Skip to main content

Regulation of cancer-associated fibroblasts for enhanced cancer immunotherapy using advanced functional nanomedicines: an updated review

A Correction to this article was published on 04 April 2025

This article has been updated

Abstract

The tumor microenvironment (TME) is a complex and dynamic ecosystem that plays a critical role in cancer progression. It comprises various cell types, including immune cells, tumor cells, and stromal cells. Among these, cancer-associated fibroblasts (CAFs) represent a heterogeneous population with diverse origins, phenotypes, and functions. Activated CAFs secrete multiple factors that promote tumor growth, migration, angiogenesis, and contribute to chemoresistance. Additionally, CAFs secrete extracellular matrix (ECM) components, such as collagen, which form a physical barrier that hinders the penetration of chemotherapeutic and immunotherapeutic agents. This ECM also influences immune cell infiltration, impeding their ability to effectively target tumor cells. As a result, modulating the activity of CAFs has emerged as a promising strategy to enhance the efficacy of tumor immunotherapy. Nano-delivery systems, constructed from various nanomaterials with high targeting specificity and biocompatibility, offer a compelling approach to deliver therapeutic agents or immunomodulatory factors directly to CAFs. This modulation can alter CAF function, reduce their tumor-promoting effects, and thereby improve the outcomes of immunotherapy. This review provides an in-depth exploration of the origins, functions, and interactions of CAFs within the TME, particularly in the context of immune suppression. Furthermore, it discusses the potential applications of functional nanocarrifers in modulating CAFs and enhancing the effectiveness of tumor immunotherapy, highlighting the significant progress and potential of nanotechnology in this area.

Graphical Abstract

Introduction

The global burden of cancer continues to rise, posing a formidable public health challenge worldwide. Despite significant advancements in the understanding of cancer biology, it is increasingly clear that cancer is not solely the result of genetic mutations in tumor cells, but rather arises within a complex and dynamic tumor microenvironment (TME) [1]. The TME comprises a heterogeneous mixture of cellular and non-cellular components, including tumor cells, immune cells, cancer-associated fibroblasts (CAFs), extracellular matrix (ECM) elements, and soluble factors such as growth factors and cytokines [2]. These components engage in intricate and reciprocal interactions that play crucial roles in regulating tumor growth, metastasis, and the tumor’s response to therapeutic interventions.

Among the key cellular players within the TME, CAFs are a highly heterogeneous population of stromal cells that significantly contribute to tumor progression. CAFs are derived from normal fibroblasts but undergo phenotypic and functional transformations in response to tumor-derived signals, including cytokines, growth factors, and ECM remodeling enzymes [3]. These activated fibroblasts reside within the TME, where they facilitate tumor progression through multiple mechanisms. CAFs promote tumor growth and metastasis by secreting immune-suppressive factors (e.g., IL-6, TGF-β, CXCL12) and by altering the physical properties of the tumor stroma [4,5,6]. They also interact with various immune cells, including macrophages, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), thereby contributing to an immunosuppressive microenvironment [3, 6]. These interactions hinder the activity of effector T cells, facilitating the tumor’s evasion of host immune surveillance [7, 8]. Given their pivotal role in shaping the immunosuppressive TME, CAFs have emerged as an important therapeutic target in cancer immunotherapy.

Current strategies to modulate CAFs primarily focus on both direct and indirect approaches to alter their activation, function, and interactions with the TME [9,10,11]. These approaches include targeting key signaling pathways such as TGF-β [12, 13], Hedgehog [14], and Wnt/β-catenin [15], which are known to regulate CAF activation and their subsequent contributions to tumor progression. Additionally, immunomodulatory strategies—such as modulation of immune checkpoint molecules and the use of CAF-targeting antibodies—aim to enhance anti-tumor immunity by disrupting the immunosuppressive microenvironment created by CAFs [16,17,18,19]. Moreover, the application of nanomaterials offers significant promise in enhancing the solubility, stability, and sustained release of therapeutic agents, while also enabling targeted delivery to CAFs and other components of the TME [20,21,22]. Nanomaterial-based drug delivery systems hold the potential to selectively target CAFs, modulate their activity within the TME, improve drug penetration, facilitate immune cell infiltration, and counteract the immunosuppressive effects exerted by CAFs [21, 23,24,25]. Although targeting CAFs has demonstrated promise in tumor therapy, several limitations persist, particularly the heterogeneity of CAFs and the complexity of the TME [26]. These challenges highlight the necessity for a more comprehensive understanding of the intricate interactions between CAFs and their surrounding environment. A deeper elucidation of these interactions is essential for the development of more effective and targeted therapeutic strategies.

In this review, we explore the complex interactions between CAFs and immune cells within the TME, with a focus on the role of CAFs in shaping the tumor immune microenvironment through various metabolic and signaling pathways. We also discuss the potential of functional nanocarriers to modulate CAF activity, enhance anti-tumor immune responses, and optimize cancer treatment outcomes by overcoming the structural and immunosuppressive barriers imposed by the TME. Strategies aimed at inhibiting CAF activation, interfering with CAF functions, and selectively depleting or killing CAFs are examined in detail. However, despite the promising therapeutic potential, these approaches face several challenges that need to be addressed to improve their clinical efficacy.

CAFs’ origin and heterogeneity

CAFs are a diverse and dynamic population of cells that contribute significantly to the TME. The origin and heterogeneity of CAFs have been extensively studied, as these cells play key roles in tumor progression, metastasis, and therapy resistance (Fig. 1). CAFs are derived from multiple sources, including resident fibroblasts, endothelial cells, perivascular cells, and bone marrow-derived mesenchymal stem cells MSCs [3]. Upon exposure to tumor-derived signals such as growth factors (e.g., TGF-β, PDGF) and cytokines (e.g., IL-6), these precursor cells undergo activation and reprogramming, acquiring a myofibroblast-like phenotype characterized by the expression of markers such as α-smooth muscle actin (α-SMA) and fibroblast activation protein (FAP) [5, 27, 28].

Fig. 1
figure 1

The origins of CAFs and their multifaceted roles in tumor development and therapy. CAFs originate from various cell types, including adipocytes, endothelial cells, chondrocytes, myoblast cells, stellate cells, fibroblasts, and mesenchymal stem cells. Upon activation by tumor-derived signals, CAFs undergo phenotypic and functional changes that enable them to contribute to various aspects of tumor progression

The heterogeneity of CAFs is a defining feature that complicates their study and therapeutic targeting. CAFs are not a uniform population, but rather consist of distinct subtypes with diverse molecular profiles and functional roles. Recent research has identified at least two major subtypes of CAFs based on their molecular signatures: the myofibroblastic CAFs (myCAFs), which are involved in ECM remodeling and tissue stiffness, and the inflammatory CAFs (iCAFs), which produce pro-inflammatory cytokines such as IL-6 and IL-8, contributing to immune modulation and tumor progression [29,30,31]. Additionally, antigen-presenting CAFs (apCAFs) have been shown to influence immune cell infiltration and anti-tumor immunity [32]. These subpopulations of CAFs exhibit different capacities to modulate tumor behavior, with some facilitating tumor growth and metastasis, while others may suppress certain aspects of malignancy. The functional diversity and plasticity of CAFs are shaped by a combination of intrinsic factors, such as genetic mutations, and extrinsic signals from the TME, including hypoxia, altered metabolism, and tumor-derived exosomes [33]. This remarkable adaptability allows CAFs to support various stages of tumor progression, including EMT, angiogenesis, and immune evasion [34].

Overall, the origin and heterogeneity of CAFs are fundamental to their diverse roles in cancer biology. The identification of distinct CAF subpopulations and their specific contributions to the TME underscores the complexity of their functions. A better understanding of the molecular mechanisms driving CAF differentiation and plasticity will be crucial for developing targeted therapies aimed at modulating CAF activity in cancer treatment [2].

Role of CAFs in the TME

CAFs secrete factors like TGF-β, IL-6, VEGF, and PDGF, which drive tumor progression by enhancing angiogenesis and creating a pro-tumorigenic microenvironment that supports tumor cell survival and migration [34]. CAFs also play a critical role in therapeutic resistance, activating signaling pathways such as STAT3 and NF-κB, which help tumor cells evade chemotherapy and radiotherapy [35, 36]. Furthermore, CAFs influence immune responses by recruiting and activating immune suppressive cells, including Tregs and MDSCs, thereby impairing the efficacy of immunotherapies [6]. They also remodel the extracellular matrix, promoting tumor invasion and further inhibiting immune cell infiltration, which contributes to immune evasion [3]. CAF-derived exosomes, which carry bioactive molecules, facilitate communication between CAFs and other TME cells, enhancing tumor progression and metastasis [37, 38]. Thus, CAFs play a multifaceted role in shaping the TME, promoting tumor growth, and limiting the effectiveness of both immune and conventional therapies. Given the substantial impact of CAFs on tumor biology, their role in the TME represents an area of intense research, with ongoing efforts aimed at understanding their functional diversity and developing therapeutic approaches to modulate their activity in the context of cancer (Fig. 2).

Fig. 2
figure 2

The Role of CAFs in the TME.CAFs support tumor growth, migration, angiogenesis, drug resistance, and immune suppression by secreting factors like TGF-β, IL-6, VEGF, and PDGF. They enhance angiogenesis, promote drug resistance via STAT3 and NF-κB, and recruit immune cells such as Tregs and MDSCs. CAFs also remodel the extracellular matrix, affecting immune cell infiltration, while CAF-derived exosomes further impact tumor progression and immunity

Promote tumor proliferation and migration

One of the major components of the TME, CAFs contribute significantly to tumor progression by promoting cell proliferation, migration, and metastasis. This is primarily achieved through the secretion of various bioactive factors, including transforming growth factor-beta (TGF-β), interleukins (IL-2, IL-6), C-X-C motif chemokine ligands (CXCL2, CXCL5, CXCL12, CXCL16), fibroblast growth factor 7 (FGF7), and hypoxia-inducible factor (HIF1A/HIF-1α) [3].For example, TGF-β induces CAF activation, thereby promoting tumor fibroplasia and facilitating tumor progression through its autocrine and paracrine effects. Studies have implicated the TGF-β pathway in the promotion of prostate cancer cell proliferation and migration [39], while overexpression of TGF-β1 and SDF-1α in cervical cancer enhances cell growth and invasiveness [40]. CAFs also modulate the Wnt/β-catenin and TGF-β/SMAD pathways, influencing AKT and MAPK signaling through the secretion of VCAM1, thereby impacting tumor growth and invasion [41, 42].Under hypoxic conditions, CAFs regulate HIF-1α-dependent glycolytic signaling, affecting ovarian cancer cell invasion and migration by modulating CRMP2 [43]. Additionally, FGF7 secreted by CAFs influences HIF-1α signaling and epithelial-mesenchymal transition EMT [44]. CAFs contribute to ECM production by secreting CXCL16 and enhancing MMP expression, which increases tumor stiffness and alters ECM structure [45].The binding of PF4/CXCL4 and PPBP/CXCL7 to CXCL12, mediated by G protein-coupled receptors, promotes EMT and the expression of chemokines and cytokines [46]. In hepatocellular carcinoma (HCC), versican (VCAN) secreted by CAFs is associated with malignant transformation and prognosis [47]. SERPINE1/PAI1 from CAFs induces the transformation of lymphatic endothelial cells into mesenchymal cells in cervical squamous cell carcinoma, facilitating cancer metastasis [48].Furthermore, CAFs secrete exosomes that actively participate in cancer cell proliferation and invasion. These exosomes contain specific miRNAs, such as miR-20a-5p and miR-421, which promote IL-6 production and pancrea1ic cancer progression, respectively, by regulating the SIRT3/H3K9ac/HIF-1α axis [37, 49].The cargo of CAF-derived exosomes is crucial for their effectiveness in cancer progression.

In summary, CAFs play multifaceted roles in promoting tumor growth, invasion, and metastasis through complex signaling networks, ECM remodeling, and exosome-mediated communication, making them critical players in cancer biology and potential targets for therapeutic interventions.

Promote tumor angiogenesis

CAFs possess the ability to secrete a variety of pro-angiogenic factors, including vascular endothelial growth factors (VEGFs), PDGFs, and Wnt family member 2 (WNT2). These factors promote endothelial cell proliferation by binding to their corresponding receptors and activating the corresponding signaling pathways, which in turn promote angiogenesis and tumor development in vivo [50]. For example, VEGF, a potent angiogenic and vascular permeability factor, can significantly affect the ECM when secreted by CAFs, promoting an increase in vascular volume and permeability, which in turn contributes to enhanced tumor metastasis [51]. In addition, PDGFs secreted by CAFs can further promote angiogenesis by influencing matrix activation [52]. In CRC, reducing WNT2 expression in CAFs significantly reduced angiogenesis, whereas overexpressing WNT2 increased vessel density and tumor volume [53]. Moreover, CAFs affect MMP degradation function by downregulating SRY-box transcription factor 4 (SOX4), further affecting angiogenesis and tumor metastasis [54]. Furthermore, CAFs can infiltrate the TME by attracting immune cells such as TAMs [55]. For example, in head and neck squamous cell carcinoma, CAFs regulate the YAP1/HIF-1α axis by delivering miR-21-5p to endothelial cells, thereby promoting tumor angiogenesis [38]

Increasing tumor drug resistance

CAFs contribute to tumor drug resistance through complex mechanisms, primarily by modulating key signaling pathways. One such pathway is the signal transducer and activator of transcription 3 (STAT3), which is often hyperactivated in cancer. This can occur via cytokine signaling (e.g., IL-6) or interactions with other pathways like nuclear factor kappa B (NF-κB) [56]. CAFs secrete IL-6, which activates STAT3 and reduces the efficacy of chemotherapies like gemcitabine [35]. Additionally, CAF-derived exosomal miR-21 enhances STAT3 activation and induces MDSC production [57], promoting cisplatin resistance in esophageal squamous cell carcinoma [58]. CAFs also influence tumor drug resistance through exosome-mediated signaling. In colorectal cancer (CRC), CAF-secreted miR-625-3p via exosomes enhances the CELF2/WWOX pathway, promoting tumor growth and chemoresistance [59]. In pancreatic cancer, CAFs release circFARP1, which activates STAT3 and augments resistance to gemcitabine [60]. Conversely, circZFR in HCC inhibits the STAT3/NF-κB pathway, promoting cisplatin resistance. CAFs further contribute to tumor resistance by secreting extracellular matrix (ECM components like HA and collagenase), which alters drug diffusion within the TME [61]. Metabolic reprogramming also plays a role, as lactic acid secreted by cancer cells transforms stromal fibroblasts into CAFs [36]. This transformation, coupled with NF-κB activation, upregulates brain-derived neurotrophic factor (BDNF) expression, which affects gastric cancer cell sensitivity to anlotinib through the NTRK2/NFE2L2 pathway [62].

These intricate mechanisms highlight the critical role of CAFs in modulating tumor progression and therapeutic resistance, providing essential insights for developing novel therapies targeting the TME.

Modulation of tumor immunity

Immune cells play a key role in the TME, including innate immune cells (e.g., macrophages, neutrophils, mast cells, MDSCs, dendritic cells (DCs), and natural killer [NK] cells) and adaptive immune cells (e.g., T and B lymphocytes). These cells play an important role in controlling and shaping tumor development [63]. CAFs interfere with T cell responses and are resistant to immunotherapy by secreting various immunosuppressive factors (e.g., IL-6, IL-10, C-X-C motif chemokine ligand 9 [CXCL9], and TGF-β) and recruiting suppressor immune cells (e.g., MDSCs and Tregs). In addition, the dense collagenous matrix produced by CAFs also affects immune cell infiltration[64, 65]. Therefore, targeting CAFs has become a new direction in anticancer immunotherapy [66, 67]. Two main strategies are currently used in cancer immunotherapy: (1) activating tumor antigens using cancer vaccines to stimulate immune cell responses to tumors and (2) enhancing endogenous antitumor activity by blocking immune checkpoint receptors, a strategy that involves targeting lymphocytes and their ligands [21].

