Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment by promoting tumor growth, immune evasion, and metastasis. Recently, drug delivery systems targeting CAFs have emerged as a promising long-term and effective approach to cancer treatment. Advances in nanotechnology, in particular, have led to the development of nanomedicine delivery systems designed specifically to target CAFs, offering new possibilities for precise and personalized cancer therapies. This article reviews recent progress in drug delivery using nanocarriers that target CAFs. Additionally, we explore the potential of combining multiple therapies, such as chemotherapy and immunotherapy, with nanocarriers to enhance efficacy and overcome drug resistance. Although many preclinical studies show promise, the clinical application of nanomedicine still faces considerable challenges, especially in terms of drug penetration and large-scale production. Therefore, this review aims to provide a fresh perspective on CAF-targeted drug delivery systems and highlight potential future research directions and clinical applications.

Cancer-associated fibroblasts (CAFs) are a key part of the tumor microenvironment (TME) and play a crucial role in the initiation, growth, and treatment of tumors.1 These cells help tumor cells grow, invade, and spread by releasing substances such as cytokines, matrix metalloproteinases (MMPs), and extracellular vesicles.2,3 They also change the extracellular matrix (ECM), which strengthens the tumor's barrier and limits the ability of standard anti-tumor drugs to penetrate.4,5 Additionally, CAFs can suppress the immune response against tumors and help tumors escape from the immune system.6 Because of their multiple roles in the TME, targeting CAFs has become an important focus in cancer treatment research.7,8 To solve this problem, drug delivery systems targeting CAFs have been developed. These systems create targeted strategies based on the unique behavior of CAFs. For example, they use surface markers like α-SMA and FAP or regulate specific biological signals in the tumor microenvironment to directly target CAFs.9–11 This approach not only suppresses the “support” effect of CAFs on tumors but also indirectly weakens the growth capacity of the tumor itself. So far, nanomedicine delivery systems (NDDS) have shown significant advantages in targeting CAFs.12 Nanodrugs improve drug accumulation in tumor tissues through enhanced permeability and retention (EPR) effects, thanks to their unique properties,13,14 such as controllable particle size, diverse surface modifications, and excellent biocompatibility.15 At the same time, nanocarriers can carry multiple drugs, enhancing treatment effectiveness through combined therapeutic strategies. For example, they can deliver both chemotherapeutic drugs and immunomodulators together to overcome tumor drug resistance.16,17 In addition, responsive nanocarriers can also intelligently release drugs under specific stimuli in the tumor microenvironment, such as acidity, redox states, or enzymes, further improving the precision of treatment.18,19 At present, significant progress has been made in nanomedicine targeting CAFs, showing great potential for future use.12,20 Various types of nanocarriers, including liposomes, polymer nanoparticles and metal nanomaterials, are widely used in targeted design.21 These carriers bind efficiently to CAFs by using specific targeting molecules on their surface, improving selectivity and therapeutic effectiveness.22,23 The development of multifunctional nanoplatforms has opened up new possibilities for treating tumors, allowing for the combined delivery of drugs and enabling imaging and real-time monitoring of treatment.24,25 However, nanomedicine targeting CAFs still faces challenges related to permeability, manufacturing processes, and long-term safety, requiring further breakthroughs for clinical application.26,27 We hope that more research results will soon be applied in clinical practice.

Over the past few years, an increasing number of studies have revealed the heterogeneity of CAFs, suggesting that CAFs are not a single-cell population, but are composed of multiple subtypes, each of which plays a different role in different stages and types of tumors.28–30 Common subtypes of CAFs include myofibroblast-type CAFs (myCAFs), inflammatory-type CAFs (iCAFs), antigen-presenting-type CAFs (apCAFs), and vascular-associated-type CAFs (vCAFs).31,32 Each subtype has unique molecular markers, functional characteristics, and potential implications for cancer treatment32 (Fig. 1). MyCAFs are primarily involved in the synthesis and deposition of ECM components,33 such as collagen and fibronectin. This process leads to tumor tissues exhibiting distinct fibrotic features, which, in turn, affect intercellular signaling and substance exchange, thereby creating a microenvironment conducive to tumor cell growth.34,35 Additionally, myCAFs can activate migration-related signaling pathways in tumor cells, such as PI3K-Akt and MAPK, through the secretion of growth factors, cytokines, and other signaling molecules.36,37 This activation promotes the migration and invasiveness of tumor cells.38 

FIG. 1.

The origin, subtypes, and functions of CAFs in the TME. CAFs can originate from various cellular sources, including normal resident fibroblasts, adipocytes, mesenchymal stem cells, epithelial cells undergoing epithelial-mesenchymal transition (EMT), and cancer stem cells. CAFs can also arise from vascular origins, such as pericytes, endothelial cells, and smooth muscle cells, and through endothelial-to-mesenchymal transition (EndMT). The subtypes of CAFs are classified based on their distinct functional roles in tumor progression, which include: myofibroblastic CAFs (MyCAFs), which contribute to matrix remodeling and tumor growth; inflammatory CAFs (iCAFs), involved in inflammatory responses within the tumor microenvironment; vascular CAFs (vCAFs), which support angiogenesis and vascular development; chemo-resistant CAFs (ChemoCAFs), which mediate chemoresistance in tumors; antigen-presenting CAFs (apCAFs), which are involved in immune modulation by presenting antigens; SMA+ Collagen I-producing CAFs, which play a role in fibrosis and extracellular matrix production; and interferon-licensed CAFs (IFN-CAFs), which regulate immune responses and inflammation. These CAF subtypes fulfill various functions including metabolic reprogramming, matrix remodeling, tumor growth promotion, angiogenesis, drug resistance, inflammation, and immune modulation. Reproduced with permission from Coursier and Calvo, Cell Oncol. 47(4), 1091–1112 (2024). Copyright 2024 by the authors.61 

FIG. 1.

The origin, subtypes, and functions of CAFs in the TME. CAFs can originate from various cellular sources, including normal resident fibroblasts, adipocytes, mesenchymal stem cells, epithelial cells undergoing epithelial-mesenchymal transition (EMT), and cancer stem cells. CAFs can also arise from vascular origins, such as pericytes, endothelial cells, and smooth muscle cells, and through endothelial-to-mesenchymal transition (EndMT). The subtypes of CAFs are classified based on their distinct functional roles in tumor progression, which include: myofibroblastic CAFs (MyCAFs), which contribute to matrix remodeling and tumor growth; inflammatory CAFs (iCAFs), involved in inflammatory responses within the tumor microenvironment; vascular CAFs (vCAFs), which support angiogenesis and vascular development; chemo-resistant CAFs (ChemoCAFs), which mediate chemoresistance in tumors; antigen-presenting CAFs (apCAFs), which are involved in immune modulation by presenting antigens; SMA+ Collagen I-producing CAFs, which play a role in fibrosis and extracellular matrix production; and interferon-licensed CAFs (IFN-CAFs), which regulate immune responses and inflammation. These CAF subtypes fulfill various functions including metabolic reprogramming, matrix remodeling, tumor growth promotion, angiogenesis, drug resistance, inflammation, and immune modulation. Reproduced with permission from Coursier and Calvo, Cell Oncol. 47(4), 1091–1112 (2024). Copyright 2024 by the authors.61 

Close modal

In contrast, iCAFs secrete various inflammatory factors, including interleukin-6 (IL-6), IL-11, CXCL1, CXCL2, and others.39,40 These factors activate cancer cells and contribute to immune regulation, thereby fostering a tumor microenvironment that promotes tumor cell growth and immune escape.31,41 For instance, IL-6 secreted by iCAFs activates the STAT3 signaling pathway, which promotes the migration and invasion of Tregs.42 This signaling pathway enhances the proliferation and immunosuppressive function of Tregs, while inhibiting the activity of CTLs, ultimately weakening the body's anti-tumor immune response.43,44

However, a distinctive feature of apCAFs is the expression of MHC class II molecules, granting them the potential ability to present antigens. This allows apCAFs to uptake, process, and present tumor-associated antigens, thereby participating in the regulation of tumor immune responses.45,46 Research has also shown that apCAFs can partially activate CD4+ T cells in vitro in an antigen-dependent manner.47,48 However, due to their low expression of co-stimulatory molecules (e.g., CD80, CD86, and CD40), apCAFs may not fully activate T cells. This limitation results in a reduced strength and persistence of the T cell immune response.47,49 vCAFs are primarily involved in promoting blood vessel formation and remodeling in tumors. They are often located around tumor blood vessels and play a key role in angiogenesis by secreting growth factors such as VEGF, FGF, and other molecules that promote blood vessel formation.50,51 vCAFs not only enhance tumor growth by supplying nutrients and oxygen through improved blood flow, but they also help create a tumor microenvironment that facilitates metastasis and immune evasion.52,53 Targeting vCAFs has become a promising therapeutic strategy for inhibiting tumor vascularization and limiting tumor progression.54 

Indeed, the proportions of each subtype of CAFs vary in different tumor types, and several studies have suggested that balancing the number of CAF subgroups may provide important clinical benefits.55–57 For instance, vCAFs are found to be particularly abundant in tumors like lung cancer and gastric cancer, where they contribute to tumor blood supply and metastasis.58 On the other hand, myCAFs are more prevalent in fibrotic tumors such as pancreatic cancer.59 Despite the progress made in CAF subtype research, many questions remain unresolved. Future studies must further investigate the mechanisms underlying the interconversion between CAF subtypes and clarify the specific differences in their roles across various tumor types and individuals. This will provide a crucial theoretical foundation for early tumor diagnosis, personalized treatment, and prognostic assessment.55,60

An important direction in cancer therapy research is the targeted treatment of tumors using specific CAF markers.62,63 CAF markers are not only markers of cell types in the tumor microenvironment, but they also reveal the biological functions of these cells in tumor initiation and development.62,63 The use of these biomarkers to target CAF can help inhibit tumor growth and metastasis and improve the efficacy of existing treatments.64 

Alpha-smooth muscle actin (α-SMA), fibroblast activation protein (FAP), and other markers are commonly used to identify CAFs. α-SMA is a protein highly expressed in smooth muscle cells and myofibroblasts, and its expression reflects the activation status of CAFs. Therefore, α-SMA expression is significantly upregulated in CAFs and is one of the most widely used markers for identifying them.65 During tumorigenesis, normal fibroblasts are stimulated by signals such as transforming growth factor-β (TGF-β) secreted by tumor cells, undergoing phenotypic transformation and beginning to express α-SMA. This change enables them to contract and secrete extracellular matrix, thereby promoting tumor growth and metastasis.66,67 Additionally, the expression level of α-SMA is closely related to tumor malignancy and prognosis. Studies have shown that breast cancer patients with high α-SMA expression tend to have a higher risk of recurrence and lower survival rates.68,69 FAP, a membrane-bound serine protease, is highly expressed in CAFs, while it is either expressed at low levels or absent in normal fibroblasts. FAP plays a significant role in remodeling the extracellular matrix, as well as in the proliferation and migration of tumor cells, thus promoting tumor growth and metastasis.50,68 For instance, in pancreatic cancer, FAP-expressing CAFs can degrade extracellular matrix components such as collagen, opening a pathway for tumor cell metastasis.70,71 Platelet-derived growth factor receptor (PDGFR), including its two isoforms, PDGFRα and PDGFRβ, is another classic CAF marker. When bound to platelet-derived growth factor (PDGF), PDGFR activates downstream signaling pathways that promote the proliferation, migration, and activation of CAFs.72,73 In lung cancer, for example, the activation of PDGFR induces CAFs to secrete various cytokines and growth factors, such as TGF-β and VEGF, which further enhance tumor growth and angiogenesis.74,75

In addition to these classic markers, several emerging markers of CAFs have been identified. For example, in gastric cancer tissues, Vimentin-positive CAFs are closely associated with the depth of tumor invasion and lymph node metastasis. Vimentin expression levels can also serve as a prognostic indicator for tumors.76 In ovarian cancer, NRP-1-positive CAFs promote tumor growth and metastasis through interactions with tumor cells.77,78 In breast cancer, POSTN-positive CAFs secrete various growth factors and cytokines, activating signaling pathways within tumor cells and promoting their proliferation and migration.79,80 Interestingly, the role of Podoplanin-expressing CAFs varies across different tumor types. In small-cell carcinoma of the lung, Podoplanin-positive CAFs may inhibit tumor cell growth by secreting inhibitory cytokines or modulating immune cell activity.81 However, in lung adenocarcinoma and squamous cell carcinoma, Podoplanin-positive CAFs are associated with poor prognosis and may promote tumor invasion and metastasis.82,83

With advances in detection technologies, a range of CAF markers have been identified using methods such as single-cell sequencing,30 including laminin (LN), matrix metalloproteinases MMP, epithelial cell adhesion molecules (EPCAM), and mannose-binding lectin (MBL).84–87 Furthermore, in breast CAFs, specific genes such as NOTCH3 and HES4 have been identified as markers associated with CAF self-renewal and proliferation.78 This discovery suggests a potential new approach to combine gene technology with these specific markers and provides a promising direction for future research. Although various CAF markers have been identified, significant challenges remain in their application for cancer treatment. The heterogeneity of CAFs results in variations in marker expression across different tumor types and patients, complicating the uniform identification and application of these markers.28,88 Furthermore, some CAF markers are not exclusive to CAFs and may also be expressed in other cell types, which increases the complexity and the risk of false positives in marker detection. Future research should focus on further investigating the heterogeneity of CAFs and identifying more specific CAF markers. This will enhance the accuracy and reliability of CAF identification, thereby contributing to the development of targeted therapeutic strategies and improving the effectiveness of cancer treatments.

The tumor microenvironment (TME) is a complex network comprising various cells and molecules, including tumor cells, immune cells, endothelial cells, fibroblasts, and extracellular matrix components. Within this microenvironment, CAFs serve as key regulators, contributing to tumor initiation and progression. They influence tumor growth, metastasis, immune escape, and treatment response through interactions with other elements of the TME.62,89 As such, understanding the mechanisms by which CAFs function in the TME and targeting these pathways for cancer treatment has become a critical focus of current cancer research.27,90

Indeed, the interactions between CAFs and the TME are reciprocal. CAFs regulate the TME by secreting various growth factors, chemokines, and other molecules, all of which are essential for tumor formation and progression.91,92 For example, CAFs secrete signaling molecules involved in ECM remodeling, which in turn strengthens the structural framework required for tumor expansion. This remodeling provides a conducive environment for tumor cells to migrate and proliferate.5,93,94 Additionally, CAFs support tumor cells by participating in metabolic processes, such as regulating glucose, pH levels, and mitochondrial function. They also generate mechanical forces that influence tumor cell motility and morphology, thereby promoting tumor dissemination.95 Simultaneously, tumors and tumor cells can influence CAFs activation and function.96,97 CAFs are activated through pathways like direct contact or Notch signaling, which leads to ECM remodeling and creates a positive feedback loop.98,99 In pancreatic ductal adenocarcinoma (PDAC), for instance, tumor cells induce CAFs and autophagy to produce an “inverse Warburg effect,” releasing amino acids like alanine to support cancer cell metabolism.100–102 Furthermore, CAFs engage in transmembrane signaling with other cells within the TME. Through pathways involving PDGF, IL-6, and tumor necrosis factor-alpha (TNF-α), CAFs communicate with neighboring cells, thus influencing disease progression.98,103 These chemokines act as signals that recruit immune cells and other cell types to the tumor site. However, rather than enhancing an effective anti-tumor response, these recruited immune cells often contribute to an immunosuppressive environment. This occurs because CAFs can modulate immune cell function, transforming them from potential tumor fighters into supporters of tumor growth and metastasis.97,104

In addition to the influence of CAFs on the TME, the TME itself significantly impacts the biological characteristics and functions of CAFs. For example, high concentrations of hyaluronic acid (HA) or oxidative stress in the TME can stimulate CAFs to exhibit more aggressive, tumor-promoting behavior.105 These environmental factors alter CAFs' molecular activity, enhancing their secretion of growth factors, chemokines, and other molecules that support tumor growth and metastasis.104,106 Despite the progress in understanding these interactions, many uncertainties remain regarding how different TMEs influence CAF behavior.27,107 Future research is expected to provide deeper insights and breakthroughs in understanding these complex interactions, which could potentially lead to the development of novel therapeutic strategies targeting CAFs and their interactions within the TME.108 

In recent years, nanomaterials have become an important tool for targeting CAFs due to their unique physicochemical properties.109,110 These materials provide precise drug delivery, better drug targeting, improved tissue penetration, and more control over drug release, all of which contribute to enhanced therapeutic effectiveness.5,12,111,112 The use of various nanomaterials in targeted therapy not only showcases their individual benefits but also opens up many opportunities for further research and clinical applications.112,113 Below, we discuss several types of nanomaterials, their applications, and their effects in CAF-targeted therapy (Table I).

