CAS  PubMed  Google Scholar 

  107. Kooijmans SA, Aleza CG, Roffler SR, van Solinge WW, Vader P, Schiffelers RM. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J Extracell Vesicles. 2016;5:31053.

    Article  PubMed  Google Scholar 

  108. Hong Y, Nam G-H, Koh E, Jeon S, Kim GB, Jeong C, et al. Exosome as a vehicle for delivery of membrane protein therapeutics, PH20, for enhanced tumor penetration and antitumor efficacy. Adv Funct Mater. 2018;28.

  109. Wang C, Wang H, Yang H, Xu C, Wang Q, Li Z, et al. Targeting cancer-associated fibroblasts with hydroxyethyl starch nanomedicine boosts cancer therapy. Nano Res. 2023;16:7323–36.

    Article  CAS  Google Scholar 

  110. Mondal J, Pillarisetti S, Junnuthula V, Saha M, Hwang SR, Park IK, et al. Hybrid exosomes, exosome-like nanovesicles and engineered exosomes for therapeutic applications. J Control Release. 2023;353:1127–49.

    Article  CAS  PubMed  Google Scholar 

  111. Kim MS, Haney MJ, Zhao Y, Yuan D, Deygen I, Klyachko NL, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomed Nanotechnol Biol Med. 2018;14:195.

    Article  CAS  Google Scholar 

  112. Yuan S, Mu W, Liu S, Liu M, Xia Z, Liang S, et al. Transforming cancer-associated fibroblast barrier into drug depots to boost chemo-immunotherapy in “shooting fish in a barrel” pattern. ACS Nano. 2023;17:13611.

    Article  CAS  PubMed  Google Scholar 

  113. Guo L, Dandan S, Dong M, Mengmeng S, Xiao S, Xiaoying Z, et al. New FH peptide-modified ultrasonic nanobubbles for delivery of doxorubicin to cancer-associated fibroblasts. Nanomedicine. 2019;14:2957.

    Article  CAS  PubMed  Google Scholar 

  114. Wolfers J, Lozier A, Raposo G, Regnault A, Théry C, Masurier C, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7:297.

    Article  CAS  PubMed  Google Scholar 

  115. Hong Y, Kim YK, Kim GB, Nam G-H, Kim SA, Park Y, et al. Degradation of tumour stromal hyaluronan by small extracellular vesicle-PH20 stimulates CD103+ dendritic cells and in combination with PD-L1 blockade boosts anti-tumour immunity. J Extracell Vesicles. 2019;8:1670893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  117. Rana S, Malinowska K, Zöller M. Exosomal tumor microRNA modulates premetastatic organ cells. Neoplasia. 2013;15:281–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Li G, Yi X, Du S, Gong L, Wu Q, Cai J, et al. Tumour-derived exosomal piR-25783 promotes omental metastasis of ovarian carcinoma by inducing the fibroblast to myofibroblast transition. Oncogene. 2023;42:421–33.

    Article  CAS  PubMed  Google Scholar 

  119. Zhao L, Gu C, Gan Y, Shao L, Chen H, Zhu H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J Control Release. 2020;318:1–15.

    Article  CAS  PubMed  Google Scholar 

  120. Yue S, Ye X, Zhou T, Gan D, Qian H, Fang W, et al. PGRN(-/-) TAMs-derived exosomes inhibit breast cancer cell invasion and migration and its mechanism exploration. Life Sci. 2021;264: 118687.

    Article  CAS  PubMed  Google Scholar 

  121. Srivastava A, Rathore S, Munshi A, Ramesh R. Organically derived exosomes as carriers of anticancer drugs and imaging agents for cancer treatment. Semin Cancer Biol. 2022;86:80–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Jing B, Gai Y, Qian R, Liu Z, Zhu Z, Gao Y, et al. Hydrophobic insertion-based engineering of tumor cell-derived exosomes for SPECT/NIRF imaging of colon cancer. J Nanobiotechnology. 2021;19:7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Hikita T, Miyata M, Watanabe R, Oneyama C. In vivo imaging of long-term accumulation of cancer-derived exosomes using a BRET-based reporter. Sci Rep. 2020;10:16616.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Bose RJC, Uday Kumar S, Zeng Y, Afjei R, Robinson E, Lau K, et al. Tumor cell-derived extracellular vesicle-coated nanocarriers: an efficient theranostic platform for the cancer-specific delivery of anti-miR-21 and imaging agents. ACS Nano. 2018;12:10817–32.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Riley RS, Day ES. Gold nanoparticle-mediated photothermal therapy: applications and opportunities for multimodal cancer treatment. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017;9.

  126. Xu Z, Zeng S, Gong Z, Yan Y. Exosome-based immunotherapy: a promising approach for cancer treatment. Mol Cancer. 2020;19:160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Thakur A, Parra DC, Motallebnejad P, Brocchi M, Chen HJ. Exosomes: Small vesicles with big roles in cancer, vaccine development, and therapeutics. Bioact Mater. 2022;10:281–94.

    CAS  PubMed  Google Scholar 

  128. Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev. 2016;97:186–203.

    Article  CAS  PubMed  Google Scholar 

  129. Kim HJ, Kim A, Miyata K, Kataoka K. Recent progress in development of siRNA delivery vehicles for cancer therapy. Adv Drug Deliv Rev. 2016;104:61–77.

    Article  CAS  PubMed  Google Scholar 

  130. Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A. 2013;110:20212–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Abello J, Nguyen TDT, Marasini R, Aryal S, Weiss ML. Biodistribution of gadolinium- and near infrared-labeled human umbilical cord mesenchymal stromal cell-derived exosomes in tumor bearing mice. Theranostics. 2019;9:2325–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, et al. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small. 2020;16: e1903916.

    Article  PubMed  Google Scholar 

  133. Sun Z, Yang J, Li H, Wang C, Fletcher C, Li J, et al. Progress in the research of nanomaterial-based exosome bioanalysis and exosome-based nanomaterials tumor therapy. Biomaterials. 2021;274: 120873.

    Article  CAS  PubMed  Google Scholar 

  134. Yang F, Ning Z, Ma L, Liu W, Shao C, Shu Y, et al. Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Mol Cancer. 2017;16.

Download references

Acknowledgements

A special thanks for the long-term subsidy mechanism from the Ministry of Finance and the Ministry of Education of PRC (People’s Republic of China) for BUCM (Beijing University of Chinese Medicine).

Funding

This work was supported by the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (No. ZYYCXTD-D-202005) and the Youth Qihuang Scholar of National Administration of Traditional Chinese Medicine (2020).

Author information

Authors and Affiliations

Authors

Contributions

XX and YL wrote and conceived the manuscript. XW, MP, LY, JQ, XL and MT collected literature. YL, CX, CL and XX revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Cheng Lu, Cheng Xiao or Yuanyan Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that there are no potential competing interests.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xue, X., Wang, X., Pang, M. et al. An exosomal strategy for targeting cancer-associated fibroblasts mediated tumors desmoplastic microenvironments. J Nanobiotechnol 22, 196 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s12951-024-02452-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12951-024-02452-1

Keywords