Interaction between CAFs and immune cells

T cells

T cells, including CD4+and CD8+ subsets, are essential components of the adaptive immune system and play a pivotal role in defending against pathogens and tumors. However, CAFs profoundly influence T cell function and immune responses [69]. CAFs alter the physical and chemical properties of the stromal ECM through the secretion of collagen, hyaluronan (HA), and other factors, which impair T cell infiltration and tumor treatment efficacy. The deletion of type I collagen in myofibroblasts leads to increased CXCL5 production in tumor cells, promoting MDSC aggregation, reducing T and B cell populations, and accelerating cancer progression [66]. In prostate cancer, the expression of forkhead box F2 (FOXF2) in the stroma correlates with CXCL5 levels, and reduced CXCL5 enhances FOXF2 expression, leading to increased T cell infiltration [67]. Furthermore, the density of CAFs impacts the localization and abundance of CD8+T cells within tumors [70]. YAP1, a transcriptional coactivator secreted by myCAFs, inhibits CD8+ T cell infiltration, and its downregulation in HCC enhances CD8+T cell presence by reducing PD-L1 expression [68]. CAFs also secrete various chemokines, such as CXCL9, CXCL10, CXCL12, IL-6, and IL-8, to modulate T cell function. IL-6 promotes PD-L1 expression via the STAT3/AKT pathway, inducing T cell apoptosis and impairing immune responses [71, 72]. Similarly, IL-8 upregulates PD-L1 through NF-κB signaling, further inhibiting CD8+T cell activity [73].

Among CAF subtypes, mesothelial cell-derived CAFs (apCAFs) exhibit immune-regulatory functions and can induce CD4+T cell conversion to regulatory T cells (Tregs) under IL-10 and TGF-β influence [32]. Additionally, CAFs with enhanced metabolic activity (meCAFs), marked by PLA2G2A, regulate CD8+T cell function via the MAPK/ERK and NF-κB pathways, serving as prognostic markers for aggressive cancers [74]. TGF-β, a potent immune regulator secreted by CAFs, upregulates PD-L1 and Foxp3 expression, promoting Treg differentiation and inhibiting antigen-specific CD4+T cell expansion, thus impeding immune rejection and affecting chemo-immunological responses [75, 76]. In smmary, CAFs play a critical role in modulating T cell activity within the TME through various signaling pathways and secreted factors, influencing both tumor progression and immunotherapy outcomes.

NK cells

NK cells are also innate immune effector cells that can rapidly recognize and destroy abnormal and virus-infected cells in the body [77]. They are characterized by their ability to distinguish between normal and pathological cells and to recognize receptors by expressing multiple cell surface proteins [77, 78]. However, the immune function of NK cells is significantly affected by TGF-β, which, for example, leads to a decrease in the number of CD16-expressing NK cells in bladder cancer [79]. In human papillomavirus infections, NK receptor cell expression can be affected by influencing NK cell activation receptors such as natural cytotoxicity triggering receptors 3 (NCR3/NKp30) and 1 (NKp46) [80]. In addition, inhibiting TGF-β in NK cells was validated in a CRC model [81]. IL-6 and IL-8 secreted by CAFs decreased granzyme B (GZMB) in NK-92 cells [82]. Gene sequencing has identified a subset of senescent myofibroblasts (myCAFs) in breast cancer. These senescent myCAFs can affect NK cell function and foster tumor growth by secreting extracellular matrix ECM [83]. In addition, CAFs promote breast cancer bone metastasis by inhibiting NK cell activation and function through dickkopf-related protein 1 (DKK1) [84]. In gastric cancer, CAFs also lead to intracellular iron overload, triggering iron death in NK cells, and inhibiting this condition helped to enhance the efficacy of chimeric antigen receptor (CAR)-NK therapy [85]. CAR-NKcells can be designed based on molecules specifically expressed on CAFs to enhance the efficacy of cancer immunotherapy. For example, the specific expression of CD70 molecule (CD70) on CAFs can serve as a target for CAR-NK cells, with the CD70-CAR-NK cell interaction enhanced by IL-15 stimulation [86]. In addition, inhibiting IL-6 secreted by CAFs promotes enhanced CAR-NK cell function [87].

TAMs

As an important component of the TME, like other immune cells, TAMs interact with CAFs, which are critical for cancer progression and metastasis. Macrophages are usually classified into M1 and M2 types. The M1 type exhibits antitumor properties, eliminating tumor cells via phagocytosis and antibody-dependent cell-mediated cytotoxicity. In contrast, the M2 type has pro-tumorigenic effects, significantly impeding T-cell-mediated antitumor responses [88]. Single-cell sequencing has shown that various factors influence the gene expression profile of TAMs. For example, modulating SMAD3 reduced the potential for progenitor CAF production from macrophages [89]. The recruitment and polarization of TAMs are typically influenced by multiple factors secreted by CAFs, such as IL-8, IL-6, colony-stimulating factor 2 (CSF2/GM-CSF), and CXCL2 [90, 91]. CAFs promote tumor invasion and metastasis by promoting the release of FGF2 by secreting insulin-like growth factor-binding protein 7 (IGFBP7), which in turn affects the polarization of TAMs towards the M2 type and their infiltration into tumor tissue [92].

Lactate-activated CAFs can influence TAM recruitment by secreting IL-8 and contribute to TAM polarization toward the M2 type [93]. Additionally, CAFs draw monocytes through the CXCL12/C-X-C motif chemokine receptor 4 (CXCR4) signaling pathway and prompt their differentiation into M2-type macrophages, further promoting the formation of M2-type TAMs in oral squamous cell carcinoma [94]. One study examining the interaction between CAFs and TAMs found that G protein-coupled receptor 30 (GPR30) promoted macrophage recruitment and infiltration by upregulating CXCL12 and polarized more TAMs toward the M2 type with high IL-10 and low IL-12 expression [95]. Osteobridging protein (OPN), an intracellularly secreted chemokine-like phosphorylated glycoprotein, is involved in the interaction between CAFs and TAMs. Reducing OPN secretion from TAMs decreased OPN secretion by CAFs, and inhibiting OPN suppressed the proliferation, invasion, and migration of cancer cells induced by TAM-derived CAFs [96]. In addition, TAMs with activated Notch signaling expressed higher levels of immunosuppressive mediators, further enhancing their immunosuppressive function in the TME [97].

CAFs in energy metabolism-driven immune evasion of tumor cells

CAFs play a pivotal role in the modulation of TME and contribute to immune evasion mechanisms by altering cellular energy metabolism [98, 99]. Recent studies have highlighted that CAFs not only support tumor growth through secretion of growth factors and extracellular matrix remodeling but also significantly influence immune responses within the TME by manipulating metabolic pathways [100]. Tumor cells often adapt to a hypoxic, nutrient-deprived environment, reprogramming their energy metabolism to favor glycolysis and other anabolic processes, a phenomenon known as the Warburg effect [101]. CAFs interact with tumor cells through metabolic crosstalk, including the transfer of metabolites such as lactate, glutamine, and pyruvate, which not only sustain the energetic needs of tumor cells but also modulate the local immune landscape [102]. By secreting factors like IL-6, TGF-β, and CXCL12, CAFs create an immunosuppressive microenvironment, facilitating the recruitment and activation of immune cells such as Tregs and MDSCs, while inhibiting the function of CTLs and NK cells [101]. Moreover, CAFs contribute to metabolic reprogramming of immune cells, further enhancing immune escape by promoting the suppression of immune surveillance and effector functions [103, 104]. The metabolic cooperation between CAFs and tumor cells thus represents a critical mechanism through which tumors evade immune detection and destruction, providing novel insights for therapeutic strategies targeting the CAF-mediated metabolic reprogramming in cancer immunotherapy.

Nanomaterials for targeting CAFs in cancer therapy

Increasing evidence underscores the pivotal role of CAFs in inducing immunosuppression within the TME, thus driving the development of CAF-targeted nanomaterials (Fig. 3). The TME is characterized by altered tissue architecture, a dense ECM, elevated interstitial fluid pressure, and hypoxia, all of which influence CAF behavior and the efficacy of nanomedicine delivery [105]. Moreover, tumor tissue stiffness, often resulting from ECM remodeling by CAFs, can impact signaling pathways and modulate the responsiveness of both CAFs and tumor cells to therapeutic interventions. Therefore, a comprehensive understanding of tumor physical properties, in combination with CAF-targeted strategies, is essential for optimizing the design of nanomedicines and enhancing therapeutic outcomes. Modulating tumor characteristics such as ECM stiffness or normalizing the vasculature may further improve the efficacy of CAF-targeted therapies [106].

Fig. 3
figure 3

Functional nanocarriers for regulating CAFs and enhancing immunotherapy applications. By leveraging their unique physical and chemical properties, such as size, surface charge, and functionalization with targeting ligands, these nanocarriers can specifically interact with CAFs in the tumor microenvironment. This targeted approach not only helps in modulating CAF activity to overcome immunosuppressive barriers but also improves the delivery and efficacy of therapeutic agents

In this section, we review various nanomaterials designed to modulate the TME for targeting CAFs and enhancing cancer treatment [107]. However, several challenges persist in utilizing nanomaterials for CAF modulation. These include difficulties in distinguishing between normal fibroblasts and CAFs, potential off-target effects, and issues with poor circulation stability, biocompatibility, and rapid clearance. To overcome these limitations, nanomaterials must be engineered with careful consideration of their physical properties (e.g., size, shape) and chemical properties (e.g., composition, surface chemistry) to enhance targeting specificity, stability, and biocompatibility. This section will explore the properties, advantages, and limitations of various nanomaterials that aim to improve cancer immunotherapy by regulating CAFs. Table 1 summarizes key nanomaterials that enhance immunotherapy outcomes through CAF targeting.

Table 1 Applications of nanomaterials in targeting CAFs to enhance immunotherapy

Organic nanomaterials

Organic nanomaterials offer significant advantages in cancer immunotherapy due to their unique physicochemical properties and tunable compositions, which make them powerful tools for enhancing the efficacy of immunotherapeutic approaches. Common examples of organic nanomaterials include liposomes composed of phospholipid bilayers, NPs made from synthetic or natural polymers, microsphere emulsions formed by crosslinked polymers, and polymeric microspheres [19, 134]. These materials not only enable targeted drug delivery within the TME, but also modulate immune responses, thereby improving the therapeutic outcomes of immunotherapy.

The physicochemical characteristics of organic nanomaterials, such as adjustable particle size, surface properties, and structure, provide precise control over drug delivery and release. By encapsulating anticancer drugs or immune modulators within these nanocarriers, they can effectively overcome biological barriers and deliver drugs directly to the tumor site. This approach can inhibit the activity of CAFs or reduce the secretion of immune-suppressive factors by CAFs, further enhancing therapeutic efficacy. A thorough understanding of these properties is crucial for designing more precise and effective therapeutic strategies.

Polymer micelles

Micelles are nanoscale colloidal particles formed through the self-assembly of amphiphilic polymers. In solution, polymer concentration plays a crucial role in micelle formation[135, 136]. Below the critical micelle concentration (CMC), polymers exist as individual molecules, whereas above the CMC, they aggregate into micelles, which have a hydrophobic core and a hydrophilic shell. Common hydrophilic components of polymeric micelles include polyethylene glycol (PEG) and polyvinyl alcohol (PVA), while the hydrophobic components are often made from materials such as polylactic acid (PLA), polycaprolactone (PCL), polyether, or polylactic-glycolic acid copolymer (PLGA) [137, 138]. These materials have found widespread application in nanoplatforms designed to modulate CAFs and enhance immunotherapy. For example, Myrofora et al. used PEG-b-poly(benzyl-L-glutamate) (PEG-PBLG) block copolymers to prepare polymeric micelles loaded with Tranilast [108]. These micelles had an average size of approximately 95 nm and demonstrated high stability, retaining their original size even after prolonged storage, with no precipitation observed in solution. Similarly, in a study by Feng, PEG-PLA was used to target CAFs [137].

To improve the affinity of polymeric micelles for CAFs and enhance the effectiveness of targeted tumor therapies, several strategies have been employed. One such strategy involves incorporating CREKA peptides, which specifically bind to fibronectin overexpressed on CAFs [137], or peptides targeting FAP-α, such as Gly-Pro-Ala-Cys and Ac-RQRQGPA-OH [111, 138]. In addition, Cheng et al. demonstrated that polymeric micelles could non-covalently bind to antibody fragments for CAF targeting [139]. Furthermore, polymeric micelles can also complex with siRNA via electrostatic interactions, thereby facilitating CAF uptake. For example, in the work of Paul et al. a triblock polymeric micelle was designed, with poly(sarcosine) (pSar) as the first block to provide stealth characteristics and avoid rapid blood clearance and immune recognition, poly(γ-benzyl-L-glutamate) (pGlu(OBn)) as the second block to stabilize the micelle structure, and poly(L-lysine) (pLys) as the third block, which complexes with siRNA through electrostatic interactions [140].

With the continuous progress in nanotechnology, an increasing number of multifunctional polymeric micelles have been developed to respond to changes in the TME, such as variations in temperature, pH, redox state, and enzyme activity. This enables precise drug release at the tumor site [141]. Hyaluronic acid (HA), a natural high-molecular-weight polysaccharide known for its excellent biocompatibility and biodegradability, is also a promising material for synthetic polymeric micelles. For instance, in the research of Liang et al., HA was deacetylated to form a zwitterionic polysaccharide, which was then hydrophobically modified by grafting dodecylamine to create a pH-sensitive zwitterionic polymer [138]. In this case, the hydrophobic modification with dodecylamine enhanced the hydrophobicity of the micelles, thereby promoting the formation of a stable micelle structure.

Liposomes

Liposomes are vesicular structures composed of one or more phospholipid bilayers, characterized by a hydrophilic interior and a hydrophobic exterior [142, 143]. These properties enable liposomes to effectively encapsulate both hydrophilic and hydrophobic drugs [144, 145]. Common materials used in liposome construction include Phosphatidylcholine, Cholesterol, and Egg yolk lecithin. These components play a critical role in drug delivery systems by contributing to the formation of stable NPs and enhancing drug bioavailability and targeting capabilities [25, 144]. The therapeutic efficacy of liposomes is closely related to their internal stability, which is influenced by several factors such as particle size, charge, the number of membrane layers, the presence of targeting ligands, and the specific materials used in their construction [145, 146].

Liposome preparation methods are relatively straightforward, with common techniques including the film hydration and ethanol injection methods [114, 144]. For example, Wang et al. developed liposomes composed of DOPC, Cholesterol, and DSPE-PEG, loading drugs using the film dispersion method. These liposomes, with an average size of approximately 100 nm, demonstrated effective biodistribution in the bloodstream and within cells, and exhibited good physical stability with minimal changes in particle size after storage at 4 °C for 20 days [147].

However, liposomes are often rapidly recognized and cleared by the mononuclear phagocyte system (MPS), which can limit their effectiveness [148]. To address this challenge, modifications are often made to evade MPS recognition. For example, Chen et al. introduced hydrophilic polymer modifications, such as Polyethylene Glycol (PEG), to prolong the circulation time of drugs [114]. In addition, researchers have further optimized liposome compositions to improve targeting. For instance, ATF peptides specifically bind to Upar [149], CFH peptides show high affinity for Tenascin-C [150], and peptides with strong binding affinity for fibronectin and type I collagen, such as FnBPA5, can also be incorporated into liposomes [151].

To better respond to the dynamic TME, researchers have incorporated stimuli-responsive materials into liposomes. These materials allow for the specific release of drugs at the tumor site, reducing exposure to normal tissues and potentially minimizing side effects. For example, Yu et al. developed pH-sensitive liposomes by combining DSPE with PEG2K [113]. These liposomes, when delivering chemotherapeutic drugs to the tumor site, suppress CAF activation, overcome tumor stromal barriers, and promote immune cell infiltration. Additionally, drug release can be triggered by temperature changes. Tan et al. utilized dipalmitoylphosphatidylcholine (DPPC) as part of thermosensitive liposomes, which encapsulated near-infrared photosensitizer IR780 iodide and BMS202 [19]. Upon laser irradiation, the liposomes undergo a gel-to-liquid crystal phase transition, accelerating the disintegration of the lipid bilayer structure and disrupting the ECM and CAFs, thus promoting the infiltration of tumor-infiltrating lymphocytes (TILs). This thermosensitive nanoliposome achieved a drug loading (DL%) of 7.0% and an encapsulation efficiency (EE%) of 77.6%.

Through optimization of liposome preparation processes, surface modifications, and the integration of multifunctional capabilities, liposomes can significantly improve drug stability, targeting precision, and the effectiveness of immunotherapies. These advancements hold great promise in overcoming the challenges posed by immunosuppressive cells, such as CAFs, in the tumor microenvironment.

Polymer NPs

Polymeric NPs have emerged as a powerful tool in cancer immunotherapy, offering significant advantages for targeted drug delivery [21, 134]. These NPs are generally classified into two main types: nanocapsules and nanospheres [152, 153]. Nanocapsules encapsulate drugs within an oily core, surrounded by a polymeric shell that controls drug release. This design reduces non-specific drug distribution and enhances targeted therapy. In contrast, nanospheres achieve drug stability by adsorbing or entrapping drugs within a polymer network. Both types are typically composed of synthetic or natural polymers, which provide excellent biocompatibility, biodegradability, accessibility, and cost-effectiveness [26, 154, 155].