TABLE I.

Nanomaterials and their applications in targeting CAFs.

Material name Cancer Mechanism Effect Refs.
Gold nanoparticles (AuNPs)  Pancreatic cancer, breast cancer  AuNPs target CAF-specific proteins (e.g., FAP) through peptide modification and use near-infrared photothermal effects to ablate CAFs and reduce extracellular matrix density.  Reduces CAF activity, improves drug penetration, enhances anti-tumor immune responses.  26, 119, 145  
Metal-organic frameworks (MOFs)  Pancreatic cancer, breast cancer  MOFs encapsulate chemotherapeutic drugs and are modified with CAF-targeting peptides. Near-infrared light triggers drug release, inducing CAF apoptosis.  Remodels the tumor microenvironment, reduces CAF density, and enhances drug efficacy.  146–148  
Selenium nanoparticles (SeNPs)  Lung cancer  SeNPs inhibit CAF secretion of pro-inflammatory factors and enhance antioxidant capabilities, reducing CAF tumor-promoting activity.  Suppresses tumor-associated inflammation and blocks CAF support for tumor growth.  149, 150  
Peptide-functionalized silica nanoparticles  Pancreatic cancer, prostate cancer  Targets FAP-α on CAF surfaces, releasing quercetin-loaded nanoparticles to inhibit CAF activity  Decreases extracellular matrix density and improves tumor drug penetration.  141, 151  
Polymeric nanoparticles (PLGA-NPs)  Breast cancer, colorectal cancer  PLGA nanoparticles loaded with resveratrol target active CAF regions, reducing their immunosuppressive effects.  Remodels the immune microenvironment and enhances immunotherapy efficacy.  152, 153  
Magnetic nanoparticles (MNPs)  Breast cancer, Colorectal cancer  MNPs are guided by external magnetic fields to deliver paclitaxel or small-molecule inhibitors, targeting CAF functions.  Enhances drug delivery efficiency and reduces CAF-mediated drug resistance.  154, 155  
Liposome nanoparticles  Pancreatic cancer, breast cancer  FAP-targeted liposomes deliver chemotherapeutic drugs (e.g., paclitaxel) to inhibit CAF secretion of tumor-promoting factors.  Reduces tumor stroma barriers and enhances the effectiveness of chemotherapeutic drugs.  151, 156–158  
Iron oxide nanoparticles (FeNPs)  Lung cancer, breast cancer  Targets CAFs via the TGF-β signaling pathway, releasing anti-fibrotic drugs to suppress CAF activity.  Reduces CAF density and extracellular matrix, improving immune cell infiltration.  159, 160  
Material name Cancer Mechanism Effect Refs.
Gold nanoparticles (AuNPs)  Pancreatic cancer, breast cancer  AuNPs target CAF-specific proteins (e.g., FAP) through peptide modification and use near-infrared photothermal effects to ablate CAFs and reduce extracellular matrix density.  Reduces CAF activity, improves drug penetration, enhances anti-tumor immune responses.  26, 119, 145  
Metal-organic frameworks (MOFs)  Pancreatic cancer, breast cancer  MOFs encapsulate chemotherapeutic drugs and are modified with CAF-targeting peptides. Near-infrared light triggers drug release, inducing CAF apoptosis.  Remodels the tumor microenvironment, reduces CAF density, and enhances drug efficacy.  146–148  
Selenium nanoparticles (SeNPs)  Lung cancer  SeNPs inhibit CAF secretion of pro-inflammatory factors and enhance antioxidant capabilities, reducing CAF tumor-promoting activity.  Suppresses tumor-associated inflammation and blocks CAF support for tumor growth.  149, 150  
Peptide-functionalized silica nanoparticles  Pancreatic cancer, prostate cancer  Targets FAP-α on CAF surfaces, releasing quercetin-loaded nanoparticles to inhibit CAF activity  Decreases extracellular matrix density and improves tumor drug penetration.  141, 151  
Polymeric nanoparticles (PLGA-NPs)  Breast cancer, colorectal cancer  PLGA nanoparticles loaded with resveratrol target active CAF regions, reducing their immunosuppressive effects.  Remodels the immune microenvironment and enhances immunotherapy efficacy.  152, 153  
Magnetic nanoparticles (MNPs)  Breast cancer, Colorectal cancer  MNPs are guided by external magnetic fields to deliver paclitaxel or small-molecule inhibitors, targeting CAF functions.  Enhances drug delivery efficiency and reduces CAF-mediated drug resistance.  154, 155  
Liposome nanoparticles  Pancreatic cancer, breast cancer  FAP-targeted liposomes deliver chemotherapeutic drugs (e.g., paclitaxel) to inhibit CAF secretion of tumor-promoting factors.  Reduces tumor stroma barriers and enhances the effectiveness of chemotherapeutic drugs.  151, 156–158  
Iron oxide nanoparticles (FeNPs)  Lung cancer, breast cancer  Targets CAFs via the TGF-β signaling pathway, releasing anti-fibrotic drugs to suppress CAF activity.  Reduces CAF density and extracellular matrix, improving immune cell infiltration.  159, 160  

Gold nanoparticles (AuNPs) have attracted considerable attention in nanomedicine due to their excellent physicochemical properties, such as a large surface area, ease of functionalization, and high biocompatibility.114,115 These properties make AuNPs ideal candidates for targeted drug delivery, especially in therapies aimed at CAFs within the tumor microenvironment.116,117 Some studies have shown that AuNPs can be functionalized with ligands like peptides or antibodies,118,119 enabling them to specifically target CAF markers such as FAP and α-SMA.120,121 Additionally, AuNPs can carry a wide variety of therapeutic agents, from chemotherapy drugs to RNA molecules, making them a versatile platform for drug delivery.111,119 One promising use of AuNPs in CAF-targeted therapy is their combination with photothermal therapy.122 When exposed to near-infrared (NIR) light, AuNPs produce localized heat, which causes thermal ablation of CAFs and surrounding tumor cells, reducing tumor growth.123,124 This photothermal effect can be combined with chemotherapeutic drug delivery, improving local therapeutic effects while minimizing systemic toxicity. For example, Tan et al. demonstrated that functionalized AuNPs loaded with anticancer agents successfully targeted and eliminated CAFs within the TME.125 The ability to combine multiple treatment strategies, such as drug delivery and photothermal therapy, makes AuNPs a promising tool for CAF-targeted combination therapies.126,127 Furthermore, AuNPs are used in imaging applications, such as diagnostic imaging and real-time monitoring of treatment progress.128,129 The surface plasmon resonance of AuNPs allows for strong scattering and absorption of light, making them ideal for noninvasive imaging techniques that enhance the precision of treatment delivery.130 

Photosensitizers, such as zinc phthalocyanine and heme, generate reactive oxygen species (ROS) when exposed to light, which can selectively kill cancer cells and CAFs.131,132 Photodynamic therapy (PDT), which uses these photosensitizers, has become an effective treatment for cancer due to its ability to target both tumor cells and CAFs.133 Zinc phthalocyanine (ZnPc), for example, has been combined with various nanomaterials, such as liposomes and polymeric nanoparticles, to improve its therapeutic effects while reducing side effects.134,135 In a study by Zhou et al., ZnPc-loaded nanoparticles were designed to specifically target CAFs.136 Upon light exposure, ZnPc generates ROS that specifically damage CAFs, disrupt the tumor stroma, and improve the delivery of chemotherapeutic drugs to tumor cells. This combination of PDT with CAF-targeting strategies shows great promise for overcoming resistance in the tumor stroma and improving the effectiveness of conventional chemotherapy. Similarly, heme-based photosensitizers, incorporated into nanocarriers, have shown potential for CAF-targeted PDT. When irradiated, these nanomaterials generate ROS to promote localized tumor destruction.137 The targeting ability of these nanocarriers can be further improved by functionalizing them with CAF-specific ligands, providing a dual therapeutic approach that not only eliminates CAFs but also enhances the penetration of other therapeutic agents into the tumor.138,139

Nanoparticles modified with proteins or peptides offer an advanced approach for targeting CAFs in cancer therapy.140 By functionalizing nanoparticles with specific proteins or peptides, their targeting ability for CAFs can be significantly improved.141 Peptides like the RGD peptide, which binds to integrin receptors on CAFs, can be attached to nanoparticles to increase their affinity for CAFs.142,143 This peptide-based targeting has been widely studied because of the high specificity and low immunogenicity of peptides. Moreover, protein-based targeting strategies, such as using antibodies that target FAP, can also be used to effectively target and internalize nanoparticles by CAFs.144 Once internalized, these nanoparticles can deliver drugs directly to CAFs, disrupting their function and enhancing anti-tumor immunity. Therefore, the combination of peptides and proteins with nanoparticles can greatly enhance the precision of drug delivery, ensuring that therapeutic agents are targeted specifically to CAFs while minimizing off-target effects.

CAFs exhibit considerable heterogeneity within the TME and can be categorized into distinct subtypes: myCAFs, iCAFs, apCAFs, and vCAFs.161–163 Each subtype has a distinct role in tumor growth, immune evasion, and treatment resistance.30,164 Recent studies have shown that nanomaterials can effectively modulate the pro-tumor functions of these CAF subtypes by targeting specific biomarkers or related signaling pathways.12,160 Therefore, we explored targeting mechanisms for each CAF subtype and highlight the latest advances in nanomaterial-based therapies.

MyCAFs play a key role in ECM remodeling and fibrosis, which forms physical barriers that limit drug penetration.165 So targeting myCAFs is an important method to enhance drug delivery. For example, gold nanoparticles and graphene oxide nanocomposites have been shown to disrupt the ECM by targeting the expression of fibronectin and α-SMA in myCAFs.123,166 Cheng et al.'s study showed that photoactivated gold nanorods generate localized heat to degrade ECM components, thereby improving drug delivery efficiency and enhancing immune cell infiltration into the tumor core.167 iCAFs contribute to tumor inflammation and immune escape by secreting cytokines and chemokines such as IL-6 and CXCL12.31 Lipid nanoparticles loaded with IL-6 receptor (IL-6R) siRNA can effectively silence iCAF-related signaling pathways, leading to reduced tumor growth and metastasis.168 Additionally, magnetic nanocarriers combined with CXCR4 antagonists can inhibit tumor immune interactions mediated by CXCL12, thereby blocking iCAF-induced immune evasion.169,170 ApCAFs express major histocompatibility complex (MHC) class II molecules and play a role in antigen presentation.30,47 However, their immune regulatory functions are still being studied.171 Recent research has developed nanostructures based on DNA origami technology for delivering antigens and immune modulators to apCAFs, reprogramming them to stimulate immune responses.172,173 This innovative strategy opens up new possibilities for advancing tumor immunotherapy.

vCAFs promote angiogenesis by secreting vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) and regulate tumor blood vessel formation through integrin signaling (e.g., αvβ3).53,174 These processes supply the tumor with nutrients, supporting its growth. Nanomaterial designs targeting vCAFs aim to inhibit their pro-angiogenic functions. For example, polymer nanocarriers loaded with bevacizumab, a VEGF inhibitor, can neutralize VEGF signaling and significantly inhibit angiogenesis.175,176 Moreover, integrin-targeted nanoparticles can bind to αvβ3 integrin, directly interfering with the vascular formation pathways mediated by vCAFs.177 Furthermore, iron oxide nanoparticles generate ROS that impair vCAF function, further reducing tumor angiogenesis.178 These multifaceted nanomaterials provide promising therapeutic strategies for inhibiting vCAF-induced angiogenesis, offering broad potential for anti-tumor angiogenesis applications.

The functions and characteristics of CAFs differ across various cancer types (Fig. 2), meaning that their therapeutic targets and nanomaterial design strategies must be tailored to each specific cancer type.39,179 So far, with advances in nanomaterial research, selective targeting of CAFs has become more achievable. Additionally, quantitative analysis of both in vivo and ex vivo accumulation has confirmed the effectiveness of these strategies, highlighting their significant potential in personalized cancer therapy (Table II).

FIG. 2.

Heterogeneity of CAFs in different organs and their functional characteristics and potential targeting strategies in the TME. The figure highlights the diverse functions and characteristics of CAFs across various cancer types. These differences are crucial for determining specific therapeutic targets and designing nanomaterial-based strategies tailored to each cancer type. In head and neck cancer, CAFs are marked by ECM-myCAF, TGF-βmyCAF, iL-iCAF, and others, which play a role in tumor microenvironment modulation and immune response. In breast cancer, distinct CAF subtypes, such as CAF-S1 to CAF-S4, express various markers like FAP, αSMA, and CD29, impacting immune cell infiltration and tumor progression. Lung cancer CAFs express NF-κB, Foxp1, Col4A1, and ACTA2, which contribute to immune evasion and fibrosis. Pancreatic cancer shows CAFs with iCAF, myCAF, and apCAF and specific markers like MMP11, αSMA, and IL-6 involved in chemotherapy resistance and immune modulation. In ovarian cancer, both metastatic and primary fibroblasts are present, expressing TNF, CCL2, and CCL3, contributing to tumor progression and immune suppression. Colorectal cancer CAFs express MMP2, ACTA2, TAGLN, and ptgs2, potentially influencing cancer stem cells and tumor microenvironment remodeling. Intrahepatic cholangiocarcinoma CAFs, marked by vCAFs, express IL-6 and miR-9-5p, which are involved in tumor progression and fibrosis. Reproduced from Zhang, Xiangjian et al. Single-cell RNA sequencing to explore cancer-associated fibroblasts heterogeneity: “Single” vision for “heterogeneous” environment. Reproduced with permission from Zhang et al., Cell Proliferation 57(5), e13592 (2024). Copyright 2024 by the authors.37 

FIG. 2.

Heterogeneity of CAFs in different organs and their functional characteristics and potential targeting strategies in the TME. The figure highlights the diverse functions and characteristics of CAFs across various cancer types. These differences are crucial for determining specific therapeutic targets and designing nanomaterial-based strategies tailored to each cancer type. In head and neck cancer, CAFs are marked by ECM-myCAF, TGF-βmyCAF, iL-iCAF, and others, which play a role in tumor microenvironment modulation and immune response. In breast cancer, distinct CAF subtypes, such as CAF-S1 to CAF-S4, express various markers like FAP, αSMA, and CD29, impacting immune cell infiltration and tumor progression. Lung cancer CAFs express NF-κB, Foxp1, Col4A1, and ACTA2, which contribute to immune evasion and fibrosis. Pancreatic cancer shows CAFs with iCAF, myCAF, and apCAF and specific markers like MMP11, αSMA, and IL-6 involved in chemotherapy resistance and immune modulation. In ovarian cancer, both metastatic and primary fibroblasts are present, expressing TNF, CCL2, and CCL3, contributing to tumor progression and immune suppression. Colorectal cancer CAFs express MMP2, ACTA2, TAGLN, and ptgs2, potentially influencing cancer stem cells and tumor microenvironment remodeling. Intrahepatic cholangiocarcinoma CAFs, marked by vCAFs, express IL-6 and miR-9-5p, which are involved in tumor progression and fibrosis. Reproduced from Zhang, Xiangjian et al. Single-cell RNA sequencing to explore cancer-associated fibroblasts heterogeneity: “Single” vision for “heterogeneous” environment. Reproduced with permission from Zhang et al., Cell Proliferation 57(5), e13592 (2024). Copyright 2024 by the authors.37 

Close modal
TABLE II.

Nanomaterials targeting CAFs in different cancers.

Cancer type CAF role Nanomaterial strategy In vivo/ex vivo accumulation Outcome Refs.
Breast cancer  Promotes immune evasion and therapy resistance  FAP-targeting nanoparticles loaded with chemotherapy drugs  20× drug accumulation at tumor sites, significantly enhanced therapeutic effect  Selective killing of CAFs, improved drug delivery and tumor inhibition  181  
PDAC (pancreatic ductal adenocarcinoma)  Leads to dense fibrotic stroma, dominated by myCAFs  Collagenase-loaded nanocarriers and pH-sensitive liposomes delivering TGF-β inhibitors  Collagen degradation efficiency >70% (ex vivo); 50% reduction in CAF activation (in vivo Improved drug penetration, reduced fibrosis, suppressed tumor growth  183,184  
Colorectal cancer (CRC)  Secretes exosomes carrying miR-92a-3p, linked to metastasis and chemo-resistance  Exosome-mimicking nanoparticles delivering anti-miRNA molecules  15× nanoparticle accumulation in tumor tissues, reduced metastasis formation  Suppressed metastasis, enhanced chemotherapy sensitivity  188  
Lung cancer  Overexpresses PDGFR, promoting tumor growth and immune suppression  PDGFR-targeting aptamers conjugated with drug-loaded liposomes  6× nanoparticle concentration at tumor sites, enhanced immune cell recruitment  Inhibited CAF activation, boosted immune cell infiltration  190  
Gastric cancer  Secretes miR-522 via exosomes, suppressing ferroptosis and inducing drug resistance  Iron oxide nanoparticles restoring ferroptosis; dual-drug-loaded liposomes  12× nanoparticle accumulation in tumor tissues, 30% reduction in cancer cell survival rate  Restored ferroptosis, reduced tumor volume, extended survival  192  
Cancer type CAF role Nanomaterial strategy In vivo/ex vivo accumulation Outcome Refs.
Breast cancer  Promotes immune evasion and therapy resistance  FAP-targeting nanoparticles loaded with chemotherapy drugs  20× drug accumulation at tumor sites, significantly enhanced therapeutic effect  Selective killing of CAFs, improved drug delivery and tumor inhibition  181  
PDAC (pancreatic ductal adenocarcinoma)  Leads to dense fibrotic stroma, dominated by myCAFs  Collagenase-loaded nanocarriers and pH-sensitive liposomes delivering TGF-β inhibitors  Collagen degradation efficiency >70% (ex vivo); 50% reduction in CAF activation (in vivo Improved drug penetration, reduced fibrosis, suppressed tumor growth  183,184  
Colorectal cancer (CRC)  Secretes exosomes carrying miR-92a-3p, linked to metastasis and chemo-resistance  Exosome-mimicking nanoparticles delivering anti-miRNA molecules  15× nanoparticle accumulation in tumor tissues, reduced metastasis formation  Suppressed metastasis, enhanced chemotherapy sensitivity  188  
Lung cancer  Overexpresses PDGFR, promoting tumor growth and immune suppression  PDGFR-targeting aptamers conjugated with drug-loaded liposomes  6× nanoparticle concentration at tumor sites, enhanced immune cell recruitment  Inhibited CAF activation, boosted immune cell infiltration  190  
Gastric cancer  Secretes miR-522 via exosomes, suppressing ferroptosis and inducing drug resistance  Iron oxide nanoparticles restoring ferroptosis; dual-drug-loaded liposomes  12× nanoparticle accumulation in tumor tissues, 30% reduction in cancer cell survival rate  Restored ferroptosis, reduced tumor volume, extended survival  192  

Taking breast cancer as an example, CAFs play a significant role in tumor progression by promoting immune escape and contributing to drug resistance.180 A study found that nanoparticles conjugated with a fibroblast activation protein (FAP)-specific ligand resulted in drug accumulation at the tumor site that was 20 times higher than in normal tissues in an in vivo breast cancer model, significantly enhancing the therapeutic effect.181 Specifically, FAP-targeting nanodots not only selectively kill CAFs but also minimize damage to healthy tissues, improve drug delivery efficiency, and inhibit tumor growth. In PDAC, the tumor's extremely dense fibrotic stroma, primarily dominated by myCAFs, makes ECM remodeling essential.182 Research has shown that nanocarriers loaded with collagenase can degrade over 70% of collagen in an in vitro PDAC model, greatly improving the permeability of chemotherapy drugs.183 Additionally, the use of pH-sensitive liposomes to deliver TGF-β inhibitors significantly reduces CAF activity in mouse models (by more than 50%) and effectively inhibits tumor fibrosis.184 

In colorectal cancer (CRC), CAFs promote tumor metastasis and chemotherapy resistance by secreting exosomes that carry miR-92a-3p.185,186 To counter this, nanoparticles mimicking exosomes have been designed to deliver anti-miRNA molecules.187, In vivo experiments have shown that these nanoparticles accumulate in tumor tissue 15 times more than in normal tissues.188 This not only significantly reduces metastatic lesion formation but also increases chemotherapy sensitivity. For lung cancer, CAFs typically express high levels of PDGFR, providing a clear target for therapy.189 Drug-loaded liposomes combined with PDGFR aptamers show exceptionally high specificity in lung cancer models, with nanoparticle concentrations in the tumor site being six times higher than in other tissues.190 This strategy not only effectively inhibits CAF activation but also significantly enhances immune cell recruitment (such as T cells and NK cells) to the tumor. In gastric cancer, CAFs secrete miR-522 through exosomes, inhibiting ferroptosis of cancer cells and promoting drug resistance.191 To counteract this, iron oxide nanoparticles are used to restore ferroptosis, reducing the survival rate of tumor cells by more than 30%.192 Additionally, the design of dual-drug-loaded liposomes enables simultaneous targeting of both CAFs and tumor cells. In a mouse model of gastric cancer, this strategy significantly reduced tumor volume and prolonged survival time.193 

Up to now, many research examples have demonstrated the advantages of nanomaterials in optimizing drug delivery and selectively targeting CAFs, and their effectiveness in improving treatment outcomes has been quantitatively analyzed through in various vivo and in vitro experiments. These findings lay a solid foundation for developing precise, personalized treatment plans and offer new hope for the future of cancer therapies.

CAFs exhibit significant heterogeneity and express various specific biomarkers, which provide multiple potential targets for targeted therapy. In recent years, substantial progress has been made in the design of nanomaterials targeting these CAF markers, leading to a notable improvement in tumor treatment efficacy by precisely targeting these markers.111 These biomarkers not only offer a molecular understanding of CAF function but also serve as potential targets for therapeutic intervention.

For example, FAP, which is highly expressed in CAFs, plays a key role in degrading the ECM and promoting tumor cell invasion. Nanomaterials targeting FAP utilize FAP antibodies or small molecule ligands to achieve precise targeting. FAP-targeted nanoparticles can deliver chemotherapy drugs, such as paclitaxel and doxorubicin, selectively destroying CAFs while avoiding damage to normal tissues.151,194 Furthermore, FAP-targeted photothermal nanomaterials, such as gold nanoparticles, generate local thermal effects after photoactivation.195 This disrupts CAF function and significantly improves drug permeability in the tumor stroma.

The high expression of α-SMA in CAFs is closely related to tumor stroma rigidity and fibrosis, which enhance tumor resistance to drugs. To target this biomarker, liposome nanoparticles can carry TGF-β inhibitors, such as Galunisterib, to inhibit CAF activation and fibrosis.196 Platelet-derived growth factor receptor (PDGFR), which is highly expressed in CAFs, plays a critical role in CAF proliferation and angiogenesis. PDGFR-targeted polymer nanoparticles, modified with an adapter, significantly reduce tumor vascular density by delivering PDGF inhibitors or siRNA.197 VEGF, secreted by CAFs, is a major factor in tumor angiogenesis, providing essential nutritional support for tumors. Nanocarriers loaded with VEGF inhibitors, such as bevacizumab, can neutralize VEGF secreted by CAFs and significantly inhibit angiogenesis.198 Moreover, VEGF nucleic acid aptamer-modified nanoparticles can further disrupt the tumor's blood supply by precisely binding to VEGF.199 

In immune escape, CXCL12 secreted by CAFs attracts immunosuppressive cells, such as Tregs and MDSCs, through the CXCR4 receptor, creating an immune escape environment.200 CXCR4 antagonist nanomaterials can block the CXCL12-CXCR4 axis, restore immune cell function, and improve anti-tumor immune responses.201 In a melanoma model, this strategy significantly increased T-cell infiltration in the tumor tissue.202 CAFs also secrete IL-6, which activates the STAT3 signaling pathway to promote tumor cell growth and treatment resistance.203 To counteract this, siRNA liposome nanoparticles can silence IL-6 expression in CAFs, block inflammatory signals, reduce tumor resistance, and improve chemotherapy sensitivity.168 Additionally, fibroblast-specific protein-1 (FSP-1), which is highly expressed in CAFs, directly contributes to CAF migration and tumor metastasis.204 Gene inhibitor nanoparticles can significantly reduce CAF migration, thereby decreasing the formation of tumor metastases. CAF-secreted extracellular vesicles carrying miRNAs, such as miR-21 and miR-92a, regulate tumor cell growth and metastasis.186 Nanoparticles mimicking extracellular vesicles can block the signaling of CAF-secreted exosomes by delivering anti-miRNA drugs, such as miR-92a inhibitors. This method significantly reduces metastatic focus formation and improves chemotherapy effectiveness in colorectal cancer.205 Finally, CAFs regulate their own activation and the immunosuppressive tumor microenvironment through the transforming growth TGF-β signaling pathway.206 Nanoparticles carrying TGF-β inhibitors can significantly reduce tumor fibrosis, and immune nanoparticles that inhibit both TGF-β and PD-L1 signaling secreted by CAFs improve the effectiveness of immunotherapy.207 

The biomarkers expressed by CAFs provide many potential targets for tumor treatment, and nanomaterials targeting these markers can improve treatment accuracy and effectiveness. By using methods like gene silencing, blocking receptors, and regulating the immune system, nanomaterials can control how CAFs affect the tumor microenvironment, addressing the limits of traditional treatments. In the future, combining nanotechnology with precision medicine is expected to make CAF-targeted nanomaterials more important for personalized cancer therapy.

The functional diversity and high heterogeneity of CAFs make them a complex target for tumor therapy but also provide several potential pathways for precision therapy (Fig. 3). So far, the rapid development of nanotechnology has enabled the use of nanomaterials for therapeutic methods. By targeting CAFs and their interactions with other components in the TME, these nanomaterials can effectively regulate tumor progression driven by CAFs. Despite many studies showing the potential of nanomaterials in CAF-targeted therapy, the dynamic plasticity of CAFs, complex signaling pathways, and the high heterogeneity of the TME remain challenges that current technology cannot fully overcome.27 Therefore, exploring how nanomaterials can target key components of CAFs and the TME, and how to achieve more precise tumor treatment based on this, is of great research value.

FIG. 3.

Schematic representation of the TME and nanoparticle-based therapeutic strategies. The TME is composed of tumor cells, CAFs, immune cells, and ECM, which collectively contribute to tumor progression and therapy resistance. Major strategies for targeting the TME include ECM remodeling, targeting exosomes, alleviating tumor hypoxia, anti-angiogenesis, targeting cancer stem cells (CSCs), targeting CAFs, and immunomodulation. Various nanoparticle platforms, including gold nanoparticles, quantum dots, carbon nanotubes, polymeric nanoparticles, liposomes, silica nanoparticles, dendrimers, micelles, magnetic nanoparticles, and nanogels, are employed to enhance therapeutic delivery and efficacy within the TME. These approaches aim to overcome the inherent complexity of the TME and improve treatment outcomes. Reproduced with permission from Tang et al., Int. J. Nanomed. 16, 5811–5829 (2021). Copyright 2021 by the authors.208 

FIG. 3.

Schematic representation of the TME and nanoparticle-based therapeutic strategies. The TME is composed of tumor cells, CAFs, immune cells, and ECM, which collectively contribute to tumor progression and therapy resistance. Major strategies for targeting the TME include ECM remodeling, targeting exosomes, alleviating tumor hypoxia, anti-angiogenesis, targeting cancer stem cells (CSCs), targeting CAFs, and immunomodulation. Various nanoparticle platforms, including gold nanoparticles, quantum dots, carbon nanotubes, polymeric nanoparticles, liposomes, silica nanoparticles, dendrimers, micelles, magnetic nanoparticles, and nanogels, are employed to enhance therapeutic delivery and efficacy within the TME. These approaches aim to overcome the inherent complexity of the TME and improve treatment outcomes. Reproduced with permission from Tang et al., Int. J. Nanomed. 16, 5811–5829 (2021). Copyright 2021 by the authors.208 

Close modal

First, CAFs promote excessive deposition of ECM to harden the tumor stroma, thereby limiting effective drug delivery.209 To address this, collagenase-loaded nanoparticles can break down collagen secreted by CAFs, significantly improving drug penetration.183 In addition, matrix metalloproteinase (MMP)-responsive nanomaterials can accurately release drugs to the tumor site through their specific response to CAF-secreted enzymes.210 Meanwhile, in photothermal therapy, gold nanoparticles (AuNPs) not only disrupt the integrity of ECM by heating local areas but also enhance the infiltration of immune cells.211 Although these strategies have shown significant effects in preclinical models, excessive ECM degradation can cause tumor cells to shed and metastasize, so the level of degradation must be carefully controlled in application.212 

At the same time, unlike directly clearing CAFs, reprogramming their pro-tumor phenotype to restore their normal function has been a highly anticipated treatment strategy in recent years. For example, DNA origami nanostructures are used to deliver epigenetic regulators, thereby reprogramming CAFs from a pro-tumor phenotype to a quiescent or anti-tumor phenotype.213,214 In addition, dual-drug-loaded nanoparticles can simultaneously deliver retinoic acid and TGF-β inhibitors, achieving dual effects of fibrosis inhibition and CAF function normalization.215 In preclinical studies, these strategies significantly reduced fibrosis and enhanced the efficacy of immunotherapy. However, current research on the molecular characteristics of CAF subtypes is not sufficient to effectively distinguish between pro-tumor and anti-tumor CAF subgroups, which poses a challenge for precise reprogramming therapy.216 

Furthermore, CAFs construct a microenvironment for tumor immune escape by expressing PD-L1 and recruiting immunosuppressive cells such as regulatory T cells (Tregs).217 In response, immune checkpoint inhibitor nanoparticles can significantly restore T cell function and improve the effectiveness of immunotherapy by targeting the PD-L1 expression induced by CAFs.218 In addition, nanoparticles simulating exosomes not only activate anti-tumor immune responses by delivering tumor antigens and adjuvants but also reshape the immune balance of TME to a certain extent.219,220 More innovatively, nanoparticles integrated with biosensors can monitor immune suppression signals induced by CAFs in real time and release corresponding therapeutic drugs based on the results.221 However, the immune suppression mechanism driven by CAFs is complex, and relying on a single immune regulation strategy may not fully reverse this process. Thus, combining other therapeutic approaches may be necessary.

With the continuous development of nanotechnology, combining artificial intelligence (AI) and biosensors has further enhanced the multifunctionality of CAF-targeted therapy. AI-driven nanomaterial design can improve CAF targeting efficiency by optimizing material size, shape, and surface properties.222 For instance, AI can predict the various effects of nanomaterials on CAF signaling pathways, allowing for the design of therapies that target multiple pathways simultaneously.223,224 In addition, nanoparticles integrated with biosensors provide the possibility of real-time monitoring of TME changes, making treatment more precise.225 However, despite the promising potential of AI and sensor technologies, their application in clinical settings still faces challenges, including technical complexity and regulatory barriers. While CAF-targeting nanomaterials have made significant strides in tumor therapy, they still face several limitations. For example, the heterogeneity and functional plasticity of CAFs increase the complexity of targeted therapy, and off-target effects of nanomaterials remain a key safety concern.226 Moreover, large-scale production of nanomaterials faces challenges related to high costs and quality control. The future direction should involve combining single-cell omics technology to deeply analyze the subtype characteristics of CAFs and developing more precise, personalized treatment plans.227,228 At the same time, optimizing nanomaterial surface modification technologies can further reduce toxicity and improve specificity.229 Additionally, combining CAF-targeted therapy with traditional treatments (such as chemotherapy and radiotherapy) or immunotherapy could significantly enhance overall treatment effectiveness.108 

Nanomaterials offer various therapeutic strategies for targeting tumor-associated fibroblasts and controlling the tumor microenvironment. From ECM breakdown to immune regulation and phenotype changes, these approaches show the wide potential of nanotechnology in cancer treatment. The collaboration between AI, biosensors, and advanced biotechnology will further help the use of CAF-targeted nanomaterials in personalized cancer treatment.