Common synthetic polymer materials include novel nucleotides PEI25k-PPhe (PF180), poly(lactic-co-glycolic acid) (PLGA), poly(ethylene glycol)-block-poly(D,L-lactic acid) (PEG-PLA), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-poly(ethylene) (DSPE-PEG) [121, 122, 155]. These amphiphilic polymers are designed to improve the TME by modulating CAFs and enhancing the efficacy of immunotherapy. For instance, PLGA, which is known for its favorable physicochemical properties, preferentially accumulates in tumor tissues while avoiding unwanted immune activation due to its immune-inert nature. PLGA NPs, with a zeta potential between − 35 mV and − 50 mV, have been shown to induce apoptosis in CAFs and alter the phenotype of TAMs [134]. PEGylated PLGA NPs provide stealth functionality, thereby extending circulation time and reducing clearance by the RES [119]. Huang et al. developed PLGA-based NPs to regulate CAF-secreted cytokines, such as CAA and CCL2, thus inhibiting tumor growth and improving the TME [156]. Similarly, Jiang et al. demonstrated that NPs made from the AEAA-PEG-PCL polymer carrier significantly enhanced drug solubility and targeting after systemic administration [157]. These NPs are spherical and relatively uniform in size.

Cationic polymers, such as PolyMet, have gained attention for their high gene transfection efficiency and low toxicity. For example, Zhang et al. showed that PolyMet can effectively deliver plasmids encoding Relaxin, protect DNA from dissociation, and improve TGF-β-induced CAF activation, thereby altering the immunosuppressive microenvironment [158]. Polydopamine, a biocompatible material, has been used as a coating for NPs, enhancing the matrix barrier mediated by CAFs through photothermal effects [159, 160].

Hydroxyethyl starch-folate conjugates (HES-FA) are amphiphilic surfactants that achieve high drug loading efficiency and tumor selectivity. By utilizing isothiocyanate-bridged doxorubicin dimer prodrugs, HES-FA can reduce CAF activity [161].Dendritic polymers also play an important role in drug delivery. For instance, Zhang et al. conjugated dasatinib (DAS) to dendritic poly(oligo(ethylene glycol) methyl ether methacrylate) (POEGMA) via a tetrapeptide linker to create dendritic polymer NPs designed for modulating CAFs. Additionally, they linked epirubicin (Epi) to dendritic POEGMA using acid-responsive hydrazone bonds to facilitate ICD induction [162].

Natural polymers, such as bovine serum albumin (BSA), heparin, gelatin, chitosan, and cellulose, are increasingly used due to their safety and stability [21, 117, 159, 160]. In a study by Hua et al., BSA encapsulated quercetin via hydrophobic interactions and hydrogen bonding to inhibit CAF activity, reshape the TME, and improve nanoparticle penetration and immune cell infiltration [117].Acetylated methylcellulose exhibits excellent biocompatibility and drug delivery properties, aiding in nanoparticle accumulation in the tumor stroma by interacting with SMA-positive fibroblasts and F4/80-positive macrophages [163]. Heparin, with its favorable compatibility, degradability, and water solubility, can target heparanase—a protein highly expressed in tumors—offering a novel strategy to alleviate CAF-mediated immunosuppression [13].

Moreover, polymeric NPs can be engineered to respond to TME-specific characteristics, thereby improving CAF function and enhancing the efficacy of immunotherapy. Examples include glutathione (GSH)-triggered, reductive-sensitive nanoparticles [118, 120, 157]. heparinase- and pH-responsive nanoparticles [115], heparinase- and pH-responsive nanoparticles [13], and FAP-α-influenced self-assembling NPs [17].

Inorganic nanomaterials

Inorganic nanomaterials exhibit unique physicochemical properties that make them highly promising for biomedical applications, particularly in cancer therapy, diagnostics, and immunotherapy. These materials, including black phosphorus (BP), silicon-based, metal-based, metal-oxide-based, metal–organic frameworks (MOFs), and self-assembled nanomaterials, offer significant advantages in drug delivery, imaging, and therapeutic targeting. For example, the layered structure of BP enhances its performance in PTT and photosensitive diagnostics. Silicon-based nanomaterials, known for their stability and tunability, improve drug delivery by enhancing biocompatibility and targeting precision. Metal-based nanomaterials, such as gold and silver, are particularly effective in optical imaging and PTT, while metal oxide nanomaterials, like manganese and iron oxide, excel in MRI imaging and thermal therapy [164]. These properties not only improve the efficacy of conventional treatments but also help overcome tumor immune evasion, offering substantial potential for advancing cancer therapy and immunotherapy.

Silicon-based nanomaterials

Silicon, one of the most abundant elements on Earth, plays a critical role in the diagnosis and treatment of cancer[165, 166]. Solid silicon dioxide and mesoporous silicon dioxide (DMSN) nanostructures, known for their excellent chemical stability, thermal stability, and mechanical robustness, have found widespread application in cancer therapeutics [167, 168, 169]. The superior biocompatibility and blood compatibility of DMSN make it an ideal candidate for constructing drug delivery systems, thereby offering multiple avenues for cancer treatment. Additionally, its large surface area and high porosity create favorable conditions for drug loading and controlled release [170]. For example, in He’s research, DMSN was combined with trypsin-like protease imprinted polymers to neutralize trypsin-like protease (TPS), resulting in changes to the phenotype of CAFs and enhancing the efficacy of tumor immunotherapy [155].

In terms of synthesis, hybrid organic–inorganic approaches can be utilized to prepare hollow mesoporous organosilica NPs. These nanostructures not only enable efficient drug loading, but with surface modifications such as diselenide coupling, they also facilitate the degradation of the silicon-based material under the influence of reactive oxygen species (ROS), thus promoting the release of encapsulated drugs. This process not only supports the effective release of therapeutic agents but also triggers a potent immunogenic response. Moreover, binding to PD-L1 on the surface of tumor cells further amplifies the systemic immune response [171]. Furthermore, DMSN has been shown to load negatively charged siRNA, effectively shielding it from degradation and thereby influencing the behavior of CAFs [121].

Carbon-based nanomaterials

Carbon-based nanomaterials, including graphene, carbon quantum dots, and carbon nanotubes, have shown great potential in cancer therapy and diagnostics. Their unique properties make them ideal for drug delivery systems in cancer immunotherapy [172]. Surface modifications, both covalent and non-covalent, can enhance drug targeting and improve anticancer efficacy [168].

Carbon-based nanomaterials exhibit excellent optical properties in the near-infrared (NIR) region, making them ideal for photothermal therapy, which can trigger strong antitumor immune responses. Their biocompatibility and water dispersibility also make them excellent fluorescence imaging probes for early cancer detection and monitoring. For instance, carbon dot(CD) nanoclusters serve as imaging agents, photothermal therapy tools, and carriers for chemotherapeutic drugs and immunological inducers, playing a key role in cancer treatment and immunotherapy [173, 174, 175].

The high surface area and porous structure of carbon dots enhance their ability to adsorb chemotherapeutic drugs and immunological adjuvants. In a study by Hou et al. DOX molecules were efficiently loaded onto CDs through π-π stacking interactions with the large conjugated π system of CD, facilitating the drug’s binding to the nanomaterial [122]. Furthermore, the study involved surface modification of CDs with AEAA-PEG-NH₂ and NH₂-PEG-NH₂, which endowed the CDs with the ability to specifically target CAFs and significantly improved their stability. Additionally, graphene-based fluorescence NPs, synthesized via hydrothermal reactions, achieved labeling efficiencies of up to 60% for CAFs [176].

BP-based nanomaterials

BP, as a member of the two-dimensional material family, is a unique metal-free layered semiconductor. Its bandgap is tunable, capable of flexibly shifting from 0.3 eV in bulk material to 2.0 eV in monolayer materials, and it also possesses ultraviolet and near-infrared light absorption characteristics. These exceptional optical properties give it immense potential in the field of cancer therapy, especially in optical treatment, drug delivery, and diagnostics [174, 177].

Compared to other two-dimensional materials such as graphene and MoS, black phosphorus nanosheets exhibit higher drug loading capacity and significant pH/photon response capability [173]. Their good biocompatibility and photostability further establish their position as an ideal choice for a multifunctional nanoplatform. Surface modification of BP through physical or chemical means can achieve multiple goals, such as active targeting, extended retention time in the body, enhanced photosensitivity, and improved photothermal conversion ability [178, 179].

In practical cancer treatment scenarios, black phosphorus has shown excellent results in both PTT and PDT. When exposed to specific laser irradiation, it not only effectively inhibits tumor growth but also strongly activates antitumor immune responses [180]. More importantly, bioactive black phosphorus can suppress the activation of CAFs, subtly promoting the interaction between tumor and stroma, thereby further enhancing the overall therapeutic effect [180]. Additionally, black phosphorus can act as a bridge to synergize the combined impact of photothermal therapy and adenosine blockade, significantly improving the physical barrier of tumor stroma and immune-suppressive factors and thereby considerably enhancing the activity of cytotoxic T cells [123]. However, BP is highly susceptible to oxidation, and it is commonly modified, for example, by loading with PEG [178].

Metal-based nanomaterials

Metal-based nanomaterials have attracted much attention due to their unique biological effects [181]. The commonly used elements are calcium (Ca2+), manganese (Mn2+), iron (Fe2+/3+), and potassium (K), which can be modified to synthesize a wide range of common metal-based nanomaterials, such as CaCO3 NPs, manganese oxide (MnO) NPs (e.g., MnO, MnO2), FeO NPs (e.g., Fe3O4), and NaCl NPs [179, 182]. These materials modulate immune responses in tumor immunotherapy by triggering and influencing key immune processes. For instance, FeO NPs contain both Fe2⁺ and Fe3⁺ ions, with Fe2⁺ promoting the innate immune response against cancer cells by catalyzing the Fenton reaction to generate ROS, which can induce classical cell death and ICD. In contrast, Fe3⁺ enhances the effects of ICD by preventing ROS clearance, a process that involves depleting excess GSH. Additionally, Fe ions can stimulate macrophages to adopt the immunostimulatory M1 phenotype [183].

Moreover, metal-based nanomaterials can also exhibit photothermal effects, which can further enhance their therapeutic potential [183, 184].For example, He et al. developed Fe/Co bimetallic nanocarriers that not only trigger the Fenton reaction to induce ferroptosis but also possess photothermal properties. This dual-functionality enables targeted therapy of both tumor cells and CAFs [128].

In another study, Zheng et al. utilized gold nanoparticles (Au NPs) modified with polyallylamine hydrochloride (PAH) to alter the surface charge of the Au NPs, converting it from negative to positive. This modification enabled the Au NPs to bind to the surface of Fe₃O₄ NPs. The combination of photothermal therapy and imaging significantly impacted CAFs, enhancing ICD and activating immune responses [124]. In addition to iron oxide, other metal elements such as palladium (Pd), platinum (Pt), and gold (Au) also play crucial roles in tumor immunotherapy. For example, Hou et al. designed PdPtAu NPs with a metal-polyphenol network on their surface [185]. These NPs, with a spherical shape and an average diameter of 75 nm, exhibited a well-defined mesoporous structure.

MOFs, as metal-based nanomaterials with three-dimensional porous structures, have garnered significant interest in recent years due to their promising applications in biomedicine. MOFs are typically composed of inorganic metal nodes or clusters, offering advantages such as excellent biocompatibility, chemical stability, tunable porosity, and versatile surface modification capabilities, making them well-suited for biomedical purposes [184, 185, 186]. These materials have the potential to elicit robust immune responses by encapsulating antigens and immune adjuvants. For example, MOF surfaces can selectively bind to cytosine-phosphate-guanosine (CpG) oligonucleotides, thereby triggering immune activation [187, 188]. Furthermore, MOFs can be used to deliver PD-L1 inhibitors, which counteract immunosuppressive signals and enhance the efficacy of immunotherapy [189].

In research by Guo et al. a MOF based on Fe₄O₃ was coated with a platelet membrane [127]. MOFs can also be combined with metals like copper (Cu) and iron (Fe). For example, Meng et al. developed Cu(II)-MOFs that encapsulated Blebbistatin and a FAP-responsive peptide (Tp), enabling targeted delivery to CAFs and photothermal triggering [125]. Huang et al. synthesized a MOF that coordinated Fe3⁺ with PLGA nanocores, forming a stable, negatively charged MOF structure on the surface. This structure reduces drug molecule collisions, delays aggregation and sedimentation of drugs in solution, and enhances colloidal stability [187]. This MOF encapsulated Erastin, forming a spherical core–shell structure with a particle size of approximately 170 nm and a shell thickness of around 30 nm. It also promoted M1 macrophage polarization while reducing TGF-β secretion.

Overall, these metal-based nanomaterials are emerging as powerful tools in cancer immunotherapy due to their ability to modulate immune responses, induce cell death, and enhance the efficacy of immunotherapeutic strategies. Their multifunctionality, along with the potential for surface modifications, positions them as promising candidates for targeted cancer therapies and immune modulation.

Natural carrier nanomaterials

Cell membranes, as natural carrier nanomaterials, consist of lipid bilayers and are adorned with proteins, glycoproteins, and lipoproteins on their surface[190]. These molecules play crucial roles in immune evasion, homing targeting, and regulation of inflammatory responses [191, 192]. Researchers have exploited these properties by camouflaging nanoparticles (NPs) with cell membranes, thus mimicking the antigenic diversity of the original cells and endowing the NPs with functionalities akin to those of the original cell membrane [193]. Cell membranes from various sources—such as erythrocytes, tumor cells, macrophages, and NK cells—have been widely utilized to coat NPs, enhancing their ability to modulate CAFs.

To further augment the functionality of these membranes, coupling methods involving amines, carboxyl groups, and thiols are often used to decorate the cell membranes with functional ligands [191, 194]. For example, Li et al. developed hyaluronic acid (HA)-polyethylene glycol (PEG)-lipid conjugates for surface engineering of ex vivo NK cells, thereby enhancing the efficacy and duration of NK cell membrane coating [192]. This approach does not induce off-target effects on human fibroblasts, boosting the effectiveness of NK cells in cancer-targeted immunotherapy. In the field of cancer nanotherapy, biomimetic cell membrane technology has been increasingly used to improve immune evasion in immunotherapy by mimicking immune cells in the inflamed tumor microenvironment [193].

Macrophage membranes, for instance, can recruit additional immune cells to inflammatory sites by releasing chemokines and cytokines, thereby activating immune responses [195]. This makes macrophage membranes an excellent choice for drug delivery, especially when targeting tumor cells, as they significantly improve drug targeting, biocompatibility, and reduce unwanted immune responses [194].Du et al. utilized macrophage membranes encapsulating α-mangostin and O2 to form biomimetic nanoprobes, enhancing the ECM created by CAFs, Improving drug penetration, and facilitating immune cell infiltration [196].

In some studies, homing targeting strategies are employed, such as coating NPs with activated fibroblast membranes to minimize immunogenic responses and effectively target CAFs, thereby improving the tumor microenvironment [197]. For example, Jia et al. constructed liposomes coated with CAF and tumor cell membranes, leveraging the inherent homing properties of these cell membranes to specifically target CAFs and tumor cells [131]. Similarly, Zang et al. used CAF and tumor cell membrane-coated solid lipid nanoparticles [131].Genetic engineering techniques can also be combined with biomimetic nanocarriers to introduce molecules that specifically bind to the FAP receptor on CAFs, enabling precise targeting and elimination of CAFs, while triggering immune responses against tumors [198].

Erythrocyte and platelet membranes, the two major types of cell membranes in blood, have also been applied to regulate CAFs. Erythrocytes are particularly advantageous for extraction and purification because they lack nuclei and other organelles. Furthermore, they are highly effective in evading phagocytosis by monocytes and macrophages, assisting NPs in targeting CAFs. Guo et al. coated MOFs with platelet membranes, which specifically recognize and adhere to collagen secreted by CAFs in the tumor microenvironment [132].

Additionally, extracellular vesicles (EVs) derived from cell membranes have gained significant attention in tumor immunotherapy. These vesicles serve as ideal drug delivery vehicles due to their role in extracellular communication between tumor cells and CAFs. For example, Yuan et al. demonstrated that extracellular vesicles help drugs cross the tumor stromal barrier and enhance the immune response against tumors [133]. Hu et al. explored exosome-like nanovesicles derived from FAP gene-engineered tumor cells [199].

Comparison of different nanoparticles

Each type of nanomaterial possesses unique advantages and limitations within drug delivery systems. When selecting nanomaterials for drug delivery, it is essential to consider their comprehensive biocompatibility, stability, drug loading capacity, targeting ability, and biosafety. Through further research and optimization, these nanomaterials hold the potential to play an increasingly significant role in future drug delivery systems. Table 2 will provide a comparison of the nanomaterials listed above.