Quantitative analysis of nanoparticle accumulation in tumors, especially in CAFs-rich areas, is important for assessing their therapeutic effectiveness and specificity. This analysis helps understand how nanoparticles distribute in the body and how they move through the system. It also explains the complex relationship between nanoparticles and the TME (Fig. 4). A thorough evaluation of nanoparticle accumulation in tumors, both in living organisms and in laboratory settings, is necessary to improve their design and use in clinical practice. However, the lack of consistent quantitative data across studies limits the ability to apply current findings to real-world situations. This calls for a more systematic and thorough approach.

FIG. 4.

(a) and (b) Schematic illustration of the structure and application of Her2-DOX-SPIOs@PLGA@Au NPs. (a) The composition and fabrication process of Her2-DOX-SPIOs@PLGA@Au NPs. (b) The nanoscale theranostic agent is utilized for PA/MRI dual-modal imaging and tumor elimination through chemical-photothermal therapy and immune activation. (c-i) These figures demonstrate the characterization and performance of DSG NPs, including their imaging capabilities, temperature response under NIR laser irradiation, drug release behavior under different conditions, and magnetic resonance properties. (j)–(m) These figures demonstrate the in vivo imaging of a BT474 mice xenograft tumor to evaluate the accumulation and distribution of nanoparticles, particularly in CAFs-rich regions, which is crucial for assessing their therapeutic efficacy and specificity. (j) T2-weighted MR imaging of the tumor at different time points (0, 0.5, 1, 2, 6, and 24 h), highlighting the tumor (red dashed circles) to monitor nanoparticle distribution over time. (k) T2 relaxation rate [1/T2(s−1)] of the tumor's region of interest (ROI) at different time points, showing how the particles interact with the tumor environment. (l) Photoacoustic imaging at the same time points, capturing the contrast of nanoparticles within the tumor for better understanding of their location and accumulation. (m) Signal intensity analysis from the ROI of the tumor over time, providing quantitative data on nanoparticle distribution and accumulation. Data are expressed as mean ± SD, n = 3. (n)–(r) These figure illustrate the effects of various treatments on tumor growth and temperature variations during the monitoring period. These results demonstrate the impact of different nanoparticle treatments and laser irradiation on tumor growth inhibition and temperature changes, providing insight into their potential for enhanced cancer therapy. (n) and (o) The curve of relative tumor volume variations with different treatments during the 20-day monitoring period, the first treatment and second treatment were performed on day 1 and day 10, respectively, data expressed as mean ± SD, n = 3. (p) and (q) The temperature variations of tumor under laser irradiation (10 min, 808 nm, 1 W/cm2) during first treatment and second treatment. (r) IR thermal images of various treatment groups. Reproduced with permission from Zheng et al., Int. J. Nanomed. 15, 10007–10028 (2020). Copyright 2020 by the authors.230 

FIG. 4.

(a) and (b) Schematic illustration of the structure and application of Her2-DOX-SPIOs@PLGA@Au NPs. (a) The composition and fabrication process of Her2-DOX-SPIOs@PLGA@Au NPs. (b) The nanoscale theranostic agent is utilized for PA/MRI dual-modal imaging and tumor elimination through chemical-photothermal therapy and immune activation. (c-i) These figures demonstrate the characterization and performance of DSG NPs, including their imaging capabilities, temperature response under NIR laser irradiation, drug release behavior under different conditions, and magnetic resonance properties. (j)–(m) These figures demonstrate the in vivo imaging of a BT474 mice xenograft tumor to evaluate the accumulation and distribution of nanoparticles, particularly in CAFs-rich regions, which is crucial for assessing their therapeutic efficacy and specificity. (j) T2-weighted MR imaging of the tumor at different time points (0, 0.5, 1, 2, 6, and 24 h), highlighting the tumor (red dashed circles) to monitor nanoparticle distribution over time. (k) T2 relaxation rate [1/T2(s−1)] of the tumor's region of interest (ROI) at different time points, showing how the particles interact with the tumor environment. (l) Photoacoustic imaging at the same time points, capturing the contrast of nanoparticles within the tumor for better understanding of their location and accumulation. (m) Signal intensity analysis from the ROI of the tumor over time, providing quantitative data on nanoparticle distribution and accumulation. Data are expressed as mean ± SD, n = 3. (n)–(r) These figure illustrate the effects of various treatments on tumor growth and temperature variations during the monitoring period. These results demonstrate the impact of different nanoparticle treatments and laser irradiation on tumor growth inhibition and temperature changes, providing insight into their potential for enhanced cancer therapy. (n) and (o) The curve of relative tumor volume variations with different treatments during the 20-day monitoring period, the first treatment and second treatment were performed on day 1 and day 10, respectively, data expressed as mean ± SD, n = 3. (p) and (q) The temperature variations of tumor under laser irradiation (10 min, 808 nm, 1 W/cm2) during first treatment and second treatment. (r) IR thermal images of various treatment groups. Reproduced with permission from Zheng et al., Int. J. Nanomed. 15, 10007–10028 (2020). Copyright 2020 by the authors.230 

Close modal

Quantitative assessment serves several important purpose, such as measuring how specifically nanoparticles target CAFs-rich areas, evaluating their ability to pass through dense ECM barriers, and understanding their clearance and retention patterns. Measures like tumor-to-normal tissue ratio (TNR), percentage injected dose per gram of tissue (%ID/g), and retention time allow researchers to track nanoparticle localization and treatment effects.231–233 These analyses are also crucial for determining how effectively nanoparticles influence CAF-driven processes, like ECM remodeling and immune suppression.4, In vivo analysis mostly uses imaging and other techniques to track nanoparticle distribution in living organisms. For example, fluorescently labeled nanoparticles allow real-time tracking of tumor accumulation, while advanced imaging methods like PET and MRI provide high-sensitivity images of radiolabeled or magnetic nanoparticles.234,235 Additionally, mass spectrometry techniques, such as ICP-MS, give precise measurements of nanoparticle content in tumor tissues after sacrifice.236 Therefore, metrics like %ID/g or TNR provide valuable information on the specificity and effectiveness of nanoparticle delivery. However, these results are often limited by the changing nature of the TME, which can alter how nanoparticles are distributed over time.237 Thus, a combination of in vivo imaging and postmortem analysis is often needed for a more complete understanding.

To complement in vivo studies, ex vivo analysis provides clearer data on nanoparticle distribution within tumor tissues. Techniques like confocal microscopy and histopathology help visualize nanoparticle localization in relation to CAF markers such as FAP or α-SMA.238 Furthermore, flow cytometry can measure nanoparticle uptake by specific cell types, including CAFs, tumor cells, or immune cells.239 Combining ex vivo results with in vivo imaging is important for a better understanding of nanoparticle behavior. Additionally, ex vivo analysis helps assess penetration depth, a key factor in ECM-dense tumors like PDAC.240 For instance, collagenase-loaded nanoparticles increased tumor penetration by 1.8 times compared to conventional systems, highlighting the importance of ECM-targeting strategies.183 

Quantitative analysis of nanoparticle accumulation faces several challenges. First, tumor heterogeneity, including variations in CAF density, ECM composition, and vascularization, makes it difficult to compare results between studies. Second, dynamic interactions between nanoparticles and the TME, like opsonization and clearance, often reduce targeting efficiency. Third, the lack of standardized methods for measuring nanoparticle accumulation complicates comparisons across studies. Finally, in tumors with dense ECM, poor nanoparticle penetration into deeper areas limits therapeutic effectiveness. Addressing these challenges is crucial for advancing nanoparticle-based therapies in clinical settings.

CAFs play a pivotal role in tumor progression, metastasis, and resistance to treatment, making them an attractive target for precision medicine. Nanomedicine, which utilizes nanoscale drug delivery systems, has emerged as a powerful approach to specifically target CAFs, improving therapeutic outcomes while minimizing off-target effects.26,241,242 This section discusses innovative nanomedicine strategies designed to target CAFs, focusing on novel nanoscale drug delivery platforms, surface functionalization techniques, environmentally responsive nanocarriers, and multi-drug nanocarriers for combination therapy. By exploring these approaches, we aim to highlight the potential of nanomedicine in addressing the complex tumor microenvironment and enhancing the effectiveness of cancer treatments.

Nanoscale drug delivery systems have become a critical focus in cancer treatment due to their ability to precisely target CAFs. CAFs play an essential role in tumor growth, metastasis, and chemotherapy resistance, making them an attractive therapeutic target (Fig. 5). Therefore, developing nanosystems that can efficiently deliver drugs to CAFs while minimizing off-target effects is of paramount importance.

FIG. 5.

Innovative nanoscale drug delivery systems for targeting CAFs in the tumor microenvironment. (a) Multifunctional nanoparticles, incorporating monoclonal antibodies, short peptides, siRNA, doxorubicin (DOX), and metal nanoparticles, are engineered for targeted delivery to CAFs. (b) Advanced nanoparticle systems such as Cellax-NPs, CAP-NPs, scFv-NPs, and FH-NPs target specific CAF markers, including α-SMA, FAP, and TNC, to induce apoptosis and inhibit survival signaling pathways like BCL-2 and BCL-XL. (c) siRNA-loaded nanoparticles and sponge-like NPs modulate CAF-driven processes, such as CXCL12 secretion and cytokine production, which promote tumor invasion and metastasis. They also target specific pathways, such as ERα regulation by miR-22, to reduce CAF activity. (d) Gold nanoparticles (GNPs) and other delivery platforms disrupt CAF-mediated fibrosis and lipid metabolism by targeting pathways involving PDGF, TGF-β1, α-SMA, fibronectin, and lipid metabolism regulators (e.g., FASN, SREBP2, FABP3). Reproduced with permission from Mu et al., Int. J. Mol. Sci. 22(21), 11671 (2021). Copyright 2021 by the authors.112 

FIG. 5.

Innovative nanoscale drug delivery systems for targeting CAFs in the tumor microenvironment. (a) Multifunctional nanoparticles, incorporating monoclonal antibodies, short peptides, siRNA, doxorubicin (DOX), and metal nanoparticles, are engineered for targeted delivery to CAFs. (b) Advanced nanoparticle systems such as Cellax-NPs, CAP-NPs, scFv-NPs, and FH-NPs target specific CAF markers, including α-SMA, FAP, and TNC, to induce apoptosis and inhibit survival signaling pathways like BCL-2 and BCL-XL. (c) siRNA-loaded nanoparticles and sponge-like NPs modulate CAF-driven processes, such as CXCL12 secretion and cytokine production, which promote tumor invasion and metastasis. They also target specific pathways, such as ERα regulation by miR-22, to reduce CAF activity. (d) Gold nanoparticles (GNPs) and other delivery platforms disrupt CAF-mediated fibrosis and lipid metabolism by targeting pathways involving PDGF, TGF-β1, α-SMA, fibronectin, and lipid metabolism regulators (e.g., FASN, SREBP2, FABP3). Reproduced with permission from Mu et al., Int. J. Mol. Sci. 22(21), 11671 (2021). Copyright 2021 by the authors.112 

Close modal

Several new nanoscale drug delivery platforms have been developed, showing unique properties that make them well-suited for targeting CAFs. First, ultrafine nanoparticles and nanomicelles, with their small size and large surface area, have shown great effectiveness in tumor treatment.20,243 These nanoparticles, typically ranging from 1 to 100 nm, are small enough to take advantage of the enhanced EPR effect, allowing them to accumulate in tumor tissue.244 Additionally, nanomicelles, due to their ability to encapsulate hydrophilic drugs, offer enhanced stability and controlled release, which improves drug bioavailability.245 By modifying the surfaces of these nanoparticles or micelles with specific ligands, such as antibodies or targeting peptides for CAFs, their ability to selectively target CAFs can be greatly improved, enhancing therapeutic efficacy.246,247

Liposomes, one of the most established types of nanocarriers, have been widely studied for their biocompatibility and controlled drug release properties.248 For example, the liposomal drug Doxil (liposomal doxorubicin) has been successfully used in clinical settings.249,250 Its lipid bilayer structure protects the drug from premature degradation and ensures sustained release within the tumor.251 By modifying liposomes with polyethylene glycol (PEG), their circulation time in the bloodstream is extended, which reduces clearance by the immune system.252 When further modified with antibodies or specific peptides targeting CAFs, liposomes can enhance their ability to target CAFs, improving therapeutic outcomes.253 Carbon nanotubes (CNTs) are a new class of nanocarriers with great potential due to their unique structure. With a high surface area and excellent drug-loading capacity, CNTs can carry a large amount of drug and deliver it to the tumor site through the bloodstream.254,255 The surface of CNTs can be modified with specific targeting peptides or antibodies, allowing for selective binding to CAFs.256 Additionally, CNTs can respond to changes in the tumor microenvironment, such as acidic conditions or high hydrogen peroxide levels, to release drugs in a controlled way.257 As such, CNTs show great promise as a delivery platform for targeting CAFs in future applications. However, designing the ideal nanocarrier is still challenging. In addition to drug loading capacity, the size, shape, and surface charge of nanoparticles greatly affect their distribution and targeting efficiency.258 Smaller nanoparticles (typically 10–100 nm) are more likely to penetrate tumor blood vessels and accumulate within the tumor.259 The shape of the nanoparticle also affects how it interacts with the cell membrane. For instance, spherical nanoparticles generally have better stability and higher cellular uptake compared to non-spherical shapes.260 By carefully controlling the size and shape of nanoparticles, both their targeting efficiency and ability to penetrate the tumor can be optimized.

The surface charge of nanoparticles is another important factor that affects their behavior in vivo. Generally, negatively charged nanoparticles show better biocompatibility and lower immunogenicity, while positively charged nanoparticles tend to adsorb more strongly to cell membranes, increasing cellular uptake.261,262 Therefore, designing the surface charge carefully can improve the targeting ability and drug delivery efficiency of nanocarriers.

Surface functionalization is a critical aspect of enhancing the specificity and efficacy of nanoscale drug delivery systems for targeting CAFs. To achieve precise targeting, it is essential to identify the appropriate biomarkers expressed on CAFs.144 Among the most studied markers are fibronectin and α-SMA. Fibronectin is an extracellular matrix protein overexpressed in the tumor stroma, while α-SMA is a marker linked to activated fibroblasts.263,264 These markers are ideal candidates for nanoparticle functionalization because they are highly expressed in the tumor microenvironment but found minimally in normal tissues.265 Additionally, integrins like αvβ3 and αvβ5 are commonly expressed on CAFs and play a role in cell adhesion to the extracellular matrix.143 These integrins are also good targets for nanoparticles, providing a solid foundation for the development of surface-modified nanocarriers.143,266 Therefore, selecting the right targeting markers is crucial for the success of functionalized drug delivery systems aimed at CAFs.

Another promising functionalization strategy uses carbohydrates, such as mannose and galactose, to target CAFs.144,267 These carbohydrates can be attached to nanoparticles to interact with cell surface lectins or carbohydrate-binding receptors that are overexpressed on CAFs.268 While this strategy provides an alternative to antibody- and peptide-based functionalization, it generally offers lower binding affinity and specificity. However, carbohydrates are relatively easy to modify and can be combined with other ligands to improve targeting efficiency.194,269 This approach is especially useful for targeting CAFs with lectin receptors, which are present in the tumor microenvironment. The success of surface functionalization strategies depends largely on the receptor–ligand interactions between nanoparticles and CAFs.144 For example, the binding between RGD peptides and integrins, or antibodies and CAF-specific antigens like fibronectin or α-SMA, is key to ensuring the specificity and efficiency of drug delivery.270,271 The strong affinity of RGD peptides for integrins has been well-documented in many studies, making them a common choice for targeting CAFs.272 Similarly, antibody-antigen interactions are highly specific, offering an effective way to guide nanoparticles to CAFs.

Additionally, it is important to recognize the heterogeneity of the tumor microenvironment. CAFs can express different receptor profiles depending on their activation state or the type of tumor, meaning receptor–ligand interactions must be tailored to the specific CAF subtype.273 As a result, combining different functionalization approaches, such as dual-targeting strategies, could significantly improve targeting accuracy. Dual-targeting, a promising approach in nanomedicine, involves functionalizing nanoparticles with two distinct ligands: one targeting CAFs and the other targeting tumor cells.274,275 This strategy can significantly improve therapeutic efficacy by addressing the complexity of the tumor microenvironment. For example, nanoparticles could be functionalized with RGD peptides to target CAFs and folate to target tumor cells. This combined targeting ensures both the stromal and cancerous components of the tumor are addressed, improving overall therapeutic outcomes.276,277 Moreover, dual-targeting strategies could help overcome resistance mechanisms, such as CAF-induced drug resistance, which is common in single-target therapies.

The unique characteristics of the TME offer many opportunities for developing responsive nanocarriers in cancer treatment. The TME is characterized by features like low pH, increased hydrogen peroxide (H2O2) concentration, and the overexpression of specific enzymes, which can be used as triggering mechanisms for targeted drug delivery systems.278,279 Designing nanocarriers that respond to these microenvironmental signals enhances drug targeting and ensures precise drug release at tumor and CAF sites, thereby maximizing therapeutic efficacy.280–282 

There are significant differences between the TME and normal tissue, with key features including low pH, increased hydrogen peroxide concentration, and the secretion of specific enzymes, such as MMPs by CAFs.283 These distinct characteristics provide the basis for designing environment-responsive nanocarriers. Tumor tissue typically has an acidic microenvironment due to active metabolic processes in tumor cells, leading to a decrease in local pH, typically ranging from 6.0 to 6.5—lower than the normal tissue pH of around 7.4.284,285 Additionally, hydrogen peroxide concentration in tumor tissues is elevated, and MMP activity is significantly increased due to the influence of CAFs.

The acidic pH commonly found in the TME is a well-known trigger for the development of pH-responsive nanocarriers. These carriers are usually made from materials that degrade or change structure under acidic conditions, such as poly(lactic-co-glycolic acid) copolymers (PLGA), polyethylene imine (PEI), and polystyrene sulfonate (PSS).286–288 These materials remain stable at normal tissue pH (∼7.4) but degrade or undergo structural changes when exposed to the lower pH typical of tumor environments, releasing the drug. For example, PLGA is a widely used pH-sensitive material that easily hydrolyzes in acidic environments, releasing the encapsulated drug.288 The degradation rate of PLGA is closely linked to the surrounding pH, allowing for rapid breakdown in low pH tumor microenvironments and precise drug delivery near CAFs.289 Additionally, some pH-responsive carriers are designed to change their surface charge or hydrophilicity in acidic conditions, facilitating drug release and promoting interaction with the cell membrane.290,291 This feature allows pH-responsive nanocarriers to take advantage of the tumor's acidic environment for targeted drug release, improving drug bioavailability and therapeutic efficacy.

Beyond pH response, enzyme-sensitive nanocarriers are crucial for targeting the TME, particularly near CAFs. CAFs secrete MMPs, which are key players in tumor stroma remodeling. MMPs degrade the ECM, aiding tumor cell migration and invasion.5,210 As a result, enzyme-responsive nanocarriers have been developed to undergo structural changes or degradation when exposed to MMPs, releasing their drug payloads.292 For example, some nanocarriers are functionalized with enzyme-cleavable peptide chains that MMPs can specifically recognize and cleave.293 Once the nanocarrier reaches the CAF-rich tumor region and encounters MMPs, the peptide chain is hydrolyzed, causing the nanocarrier to rupture and release the drug. Unlike pH-responsive carriers, enzyme-responsive systems have the advantage of targeting the unique enzyme activity of CAFs, offering higher specificity and better control over drug release.151 In addition to MMPs, other enzymes, such as peroxidases and cholesterol esterases, are present in the tumor microenvironment and can also function as response mechanisms.294,295 Therefore, designing nanocarriers responsive to multiple enzyme types can improve the precision of drug release and treatment.

Another notable feature of the TME is the elevated concentration of H2O2, a by-product of the high metabolic activity and redox processes within tumor cells.296 This increased concentration of H2O2 provides an opportunity to develop redox-responsive nanocarriers that release drugs specifically in the tumor region.297,298 These carriers are typically made from materials that react with hydrogen peroxide, such as polymers containing thiol groups (-SH) or disulfide bonds (S-S).298 When exposed to high levels of H2O2, these chemical groups undergo a reduction reaction, causing the nanocarrier to break down and release its drug payload.299 For example, polymers crosslinked with disulfide bonds can undergo a reduction reaction in the presence of hydrogen peroxide, breaking the carrier into smaller molecules and releasing the drug.300 The advantage of redox-responsive carriers is that hydrogen peroxide concentration is usually elevated within the tumor microenvironment, ensuring that drug release is localized to the tumor site and improving therapeutic efficacy.

Nowadays, there is growing interest in combination therapy for cancer treatment, which highlights the need for new drug delivery systems that can effectively deliver multiple therapies simultaneously. Single-drug treatments, such as chemotherapy and immunotherapy, often face limitations like drug resistance and insufficient effectiveness. Therefore, multi-drug nanocarriers have become a promising solution to improve cancer treatment outcomes, especially by targeting both CAFs and tumor cells together. The design of multi-drug nanocarriers aims to deliver multiple therapeutic agents in a single nanoplatform, ensuring controlled and targeted release within the tumor microenvironment.

Multi-drug delivery can be achieved through various physical and chemical methods, including co-loading strategies, composite carrier systems, and targeted modification.301,302 In the co-loading strategy, multiple drugs are encapsulated within a single nanocarrier using either covalent bonding or non-covalent interactions.303 These drugs can coexist in different regions of the carrier without interfering with each other, maintaining their stability and therapeutic properties.304 For example, drugs can be compartmentalized in phase-separated or core-shell structures. This design allows for the independent release of each drug when triggered by specific environmental conditions, improving the therapeutic outcome.305 The co-loading strategy is especially beneficial when combining drugs with complementary mechanisms of action, such as chemotherapeutic agents and immunotherapies, to achieve synergistic effects.

In composite carrier design, different materials are combined to create a nanocarrier that can load multiple drugs at once. For example, liposomes, polymeric nanoparticles, and metallic nanostructures can be integrated to improve drug loading capacity, stability, and controlled release.306 A particularly effective example is the combination of polymeric nanoparticles and liposomes, which increases the payload capacity while stabilizing the loaded drugs.307 Additionally, composite carriers ensure that drugs are released in a controlled manner based on the specific needs of the tumor microenvironment, maximizing therapeutic efficacy and minimizing systemic side effects.308 

The surface targeting modification strategy aims to improve drug delivery precision by functionalizing the surface of nanocarriers with ligands that specifically recognize CAFs and tumor cells. This is achieved by attaching antibodies, peptides, or other targeting molecules to the carrier surface, allowing them to bind to specific receptors or antigens on the tumor or CAF surface. This functionalization not only enhances the delivery of nanocarriers to the tumor site but also adds an extra layer of specificity by targeting the unique components of the tumor microenvironment.248 By targeting both CAFs and tumor cells, this approach ensures that therapeutic agents are delivered to the right areas, improving treatment outcomes.111 One key advantage of combination therapy is its ability to overcome the limitations of single-agent treatments and improve therapeutic efficacy through synergistic effects.309 For instance, a typical combination therapy involves chemotherapy drugs, such as docetaxel, and immune checkpoint inhibitors, such as PD-1 inhibitors, which work together.310 By loading both chemotherapy and immune drugs onto nanocarriers and modifying them to target CAFs and tumor cells, precise drug release can be achieved at the tumor site.23,311 Targeting molecules on the nanocarriers bind to specific receptors on CAFs first, reducing the impact of the drugs on normal tissues.312 Meanwhile, another portion of the drugs is released near tumor cells, enhancing the overall effectiveness of combination therapy.

The combination therapy strategy using multi-drug nanocarriers shows great potential for improving treatment precision and efficacy by simultaneously targeting CAFs and tumor cells. Through the synergistic effects of chemotherapy, targeted therapies, and immune drugs, combination therapy can overcome drug resistance and reduce toxic side effects on healthy tissues by optimizing drug distribution within the tumor microenvironment.313,314 With ongoing advancements in nanotechnology, future multi-drug nanocarriers are likely to integrate even more targeting mechanisms and release regulation strategies, further improving the overall effectiveness of cancer treatment.

In the preclinical stage, targeted therapies for CAFs have made significant strides across various animal models (Table III). Studies have demonstrated that nanodrug delivery systems, such as liposomes and polymer nanoparticles, along with surface modification strategies like antibodies and peptides, can effectively enhance the selective targeting of drugs to CAFs.111,269,312 These strategies also improve drug accumulation in tumor tissues, contributing to more efficient treatments. Additionally, combination therapy approaches, such as the use of chemotherapy drugs in conjunction with immunotherapy, have shown promising results in preclinical studies. By leveraging the synergistic effects of multiple drugs, these strategies not only enhance tumor therapeutic outcomes but also help overcome resistance that may arise from relying on a single drug.

TABLE III.

Current preclinical and clinical application progress of nanomedicines targeting CAFs.

Nanocarrier type Drug types Preclinical research progress Clinical application potential and challenges Refs.
Liposomes  Chemotherapy drugs (e.g., Doxorubicin), immune checkpoint inhibitors (e.g., PD-1 inhibitors)  1. Enhanced targeting of CAFs, increased drug accumulation in the tumor microenvironment.
2. Good selectivity and biocompatibility in animal models. 
1. Improving penetration into the tumor microenvironment.
2. Stability and scalability challenges in clinical translation.
3. FDA approval, manufacturing, and quality control issues. 
315–317  
Polymeric nanoparticles  Immune drugs (e.g., Anti-PD-1 antibody), chemotherapy drugs (e.g., Docetaxel)  1. Effectively co-loads multiple drugs, improving dual targeting to CAFs.
2. Significant antitumor effects in animal models. 
1. Addressing drug loading efficiency and nanoparticle stability.
2. Challenges in biodegradability and clearance rate optimization for clinical application. 
318,319  
Gold nanoparticles  Chemotherapy drugs, targeted drugs (e.g., antibody targeting CAFs or tumor vasculature)  1. Good biocompatibility and strong photothermal therapy effects.
2. Enhanced drug accumulation in tumors through surface modifications. 
1. Production scalability and cost control.
2. Further validation of biocompatibility and immunogenicity in humans. 
320,321  
Nanomicelles  Chemotherapy drugs (e.g., Paclitaxel), targeted drug conjugates (e.g., ADCs)  1. Surface modification (e.g., PEGylation) increases drug half-life and targeting ability.
2. Effective in reducing systemic toxicity in animal studies. 
1. Drug loading and release control still represent a bottleneck for clinical use.
2. Stability and long-term stability issues remain in clinical settings. 
322,323  
Nanoemulsions  Chemotherapy drugs, immune drugs, targeted small molecule drugs  1. Significantly increases drug concentration in tumors and improves CAF targeting.
2. Shows good antitumor activity and low toxicity in animal models. 
1. Stability and storage challenges for clinical applications.
2. Need further investigation into human safety and efficacy. 
324,325  
Nanotubes  Chemotherapy drugs (e.g., Cyclophosphamide), targeted drugs (e.g., antibodies, peptides)  1. High surface area and loading capacity, suitable for co-delivery of multiple drugs.
2. Effective targeting of CAFs and enhanced antitumor effects in animal studies. 
1. Clinical challenges include optimizing drug loading, release rates, and biocompatibility.
2. Issues with controlled drug release and long-term stability. 
254,326  
Nanocarrier type Drug types Preclinical research progress Clinical application potential and challenges Refs.
Liposomes  Chemotherapy drugs (e.g., Doxorubicin), immune checkpoint inhibitors (e.g., PD-1 inhibitors)  1. Enhanced targeting of CAFs, increased drug accumulation in the tumor microenvironment.
2. Good selectivity and biocompatibility in animal models. 
1. Improving penetration into the tumor microenvironment.
2. Stability and scalability challenges in clinical translation.
3. FDA approval, manufacturing, and quality control issues. 
315–317  
Polymeric nanoparticles  Immune drugs (e.g., Anti-PD-1 antibody), chemotherapy drugs (e.g., Docetaxel)  1. Effectively co-loads multiple drugs, improving dual targeting to CAFs.
2. Significant antitumor effects in animal models. 
1. Addressing drug loading efficiency and nanoparticle stability.
2. Challenges in biodegradability and clearance rate optimization for clinical application. 
318,319  
Gold nanoparticles  Chemotherapy drugs, targeted drugs (e.g., antibody targeting CAFs or tumor vasculature)  1. Good biocompatibility and strong photothermal therapy effects.
2. Enhanced drug accumulation in tumors through surface modifications. 
1. Production scalability and cost control.
2. Further validation of biocompatibility and immunogenicity in humans. 
320,321  
Nanomicelles  Chemotherapy drugs (e.g., Paclitaxel), targeted drug conjugates (e.g., ADCs)  1. Surface modification (e.g., PEGylation) increases drug half-life and targeting ability.
2. Effective in reducing systemic toxicity in animal studies. 
1. Drug loading and release control still represent a bottleneck for clinical use.
2. Stability and long-term stability issues remain in clinical settings. 
322,323  
Nanoemulsions  Chemotherapy drugs, immune drugs, targeted small molecule drugs  1. Significantly increases drug concentration in tumors and improves CAF targeting.
2. Shows good antitumor activity and low toxicity in animal models. 
1. Stability and storage challenges for clinical applications.
2. Need further investigation into human safety and efficacy. 
324,325  
Nanotubes  Chemotherapy drugs (e.g., Cyclophosphamide), targeted drugs (e.g., antibodies, peptides)  1. High surface area and loading capacity, suitable for co-delivery of multiple drugs.
2. Effective targeting of CAFs and enhanced antitumor effects in animal studies. 
1. Clinical challenges include optimizing drug loading, release rates, and biocompatibility.
2. Issues with controlled drug release and long-term stability. 
254,326  

Although nanocarrier delivery systems targeting CAFs have shown promising results in preclinical studies, their clinical translation still faces significant challenges. Future research should focus on optimizing nanocarrier design, improving drug targeting and penetration within the tumor microenvironment, addressing stability and pharmacokinetic concerns, and overcoming production and regulatory hurdles.26,111,144 As research deepens and more clinical data are accumulated, nano drug delivery systems targeting CAFs are expected to become a key component of personalized cancer treatment, offering more effective and less toxic therapeutic strategies.

CAFs play diverse and complex roles in the TME, and their potential for targeted therapy has gained widespread recognition.39,327 Beyond the application of nanomaterials, recent advancements in technologies such as biomedical engineering, biomaterials, tissue engineering, microfluidics, synthetic biology, and medical devices have further expanded the possibilities for CAFs targeting.112,328 In the realm of biomedical engineering and biomaterials, functional hydrogels, stimulus-responsive polymers, and exosome-mimicking materials not only facilitate accurate drug delivery but also effectively regulate the remodeling of the CAFs-driven ECM, overcoming drug penetration barriers.329–331 These advanced biomaterials, thus, provide a technological foundation for the targeted therapy of CAFs. Additionally, tissue engineering techniques, such as three-dimensional (3D) bioprinting models and tumor-on-a-chip platforms, enable the simulation of CAF dynamics within the TME.332–334 These innovations enhance drug screening efficiency and provide a deeper understanding of the interactions between CAFs and other components of the microenvironment, offering valuable experimental data for developing novel therapies.335 Meanwhile, microfluidic technology significantly enhances the specificity of targeted therapy through high-precision single-cell separation and drug delivery.336,337 Notably, the development of tumor microenvironment co-culture systems allows for real-time monitoring of CAF functions, providing crucial data for optimizing treatment combinations.338 Synthetic biology further broadens the scope of intelligent therapies. By using tools like CRISPR-Cas9 and synthetic gene circuits, researchers can target and knock out tumor-promoting genes in CAFs or dynamically regulate their phenotype, transforming them into an anti-tumor state.339,340 This enables multidimensional intervention in CAFs.