Table 2 Comparison of Different Nanomaterials

Mechanisms of targeting distinct CAFs

A comprehensive understanding of the regulatory mechanisms that govern CAF functions is crucial for the development of more effective therapeutic strategies (Fig. 4). Furthermore, the integration of nanodrugs with complementary modalities, such as photothermal therapy (PTT) and photodynamic therapy (PDT), offers significant promise for enhancing therapeutic outcomes. In this section, we examine the current mechanisms by which targeting CAFs can be utilized to advance cancer treatment, providing valuable insights into the development of more efficient therapeutic approaches. Table 3 outlines the mechanisms through which targeting CAFs can optimize tumor immunotherapy outcomes.

Fig. 4
figure 4

The current main regulatory approaches of CAFs and combination therapies

Table 3 Mechanism of action of nanomedicines targeting CAFs

Inhibition of CAF activation

CAF activation is influenced by multiple factors and signaling pathways in the TME, and these activated CAFs play an important role in tumorigenesis and progression. Several methods can usually be used to inhibit CAF activation, such as TGF-β receptor inhibitors (e.g.,Galunisertib) [9], FAP inhibitors(e.g.,Talabostat mesylate) [110], Fibroblast adhesion patch kinase (FAK) inhibitors(e.g.,IN10018) [212], Angiotensin II Receptor Antagonists (ARBs) inhibitors(e.g., Valsartan、Losartan) [110, 138], tyrosine kinase (TK) inhibitors(e.g.,Sunitinib、Nintedanib)[213, 214], NF-κB inhibitors(e.g.,Silybin) [121, 140], and TGF-β/SMAD signaling pathway inhibitors(e.g.,Baicalein、Salvianolic acid B) [24]. In addition, CAF activation was effectively inhibited by reversing EMT [127].

These inhibitory strategies often rely on the encapsulation of the drugs in nanomaterials for improved delivery and efficacy. For instance, Chen et al. developed PEGylated liposomes loaded with SAB (PEG-SAB-Lip). These liposomes remodel the TME by inhibiting the TGF-β1/Smad signaling pathway in activated fibroblasts [114]. Furthermore, the impact of PEG-SAB-Lip on key proteins such as α-SMA, TGF-β1, Smad2, Smad3, and pSmad2 was found to be significantly stronger than that of the SAB group (Fig. 5A, B). In addition to targeting CAFs, PEG-SAB-Lip also enhances the anti-tumor effects of docetaxel and significantly increases the expression of CD4+ and CD8+T cells (Fig. 5C). In another study, Liang et al. encapsulated Valsartan and doxorubicin (DOX) in polymer micelles. Valsartan inhibits CAF activity, while DOX penetrates deep into tumor cells, inducing cellular senescence and recruiting effector immune cells [138].

Fig. 5
figure 5

© 2023 American Chemical Society.)

A Western blot bands and semi-quantitative analysis of the effects of SAB and PEG-SAB-Lip on the TGF-β1/Smad signaling in TGF-β1-activated NIH3T3 cells. B Western blot analysis of the levels of tumor proteins after different treatments. 1: NS, 2: PEG-B-Lip, 3: SAB, 4: PEG-SAB-Lip. C Immunofluorescence staining and the corresponding quantitative analysis of CD4+T and CD8+T immune functional cells in the TME under the combined therapy of PEG-SAB-Lip and PEG-DTX-Lip. Adapted with permission from [114] (copyright

Reprogramming of CAFs

Currently, one of the recommended strategies to inhibit tumor development is to modify CAFs to restore their normal functions, transforming them from “tumor-promoting” to “tumor-suppressing” cells. To achieve this, various molecules have been integrated into nano-delivery systems to regulate CAFs and enhance the efficacy of cancer immunotherapy. Notable examples include Calcipotriol [207], Retinoic Acid (RA) [209], All trans-retinoic acid (ATRA), the bromodomain-containing protein 4 (BRD4) inhibitor (e.g., JQ1) [133, 197], and melittin, the principal component of Apis mellifera venom [215]. For instance, Yuan et al. encapsulated JQ1, interleukin-12 (IL-12), and Elaidate in lipid nanoparticles, which exerted combined chemo-immunological effects (Fig. 6A). These lipid nanoparticles significantly reduced the expression of α-SMA and collagen I (Fig. 6B, C), while promoting the generation of lipid droplets (Fig. 6D), thereby facilitating CAF normalization. By decreasing ECM deposition, the lipid nanoparticles also enhanced immune cell infiltration (Fig. 6E–G) [133]. In another study, Zhang et al. created a biomimetic delivery system by coating liposomes with CAF membranes and co-encapsulating Pirfenidone and JQ1. This liposomal system specifically targeted CAFs, decreasing their activity and collagen production [197].

Fig. 6
figure 6

© 2023 American Chemical Society.)

A Mechanism of Action of Lipid Nanoparticles (B) Immunofluorescence (IF) Staining of α-SMA and collagen I, alongside BODIPY 493/503 staining of lipid droplets in CAFs after 48 h of culture with the formulation containing JQ1. Scale bar = 40 μm. C Western Blot Analysis of α-SMA and collagen I in CAF lysates following 48-h treatment with the formulation containing JQ1. D Quantification of Lipid Droplets: The average number of lipid droplets per 10 CAFs. EG Flow Cytometry (FCM) Analysis of CD4+ T cells, CD8+ T cells, and Tregs within the TME. Adapted with permission from [133] (copyright

In addition, promoting the normalization of CAFs' functions has shown potential in improving the treatment of desmoplastic tumors. These tumors are characterized by intratumoral stiffness and mechanical forces that compress tumor blood vessels, reducing perfusion and hindering the infiltration of both drugs and immune cells [105]. Strategies to address this issue include directly targeting the tumor stroma or modulating CAF activity. Common therapeutic agents used for this purpose include antifibrotic drugs, antihypertensive agents, and corticosteroids. For example, Myrofora et al. encapsulated Tranilast in polymer micelles to control fibrotic signaling in CAFs. This approach modulated the mechanical properties of the TME, improved tumor perfusion, and significantly enhanced the uniform accumulation of cytotoxic nanodrugs, leading to stronger anti-tumor effects. Furthermore, these polymer micelles also promoted T-cell infiltration and induced long-term immune memory [108]. Similarly, Zhu et al. encapsulated Sunitinib in lipid-calcium sulfate nanoparticles and combined it with vaccination immunotherapy to improve the immunosuppressive environment of fibrotic, collagen-rich desmoplastic melanoma [214]. Additionally, remodeling the stromal environment can be achieved by targeting key downstream mediators of pro-fibrotic pathways, such as reactive oxygen species (ROS). Xu et al. developed a novel puerarin nanoemulsion that downregulated intratumoral ROS levels, thereby inactivating CAFs, reducing collagen deposition, and alleviating desmoplasia [133].

Depletion of CAFs

Enhancing cancer immunotherapy can also be achieved by targeting and depleting CAFs. Small interfering RNA (siRNA) technology can be used to affect the interaction between CAFs and cancer cells by reducing the expression of specific target genes such as hepatocyte growth factor (HGF). The combined use of siRNA and chemotherapeutic agents like doxorubicin (DOX) has been shown to effectively deplete CAFs and reduce their secretion of HGF, thereby inhibiting their tumor-promoting role [216, 217]. Similarly, applying a TGF-β siRNA (siTGF-β) to tumor cells and CAFs significantly reduced their expression of TGF-β, helping to normalize the vascular system of the tumors, inhibiting EMT, and increasing the infiltration of immune cells [149]. Moreover, activated CAFs are sensitive to nitric oxide (NO), a property that has also been utilized to improve CAF-mediated tumor immunosuppression [211]. However, excessive depletion or killing of CAFs could potentially disrupt the functions of normal tissues. Therefore, researchers are working on refining strategies to ensure both safety and efficacy. For instance, Akai et al. targeted FAP and developed a near-infrared photoimmunotherapy approach, which successfully and selectively depleted CAFs without affecting normal cells. This strategy effectively eliminated the immunosuppressive CAFs and remodeled the local tumor immune microenvironment [217].

Targeted CAFs therapy in combination with adjunctive treatment modalities

Combination with photothermal therapy (PTT)

Photothermal therapy (PTT) treats tumors by irradiating targeted photothermal materials or photosensitizers with near-infrared (NIR) light, inducing localized thermal effects that can effectively kill tumor cells and modulate the immune response by altering the tumor microenvironment [218]. However, the presence of CAFs significantly hinders the efficacy of PTT, as CAFs secrete ECM components, such as collagen and hyaluronic acid, which create physical barriers that limit therapeutic effectiveness. Existing research has shown that PTT can alleviate tumor hypoxia and enhance the efficacy of PDT [219]. A range of photosensitizers, such as indocyanine green (ICG), and photothermal materials, including PDA and gold nanomaterials, have been successfully targeted and delivered to tumor sites. Upon external light irradiation, these materials generate localized hyperthermia, directly heating the tumor tissue [124, 201, 206, 219]. Wang et al. developed polydopamine (PDA) particles coated with CAF membranes and tumor cell membranes. Through the PDA-mediated photothermal effect, the tumor stroma was relaxed, thereby disrupting the physical barriers formed by the ECM and facilitating deeper drug penetration and targeting of tumor cells [158]. Similarly, Li et al. encapsulated calcipotriol and indocyanine green (ICG) in microparticles (MPs) to create Cal/ICG@MPs. Upon 808 nm laser irradiation, this formulation induced a potent photothermal effect that triggered immunogenic cell death (ICD) in tumor cells (Fig. 7A, B) [207]. Notably, the percentage of calreticulin (CRT)-positive cells in the ICG@MPs and Cal/ICG@MPs treatment groups was significantly higher than in the free ICG and Cal/ICG groups, with enhanced secretion of HMGB1 and release of M (Fig. 7C, D). Moreover, the Cal/ICG@MPs treatment promoted the maturation of DCs and the activation of CD8+ T cells (Fig. 7E, F). Importantly, it also increased the infiltration of CD8+ T cells into the deeper regions of the tumor by modulating CAF activity (Fig. 7G).

Fig. 7
figure 7

© 2022 Springer Nature Limited)

A Mechanism diagram of Cal/ICG@MPs. B TME images of MPs, Cal@MPs, ICG@MPs, and Cal/ICG@MPs. C Cell viability under different treatment groups with or without 808 nm laser irradiation. D Analysis of CRT, HMGB1, and ATP levels. E Flow cytometric analysis of BMDCs. F Semi-quantitative analysis. G Immunofluorescence staining for α-SMA, fibronectin, and collagen-I, along with Masson’s trichrome staining. Adapted with permission from [207] (copyright

Combination with photodynamic therapy (PDT)

Photodynamic therapy (PDT) utilizes photosensitizers to generate reactive oxygen species (ROS) under specific light irradiation, which can not only induce tumor cell death but also enhance tumor immunotherapy through mechanisms such as the release of tumor antigens and immune activation [220]. The ROS produced during PDT can also modulate the phenotypes and functions of cancer-associated fibroblasts (CAFs), thereby influencing their role in promoting tumor progression. PDT's therapeutic efficacy can be augmented either by optimizing photosensitizer use or by leveraging the intrinsic properties of nanomaterials to increase intracellular ROS levels. Tian et al. developed an alginate (ALG)-based hydrogel drug delivery platform (ALG@TPN), which incorporates TBP-2, Pt (0), and Nintedanib. The combined effect of PDT and chemotherapy activates the cGAS/STING pathway, leading to CAF inhibition and remodeling of the tumor immunosuppressive microenvironment [221]. Moreover, Zhou et al. engineered ferritin nanoparticles (αFAP-Z@FRTs) that conjugate a photosensitizer (ZnF+16Pc) with a fibroblast activation protein (FAP)-specific single-chain antibody (Fig. 8A) [222]. This design enables targeted delivery of PDT to CAFs, significantly enhancing accumulation at the tumor site (Fig. 8B). In both the PDT and combination therapy groups, the number of damaged CAFs surpasses that of 4T1 cancer cells, suggesting that the treatment may trigger an immune response against CAFs (Fig. 8C). Furthermore, the frequencies of effector T cells targeting both 4T1 tumor cells and CAFs are markedly elevated, indicating that these therapeutic strategies can activate the immune system, leading to an increased generation of effector T cells. This, in turn, may enhance the immune response against both cancer cells and CAFs (Fig. 8D).

Fig. 8
figure 8

© 2020 Wiley–VCH GmbH.)

A TEM images of ferritin (FRT) and DLS analysis of αFAP-FRT, Z@FRT, and αFAP-Z@FRT. B PET imaging of 64Cu-labeled αFAP-FRT injected into 4T1-bearing mice, with images taken at 1, 5, and 24 h post intravenous injection. C Cell-specific cytotoxicity analysis for different groups. D Enzyme-linked immunospot (ELISpot) analysis for different groups.Adapted with permission from [222] (copyright

Combination with immune checkpoint inhibition therapy

Direct interactions between cancer cells and CAFs have been shown to promote the upregulation of programmed death-ligand 1 (PD-L1) expression. This upregulation can be targeted by anti-PD-L1 antibodies, which not only enhance tumor cell death but also induce apoptosis in CAFs [224]. Programmed death 1 (PD-1) inhibitors, a class of immune checkpoint inhibitors (ICIs), work by reactivating the anti-tumor activity of TILs. They achieve this by blocking the PD-1/PD-L1 interaction, which suppresses TIL function under normal conditions. For instance, Guo et al. developed a system to encapsulate Oxymatrine and Astragaloside IV in MOFs. Oxymatrine (Om) was employed to inhibit CAF activation, thereby increasing the presence of TILs in the tumor microenvironment. Meanwhile, Astragaloside IV (As) enhanced the anti-tumor efficacy of TILs by improving their mitochondrial function. When combined with α-PD-1 antibodies, this therapeutic system significantly improved the anti-hepatocellular carcinoma(HCC) response, resulting in a tumor inhibition rate of 84.15% and extending the survival time of tumor-bearing mice [127].

Furthermore, Yang et al. developed an innovative polymeric vesicle-based prodrug nanoplatform (TRPP/Tab) designed to simultaneously target CAFs and induce ICD, thereby enhancing the efficacy of cancer immunotherapy. This nanoplatform encapsulated three active compounds—DPPA-1, doxorubicin (DOX), and Talabostat mesylate—within polymeric micelles (Fig. 9A). In vivo, Talabostat mesylate effectively inhibited CAFs and reduced the accumulation of excessive reactive oxygen species (ROS), while DPPA-1 blocked immune checkpoints and doxorubicin (DOX) triggered ICD. Experimental results demonstrated that the DPPA-TRPP/Tab nanoplatform not only increased tumor accumulation but also suppressed CAF formation (Fig. 9C) and enhanced the activation of CD4+ and CD8+ T cells (Fig. 9B). As a result, this combination therapy achieved a 60% complete tumor regression rate and induced a durable immune memory response in the mouse model (Fig. 9D, E) [110].

Fig. 9
figure 9

A Preparation and mechanism of action of PPA-TRPP/Tab. B Quantitative analysis of representative CD8+、CD4+T cell percentages after treatment in different groups and measurement of tumor tissues after different treatments using flow cytometry. C The expression of α—SMA after treatment in different groups is used to characterize CAFs (D) The average tumor growth curve (E) weight curve, F and survival curve. Adapted with permission from [110] (copyright ©2023 Ivyspring International Publisher.)

Combination with CAR-T cell therapy

CAR-T cell therapy has the characteristics of self-expansion and higher sensitivity to low antigen expression, thus having the potential to become a promising alternative strategy for targeting fibroblast activation protein (FAP) tumor stromal cells. For example, Das et al. designed chimeric antigen receptor (CAR) T cells targeting fibroblast activation protein (FAP) through the transcription activator-like effector nuclease (TALEN) gene editing platform, which can effectively deplete cancer-associated fibroblasts CAFs in the tumor microenvironment TME and reduce the physical barriers of the stroma, thereby facilitating the infiltration of T cells [225].

Combination with microorganisms

Bacteria and their components have demonstrated significant potential as immunomodulators [226]. For instance, Bacillus Calmette-Guérin (BCG) has been established as the first successful immunotherapy. Research has shown that Trehalose Dimycolate (TDM), the most abundant hydrophobic glycolipid on the cell wall of BCG, possesses notable anti-tumor properties along with various immunostimulatory activities. Li et al. have explored the use of mesoporous silica nanoparticles loaded with TDM to enhance tumor fibrosis [227].

Moreover, engineered bacteria have emerged as powerful tools for augmenting cancer immunotherapy. Zhang et al. designed a genetically engineered bacterium capable of secreting nattokinase (NKase) [228]. This bacterium was developed by attenuating the Salmonella typhimurium strain VNP20009 and incorporating a gene encoding nattokinase into its plasmid via genetic engineering. The engineered bacterium can reprogram CAFs from an activated to a quiescent state by secreting substantial amounts of NKase, which degrades fibronectin within the tumor microenvironment. This modification alters the ECM and results in a reduction in tumor tissue stiffness, solid stress, and interstitial fluid pressure, thereby enhancing the efficacy of subsequent radiotherapy and immunotherapy.

Future perspectives

CAFs are critical drivers of tumor progression, shaping the TME and influencing multiple aspects of cancer biology. Beyond supporting tumor growth and metastasis, CAFs contribute to immune evasion and the establishment of a pro-inflammatory milieu. They secrete a variety of cytokines, growth factors, and ECM components that promote tumor cell proliferation, survival, and migration. CAFs also facilitate ECM remodeling, enabling tumor cell invasion and metastasis. Recent insights into CAFs' role in EMT and the activation of key signaling pathways, such as TGF-β and Wnt/β-catenin, have deepened our understanding of the molecular mechanisms driving tumor progression. Moreover, CAF-immune cell interactions further complicate tumor immunology, modulating immune surveillance and influencing therapeutic responses. Looking ahead, targeting CAFs represents a promising therapeutic strategy for cancer treatment. However, the heterogeneity of CAFs, their dynamic behavior, and the complexity of their interactions with the tumor microenvironment present significant challenges for the development of effective therapies. Future research should focus on unraveling the precise molecular mechanisms underlying CAF function, as well as identifying specific biomarkers that can be used to selectively target CAFs without disrupting normal tissue homeostasis. Additionally, it is necessary to optimize these strategies for clinical application, addressing challenges related to specificity, safety, and tumor heterogeneity. The integration of CAF-targeted approaches into personalized cancer treatment regimens will likely be an important step toward achieving more effective and durable therapeutic responses. Thus, a deeper understanding of CAF biology will be essential for the development of novel, targeted therapies aimed at improving patient outcomes in cancer.

Conclusions

This review provides an overview of recent advancements in CAF-targeted therapies and explores emerging techniques for the molecular targeting of CAFs. Despite being a predominant cell type in the tumor microenvironment, CAFs present a significant challenge for precise therapeutic targeting due to their inherent heterogeneity. Overcoming these challenges is essential to translating research findings from the laboratory to clinical applications. The origins of CAFs across different cancer types remain unclear, as does the full understanding of their subtypes and functional diversity. Additionally, minimizing off-target and systemic effects continues to be a major hurdle. Furthermore, combining CAF-targeted immunotherapies with existing therapeutic strategies holds great promise and is an active area of investigation. These approaches not only deepen our understanding of CAF biology but also have the potential to enhance the precision of cancer therapies and improve patient outcomes.

Availability of data and materials

No datasets were generated or analysed during the current study.

Change history

Abbreviations

IL1R1/IL-1R:

The IL-1/IL-1 receptor type 1

PKM/PKM2:

Pyruvate kinase M1/2 (PKM/PKM2)

(NTRK2/TRKB)/NFE2:

The neurotrophic receptor tyrosine kinase 2

ACTA2/α-SMA:

Actin alpha 2, smooth muscle

ADORA2A:

Adenosine A2A receptor

AKT:

Protein kinase B

ALB:

Albumin

ApCAFs:

Antigen-presenting CAFs

BCG:

Bacillus Calmette-Guérin

BDNF:

Brain-derived neurotrophic factor

BP:

Black phosphorus

BSA:

Bovine serum albumin

CaCO3 :

Calcium carbonate

CAFs:

Cancer-associated fibroblasts

CELF2:

The CUGBP Elav-like family member 2

circFARP1:

Pleckstrin domain protein 1

circZFR:

The zinc finger RNA binding protein

CMC:

Critical micelle concentration

CPG:

Cytosine-phosphate-guanine

CRC:

Colorectal cancer

CRMP2:

Collapsin response-mediated protein 2

CSF2/GM-CSF:

Colony-stimulating factor 2

CTLs:

Cytotoxic T lymphocytes

CXCL2:

C-X-C motif chemokine ligand 2

CXCL5:

C-X-C motif chemokine ligand 5

CXCL9:

C-X-C motif chemokine ligand 9

CXCL10:

C-X-C motif chemokine ligand 10

CXCL12:

C-X-C motif chemokine ligand 12

CXCL16:

C-X-C motif chemokine ligand 16

CXCR4:

C-X-C motif chemokine receptor 4

DAS:

Dasatinib

DCs:

Dendritic cells

DPPC:

Dipalmitoylphosphatidylcholine

DSPE-PEG:

1, 2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-PEG

ECM:

Extracellular matrix

EGF:

Epidermal growth factor

EMT:

Epithelial-mesenchymal transition

EVs:

Extracellular vesicles

FAK:

Fibroblast adhesion patch kinase

FAP/FAP-α:

Fibroblast activation protein alpha

FGF2:

Fibroblast growth factor 2

FGF7:

Fibroblast growth factor 7

FOXF2:

Forkhead box F2

Foxp3:

Forkhead box P3

GPR30:

G protein-coupled receptor 30

HA:

Hyaluronic acid

HAS2:

Hyaluronan synthase 2

HCC:

Hepatocellular carcinoma

HES-FA:

Hydroxyethyl starch-folate conjugates

HIF1A/HIF-1α:

Hypoxia-inducible factor

HPSE:

Heparanase

iCAFs:

Inflammatory CAFs

ICD:

Immune cell death

IGFBP7:

Insulin-like growth factor-binding protein 7

JAKs:

Janus kinases

MAPK:

Mitogen-activated protein kinase

MCT1:

Monocarboxylate transporter1

MDSC:

Mononuclear myeloid-derived suppressor cell

MMP:

Matrix metalloproteinase

MOF:

Metal–organic framework

myCAFs:

Myofibroblasts

NF-κB:

Nuclear factor kappa B

NKs:

Natural killer cells

NO:

Nitric oxide

NPs:

Nanoparticles

OPN:

Osteobridging protein

OV:

Oncolytic virus

p62:

Encoded by the SQSTM1 gene

PCL:

Polycaprolactone

PDGF:

Platelet-derived growth factor

PDGFRA/PDGFR-α:

PDGF receptor alpha

PEG:

Polyethylene glycol

PEG-PBLG:

PEG-b-poly(benzyl-L-glutamate)

PF4/CXCL4:

G protein-coupled receptor-mediated binding of platelet factor 4

PLA:

Polylactic acid

PLA2G2A:

Phospholipase A2 group IIA

PLGA:

Biodegradable poly(lactic-co-glycolic) acid

POEGMA:

Poly(oligo(ethylene glycol) methyl ether methacrylate)

PPBP/CXCL7:

Pro-platelet basic protein

PSC:

Pancreatic stellate cell

PVA:

Polyvinyl alcohol

RA:

Retinoic acid

ROS:

Reactive oxygen species

SAB:

Salvianolic acid B

SERPINE1/PAI1:

Serpin family E member 1

sFRP4:

Secreted frizzled-related protein

siRNA:

Small interfering RNA

SOX4:

SRY-box transcription factor 4

STAT3:

Signal transducer and activator of transcription 3

TAMs:

Tumor-associated macrophages

TDM:

Trehalose Dimycolate

TGF-β:

Transforming growth factor-beta

TPS:

Trypsin-like protease

TILs:

Tumor-infiltrating lymphocytes 4

TME:

Tumor microenvironment

Tregs:

Regulatory T cells

uPAR:

The urokinase plasminogen activator receptor

VCAM1:

Vascular cell adhesion molecule 1

VCAN:

Versican

VEGFs:

Vascular endothelial growth factors

WNT2:

Wnt family member 2

WWOX:

WW domain-containing oxidoreductase

References

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

    Article  PubMed  Google Scholar 

  2. Yang D, Liu J, Qian H, Zhuang Q. Cancer-associated fibroblasts: from basic science to anticancer therapy. Exp Mol Med. 2023;55:1322–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. 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. 2022;42:401–34.

    Article  Google Scholar 

  4. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021;20:1–30.

    Article  Google Scholar 

  5. Biffi G, Tuveson DA. Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 2021;101:147–76.

    Article  PubMed  CAS  Google Scholar 

  6. Kennel KB, Bozlar M, De Valk AF, Greten FR. Cancer-associated fibroblasts in inflammation and antitumor immunity. Clin Cancer Res. 2023;29:1009–16.

    Article  PubMed  CAS  Google Scholar 

  7. Guo S, Yuan J, Meng X, Feng X, Ma D, Han Y, Li K. Cancer-associated fibroblasts: Just on the opposite side of antitumour immunity? Int Immunopharmacol. 2023;122:110601.

    Article  PubMed  CAS  Google Scholar 

  8. Chen M, Chen F, Gao Z, Li X, Hu L, Yang S, Zhao S, Song Z. CAFs and T cells interplay: the emergence of a new arena in cancer combat. Biomed Pharmacother. 2024;177:117045.

    Article  PubMed  CAS  Google Scholar 

  9. Huang S, Ding D, Lan T, He G, Ren J, Liang R, et al. Multifunctional nanodrug performs sonodynamic therapy and inhibits TGF-β to boost immune response against colorectal cancer and liver metastasis. Acta Biomater. 2023;164:538–52.

    Article  PubMed  CAS  Google Scholar 

  10. Froeling FEM, Feig C, Chelala C, Dobson R, Mein CE, Tuveson DA, et al. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt–β-catenin signaling to slow tumor progression. Gastroenterology. 2011;141:1486-1497.e14.

    Article  PubMed  CAS  Google Scholar 

  11. Liu S, Li F, Ma Q, Du M, Wang H, Zhu Y, et al. OX40L-armed oncolytic virus boosts T-cell response and remodels tumor microenvironment for pancreatic cancer treatment. Theranostics. 2023;13:4016–29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Fang Z, Meng Q, Xu J, Wang W, Zhang B, Liu J, et al. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. Cancer Commun. 2023;43:3–41.

    Article  Google Scholar 

  13. Zhang J, Zuo T, Yang J, Hu Z, Wang Z, Xu R, et al. Hierarchically releasing bio-responsive nanoparticles for complete tumor microenvironment modulation via TGF-beta pathway inhibition and TAF reduction. ACS Appl Mater Interfaces. 2021;13:2256–68.

    Article  PubMed  CAS  Google Scholar 

  14. Onishi H, Katano M. Hedgehog signaling pathway as a new therapeutic target in pancreatic cancer. World J Gastroenterol. 2014;20:2335–42.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Arensman MD, Nguyen P, Kershaw KM, Lay AR, Ostertag-Hill CA, Sherman MH, et al. Calcipotriol targets LRP6 to inhibit Wnt signaling in pancreatic cancer. Mol Cancer Res. 2015;13:1509–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15:28.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Sun M, Yao S, Fan L, Fang Z, Miao W, Hu Z, et al. Fibroblast activation protein-α responsive peptide assembling prodrug nanoparticles for remodeling the immunosuppressive microenvironment and boosting cancer immunotherapy. Small. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/smll.202106296.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Sitia L, Bonizzi A, Mazzucchelli S, Negri S, Sottani C, Grignani E, et al. Selective targeting of cancer-associated fibroblasts by engineered H-ferritin nanocages loaded with navitoclax. Cells. 2021;10:328.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Tan Y-N, Li Y-P, Huang J-D, Luo M, Li S-S, Lee AW-M, et al. Thermal-sensitive lipid nanoparticles potentiate anti-PD therapy through enhancing drug penetration and T lymphocytes infiltration in metastatic tumor. Cancer Lett. 2021;522:238–54.

    Article  PubMed  CAS  Google Scholar 

  20. Yang B, Zhang Y, Sun L, Wang J, Zhao Z, Huang Z, et al. Modulated ultrasmall γ-Fe2O3 nanocrystal assemblies for switchable magnetic resonance imaging and photothermal-ferroptotic-chemical synergistic cancer therapy. Adv Func Mater. 2023;33:2211251.

    Article  CAS  Google Scholar 

  21. Liang X, Yang Y, Huang C, Ye Z, Lai W, Luo J, et al. cRGD-targeted heparin nanoparticles for effective dual drug treatment of cisplatin-resistant ovarian cancer. J Control Release. 2023;356:691–701.

    Article  PubMed  CAS  Google Scholar 

  22. Jang H, Kim EH, Chi S-G, Kim SH, Yang Y. Nanoparticles targeting innate immune cells in tumor microenvironment. Int J Mol Sci. 2021;22:10009.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  24. Li Z, Shi H, Xie H, Yang Y, Zheng Y, Chen H, et al. Tri-component programmable nanoregulator with three-pronged penetration boosts immunotherapy of triple-negative breast cancer. Chem Eng J. 2022;439:135712.

    Article  CAS  Google Scholar 

  25. Wu S, Liu D, Li W, Song B, Chen C, Chen D, et al. Enhancing TNBC Chemo-immunotherapy via combination reprogramming tumor immune microenvironment with Immunogenic cell death. Int J Pharm. 2021;598:120333.

    Article  PubMed  CAS  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  PubMed  PubMed Central  CAS  Google Scholar 

  27. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Nurmik M, Ullmann P, Rodriguez F, Haan S, Letellier E. In search of definitions: cancer-associated fibroblasts and their markers. Int J Cancer. 2020;146:895–905.

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Piersma B, Hayward M-K, Weaver VM. Fibrosis and cancer: a strained relationship. Biochimica et Biophysica Acta (BBA) Rev Cancer. 2020;1873:188356.

    Article  CAS  Google Scholar 

  31. Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019;9:1102–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R, et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell. 2022;40:656-673.e7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Cirri P, Chiarugi P. Cancer associated fibroblasts: the dark side of the coin. Am J Cancer Res. 2011;1:482–97.

    PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  35. Kittirat Y, Suksawat M, Thongchot S, Padthaisong S, Phetcharaburanin J, Wangwiwatsin A, et al. Interleukin-6-derived cancer-associated fibroblasts activate STAT3 pathway contributing to gemcitabine resistance in cholangiocarcinoma. Front Pharmacol. 2022;13:897368.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Zhou Y, Tang W, Zhuo H, Zhu D, Rong D, Sun J, et al. Cancer-associated fibroblast exosomes promote chemoresistance to cisplatin in hepatocellular carcinoma through circZFR targeting signal transducers and activators of transcription (STAT3)/ nuclear factor -kappa B (NF-κB) pathway. Bioengineered. 2022;13:4786–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Zhou B, Lei J-H, Wang Q, Qu T-F, Cha L-C, Zhan H-X, et al. Cancer-associated fibroblast-secreted miR-421 promotes pancreatic cancer by regulating the SIRT3/H3K9Ac/HIF-1α axis. Kaohsiung J Med Sci. 2022;38:1080–92.

    Article  PubMed  CAS  Google Scholar 

  38. Yan Q, Liu J, Liu Y, Wen Z, Jin D, Wang F, et al. Tumor-associated macrophage-derived exosomal miR21-5p promotes tumor angiogenesis by regulating YAP1/HIF-1α axis in head and neck squamous cell carcinoma. Cell Mol Life Sci. 2024;81:179.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Sun D-Y, Wu J-Q, He Z-H, He M-F, Sun H-B. Cancer-associated fibroblast regulate proliferation and migration of prostatecancer cells through TGF-β signaling pathway. Life Sci. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2019.116791.

    Article  PubMed  Google Scholar 

  40. Xiao L, Zhu H, Shu J, Gong D, Zheng D, Gao J. Overexpression of TGF-β1 and SDF-1 in cervical cancer-associated fibroblasts promotes cell growth, invasion and migration. Arch Gynecol Obstet. 2022;305:179–92.

    Article  PubMed  CAS  Google Scholar 

  41. Liang D, Liu L, Zheng Q, Zhao M, Zhang G, Tang S, et al. Chelerythrine chloride inhibits the progression of colorectal cancer by targeting cancer-associated fibroblasts through intervention with WNT10B/β-catenin and TGFβ2/Smad2/3 axis. Phytother Res. 2023;37:4674–86.

    Article  PubMed  CAS  Google Scholar 

  42. Zhou Z. VCAM-1 secreted from cancer-associated fibroblasts enhances the growth and invasion of lung cancer cells through AKT and MAPK signaling. Cancer Lett. 2020;473:62–73.

    Article  PubMed  CAS  Google Scholar 

  43. Jin Y, Bian S, Wang H, Mo J, Fei H, Li L, et al. CRMP2 derived from cancer associated fibroblasts facilitates progression of ovarian cancer via HIF-1α-glycolysis signaling pathway. Cell Death Dis. 2022;13:1–13.