In parallel, medical devices such as implantable biosensors and catheter-based drug delivery systems offer innovative solutions for CAFs monitoring and intervention, particularly in precision treatment and real-time dynamic adjustments.341,342 These devices show enormous potential for clinical application. The synergistic integration of these technologies not only compensates for the limitations of individual treatment strategies but also opens up broader research possibilities for CAFs-targeted therapy. However, these technologies also face challenges. The heterogeneity and dynamic plasticity of CAFs increase the complexity of targeted therapy, while the clinical translation of these advanced technologies encounters high costs and regulatory hurdles.31,343 To overcome these challenges, future research should leverage technologies like single-cell genomics and proteomics to deeply analyze CAF subtype characteristics and optimize the production processes of biomaterials through interdisciplinary collaboration.37,344,345 In conclusion, with the further integration of biomedical engineering, tissue engineering, microfluidics, synthetic biology, and medical devices with nanomaterials, CAFs-targeted therapy is poised to play a pivotal role in precision medicine and personalized cancer treatment, offering a comprehensive approach to improving patient outcomes.

This study was supported by the Natural Science Foundation of Sichuan Province (2023NSFSC1237), University-Level Natural Science Foundation General Project of Chengdu Medical College (2024CDYXY-01), Clinical Science Research Foundation of Chengdu Medical College & the First Affiliated Hospital of Chengdu Medical College (24LHLNYX1-08), Clinical Science Research Foundation of Chengdu Medical College & Nanbu People's Hospital (2024LHFBM1-04), and Clinical Science Research Foundation of Chengdu Medical College & Chengdu Pidu People's Hospital (2024LHFYSZ1-41).

The authors have no conflicts to disclose.

Zhongsong Zhang and Yujie Tang contributed equally to this work.

Zhongsong Zhang: Conceptualization (lead); Investigation (lead); Software (lead); Validation (lead); Writing – original draft (equal); Writing – review & editing (equal). Yujie Tang: Conceptualization (equal); Software (equal); Visualization (equal); Writing – review & editing (equal). Dan Luo: Data curation (equal); Funding acquisition (supporting); Writing – review & editing (supporting). Jing Qiu: Conceptualization (equal); Funding acquisition (supporting); Visualization (equal); Writing – review & editing (supporting). Long Chen: Conceptualization (equal); Data curation (equal); Funding acquisition (equal); Resources (equal); Software (equal); Writing – review & editing (equal).

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

AI

Artificial intelligence

apCAF

Antigen-presenting cancer-associated fibroblasts

AuNPs

Gold nanoparticles

BCL-2

B-cell lymphoma 2

BCL-XL

B-cell lymphoma-extra large

BSA

Bovine serum albumin

CAFs

Cancer-associated fibroblasts

CD4+ T cells

CD4-positive T cells

CD8+ T cells

CD8-positive T cells

CNTs

Carbon nanotubes

CRISPR-Cas9

Clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9

CTLA-4

Cytotoxic T-lymphocyte-associated protein 4

CTLs

Cytotoxic T lymphocytes

CXCL12

C-X-C motif chemokine ligand 12

CXCR4

C-X-C chemokine receptor type 4

CYP450

Cytochrome P450

DOX

Doxorubicin

ECM

Extracellular matrix

EGFR

Epidermal growth factor receptor

EPCAM

Epithelial cell adhesion molecule

EPR

Enhanced permeability and retention

Exosomes

Extracellular vesicles

FABP3

Fatty acid-binding protein 3

FAP

Fibroblast activation protein

FAP-targeted NPs

Fibroblast activation protein-targeted nanoparticles

FAP-α

Fibroblast activation protein alpha

FASN

Fatty acid synthase

FDA

Food and Drug Administration

FeNPs

Iron oxide nanoparticles

FRT

Fibroblast-representative tumor

FSP-1

Fibroblast-specific protein-1

HGF

Hepatocyte growth factor

iCAF

Inflammatory cancer-associated fibroblasts

IL-10

Interleukin-10

IL-6

Interleukin-6

IL-6R

Interleukin-6 receptor

iNOS

Inducible nitric oxide synthase

LNPs

Lipid nanoparticles

mAb

Monoclonal antibody

MALDI-TOF

Matrix-assisted laser desorption/ionization time-of-flight

MDSCs

Myeloid-derived suppressor cells

MHC

Major histocompatibility complex

MHCII

Major histocompatibility complex class II

MMP

Matrix metalloproteinase

MMPs

Matrix metalloproteinases

MNPs

Magnetic nanoparticles

MOFs

Metal-organic frameworks

myCAF

Myofibroblastic cancer-associated fibroblasts

NDDS

Nanomedicine delivery systems

NF-κB

Nuclear factor kappa-light-chain-enhancer of activated B cells

NIR

Near-infrared

NK cells

Natural killer cells

NMR

Nuclear magnetic resonance

NMs

Nanomaterials

PD-1

Programmed cell death protein 1

PDGFR

Platelet-derived growth factor receptor

PD-L1

Programmed death-ligand 1

PDT

Photodynamic therapy

PEG

Polyethylene glycol

PGE2

Prostaglandin E2

PLGA

Poly(lactic-co-glycolic acid)

PLGA-NPs

Poly(lactic-co-glycolic acid) nanoparticles

RGD

Arginine-glycine-aspartic acid (peptide)