    Article  Google Scholar 

  44. Feng S, Ding B, Dai Z, Yin H, Ding Y, Liu S, et al. Cancer-associated fibroblast-secreted FGF7 as an ovarian cancer progression promoter. J Transl Med. 2024;22:280.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Asif PJ, Longobardi C, Hahne M, Medema JP. The role of cancer-associated fibroblasts in cancer invasion and metastasis. Cancers. 2021;13:4720.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Sobolik T, Su Y-J, Wells S, Ayers GD, Cook RS, Richmond A. CXCR4 drives the metastatic phenotype in breast cancer through induction of CXCR2 and activation of MEK and PI3K pathways. Mol Biol Cell. 2014;25:566–82.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Kato K, Fukai M, Hatanaka KC, Takasawa A, Aoyama T, Hayasaka T, et al. Versican secreted by cancer-associated fibroblasts is a poor prognostic factor in hepatocellular carcinoma. Ann Surg Oncol. 2022;29:7135–46.

    Article  PubMed  Google Scholar 

  48. Yuan Z, Hu H, Zhu Y, Zhang W, Fang Q, Qiao T, et al. Colorectal cancer cell intrinsic fibroblast activation protein alpha binds to Enolase1 and activates NF-κB pathway to promote metastasis. Cell Death Dis. 2021;12:543.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Ghofrani-Shahpar M, Pakravan K, Razmara E, Amooie F, Mahmoudian M, Heshmati M, et al. Cancer-associated fibroblasts drive colorectal cancer cell progression through exosomal miR-20a-5p-mediated targeting of PTEN and stimulating interleukin-6 production. BMC Cancer. 2024;24:400.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Wan X, Guan S, Hou Y, Qin Y, Zeng H, Yang L, et al. FOSL2 promotes VEGF-independent angiogenesis by transcriptionnally activating Wnt5a in breast cancer-associated fibroblasts. Theranostics. 2021;11:4975–91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Goggins E, Mironchik Y, Kakkad S, Jacob D, Wildes F, Bhujwalla ZM, et al. Reprogramming of VEGF-mediated extracellular matrix changes through autocrine signaling. Cancer Biol Ther. 2023;24:2184145.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Li J, Zhi X, Sun Y, Chen M, Yao L. The PDGF family is associated with activated tumor stroma and poor prognosis in ovarian cancer. Dis Mark. 2022;2022:5940049.

    Google Scholar 

  53. Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis. 2020;23:159–77.

    Article  PubMed  CAS  Google Scholar 

  54. Ge F, Zeng C, Wang J, Liu X, Zheng C, Zhang H, et al. Cancer-associated fibroblasts drive early pancreatic cancer cell invasion via the SOX4/MMP11 signalling axis. Biochimica et Biophysica Acta (BBA) Mol Basis of Dis. 2024;1870:166852.

    Article  CAS  Google Scholar 

  55. Banerjee K, Kerzel T, Bekkhus T, de Souza FS, Wallmann T, Wallerius M, et al. VEGF-C-expressing TAMs rewire the metastatic fate of breast cancer cells. Cell Rep. 2023;42:113507.

    Article  PubMed  CAS  Google Scholar 

  56. Zou S, Tong Q, Liu B, Huang W, Tian Y, Fu X. Targeting STAT3 in cancer immunotherapy. Mol Cancer. 2020;19:145.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Zhao Q, Huang L, Qin G, Qiao Y, Ren F, Shen C, et al. Cancer-associated fibroblasts induce monocytic myeloid-derived suppressor cell generation via IL-6/exosomal miR-21-activated STAT3 signaling to promote cisplatin resistance in esophageal squamous cell carcinoma. Cancer Lett. 2021;518:35–48.

    Article  PubMed  CAS  Google Scholar 

  58. Li Z, Low V, Luga V, Sun J, Earlie E, Parang B, et al. Tumor-produced and aging-associated oncometabolite methylmalonic acid promotes cancer-associated fibroblast activation to drive metastatic progression. Nat Commun. 2022;13:6239.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Zhang Y, Yin C, Wei C, Xia S, Qiao Z, Zhang X, et al. Exosomal miR-625-3p secreted by cancer-associated fibroblasts in colorectal cancer promotes EMT and chemotherapeutic resistance by blocking the CELF2/WWOX pathway. Pharmacol Res. 2022;186:106534.

    Article  PubMed  CAS  Google Scholar 

  60. Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, et al. The landscape of circular RNA in cancer. Cell. 2019;176:869-881.e13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Hu C, Xia R, Zhang X, Li T, Ye Y, Li G, et al. circFARP1 enables cancer-associated fibroblasts to promote gemcitabine resistance in pancreatic cancer via the LIF/STAT3 axis. Mol Cancer. 2022;21:24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Jin Z, Lu Y, Wu X, Pan T, Yu Z, Hou J, et al. The cross-talk between tumor cells and activated fibroblasts mediated by lactate/BDNF/TrkB signaling promotes acquired resistance to anlotinib in human gastric cancer. Redox Biol. 2021;46:102076.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–99.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Liu T, Han C, Wang S, Fang P, Ma Z, Xu L, et al. Cancer-associated fibroblasts: an emerging target of anti-cancer immunotherapy. J Hematol Oncol. 2019;12:86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Chen Y, Kim J, Yang S, Wang H, Wu C-J, Sugimoto H, et al. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell. 2021;39:548-565.e6

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. ia D, Zhou Z, Kwon O-J, Zhang L, Wei X, Zhang Y, et al. Stromal FOXF2 suppresses prostate cancer progression and metastasis by enhancing antitumor immunity. Nat Commun. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-022-34665-z.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Peng Q, Li S, Shi X, Guo Y, Hao L, Zhang Z, et al. Dihydroartemisinin broke the tumor immunosuppressive microenvironment by inhibiting YAP1 expression to enhance anti-PD-1 efficacy. Phytother Res. 2023;37:1740–53.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Lim SA, Su W, Chapman NM, Chi H. Lipid metabolism in T cell signaling and function. Nat Chem Biol. 2022;18:470–81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Guo S, Chen P, Yang Y, Wei W, Pan Y, Zeng F, et al. Tumor-to-stroma cd8+ t cells ratio combined with cancer-associated fibroblasts: an innovative approach to predicting lymph node metastases of cervical cancer. J Cancer Res Clin Oncol. 2024;150:93.

    Article  PubMed  CAS  Google Scholar 

  71. Yan R, Moresco P, Gegenhuber B, Fearon DT. T cell–mediated development of stromal fibroblasts with an immune-enhancing chemokine profile. Cancer Immunol Res. 2023;11:1044–54.

    Article  CAS  Google Scholar 

  72. Li C, Guo H, Zhai P, Yan M, Liu C, Wang X, et al. Spatial and single-cell transcriptomics reveal a cancer-associated fibroblast subset in HNSCC that restricts infiltration and antitumor activity of CD8+ T cells. Can Res. 2024;84:258–75.

    Article  CAS  Google Scholar 

  73. Lou M, Iwatsuki M, Wu X, Zhang W, Matsumoto C, Baba H. Cancer-associated fibroblast-derived IL-8 upregulates PD-L1 expression in gastric cancer through the NF-κB pathway. Ann Surg Oncol. 2024;31:2983–95.

    Article  PubMed  Google Scholar 

  74. Ge W, Yue M, Lin R, Zhou T, Xu H, Wang Y, et al. PLA2G2A+ cancer-associated fibroblasts mediate pancreatic cancer immune escape via impeding antitumor immune response of CD8+ cytotoxic T cells. Cancer Lett. 2023;558:216095.

    Article  PubMed  CAS  Google Scholar 

  75. Vienot A, Pallandre J-R, Renaude E, Viot J, Bouard A, Spehner L, et al. Chemokine switch regulated by TGF-β1 in cancer-associated fibroblast subsets determines the efficacy of chemo-immunotherapy. Oncoimmunology. 2022;11:2144669.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Chen W, Jin W, Hardegen N, Lei K-J, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Gong Y, Klein Wolterink RGJ, Wang J, Bos GMJ, Germeraad WTV. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. J Hematol Oncol. 2021;14:73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Vivier E, Rebuffet L, Narni-Mancinelli E, Cornen S, Igarashi RY, Fantin VR. Natural killer cell therapies. Nature. 2024;626:727–36.

    Article  PubMed  CAS  Google Scholar 

  79. Wong JKM, McCulloch TR, Alim L, Omer N, Mehdi AM, Tuong ZK, et al. TGF-β signalling limits effector function capacity of NK cell anti-tumour immunity in human bladder cancer. EBioMedicine. 2024;104:105176.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Wu X, Xiao Y, Guo D, Zhang Z, Liu M. Reduced NK cell cytotoxicity by papillomatosis-derived TGF-β contributing to low-risk HPV persistence in JORRP patients. Front Immunol. 2022;13:849493.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Otegbeye F, Ojo E, Moreton S, Mackowski N, Lee DA, de Lima M, et al. Inhibiting TGF-beta signaling preserves the function of highly activated, in vitro expanded natural killer cells in AML and colon cancer models. PLoS ONE. 2018;13: e0191358.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Kim H-A, Kim H, Nam M-K, Park JK, Lee M-Y, Chung S, et al. Suppression of the antitumoral activity of natural killer cells under indirect coculture with cancer-associated fibroblasts in a pancreatic TIME-on-chip model. Cancer Cell Int. 2023;23:219.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Ye J, Baer JM, Faget DV, Morikis VA, Ren Q, Melam A, et al. Senescent CAFs mediate immunosuppression and drive breast cancer progression. Cancer Discov. 2024;14:1302–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Lee S, Ricci B, Faccio R. Cancer-associated fibroblast-derived Dickkopf-1 impairs anti-tumor immunity in breast cancer by suppressing NK cell-mediated cytotoxicity. Cancer Res. 2024;84(6_Supplement):3925–3925.

    Article  Google Scholar 

  85. Yao L, Hou J, Wu X, Lu Y, Jin Z, Yu Z, et al. Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation. Redox Biol. 2023;67:102923.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Van den Eynde A, Gehrcken L, Verhezen T, Lau HW, Hermans C, Lambrechts H, et al. IL-15-secreting CAR natural killer cells directed toward the pan-cancer target CD70 eliminate both cancer cells and cancer-associated fibroblasts. J Hematol Oncol. 2024;17:8.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Lee YE, Go G-Y, Koh E-Y, Yoon H-N, Seo M, Hong S-M, et al. Synergistic therapeutic combination with a CAF inhibitor enhances CAR-NK-mediated cytotoxicity via reduction of CAF-released IL-6. J Immunother Cancer. 2023;11: e006130.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zhang W, Wang M, Ji C, Liu X, Gu B, Dong T. Macrophage polarization in the tumor microenvironment: emerging roles and therapeutic potentials. Biomed Pharmacother. 2024;177:116930.

    Article  PubMed  CAS  Google Scholar 

  89. Tang PC-T, Chung JY-F, Xue VW, Xiao J, Meng X-M, Huang X-R, et al. Smad3 promotes cancer-associated fibroblasts generation via macrophage-myofibroblast transition. Adv Sci. 2022;9:2101235.

    Article  CAS  Google Scholar 

  90. Cho H, Seo Y, Loke KM, Kim S-W, Oh S-M, Kim J-H, et al. Cancer-stimulated CAFs enhance monocyte differentiation and protumoral TAM activation via IL6 and GM-CSF secretion. Clin Cancer Res. 2018;24:5407–21.

    Article  PubMed  CAS  Google Scholar 

  91. Raskov H, Orhan A, Gaggar S, Gögenur I. Cancer-associated fibroblasts and tumor-associated macrophages in cancer and cancer immunotherapy. Front Oncol. 2021;11:668731.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, et al. Cancer-associated fibroblast-secreted IGFBP7 promotes gastric cancer by enhancing tumor associated macrophage infiltration via FGF2/FGFR1/PI3K/AKT axis. Cell Death Discov. 2023;9:17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Gu X, Zhu Y, Su J, Wang S, Su X, Ding X, Jiang L, Fei X, Zhang W. Lactate-induced activation of tumor-associated fibroblasts and IL-8-mediated macrophage recruitment promote lung cancer progression. Redox Biol. 2024;74:103209.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Li X, Bu W, Meng L, Liu X, Wang S, Jiang L, et al. CXCL12/CXCR4 pathway orchestrates CSC-like properties by CAF recruited tumor associated macrophage in OSCC. Exp Cell Res. 2019;378:131–8.

    Article  PubMed  CAS  Google Scholar 

  95. Zhang R, Zong J, Peng Y, Shi J, Du X, Liu H, et al. GPR30 knockdown weakens the capacity of CAF in promoting prostate cancer cell invasion via reducing macrophage infiltration and M2 polarization. J Cell Biochem. 2021;122:1173–91.

    Article  CAS  Google Scholar 

  96. Tokuda K, Morine Y, Miyazaki K, Yamada S, Saito Y, Nishi M, et al. The interaction between cancer associated fibroblasts and tumor associated macrophages via the osteopontin pathway in the tumor microenvironment of hepatocellular carcinoma. Oncotarget. 2021;12:333–43.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Yan W, Menjivar RE, Bonilla ME, Steele NG, Kemp SB, Du W, et al. Notch signaling regulates immunosuppressive tumor-associated macrophage function in pancreatic cancer. Cancer Immunol Res. 2024;12:91–106.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Maia A, Schöllhorn A, Schuhmacher J, Gouttefangeas C. CAF-immune cell crosstalk and its impact in immunotherapy. Semin Immunopathol. 2023;45:203–14.

    Article  PubMed  Google Scholar 

  99. Barrett RL, Puré E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. Elife. 2020;9:e57243–e57243.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Van Wilpe S, Koornstra R, Den Brok M, De Groot JW, Blank C, De Vries J, et al. Lactate dehydrogenase: a marker of diminished antitumor immunity. OncoImmunology. 2020;9:1731942.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Siska PJ, Singer K, Evert K, Renner K, Kreutz M. The immunological Warburg effect: can a metabolic-tumor-stroma score (MeTS) guide cancer immunotherapy? Immunol Rev. 2020;295:187–202.

    Article  PubMed  CAS  Google Scholar 

  102. Mostafavi S, Zalpoor H, Hassan ZM. The promising therapeutic effects of metformin on metabolic reprogramming of cancer-associated fibroblasts in solid tumors. Cell Mol Biol Lett. 2022;27:58.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Liang L, Li W, Li X, Jin X, Liao Q, Li Y, et al. ‘Reverse Warburg effect’ of cancer-associated fibroblasts (review). Int J Oncol. 2022;60:67.

    Article  PubMed  CAS  Google Scholar 

  104. Ippolito L, Comito G, Parri M, Iozzo M, Duatti A, Virgilio F, et al. Lactate rewires lipid metabolism and sustains a metabolic-epigenetic axis in prostate cancer. Can Res. 2022;82:1267–82.

    Article  CAS  Google Scholar 

  105. Nia HT, Munn LL, Jain RK. Physical traits of cancer. Science. 2020;370: eaaz0868.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, et al. The application of nanoparticles targeting cancer-associated fibroblasts. IJN. 2024;19:3333–65.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater. 2021;6:1973–87.

    PubMed  CAS  Google Scholar 

  108. Panagi M, Mpekris F, Chen P, Voutouri C, Nakagawa Y, Martin JD, et al. Polymeric micelles effectively reprogram the tumor microenvironment to potentiate nano-immunotherapy in mouse breast cancer models. Nat Commun. 2022;13:7165.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Zhang Y, Han X, Wang K, Liu D, Ding X, Hu Z, et al. Co-delivery nanomicelles for potentiating TNBC immunotherapy by synergetically reshaping CAFs-mediated tumor stroma and reprogramming immunosuppressive microenvironment. Int J Nanomed. 2023;18:4329–46.

    Article  CAS  Google Scholar 

  110. Yang W, Yi J, Zhu R, Guo Y, Zhang K, Cao Y, et al. Transformable prodrug nanoplatform via tumor microenvironment modulation and immune checkpoint blockade potentiates immunogenic cell death mediated cancer immunotherapy. Theranostics. 2023;13:1906–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Pan J, Lai Y, Zhang S, Zhang H, Shan Y, Huang L, et al. Self-adaptive nanoregulator to mitigate dynamic immune evasion of pancreatic cancer. Adv Mater. 2023;35:2305798.

    Article  CAS  Google Scholar 

  112. Li X, Luo Y, Huang Z, Wang Y, Wu J, Zhou S. Multifunctional liposomes remodeling tumor immune microenvironment for tumor chemoimmunotherapy. Small Methods. 2023;7:2201327.

    Article  CAS  Google Scholar 

  113. Yu H, Zhu W, Lin C, Jia M, Tan X, Yuan Z, et al. Stromal and tumor immune microenvironment reprogramming through multifunctional cisplatin-based liposomes boosts the efficacy of anti-PD-1 immunotherapy in pancreatic cancer. Biomater Sci. 2024;12:116–33.

    Article  CAS  Google Scholar 

  114. Chen Y, Hu M, Wang S, Wang Q, Lu H, Wang F, et al. Nano-delivery of salvianolic acid B induces the quiescence of tumor-associated fibroblasts via interfering with TGF-β1/Smad signaling to facilitate chemo- and immunotherapy in desmoplastic tumor. Int J Pharm. 2022;623:121953.

    Article  PubMed  CAS  Google Scholar 

  115. Dong S, Zhang Y, Guo X, Zhang C, Wang Z, Yu J, et al. Glutathione pulse therapy: promote spatiotemporal delivery of reduction-sensitive nanoparticles at the “cellular level” and synergize PD-1 blockade therapy. Adv Sci. 2022;9:2202744.

    Article  CAS  Google Scholar 

  116. Yu Q, Tang X, Zhao W, Qiu Y, He J, Wan D, et al. Mild hyperthermia promotes immune checkpoint blockade-based immunotherapy against metastatic pancreatic cancer using size-adjustable nanoparticles. Acta Biomater. 2021;133:244–56.

    Article  PubMed  CAS  Google Scholar 

  117. Hua P, Jiang D, Guo Z, Tian H, Chen X, Chen M. Amplified cancer immunotherapy of PD-L1 blockade by sequential tumor microenvironment reshaping and DC maturation. Chem Eng J. 2023;453:139795.

    Article  CAS  Google Scholar 

  118. Ramzy A, Soliman AH, Hassanein SI, Sebak AA. Multitarget, multiagent PLGA nanoparticles for simultaneous tumor eradication and TME remodeling in a melanoma mouse model. Drug Deliv Transl Res. 2023;14:491–509.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Zheng F, Luo Y, Liu Y, Gao Y, Chen W, Wei K. Nano-baicalein facilitates chemotherapy in breast cancer by targeting tumor microenvironment. Int J Pharm. 2023;635:122778.

    Article  PubMed  CAS  Google Scholar 

  120. Tong Q-S, Miao W-M, Huang H, Luo J-Q, Liu R, Huang Y-C, et al. A tumor-penetrating nanomedicine improves the chemoimmunotherapy of pancreatic cancer. Small. 2021;17:2101208.

    Article  CAS  Google Scholar 

  121. Guo D, Ji X, Xie H, Ma J, Xu C, Zhou Y, Chen N, Wang H, Fan C, Song H, et al. Targeted reprogramming of vitamin B3 metabolism as a nanotherapeutic strategy towards chemoresistant cancers. Adv Mater. 2023;35:2301257.

    Article  CAS  Google Scholar 

  122. Hou L, Chen D, Wang R, Wang R, Zhang H, Zhang Z, et al. Transformable honeycomb-like nanoassemblies of carbon dots for regulated multisite delivery and enhanced antitumor chemoimmunotherapy. Angew Chem-Int Edit. 2021;60:6581–92.

    Article  CAS  Google Scholar 

  123. Zhao Y, Xie Z, Deng Y, Huang A, He Y, Wen B, et al. Photothermal nanobomb blocking metabolic adenosine-A2AR potentiates infiltration and activity of T cells for robust antitumor immunotherapy. Chem Eng J. 2022;450:138139.

    Article  CAS  Google Scholar 

  124. Zheng D, Wan C, Yang H, Xu L, Dong Q, Du C, Du J, Li F, et al. Her2-targeted multifunctional nano-theranostic platform mediates tumor microenvironment remodeling and immune activation for breast cancer treatment. Int J Nanomed. 2020;15:10007–28.

    Article  CAS  Google Scholar 

  125. Meng J, Bao W, Liu M, Ma Z, Tian Z. MOFs-based nanoagents enable sequential damage to cancer-associated fibroblast and tumor cells for phototriggered tumor microenvironment regulation. Small. 2024;20:2304491.

    Article  CAS  Google Scholar 

  126. Ni K, Xu Z, Culbert A, Luo T, Guo N, Yang K, et al. Synergistic checkpoint-blockade and radiotherapy–radiodynamic therapy via an immunomodulatory nanoscale metal–organic framework. Nat Biomed Eng. 2022;6:144–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Guo H, Liu Y, Li X, Wang H, Mao D, Wei L, et al. Magnetic metal-organic framework-based nanoplatform with platelet membrane coating as a synergistic programmed cell death protein 1 inhibitor against hepatocellular carcinoma. ACS Nano. 2023;17:23829–49.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. He Z, Zhou H, Feng Q, Zhang Y, Gao S, Liu S, et al. A bimetallic dual-targeting nanoplatform for combinational ferroptosis activation/epigenetic regulation/photothermal therapy against breast cancer and tumor microenvironment remodeling. Chem Eng J. 2024;479:147466.

    Article  CAS  Google Scholar 

  129. Jin J, Krishnamachary B, Barnett JD, Chatterjee S, Chang D, Mironchik Y, et al. Human cancer cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated invasion and metastasis and induce T-cells. ACS Appl Mater Interfaces. 2019;11:7850–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Zang S, Huang K, Li J, Ren K, Li T, He X, et al. Metabolic reprogramming by dual-targeting biomimetic nanoparticles for enhanced tumor chemo-immunotherapy. Acta Biomater. 2022;148:181–93.

    Article  PubMed  CAS  Google Scholar 

  131. Jia N, Wang Q, Li W, Chen D, Hu H. Membrane fusion liposomes deliver antifibrotic and chemotherapeutic drugs sequentially to enhance tumor treatment efficacy by reshaping tumor microenvironment. Adv Healthcare Mater. 2024;13:2400219.

    Article  CAS  Google Scholar 

  132. Wu Y, Chen R, Ni S, Hu K. Biomimetic, “nano-spears” for CAFs-targeting: splintered three “shields” with enhanced cisplatin anti-TNBC efficiency. J Control Release. 2024;370:556–69.

    Article  PubMed  CAS  Google Scholar 

  133. 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;17:13611–26.

    Article  PubMed  CAS  Google Scholar 

  134. Donthireddy L, Vonteddu P, Murthy T, Kwak T, Eraslan R-N, Podojil JR, et al. ONP-302 nanoparticles inhibit tumor growth by altering tumor-associated macrophages and cancer-associated fibroblasts. J Cancer. 2022;13:1933–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Hwang D, Ramsey JD, Kabanov AV. Polymeric micelles for the delivery of poorly soluble drugs: from nanoformulation to clinical approval. Adv Drug Deliv Rev. 2020;156:80–118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Ghezzi M, Pescina S, Padula C, Santi P, Del Favero E, Cantù L, et al. Polymeric micelles in drug delivery: an insight of the techniques for their characterization and assessment in biorelevant conditions. J Control Release. 2021;332:312–36.

    Article  PubMed  CAS  Google Scholar 

  137. Feng J, Xu M, Wang J, Zhou S, Liu Y, Liu S, et al. Sequential delivery of nanoformulated α-mangostin and triptolide overcomes permeation obstacles and improves therapeutic effects in pancreatic cancer. Biomaterials. 2020;241:119907.

    Article  PubMed  CAS  Google Scholar 

  138. Liang C, Zhang G, Guo L, Ding X, Yang H, Zhang H, et al. Spatiotemporal transformable nano-assembly for on-demand drug delivery to enhance anti-tumor immunotherapy. Asian J Pharm Sci. 2024;19:100888.

    PubMed  PubMed Central  Google Scholar 

  139. Cheng W-J, Lin S-Y, Chen M, Chen L-C, Ho H-O, Chuang K-H, et al. Active tumoral/tumor environmental dual-targeting by non-covalently arming with trispecific antibodies or dual-bispecific antibodies on docetaxel-loaded mPEGylated nanocarriers to enhance chemotherapeutic efficacy and minimize systemic toxicity. IJN. 2021;16:4017–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Schneider P, Zhang H, Simic L, Dai Z, Schrörs B, Akilli-Öztürk Ö, et al. Multicompartment polyion complex micelles based on triblock Polypept(o)ides mediate efficient siRNA delivery to cancer-associated fibroblasts for antistromal therapy of hepatocellular carcinoma. Adv Mater. 2024;36:2404784.

    Article  CAS  Google Scholar 

  141. Hari SK, Gauba A, Shrivastava N, Tripathi RM, Jain SK, Pandey AK. Polymeric micelles and cancer therapy: an ingenious multimodal tumor-targeted drug delivery system. Drug Deliv Transl Res. 2023;13:135–63.

    Article  PubMed  Google Scholar 

  142. Nsairat H, Khater D, Sayed U, Odeh F, Bawab AA, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon. 2022;8: e09394.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Guimarães D, Cavaco-Paulo A, Nogueira E. Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm. 2021;601:120571.

    Article  PubMed  Google Scholar 

  144. Wang A, Li S, Zhang R, Chen X, Zhu Y, Xia J, et al. Senescence-associated secretory phenotype regulation by dual drug delivery biomimetic nanoplatform for enhanced tumor chemotherapy. Mol Ther Oncol. 2024;32:200856.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Saraf S, Jain A, Tiwari A, Verma A, Panda PK, Jain SK. Advances in liposomal drug delivery to cancer: an overview. J Drug Deliv Sci Technol. 2020;56:101549.

    Article  CAS  Google Scholar 

  147. Wang S, Miao J, Zhu P, Xu L. Co-delivery of liposomal ketoconazole and bevacizumab for synergistical inhibition of angiogenesis against endometrial cancer. Mol Biotechnol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12033-024-01227-1.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016;99:28–51.

    Article  PubMed  CAS  Google Scholar 

  149. Yang M, Qin C, Tao L, Cheng G, Li J, Lv F, et al. Synchronous targeted delivery of TGF-β siRNA to stromal and tumor cells elicits robust antitumor immunity against triple-negative breast cancer by comprehensively remodeling the tumor microenvironment. Biomaterials. 2023;301:122253.

    Article  PubMed  CAS  Google Scholar 

  150. Guo J, Zeng H, Shi X, Han T, Liu Y, Liu Y, et al. A CFH peptide-decorated liposomal oxymatrine inactivates cancer-associated fibroblasts of hepatocellular carcinoma through epithelial-mesenchymal transition reversion. J Nanobiotechnol. 2022;20:114.

    Article  CAS  Google Scholar 

  151. Zhao T, Zhou H, Lei L, Guo C, Yang Q, Gong T, et al. A new tandem peptide modified liposomal doxorubicin for tumor “ecological therapy.” Nanoscale. 2020;12:3359–69.

    Article  PubMed  CAS  Google Scholar 

  152. Yang Z, Zhang L, Zhu H, Zhou K, Wang H, Wang Y, et al. Nanoparticle formulation of mycophenolate mofetil achieves enhanced efficacy against hepatocellular carcinoma by targeting tumour-associated fibroblast. J Cell Mol Med. 2021;25:3511–23.

    Google Scholar 

  153. Zielińska A, Carreiró F, Oliveira AM, Neves A, Pires B, Venkatesh DN, et al. Polymeric nanoparticles: production, characterization. Toxicol Ecotoxicol Mol. 2020;25:3731.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Wang L, Wu M, Pan Y, Xie D, Hong C, Li J, et al. Sequential targeting biomimetic nano platform for enhanced mild photothermal therapy and chemotherapy of tumor. Comput Struct Biotechnol J. 2023;21:2780–91.

    Article  PubMed  CAS  Google Scholar 

  155. He Y, Wu S, Yuan Y, Sun Y, Ai Q, Zhou R, et al. Remodeling tumor immunosuppression with molecularly imprinted nanoparticles to enhance immunogenic cell death for cancer immunotherapy. J Control Release. 2023;362:44–57.

    Article  PubMed  CAS  Google Scholar 

  156. Huang J, Zhang H, Ma L, Ma N, Luo N, Jin W, et al. Rhein and hesperidin nanoparticles remodel tumor immune microenvironment by reducing CAFs and CCL2 secreted by CAAs for efficient triple-negative breast cancer therapy. Int Immunopharmacol. 2024;141:113001.

    PubMed  PubMed Central  Google Scholar 

  157. Jiang M, He K, Qiu T, Sun J, Liu Q, Zhang X, et al. Tumor-targeted delivery of silibinin and IPI-549 synergistically inhibit breast cancer by remodeling the microenvironment. Int J Pharm. 2020;581:119239.

    Article  PubMed  CAS  Google Scholar 

  158. Zhang, H,Chen L,Zhao Y,Luo N,Shi J,Xu S, et al.Relaxin-encapsulated polymeric metformin nanoparticles remodel tumor immune microenvironment by reducing CAFs for efficient triple-negative breast cancer immunotherapy.Asian J Pharm Sci 18,2023;100796 .

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Wu Z, Tang Y, Liu Y, Chen Z, Feng Y, Hu H, et al. Co-delivery of fucoxanthin and Twist siRNA using hydroxyethyl starch-cholesterol self-assembled polymer nanoparticles for triple-negative breast cancer synergistic therapy. J Adv Res. 2024;S2090–1232(24):00160–7.

    Article  PubMed  Google Scholar 

  160. George A, Shah PA, Shrivastav PS. Natural biodegradable polymers based nano-formulations for drug delivery: a review. Int J Pharm. 2019;561:244–64.

    Google Scholar 

  161. Wang C, Wang Q, Wang H, Li Z, Chen J, Zhang Z, et al. Hydroxyethyl starch-folic acid conjugates stabilized theranostic nanoparticles for cancer therapy. J Control Release. 2022;353:391–410.

    Article  PubMed  CAS  Google Scholar 

  162. Y Zhang Z Fang D Pan Y Li J Zhou H Chen 2024 Dendritic polymer-based nanomedicines remodel the tumor stroma: improve drug penetration and enhance antitumor immune response Adv Mater 36 2401304 Zhang Y, Fang Z, Pan D, Li Y, Zhou J, Chen H, et al. Dendritic polymer-based nanomedicines remodel the tumor stroma: improve drug penetration and enhance antitumor immune response. Adv Mater. 2024;36:2401304.

    Article  CAS  Google Scholar 

  163. Ernsting M, Hoang B,Loshe I, UNdzys E, Gao P,Do T et al. Targeting of metastasis-promoting tumor-associated fibroblasts and modulation of pancreatic tumor-associated stroma with a carboxymethylcellulose-docetaxel nanoparticle.Journal of Controlled Release.2015;28:206:122-30.

    Article  Google Scholar 

  164. Tang L, Zhang A, Zhang Z, Zhao Q, Li J, Mei Y, et al. Multifunctional inorganic nanomaterials for cancer photoimmunotherapy. Cancer Commun. 2022;42:141–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. Chen Y-C, Huang X-C, Luo Y-L, Chang Y-C, Hsieh Y-Z, Hsu H-Y. Non-metallic nanomaterials in cancer theranostics: a review of silica- and carbon-based drug delivery systems. Sci Technol Adv Mater. 2013;14:044407.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Shahbazi M-A, Herranz B, Santos HA. Nanostructured porous Si-based nanoparticles for targeted drug delivery. Biomatter. 2012;2:296–312.

    Article  CAS  Google Scholar 

  167. Liu W, Wu B, Sun W, Liu W, Gu H, Du J, et al. Near-infrared II fluorescent carbon dots for differential imaging of drug-resistant bacteria and dynamic monitoring of immune system defense against bacterial infection in vivo. Chem Eng J. 2023;471:144530.

    Article  PubMed  CAS  Google Scholar 

  168. Battigelli A, Ménard-Moyon C, Bianco A. Carbon nanomaterials as new tools for immunotherapeutic applications. J Mater Chem B. 2014;2:6144–56.

    Article  PubMed  CAS  Google Scholar 

  169. Duan F, Wang J, Li Z, Zhang T, Li Z, Zhou X. pH-responsive metal-organic framework-coated mesoporous silica nanoparticles for immunotherapy. ACS Appl Nano Mater. 2021;4:13398–404.

    Article  CAS  Google Scholar 

  170. Argyo C, Weiss V, Bräuchle C, Bein T. Multifunctional mesoporous silica nanoparticles as a universal platform for drug delivery. Chem Mater. 2014;26:435–51.

    Article  PubMed  CAS  Google Scholar 

  171. Yang Y, Chen F, Xu N, Yao Q, Wang R, Xie X, et al. Red-light-triggered self-destructive mesoporous silica nanoparticles for cascade-amplifying chemo-photodynamic therapy favoring antitumor immune responses. Biomaterials. 2022;281:121368.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Guo Y, Fan Y, Li G, Wang Z, Shi X, Shen M. “Cluster bomb” based on redox-responsive carbon dot nanoclusters coated with cell membranes for enhanced tumor theranostics. ACS Appl Mater Interfaces. 2021;13:55815–26.