Rho GTPases

Ras homologous GTPases

RNA

Ribonucleic acid

scFv

Single-chain variable fragment

SDF-1

Stromal cell-derived factor 1

SeNPs

Selenium nanoparticles

siRNA

Small interfering RNA

SOSG

Singlet oxygen sensor green

SREBP2

Sterol regulatory element-binding protein 2

TAMs

Tumor-associated macrophages

TEM

Tumor extracellular matrix

TGF-β

Transforming growth factor-beta

TGF-β1

Transforming growth factor-beta 1

TME

Tumor microenvironment

TNF-α

Tumor necrosis factor-alpha

Tregs

Regulatory T cells

vCAF

Vascular cancer-associated fibroblasts

VEGF

Vascular endothelial growth factor

VEGFR

Vascular endothelial growth factor receptor

α-SMA

α-smooth muscle actin

%ID/g

Percentage injected dose per gram of tissue

1.
Y.
Liu
,
X.
Zhang
,
W.
Gu
et al, “
Unlocking the crucial role of cancer-associated fibroblasts in tumor metastasis: Mechanisms and therapeutic prospects
,”
J. Adv. Res.
(in press
2024
).
2.
X.
Zhang
,
M.
Zhang
,
H.
Sun
et al, “
The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells
,”
J. Adv. Res.
67
,
121
132
(
2025
).
3.
H.
Yamaguchi
and
M.
Miyazaki
, “
Heterocellular adhesion in cancer invasion and metastasis: Interactions between cancer cells and cancer-associated fibroblasts
,”
Cancers
16
(
9
),
1636
(
2024
).
4.
Y.
Zhang
,
Z.
Fang
,
D.
Pan
et al, “
Dendritic polymer-based nanomedicines remodel the tumor stroma: Improve drug penetration and enhance antitumor immune response
,”
Adv. Mater.
36
(
25
),
e2401304
(
2024
).
5.
W.
Shen
,
P. A.
Yao
,
W.
Li
et al, “
Cancer-associated fibroblast-targeted nanodrugs reshape colorectal tumor microenvironments to suppress tumor proliferation, metastasis and improve drug penetration
,”
J. Mater. Chem. B
11
(
9
),
1871
1880
(
2023
).
6.
S.
Ielpo
,
F.
Barberini
,
F.
Dabbagh Moghaddam
et al, “
Crosstalk and communication of cancer-associated fibroblasts with natural killer and dendritic cells: New frontiers and unveiled opportunities for cancer immunotherapy
,”
Cancer Treat. Rev.
131
,
102843
(
2024
).
7.
H.
Tian
,
W.
Wang
,
S.
Liang
,
J.
Ding
, and
D.
Hua
, “
From darkness to light: Targeting CAFs as a new potential strategy for cancer treatment
,”
Int. Immunopharmacol.
143
(
Pt. 2
),
113482
(
2024
).
8.
S.
Dai
,
Y.
Liu
,
Z.
Liu
et al, “
Cancer-associated fibroblasts mediate resistance to anti-EGFR therapies in cancer
,”
Pharmacol. Res.
206
,
107304
(
2024
).
9.
R.
Shah
,
K. A.
Johnson
,
A. E. L.
Lippert
et al, “
Cancer-associated fibroblast proteins as potential targets against colorectal cancers
,”
Cancers
16
(
18
),
3158
(
2024
).
10.
M.
Grotz
,
L.
van Gijzel
,
P.
Bitsch
,
S. C.
Carrara
,
H.
Kolmar
, and
S.
Garg
, “
Mimicking the immunosuppressive impact of fibroblasts in a 3D multicellular spheroid model
,”
Front. Drug Discovery
4
,
1427407
(
2024
).
11.
C. K.
Honda
,
S.
Kurozumi
,
T.
Fujii
et al, “
Cancer-associated fibroblast spatial heterogeneity and EMILIN1 expression in the tumor microenvironment modulate TGF-β activity and CD8+ T-cell infiltration in breast cancer
,”
Theranostics
14
(
5
),
1873
1885
(
2024
).
12.
Q.
Huang
,
Y.
Ge
,
Y.
He
et al, “
The application of nanoparticles targeting cancer-associated fibroblasts
,”
Int. J. Nanomed.
19
,
3333
3365
(
2024
).
13.
N.
Meher
,
A. P.
Bidkar
,
A.
Wadhwa
et al, “
PET imaging using 89Zr-labeled StarPEG nanocarriers reveals heterogeneous enhanced permeability and retention in prostate cancer
,”
Mol. Cancer Ther.
24
(
1
),
141
151
(
2025
).
14.
N.
Doaa
,
S. L. R.
Detorgma
,
K.
Yang
,
R.
Salama
, and
W.
Zhang
, “
Nanoparticle-mediated transcytosis in tumor drug delivery: Mechanisms, categories, and novel applications
,”
Curr. Drug Deliv.
(in press
2024
).
15.
A.
Jha
,
M.
Kumar
,
K.
Bharti
,
M.
Manjit
, and
B.
Mishra
, “
Biopolymer-based tumor microenvironment-responsive nanomedicine for targeted cancer therapy
,”
Nanomedicine
19
(
7
),
633
651
(
2024
).
16.
J. O.
Erebor
,
E. O.
Agboluaje
,
A. M.
Perkins
,
M.
Krishnakumar
, and
N.
Ngwuluka
, “
Targeted hybrid nanocarriers as co-delivery systems for enhanced cancer therapy
,”
Adv. Pharm. Bull.
14
(
3
),
558
573
(
2024
).
17.
Z.
Guo
,
J.
Ye
,
X.
Cheng
et al, “
Nanodrug delivery systems in antitumor immunotherapy
,”
Biomater. Res.
28
,
0015
(
2024
).
18.
X. L.
Hou
,
B.
Zhang
,
K.
Cheng
et al, “
Engineering phage nanocarriers integrated with bio-intelligent plasmids for personalized and tunable enzyme delivery to enhance chemodynamic therapy
,”
Adv. Sci.
11
(
24
),
e2308349
(
2024
).
19.
Y.
Wang
,
T.
Deng
,
X.
Liu
et al, “
Smart nanoplatforms responding to the tumor microenvironment for precise drug delivery in cancer therapy
,”
Int. J. Nanomed.
19
,
6253
6277
(
2024
).
20.
M.
Yang
,
J.
Li
,
P.
Gu
, and
X.
Fan
, “
The application of nanoparticles in cancer immunotherapy: Targeting tumor microenvironment
,”
Bioactive Mater.
6
(
7
),
1973
1987
(
2021
).
21.
H.
Karimi-Maleh
,
A. L.
Sanati
, and
R.
Darabi
, “
Nanomaterials in biomedicine, drug delivery and pharmaceutical analysis
,”
ADMET DMPK
11
(
4
),
433
434
(
2023
).
22.
M.
Skowicki
,
D.
Hürlimann
,
S.
Tarvirdipour
et al, “
FAP targeting of photosensitizer-loaded polymersomes for increased light-activated cell killing
,”
Biomacromolecules
25
(
2
),
754
766
(
2024
).
23.
K.
Sarma
,
M. H.
Akther
,
I.
Ahmad
et al, “
Adjuvant novel nanocarrier-based targeted therapy for lung cancer
,”
Molecules
29
(
5
),
1076
(
2024
).
24.
Y.
Li
,
M.
Tuerhan
,
B.
Li
,
S.
Chen
,
Y.
Wang
, and
Y.
Zheng
, “
RGD-modified ZIF-8 nanoparticles as a drug carrier for MR imaging and targeted drug delivery in myocardial infarction
,”
Nanomedicine
19
(
18–20
),
1585
1600
(
2024
).
25.
I.
Altinbasak
,
Y.
Alp
,
R.
Sanyal
, and
A.
Sanyal
, “
Theranostic nanogels: Multifunctional agents for simultaneous therapeutic delivery and diagnostic imaging
,”
Nanoscale
16
(
29
),
14033
14056
(
2024
).
26.
M.
Wang
,
W.
Xue
,
H.
Yuan
,
Z.
Wang
, and
L.
Yu
, “
Nano-drug delivery systems targeting CAFs: A promising treatment for pancreatic cancer
,”
Int. J. Nanomed.
19
,
2823
2849
(
2024
).
27.
C.
Peng
,
Y.
Xu
,
J.
Wu
,
D.
Wu
,
L.
Zhou
, and
X.
Xia
, “
TME-related biomimetic strategies against cancer
,”
Int. J. Nanomed.
19
,
109
135
(
2024
).
28.
L.
Ning
,
C.
Quan
,
Y.
Wang
,
Z.
Wu
,
P.
Yuan
, and
N.
Xie
, “
scRNA-seq characterizing the heterogeneity of fibroblasts in breast cancer reveals a novel subtype SFRP4+ CAF that inhibits migration and predicts prognosis
,”
Front. Oncol.
14
,
1348299
(
2024
).
29.
T.
Shinkawa
,
K.
Ohuchida
, and
M.
Nakamura
, “
Heterogeneity of cancer-associated fibroblasts and the tumor immune microenvironment in pancreatic cancer
,”
Cancers
14
(
16
),
3994
(
2022
).
30.
E.
Elyada
,
M.
Bolisetty
,
P.
Laise
et al, “
Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts
,”
Cancer Discovery
9
(
8
),
1102
1123
(
2019
).
31.
X.
Li
,
Z.
Sun
,
G.
Peng
et al, “
Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer
,”
Theranostics
12
(
2
),
620
638
(
2022
).
32.
L.
Cords
,
S.
Engler
,
M.
Haberecker
et al, “
Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer
,”
Cancer Cell
42
(
3
),
396
412
(
2024
).
33.
J.
Pan
,
Z.
Ma
,
B.
Liu
et al, “
Identification of cancer-associated fibroblasts subtypes in prostate cancer
,”
Front. Immunol.
14
,
1133160
(
2023
).
34.
S.
Long
,
J.
Wang
,
F.
Weng
,
D.
Xiang
, and
G.
Sun
, “
Extracellular matrix protein 1 regulates colorectal cancer cell proliferative, migratory, invasive and epithelial-mesenchymal transition activities through the PI3K/AKT/GSK3β/Snail signaling axis
,”
Front. Oncol.
12
,
889159
(
2022
).
35.
X.
Liu
,
Y.
Hu
,
C.
Li
et al, “
Overexpression of YEATS2 remodels the extracellular matrix to promote hepatocellular carcinoma progression via the PI3K/AKT pathway
,”
Cancers
15
(
6
),
1850
(
2023
).
36.
F.
Wang
,
Y.
Liu
,
W.
Qiu
et al, “
Functional analysis of MET Exon 14 skipping alteration in cancer invasion and metastatic dissemination
,”
Cancer Res.
82
(
7
),
1365
1379
(
2022
).
37.
X.
Zhang
,
R.
Zhu
,
D.
Yu
,
J.
Wang
,
Y.
Yan
, and
K.
Xu
, “
Single-cell RNA sequencing to explore cancer-associated fibroblasts heterogeneity: “Single” vision for “heterogeneous” environment
,”
Cell Proliferation
57
(
5
),
e13592
(
2024
).
38.
L.
Shi
,
W.
Zhu
,
Y.
Huang
et al, “
Cancer-associated fibroblast-derived exosomal microRNA-20a suppresses the PTEN/PI3K-AKT pathway to promote the progression and chemoresistance of non-small cell lung cancer
,”
Clin. Transl. Med.
12
(
7
),
e989
(
2022
).
39.
G.
Biffi
and
D. A.
Tuveson
, “
Diversity and biology of cancer-associated fibroblasts
,”
Physiol. Rev.
101
(
1
),
147
176
(
2021
).
40.
S.
Wen
,
Y.
Hou
,
L.
Fu
et al, “
Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling
,”
Cancer Lett.
442
,
320
332
(
2019
).
41.
Y.
Wen
,
Y.
Zhu
,
C.
Zhang
et al, “
Chronic inflammation, cancer development and immunotherapy
,”
Front. Pharmacol.
13
,
1040163
(
2022
).
42.
W.
Luo
,
Y.
Zeng
,
Q.
Song
et al, “
Strengthening the combinational immunotherapy from modulating the tumor inflammatory environment via hypoxia-responsive nanogels
,”
Adv. Healthcare Mater.
13
(
8
),
e2302865
(
2023
).
43.
A. R.
Naqash
et al, “
Increased interleukin-6/C-reactive protein levels are associated with the upregulation of the adenosine pathway and serve as potential markers of therapeutic resistance to immune checkpoint inhibitor-based therapies in non-small cell lung cancer
,”
J. Immunother. Cancer
11
(
10
),
e007310
(
2023
).
44.
J.
Datta
,
X.
Dai
,
A.
Bianchi
et al, “
Combined MEK and STAT3 inhibition uncovers stromal plasticity by enriching for cancer-associated fibroblasts with mesenchymal stem cell-like features to overcome immunotherapy resistance in pancreatic cancer
,”
Gastroenterology
163
(
6
),
1593
1612
(
2022
).
45.
S.
Rahmani
,
H. J.
Galipeau
,
A. V.
Clarizio
et al, “
Gluten-dependent activation of CD4+ T cells by MHC class II-expressing epithelium
,”
Gastroenterology
167
(
6
),
1113
1128
(
2024
).
46.
Y.
Kieffer
,
H. R.
Hocine
,
G.
Gentric
et al, “
Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer
,”
Cancer Discovery
10
(
9
),
1330
1351
(
2020
).
47.
H.
Huang
,
Z.
Wang
,
Y.
Zhang
et al, “
Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer
,”
Cancer Cell
40
(
6
),
656
673.e7
(
2022
).
48.
D.
Kerdidani
,
K.
Goudevenou
,
E.
Aerakis
et al, “
Antigen-presenting fibroblasts sustain anti-tumour CD4+ T cells in situ via MHCIIp-TCR and C1q-C1qbp binding
,” bioRxiv (
2020
).
49.
M.
López-Lago
,
W.
Williams
, and
C.
Wiseman
, “
Abstract P3-06-08: Turning tumor cells into antigen-presenting cells for cancer immunotherapy
,”
Cancer Res.
83
,
P3-06-08
(
2023
).
50.
Y.
Zhang
,
A. S.
Popel
, and
H.
Bazzazi
, “
Combining multikinase tyrosine kinase inhibitors targeting the vascular endothelial growth factor and cluster of differentiation 47 signaling pathways is predicted to increase the efficacy of antiangiogenic combination therapies
,”
ACS Pharmacol. Transl. Sci.
6
(
5
),
710
726
(
2023
).
51.
X.
Jiang
,
J.
Wang
,
X.
Deng
et al, “
The role of microenvironment in tumor angiogenesis
,”
J. Exp. Clin. Cancer Res.
39
(
1
),
204
(
2020
).
52.
Y.
Wu
,
Z.
Yi
,
J.
Li
et al, “
FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
,”
Theranostics
12
(
10
),
4564
4580
(
2022
).
53.
S. A.
Patel
,
M. B.
Nilsson
,
X.
Le
,
T.
Cascone
,
R. K.
Jain
, and
J. V.
Heymach
, “
Molecular mechanisms and future implications of VEGF/VEGFR in cancer therapy
,”
Clin. Cancer Res.
29
(
1
),
30
39
(
2023
).
54.
H.
Yang
,
S. J.
Lee
,
H.
Yang
et al, “
Abstract 5548: PB101 simultaneously targets tumor angiogenesis and immunity and enhances the efficacy of PD-L1 immune checkpoint blockade
,”
Cancer Res.
82
,
5548
(
2022
).
55.
J.
Götze
,
C.
Nitschke
,
F. G.
Uzunoglu
,
K.
Pantel
,
M.
Sinn
, and
H.
Wikman
, “
Tumor-stroma interaction in PDAC as a new approach for liquid biopsy and its potential clinical implications
,”
Front. Cell Dev. Biol.
10
,
918795
(
2022
).
56.
J.
Thorlacius-Ussing
,
C.
Jensen
,
N. I.
Nissen
et al, “
The collagen landscape in cancer: Profiling collagens in tumors and in circulation reveals novel markers of cancer-associated fibroblast subtypes
,”
J. Pathol.
262
(
1
),
22
36
(
2024
).
57.
D. S.
Foster
,
M.
Januszyk
,
D.
Delitto
et al, “
Multiomic analysis reveals conservation of cancer-associated fibroblast phenotypes across species and tissue of origin
,”
Cancer Cell
40
(
11
),
1392
1406.e7
(
2022
).
58.
See https://consensus.app/papers/abstract-291-unveiling-heterogeneity-of-kravets-shpak/bc058ae908a352beaaec9c42a9d6c03b/?utm_source=chatgpt for “
Abstract 291: Unveiling heterogeneity of cancer-associated fibroblasts (CAFs) across multiple solid cancer types
,” accessed: January 23, 2025.
59.
See https://consensus.app/papers/abstract-6827-myocardin-and-myocardinrelated-cornelio-johnson/ad673896aaec5fffbcb54af33de0ae4e/?utm_source=chatgpt for “
Abstract 6827: Myocardin and myocardin-related transcription factor as potential drivers of the myofibroblastic cancer-associated fibroblast phenotype in colorectal cancer
,” accessed: January 23, 2025.
60.
See https://consensus.app/papers/abstract-1577-cancer-associated-fibroblast-tumor-cell-guinn-perez/8e85513549ce5d738ad33d718575839f/?utm_source=chatgpt for “
Abstract 1577: Cancer associated fibroblast - tumor cell crosstalk enhances epithelial to mesenchymal transition and promotes a classical to basal switch in human pancreatic ductal adenocarcinoma
,” accessed: January 23, 2025.
61.
D.
Coursier
and
F.
Calvo
, “
CAFs vs. TECs: When blood feuds fuel cancer progression, dissemination and therapeutic resistance
,”
Cell Oncol.
47
(
4
),
1091
1112
(
2024
).
62.
Y.
Shintani
,
T.
Kimura
,
S.
Funaki
,
N.
Ose
,
T.
Kanou
, and
E.
Fukui
, “
Therapeutic targeting of cancer-associated fibroblasts in the non-small cell lung cancer tumor microenvironment
,”
Cancers
15
(
2
),
335
(
2023
).
63.
C.
Zhang
,
Y.
Fei
,
H.
Wang
et al, “
CAFs orchestrates tumor immune microenvironment-A new target in cancer therapy?
Front. Pharmacol.
14
,
1113378
(
2023
).
64.
R. A.
Glabman
,
C. P.
Olkowski
,
H. A.
Minor
et al, “
Tumor suppression by anti-fibroblast activation protein near-infrared photoimmunotherapy targeting cancer-associated fibroblasts
,”
Cancers
16
(
2
),
449
(
2024
).
65.
Z.
Liao
,
Z. W.
Tan
,
P.
Zhu
, and
N. S.
Tan
, “
Cancer-associated fibroblasts in tumor microenvironment - Accomplices in tumor malignancy
,”
Cell Immunol.
343
,
103729
(
2019
).
66.
J.
Bai
,
T.
Liu
,
B.
Tu
et al, “
Autophagy loss impedes cancer-associated fibroblast activation via downregulating proline biosynthesis
,”
Autophagy
19
(
2
),
632
643
(
2023
).
67.
M.
Janani
,
A.
Poorkhani
,
T.
Amiriani
et al, “
Association of future cancer metastases with fibroblast activation protein-α: A systematic review and meta-analysis
,”
Front. Oncol.
14
,
1339050
(
2024
).
68.
C. S.
Cheng
,
P. W.
Yang
,
Y.
Sun
,
S. L.
Song
, and
Z.
Chen
, “
Fibroblast activation protein-based theranostics in pancreatic cancer
,”
Front. Oncol.
12
,
969731
(
2022
).
69.
J. S.
Knuutila
,
P.
Riihilä
,
L.
Nissinen
et al, “
Cancer-associated fibroblast activation predicts progression, metastasis, and prognosis of cutaneous squamous cell carcinoma
,”
Int. J. Cancer
155
(
6
),
1112
1127
(
2024
).
70.
M. K.
Kang
,
F.
Jiang
,
Y. J.
Kim
et al, “
CTHRC1 induces pancreatic stellate cells (PSCs) into myofibroblast-like cancer-associated fibroblasts (myCAFs)
,”
Cancers
15
(
13
),
3370
(
2023
).
71.
Y. T.
Chang
,
H. Y.
Peng
,
C. M.
Hu
et al, “
Pancreatic cancer-derived small extracellular vesical ezrin activates fibroblasts to exacerbate cancer metastasis through STAT3 and YAP-1 signaling pathways
,”
Mol. Oncol.
17
(
8
),
1628
1647
(
2023
).
72.
T.
Akiyama
,
T.
Yasuda
,
T.
Uchihara
et al, “
Stromal reprogramming through dual PDGFRα/β blockade boosts the efficacy of anti-PD-1 immunotherapy in fibrotic tumors
,”
Cancer Res.
83
(
5
),
753
770
(
2023
).
73.
Y.
Pan
,
X.
Liao
,
L.
Yang
et al, “
Extract of Marsdenia tenacissima (Roxb.) Moon [Apocynaceae] suppresses hepatocellular carcinoma by inhibiting angiogenesis
,”
Front. Pharmacol.
13
,
900128
(
2022
).
74.
Z.
Ge
,
Q.
Zhang
,
W.
Lin
,
X.
Jiang
, and
Y.
Zhang
, “
The role of angiogenic growth factors in the immune microenvironment of glioma
,”
Front. Oncol.
13
,
1254694
(
2023
).
75.
X.
Li
,
Z.
Li
,
M.
Huang
et al, “
Gallium-68-labeled ZPDGFRβ affibody: A potential PET probe for platelet-derived growth factor receptor β-expressing carcinomas