    Article  Google Scholar 

  173. Chen W, Ouyang J, Liu H, Chen M, Zeng K, Sheng J, et al. Black phosphorus nanosheet-based drug delivery system for synergistic photodynamic/photothermal/chemotherapy of cancer. Adv Mater. 2017;29:1603864.

    Article  Google Scholar 

  174. Tao W, Zhu X, Yu X, Zeng X, Xiao Q, Zhang X, et al. Black phosphorus nanosheets as a robust delivery platform for cancer theranostics. Adv Mater. 2017;29:1603276.

    Article  Google Scholar 

  175. Geng B, Shen W, Li P, Fang F, Qin H, Li XK, et al. Carbon dot-passivated black phosphorus nanosheet hybrids for synergistic cancer therapy in the NIR-II window. ACS Appl Mater Interfaces. 2019;11:44949–60.

    Article  PubMed  CAS  Google Scholar 

  176. Huang Y, Wang Y, Zheng T, Nie S, Wang Y, Shen H, et al. Development of Dual Diagnostic-Therapeutic Nanoformulation Effective Against Pancreatic Cancer in Animal Models. Int J Nanomedicine.2024;6:19:9121-9143

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Luo M, Fan T, Zhou Y, Zhang H, Mei L. 2D Black Phosphorus–Based Biomedical Applications. Advanced Functional Materials.2019;29:1808306.

    Article  CAS  Google Scholar 

  178. Yang H, Mu W, Yuan S, Yang H, Chang L, Sang X, et al. Self-delivery photothermal-boosted-nanobike multi-overcoming immune escape by photothermal/chemical/immune synergistic therapy against HCC. J Nanobiotechnol. 2024;22:137.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  179. Lei H, Pei Z, Jiang C, Cheng L. Recent progress of metal-based nanomaterials with anti-tumor biological effects for enhanced cancer therapy. Exploration. 2023;3:20220001.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Liu H, Mei Y, Zhao Q, Zhang A, Tang L, Gao H, et al. Black phosphorus, an emerging versatile nanoplatform for cancer immunotherapy. Pharmaceutics. 2021;13:1344.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Fang T, Zhang J, Zuo T, Wu G, Xu Y, Yang Y, et al. Chemo-photothermal combination cancer therapy with ROS scavenging, extracellular matrix depletion, and tumor immune activation by telmisartan and diselenide-paclitaxel prodrug loaded nanoparticles. ACS Appl Mater Interfaces. 2020;12:31292–308.

    Article  PubMed  CAS  Google Scholar 

  182. Kumar S, Shukla MK, Sharma AK, Jayaprakash GK, Tonk RK, Chellappan DK, et al. Metal-based nanomaterials and nanocomposites as promising frontier in cancer chemotherapy. MedComm. 2023;4: e253.

    Article  PubMed  CAS  Google Scholar 

  183. Luo Y, He X, Du Q, Xu L, Xu J, Wang J, et al. Metal-based smart nanosystems in cancer immunotherapy. Exploration. 2024;4:20230134.

    Article  CAS  Google Scholar 

  184. Liu J, Huang J, Zhang L, Lei J. Multifunctional metal–organic framework heterostructures for enhanced cancer therapy. Chem Soc Rev. 2021;50:1188–218.

    Article  PubMed  CAS  Google Scholar 

  185. Feng L, Wang K-Y, Day GS, Zhou H-C. The chemistry of multi-component and hierarchical framework compounds. Chem Soc Rev. 2019;48:4823–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Bindra AK, Wang D, Zhao Y. Metal-organic frameworks meet polymers: from synthesis strategies to healthcare applications. Adv Mater. 2023;35:2300700.

    Article  CAS  Google Scholar 

  187. Huang A, Li Q, Shi X, Gao J, Ma Y, Ding J, et al. An iron-containing nanomedicine for inducing deep tumor penetration and synergistic ferroptosis in enhanced pancreatic cancer therapy. Mater Today Bio. 2024;27:101132.

    Article  PubMed  CAS  Google Scholar 

  188. Shen M, Wu X, Zhu M, Yi X. Recent advances in biological membrane-based nanomaterials for cancer therapy. Biomater Sci. 2022;10:5756–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. Wei Y, Qin G, Wang Z, Zhao C, Ren J, Qu X. Bioorthogonal activation of TLR7 Agonists provokes innate immunity to reinforce aptamer-based checkpoint blockade. ACS Nano. 2023;17:5808–20.

    Article  PubMed  CAS  Google Scholar 

  190. Goñi FM. The basic structure and dynamics of cell membranes: an update of the singer-nicolson model. Biochimica et Biophysica Acta (BBA) Biomembranes. 2014;1838:1467–76.

    Article  PubMed  Google Scholar 

  191. Oroojalian F, Beygi M, Baradaran B, Mokhtarzadeh A, Shahbazi M-A. Immune cell membrane-coated biomimetic nanoparticles for targeted cancer therapy. Small. 2021;17:2006484.

    Article  CAS  Google Scholar 

  192. Ai X, Wang S, Duan Y, Zhang Q, Chen MS, Gao W, et al. Emerging approaches to functionalizing cell membrane-coated nanoparticles. Biochemistry. 2021;60:941–55.

    Article  PubMed  CAS  Google Scholar 

  193. Lee CE, Kim S, Park HW, Lee W, Jangid AK, Choi Y, et al. Tailoring tumor-recognizable hyaluronic acid-lipid conjugates to enhance anticancer efficacies of surface-engineered natural killer cells. Nano Converg. 2023;10:56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  194. Wu Y, Wan S, Yang S, Hu H, Zhang C, Lai J, et al. Macrophage cell membrane-based nanoparticles: a new promising biomimetic platform for targeted delivery and treatment. J Nanobiotechnol. 2022;20:542.

    Article  CAS  Google Scholar 

  195. Pang L, Qin J, Han L, Zhao W, Liang J, Xie Z, et al. Exploiting macrophages as targeted carrier to guide nanoparticles into glioma. Oncotarget. 2016;7:37081–91.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Du J, Chen X, Xu X, Que Z, Zhai M, Xiang Q, et al. Enhancing the tissue penetration to improve sonodynamic immunotherapy for pancreatic ductal adenocarcinoma using membrane-camouflaged nanoplatform. Eur J Nucl Med Mol Imaging. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00259-024-06952-y.

    Article  PubMed  Google Scholar 

  197. Zhang Y, Yu R, Zhao C, Liang J, Zhang Y, Su H, et al. CAFs homologous biomimetic liposome bearing BET inhibitor and pirfenidone synergistically promoting antitumor efficacy in pancreatic ductal adenocarcinoma. Adv Sci. 2024;11:2305279.

    Article  CAS  Google Scholar 

  198. Jian C, Wu T, Wang L, Gao C, Fu Z, Zhang Q, et al. Biomimetic nanoplatform for dual-targeted clearance of activated and senescent cancer-associated fibroblasts to improve radiation resistance in breast cancer. Small. 2024;20:2309279.

    Article  CAS  Google Scholar 

  199. 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  PubMed  CAS  Google Scholar 

  200. Zheng S, Wang J, Ding N, Chen W, Chen H, Xue M, et al. Prodrug polymeric micelles integrating cancer-associated fibroblasts deactivation and synergistic chemotherapy for gastric cancer. J Nanobiotechnol. 2021;19:381.

    Article  CAS  Google Scholar 

  201. Cao Y, Wen E, Chen Q, Li X, Wang Z. Multifunctional ICG-SB@Lip-ZA nanosystem focuses on remodeling the inflammatory-immunosuppressive microenvironment after photothermal therapy to potentiate cancer photothermal immunotherapy. Adv Healthc Mater. 2024;14: e2402211.

    Article  PubMed  Google Scholar 

  202. Zhu Y, Wang A, Zhang S, Kim J, Xia J, Zhang F, et al. Paclitaxel-loaded ginsenoside Rg3 liposomes for drug-resistant cancer therapy by dual targeting of the tumor microenvironment and cancer cells. J Adv Res. 2023;49:159–73.

    Article  PubMed  CAS  Google Scholar 

  203. Zhu X, Chen Q, Xie L, Chen W, Jiang Y, Song E, et al. Iron ion and sulfasalazine-loaded polydopamine nanoparticles for Fenton reaction and glutathione peroxidase 4 inactivation for enhanced cancer ferrotherapy. Acta Biomater. 2022;145:210–21.

    Article  PubMed  CAS  Google Scholar 

  204. Son S, Shin JM, Shin S, Kim CH, Lee JA, Ko H, et al. Repurposing macitentan with nanoparticle modulates tumor microenvironment to potentiate immune checkpoint blockade. Biomaterials. 2021;276:121058.

    Article  PubMed  CAS  Google Scholar 

  205. Liu S, Zhang M, Yu H, Sun X, Li Q, Yang M, et al. Immunoinducible carbon dot-incorporated hydrogels as a photothermal-derived antigen depot to trigger a robust antitumor immune response. ACS Appl Mater Interfaces. 2023;15:7700–12.

    Article  PubMed  CAS  Google Scholar 

  206. Zhong X, Sun X. Nanomedicines based on nanoscale metal-organic frameworks for cancer immunotherapy. Acta Pharmacol Sin. 2020;41:928–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. Li X, Yong T, Wei Z, Bie N, Zhang X, Zhan G, et al. Reversing insufficient photothermal therapy-induced tumor relapse and metastasis by regulating cancer-associated fibroblasts. Nat Commun. 2022;13:2794.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Xu H, Hu M, Liu M, An S, Guan K, Wang M, et al. Nano-puerarin regulates tumor microenvironment and facilitates chemo- and immunotherapy in murine triple negative breast cancer model. Biomaterials. 2020;235:119769.

    Article  CAS  Google Scholar 

  209. ei B, Mo Z, Yang J, Wang Z, Li S. Nanodrugs reprogram cancer-associated fibroblasts and normalize tumor vasculatures for sequentially enhancing photodynamic therapy of hepatocellular carcinoma. Int J Nanomed. 2023;18:6379–91.

    Article  CAS  Google Scholar 

  210. Xia J, Zhang S, Zhang R, Wang A, Zhu Y, Dong M, et al. Targeting therapy and tumor microenvironment remodeling of triple-negative breast cancer by ginsenoside Rg3 based liposomes. J Nanobiotechnol. 2022;20:414.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Shen S, Zhang Z, Huang H, Yang J, Tao X, Meng Z, et al. Copper-induced injectable hydrogel with nitric oxide for enhanced immunotherapy by amplifying immunogenic cell death and regulating cancer associated fibroblasts. Biomater Res. 2023;27:44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  212. Qiao M, Zhou F, Liu X, Jiang T, Wang H, Li X, et al. Targeting focal adhesion kinase boosts immune response in KRAS/LKB1 co-mutated lung adenocarcinoma via remodeling the tumor microenvironment. Exp Hematol Oncol. 2024;13:11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Liao S, Li J, Gao S, Han Y, Han X, Wu Y, et al. Sulfatinib, a novel multi-targeted tyrosine kinase inhibitor of FGFR1, CSF1R, and VEGFR1–3, suppresses osteosarcoma proliferation and invasion via dual role in tumor cells and tumor microenvironment. Front Oncol. 2023;13:1158857.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  214. Zhu H, Liu Q, Miao L, Musetti S, Huo M, Huang L. Remodeling the fibrotic tumor microenvironment of desmoplastic melanoma to facilitate vaccine immunotherapy. Nanoscale. 2020;12:3400–10.

    Article  CAS  Google Scholar 

  215. Shen W, Li Y, Yang Z, Li W, Cao Y, Liu Y, et al. Tumor microenvironment reprogramming combined with immunogenic enhancement by nanoemulsions potentiates immunotherapy. J Nanobiotechnol. 2024;22:154.

    Article  PubMed  CAS  Google Scholar 

  216. Shen W, Yao P, Li W, Gu C, Gao T, Cao Y, et al. Cancer-associated fibroblast-targeted nanodrugs reshape colorectal tumor microenvironments to suppress tumor proliferation, metastasis and improve drug penetration. J Mater Chem B. 2023;11:1871–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Akai M, Noma K, Kato T, Nishimura S, Matsumoto H, Kawasaki K, et al. Fibroblast activation protein-targeted near-infrared photoimmunotherapy depletes immunosuppressive cancer-associated fibroblasts and remodels local tumor immunity. Br J Cancer. 2024;130:1647–58.

    Article  PubMed  Google Scholar 

  218. Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol. 2020;17:657–74.

    Article  PubMed  Google Scholar 

  219. Zhang Z, Wang Z, Xiong Y, Wang C, Deng Q, Yang T, et al. A two-pronged strategy to alleviate tumor hypoxia and potentiate photodynamic therapy by mild hyperthermia. Biomater Sci. 2022;11:108–18.

    Article  CAS  Google Scholar 

  220. Liu J, Movahedi F, Sun B, Sun L, Zhang B, Wang J, et al. Immunostimulatory photochemotherapeutic nanocapsule for enhanced colon cancer treatment. Nanophotonics. 2021;10:3321–37.

    Article  PubMed  CAS  Google Scholar 

  221. Donohoe C, Senge MO, Arnaut LG, Gomes-da-Silva LC. Cell death in photodynamic therapy: from oxidative stress to anti-tumor immunity. Biochim Biophys Acta Rev Cancer. 2019;1872:188308.

    Article  PubMed  Google Scholar 

  222. Tian H, Zhu N, Wang H, Li Y, Yang Q, Chen H, et al. Self-oxygenated hydrogel enhances immune cell response and infiltration via triggering dual DNA damage to activate cGAS-STING and inhibiting CAFs. Small. 2024;20: e2403428.

    Article  PubMed  CAS  Google Scholar 

  223. Zhou S, Zhen Z, Paschall AV, Xue L, Yang X, Bebin-Blackwell A-G, et al. FAP-targeted photodynamic therapy mediated by ferritin nanoparticles elicits an immune response against cancer cells and cancer associated fibroblasts. Adv Funct Mater. 2021;31:2007017.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  224. Kawasaki K, Noma K, Kato T, Ohara T, Tanabe S, Takeda Y, et al. PD-L1-expressing cancer-associated fibroblasts induce tumor immunosuppression and contribute to poor clinical outcome in esophageal cancer. Cancer Immunol Immunother. 2023;72:3787–802.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Liu J, Li B, Li L, Ming X, Xu ZP. Advances in nanomaterials for immunotherapeutic improvement of cancer chemotherapy. Small. 2024;20: e2403024.

    Article  PubMed  Google Scholar 

  226. Das S, Valton J, Duchateau P, Poirot L. Stromal depletion by TALEN-edited universal hypoimmunogenic FAP-CAR T cells enables infiltration and anti-tumor cytotoxicity of tumor antigen-targeted CAR-T immunotherapy. Front Immunol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1172681.

    Article  CAS  Google Scholar 

  227. Li Y, Huang X, Li Y, Qiao Q, Chen C, Chen Y, et al. WRN nuclease-mediated EcDNA clearance enhances antitumor therapy in conjunction with trehalose dimycolate/mesoporous silica nanoparticles. Adv Sci. 2024;11:2407026.

    Article  PubMed  CAS  Google Scholar 

  228. Zhang Y, Liu Y, Li T, Yang X, Lang S, Pei P, et al. Engineered bacteria breach tumor physical barriers to enhance radio-immunotherapy. J Control Release. 2024;373:867–78.

Download references

Acknowledgements

This work was supported by the “Pioneer” and “Leading Goose” R&D Program of Zhejiang (2023C03062); Zhejiang Provincial Department of Health and Medical Affairs Project (2023KY422); Key Research and Development Plan of Lishui (2023zdyf14).

Funding

This work was supported by the “Pioneer” and “Leading Goose” R&D Program of Zhejiang (2023C03062); Zhejiang Provincial Department of Health and Medical Affairs Project (2023KY422); Key Research and Development Plan of Lishui (2023zdyf14).

Author information

Authors and Affiliations

Authors

Contributions

TL collected relevant literature, drafted manuscripts; XC and FQ prepared figures; XZ、FW、MX、MC reviewed the manuscript; JS、QS、JJ guided the preparation of this manuscript and provides funding support. All authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Jia-Wei Shen, Qiying Shen or Jiansong Ji.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors of this study agreed to publish.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

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

The original online version of this article was revised: Chinese characters were removed and the article has been updated.

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liao, T., Chen, X., Qiu, F. et al. Regulation of cancer-associated fibroblasts for enhanced cancer immunotherapy using advanced functional nanomedicines: an updated review. J Nanobiotechnol 23, 166 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-025-03217-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-025-03217-0

Keywords