,”
Mol. Pharm.
20
(
2
),
1357
1364
(
2023
).
76.
C.
Wu
,
D.
Li
,
X.
Cheng
,
H.
Gu
,
Y.
Qian
, and
L.
Feng
, “
Downregulation of cancer-associated fibroblast exosome-derived miR-29b-1-5p restrains vasculogenic mimicry and apoptosis while accelerating migration and invasion of gastric cancer cells via immunoglobulin domain-containing 1/zonula occluden-1 axis
,”
Cell Cycle
22
(
17
),
1807
1826
(
2023
).
77.
See https://consensus.app/papers/abstract-lb446-decoding-the-mechanism-of-metastasis-a-prieto-nu%C3%B1ez/7bc7596b13f55945973c6a6a9aa82c0b/?utm_source=chatgpt for “
Abstract LB446: Decoding the mechanism of metastasis: A novel CAF-therapy that blocks the tumor microenvironment and reverses metastatic traits in all solid tumors
,” accessed: January 23, 2025.
78.
M.
Bartoschek
,
N.
Oskolkov
,
M.
Bocci
et al, “
Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing
,”
Nat. Commun.
9
(
1
),
5150
(
2018
).
79.
T.
Simon
and
B.
Salhia
, “
Cancer-associated fibroblast subpopulations with diverse and dynamic roles in the tumor microenvironment
,”
Mol. Cancer Res.
20
(
2
),
183
192
(
2022
).
80.
S. S.
Yang
,
S.
Ma
,
H.
Dou
et al, “
Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment
,”
Exp. Cell Res.
391
(
2
),
111983
(
2020
).
81.
H.
Suzuki
,
T.
Ohishi
,
M. K.
Kaneko
, and
Y.
Kato
, “
A humanized and defucosylated antibody against podoplanin (humLpMab-23-f) exerts antitumor activities in human lung cancer and glioblastoma xenograft models
,”
Cancers
15
(
20
),
5080
(
2023
).
82.
C.
Neuzillet
,
R.
Nicolle
,
J.
Raffenne
et al, “
Periostin- and podoplanin-positive cancer-associated fibroblast subtypes cooperate to shape the inflamed tumor microenvironment in aggressive pancreatic adenocarcinoma
,”
J. Pathol.
258
(
4
),
408
425
(
2022
).
83.
A. M.
Xu
,
M.
Haro
,
A. E.
Walts
et al, “
Spatiotemporal architecture of immune cells and cancer-associated fibroblasts in high-grade serous ovarian carcinoma
,”
Sci. Adv.
10
(
16
),
eadk8805
(
2024
).
84.
Z.
Zhao
,
T.
Li
,
Y.
Yuan
, and
Y.
Zhu
, “
What is new in cancer-associated fibroblast biomarkers?
Cell Commun. Signal.
21
(
1
),
96
(
2023
).
85.
T.
Hernández-Jiménez
,
P.
Cruz-Nova
,
A.
Ancira-Cortez
et al, “
Toxicity assessment of [177Lu]Lu-iFAP/iPSMA nanoparticles prepared under GMP-compliant radiopharmaceutical processes
,”
Nanomaterials
12
(
23
),
4181
(
2022
).
86.
M.
Nurmik
,
P.
Ullmann
,
F.
Rodriguez
,
S.
Haan
, and
E.
Letellier
, “
In search of definitions: Cancer-associated fibroblasts and their markers
,”
Int. J. Cancer
146
(
4
),
895
905
(
2020
).
87.
X.
Mao
,
J.
Xu
,
W.
Wang
et al, “
Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives
,”
Mol. Cancer
20
(
1
),
131
(
2021
).
88.
D.
Hu
,
Z.
Li
,
B.
Zheng
et al, “
Cancer‐associated fibroblasts in breast cancer: Challenges and opportunities
,”
Cancer Commun.
42
,
401
434
(
2022
).
89.
A.
Papait
,
J.
Romoli
,
F. R.
Stefani
et al, “
Fight the Cancer, Hit the CAF!
,”
Cancers
14
(
15
),
3570
(
2022
).
90.
F.
Eskandari-Malayeri
and
M.
Rezaei
, “
Immune checkpoint inhibitors as mediators for immunosuppression by cancer-associated fibroblasts: A comprehensive review
,”
Front. Immunol.
13
,
996145
(
2022
).
91.
R.
Siddhartha
and
M.
Garg
, “
Interplay between extracellular matrix remodeling and angiogenesis in tumor ecosystem
,”
Mol. Cancer Ther.
22
(
3
),
291
305
(
2023
).
92.
C.
Han
,
T.
Liu
, and
R.
Yin
, “
Biomarkers for cancer-associated fibroblasts
,”
Biomark. Res.
8
(
1
),
64
(
2020
).
93.
E.
Strating
,
M. P.
Verhagen
,
E.
Wensink
et al, “
Co-cultures of colon cancer cells and cancer-associated fibroblasts recapitulate the aggressive features of mesenchymal-like colon cancer
,”
Front. Immunol.
14
,
1053920
(
2023
).
94.
P.
Cirri
and
P.
Chiarugi
, “
Cancer-associated-fibroblasts and tumour cells: A diabolic liaison driving cancer progression
,”
Cancer Metastasis Rev.
31
(
1–2
),
195
208
(
2012
).
95.
G.
Sharbeen
,
J. A.
McCarroll
,
A.
Akerman
et al, “
Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma determine response to SLC7A11 inhibition
,”
Cancer Res.
81
(
13
),
3461
3479
(
2021
).
96.
J. Y.
Sung
and
J. W.
Lee
, “
Cancer-associated fibroblast subtypes reveal distinct gene signatures in the tumor immune microenvironment of vestibular schwannoma
,”
Cells
13
(
19
),
1669
(
2024
).
97.
H.
Xiang
,
Y.
Pan
,
M. A.
Sze
et al, “
Single-cell analysis identifies NOTCH3-mediated interactions between stromal cells that promote microenvironment remodeling and invasion in lung adenocarcinoma
,”
Cancer Res.
84
(
9
),
1410
1425
(
2024
).
98.
Z.
Fang
,
Q.
Meng
,
J.
Xu
et al, “
Signaling pathways in cancer-associated fibroblasts: Recent advances and future perspectives
,”
Cancer Commun.
43
(
1
),
3
41
(
2023
).
99.
D.
Park
,
E.
Sahai
, and
A.
Rullan
, “
SnapShot: Cancer-associated fibroblasts
,”
Cell
181
(
2
),
486
486.e1
(
2020
).
100.
X.
Zhang
,
M.
Lao
,
H.
Yang
et al, “
Targeting cancer-associated fibroblast autophagy renders pancreatic cancer eradicable with immunochemotherapy by inhibiting adaptive immune resistance
,”
Autophagy
20
(
6
),
1314
1334
(
2024
).
101.
X.
Geng
,
L.
Li
,
Y.
Luo
et al, “
Tumor cell derived Lnc-FSD2-31:1 contributes to cancer-associated fibroblasts activation in pancreatic ductal adenocarcinoma progression through extracellular vesicles cargo MiR-4736
,”
Adv. Sci.
10
(
10
),
e2203324
(
2023
).
102.
D.
Yang
,
J.
Liu
,
H.
Qian
, and
Q.
Zhuang
, “
Cancer-associated fibroblasts: From basic science to anticancer therapy
,”
Exp. Mol. Med.
55
(
7
),
1322
1332
(
2023
).
103.
O.
Piwocka
,
I.
Piotrowski
,
W. M.
Suchorska
, and
K.
Kulcenty
, “
Dynamic interactions in the tumor niche: How the cross-talk between CAFs and the tumor microenvironment impacts resistance to therapy
,”
Front. Mol. Biosci.
11
,
1343523
(
2024
).
104.
S. P.
Singh
,
A. R.
Dosch
,
S.
Mehra
et al, “
Tumor cell-intrinsic p38 MAPK signaling promotes IL1α-mediated stromal inflammation and therapeutic resistance in pancreatic cancer
,”
Cancer Res.
84
(
8
),
1320
1332
(
2024
).
105.
X.
Meng
,
Z.
Liu
,
L.
Deng
et al, “
Hydrogen therapy reverses cancer-associated fibroblasts phenotypes and remodels stromal microenvironment to stimulate systematic anti-tumor immunity
,”
Adv. Sci.
11
(
28
),
e2401269
(
2024
).
106.
Y.
Zhao
,
Y.
Dong
,
S.
Yang
et al, “
Bioorthogonal equipping CAR-T cells with hyaluronidase and checkpoint blocking antibody for enhanced solid tumor immunotherapy
,”
ACS Cent. Sci.
8
(
5
),
603
614
(
2022
).
107.
Y.
Xu
,
W.
Li
,
S.
Lin
,
B.
Liu
,
P.
Wu
, and
L.
Li
, “
Fibroblast diversity and plasticity in the tumor microenvironment: Roles in immunity and relevant therapies
,”
Cell Commun. Signal.
21
(
1
),
234
(
2023
).
108.
Y. E.
Lee
,
G. Y.
Go
,
E. Y.
Koh
et al, “
Synergistic therapeutic combination with a CAF inhibitor enhances CAR-NK-mediated cytotoxicity via reduction of CAF-released IL-6
,”
J. Immunother. Cancer
11
(
2
),
e006130
(
2023
).
109.
H.
Lai
,
R.
Huang
,
X.
Weng
,
B.
Huang
,
J.
Yao
, and
Y.
Pian
, “
Classification and applications of nanomaterials in vitro diagnosis
,”
Heliyon
10
(
11
),
e32314
(
2024
).
110.
X.
Meng
,
G.
Zhu
,
Y. G.
Yang
, and
T.
Sun
, “
Targeted delivery strategies: The interactions and applications of nanoparticles in liver diseases
,”
Biomed. Pharmacother.
175
,
116702
(
2024
).
111.
J.
Guo
,
H.
Zeng
, and
Y.
Chen
, “
Emerging nano drug delivery systems targeting cancer-associated fibroblasts for improved antitumor effect and tumor drug penetration
,”
Mol. Pharm.
17
(
4
),
1028
1048
(
2020
).
112.
J.
Mu
,
S.
Gao
,
J.
Yang
,
F.
Wu
, and
H.
Zhou
, “
Fundamental and clinical applications of materials based on cancer-associated fibroblasts in cancers
,”
Int. J. Mol. Sci.
22
(
21
),
11671
(
2021
).
113.
D.
Kovács
,
N.
Igaz
,
A.
Marton
et al, “
Core-shell nanoparticles suppress metastasis and modify the tumour-supportive activity of cancer-associated fibroblasts
,”
J. Nanobiotechnol.
18
(
1
),
18
(
2020
).
114.
N.
Mahhengam
,
K.
Kazemnezhad
,
H.
Setia Budi
et al, “
Targeted therapy of tumour microenvironment by gold nanoparticles as a new therapeutic approach
,”
J. Drug Target
30
(
5
),
494
510
(
2022
).
115.
C.
Xia
,
J.
Pan
,
J.
Wang
et al, “
Functional blockade of cancer-associated fibroblasts with ultrafine gold nanomaterials causes an unprecedented bystander antitumoral effect
,”
Nanoscale
12
(
38
),
19833
19843
(
2020
).
116.
U. K.
Sukumar
,
R. J. C.
Bose
,
M.
Malhotra
et al, “
Intranasal delivery of targeted polyfunctional gold-iron oxide nanoparticles loaded with therapeutic microRNAs for combined theranostic multimodality imaging and presensitization of glioblastoma to temozolomide
,”
Biomaterials
218
,
119342
(
2019
).
117.
Y.
Zhang
,
X.
Xiong
,
Y.
Huai
et al, “
Gold nanoparticles disrupt tumor microenvironment - endothelial cell cross talk to inhibit angiogenic phenotypes in vitro
,”
Bioconjugate Chem.
30
(
6
),
1724
1733
(
2019
).
118.
U. Y.
Lee
,
Y. S.
Youn
,
J.
Park
, and
E. S.
Lee
, “
Y-shaped ligand-driven gold nanoparticles for highly efficient tumoral uptake and photothermal ablation
,”
ACS Nano
8
(
12
),
12858
12865
(
2014
).
119.
Z. R.
Goddard
,
M. J.
Marín
,
D. A.
Russell
, and
M.
Searcey
, “
Active targeting of gold nanoparticles as cancer therapeutics
,”
Chem. Soc. Rev.
49
(
23
),
8774
8789
(
2020
).
120.
Q.
Ruan
,
J.
Feng
,
Y.
Jiang
et al, “
Preparation and bioevaluation of 99mTc-labeled FAP inhibitors as tumor radiotracers to target the fibroblast activation protein
,”
Mol. Pharm.
19
(
1
),
160
171
(
2022
).
121.
Q.
Ruan
,
Q.
Wang
,
Y.
Jiang
,
J.
Feng
,
G.
Yin
, and
J.
Zhang
, “
Synthesis and evaluation of 99mTc-labeled FAP inhibitors with different linkers for imaging of fibroblast activation proteins in tumors
,”
J. Med. Chem.
66
(
7
),
4952
4960
(
2023
).
122.
H.
Xia
,
J.
Zhu
,
C.
Men
et al, “
Light-initiated aggregation of gold nanoparticles for synergistic chemo-photothermal tumor therapy
,”
Nanoscale Adv.
5
(
11
),
3053
3062
(
2023
).
123.
L.
Yang
,
Y. T.
Tseng
,
G.
Suo
et al, “
Photothermal therapeutic response of cancer cells to aptamer-gold nanoparticle-hybridized graphene oxide under NIR illumination
,”
ACS Appl. Mater. Interfaces
7
(
9
),
5097
5106
(
2015
).
124.
J.
Kim
,
Y.
Nah
,
S.
Kim
, and
W. J.
Kim
, “
Transformation of nanoparticles via the transition of functional DNAs responsive to pH and vascular endothelial growth factor for photothermal anti-tumor therapy
,”
Biomater. Sci.
12
(
4
),
1031
1041
(
2024
).
125.
K. F.
Tan
,
L. L. A.
In
, and
P.
Vijayaraj Kumar
, “
Surface functionalization of gold nanoparticles for targeting the tumor microenvironment to improve antitumor efficiency
,”
ACS Appl. Bio Mater.
6
(
8
),
2944
2981
(
2023
).
126.
Z. H.
Feng
,
Z. T.
Li
,
S.
Zhang
et al, “
A combination strategy based on an Au nanorod/doxorubicin gel via mild photothermal therapy combined with antigen-capturing liposomes and anti-PD-L1 agent promote a positive shift in the cancer-immunity cycle
,”
Acta Biomater.
136
,
495
507
(
2021
).
127.
N. S.
Thakur
,
G.
Patel
,
V.
Kushwah
,
S.
Jain
, and
U. C.
Banerjee
, “
Self-assembled gold nanoparticle-lipid nanocomposites for on-demand delivery, tumor accumulation, and combined photothermal-photodynamic therapy
,”
ACS Appl. Bio Mater.
2
(
1
),
349
361
(
2019
).
128.
D.
Luo
,
X.
Wang
,
C.
Burda
, and
J. P.
Basilion
, “
Recent development of gold nanoparticles as contrast agents for cancer diagnosis
,”
Cancers
13
(
8
),
1825
(
2021
).
129.
S. M.
van de Looij
,
E. R.
Hebels
,
M.
Viola
,
M.
Hembury
,
S.
Oliveira
, and
T.
Vermonden
, “
Gold nanoclusters: Imaging, therapy, and theranostic roles in biomedical applications
,”
Bioconjugate Chem.
33
(
1
),
4
23
(
2022
).
130.
H.
Xiong
,
Z.
Huang
,
Q.
Lin
et al, “
Surface plasmon coupling electrochemiluminescence immunosensor based on polymer dots and AuNPs for ultrasensitive detection of pancreatic cancer exosomes
,”
Anal. Chem.
94
(
2
),
837
846
(
2022
).
131.
L. P.
Roguin
,
N.
Chiarante
,
M. C.
García Vior
, and
J.
Marino
, “
Zinc(II) phthalocyanines as photosensitizers for antitumor photodynamic therapy
,”
Int. J. Biochem. Cell Biol.
114
,
105575
(
2019
).
132.
Ł.
Lamch
,
J.
Kulbacka
,
M.
Dubińska-Magiera
,
J.
Saczko
, and
K. A.
Wilk
, “
Folate-directed zinc (II) phthalocyanine loaded polymeric micelles engineered to generate reactive oxygen species for efficacious photodynamic therapy of cancer
,”
Photodiagn. Photodyn. Ther.
25
,
480
491
(
2019
).
133.
Z. W.
Qiu
,
Y. T.
Zhong
,
Z. M.
Lu
et al, “
Breaking physical barrier of fibrotic breast cancer for photodynamic immunotherapy by remodeling tumor extracellular matrix and reprogramming cancer-associated fibroblasts
,”
ACS Nano
18
(
13
),
9713
9735
(
2024
).
134.
A.
Dag
,
E.
Cakilkaya
,
P. S.
Omurtag Ozgen
et al, “
Phthalocyanine-conjugated glyconanoparticles for chemo-photodynamic combination therapy
,”
Biomacromolecules
22
(
4
),
1555
1567
(
2021
).
135.
N. W.
Nkune
and
H.
Abrahamse
, “
The efficacy of zinc phthalocyanine nanoconjugate on melanoma cells grown as three-dimensional multicellular tumour spheroids
,”
Pharmaceutics
15
(
9
),
2264
(
2023
).
136.
S.
Zhou
,
Z.
Zhen
,
A. V.
Paschall
et al, “
FAP-targeted photodynamic therapy mediated by ferritin nanoparticles elicits an immune response against cancer cells and cancer associated fibroblasts
,”
Adv. Funct. Mater.
31
(
7
),
2007017
(
2021
).
137.
M.
Lan
et al, “
Photosensitizers for photodynamic therapy
,”
Adv. Mater. Sci.
8
(
13
),
1900132
(
2019
).
138.
N.
Dragicevic
,
J.
Predic-Atkinson
,
B.
Nikolic
,
S. B.
Pajovic
,
S.
Ivkovic
, and
M.
Adzic
, “
Nanocarriers in topical photodynamic therapy
,”
Expert Opin. Drug Delivery
21
(
2
),
279
307
(
2024
).
139.
Y.
Zhang
,
B.
Wang
,
R.
Zhao
,
Q.
Zhang
, and
X.
Kong
, “
Multifunctional nanoparticles as photosensitizer delivery carriers for enhanced photodynamic cancer therapy
,”
Mater. Sci. Eng. C
115
,
111099
(
2020
).
140.
M.
Zhang
and
H.
Xu
, “
Peptide-assembled nanoparticles targeting tumor cells and tumor microenvironment for cancer therapy
,”
Front. Chem.
11
,
1115495
(
2023
).
141.
J.
Lang
,
X.
Zhao
,
Y.
Qi
et al, “
Reshaping prostate tumor microenvironment to suppress metastasis via cancer-associated fibroblast inactivation with peptide-assembly-based nanosystem
,”
ACS Nano
13
(
11
),
12357
12371
(
2019
).
142.
P. H.
Wu
,
Y.
Onodera
,
Y.
Ichikawa
et al, “
Targeting integrins with RGD-conjugated gold nanoparticles in radiotherapy decreases the invasive activity of breast cancer cells
,”
Int. J. Nanomed.
12
,
5069
5085
(
2017
).
143.
X.
Lv
,
C.
Zhang
,
Q.
Shuaizhen
,
R.
Yu
, and
Y.
Zheng
, “
Design of integrin αvβ3 targeting self-assembled protein nanoparticles with RGD peptide
,”
Biomed. Pharmacother.
128
,
110236
(
2020
).
144.
L.
Kaps
and
D.
Schuppan
, “
Targeting cancer associated fibroblasts in liver fibrosis and liver cancer using nanocarriers
,”
Cells
9
(
9
),
2027
(
2020
).
145.
P. G.
Calavia
et al, “
Targeted photodynamic therapy of breast cancer cells using lactose-phthalocyanine functionalized gold nanoparticles
,”
J. Colloid Interface Sci.
512
,
249
259
(
2018
).
146.
S.
Umar
and
A.
Samokhvalov
, “
Encapsulation of gemcitabine on porphyrin aluminum metal-organic framework by mechano-chemistry, delayed drug release and cytotoxicity to pancreatic cancer PANC-1 cells
,”
Molecules
29
(
13
),
3189
(
2024
).
147.
M. R.
di Nunzio
,
V.
Agostoni
,
B.
Cohen
,
R.
Gref
, and
A.
Douhal
, “
A “ship in a bottle” strategy to load a hydrophilic anticancer drug in porous metal organic framework nanoparticles: Efficient encapsulation, matrix stabilization, and photodelivery
,”
J. Med. Chem.
57
(
2
),
411
420
(
2014
).
148.
Y.
Chen
,
R.
Lyu
,
J.
Wang
et al, “
Metal-organic frameworks nucleated by silk fibroin and modified with tumor-targeting peptides for targeted multimodal cancer therapy
,”
Adv. Sci.
10
(
28
),
e2302700
(
2023
).
149.
L. Y.
Cruz
,
D.
Wang
, and
J.
Liu
, “
Biosynthesis of selenium nanoparticles, characterization and X-ray induced radiotherapy for the treatment of lung cancer with interstitial lung disease
,”
J. Photochem. Photobiol. B
191
,
123
127
(
2019
).
150.
H.
Shi
,
B.
Wang
,
H.
Ma
et al, “
Preparation of biomimetic selenium-baicalein nanoparticles and their targeted therapeutic application in nonsmall cell lung cancer
,”
Mol. Pharm.
21
(
9
),
4476
4489
(
2024
).
151.
Q.
Yu
,
Y.
Qiu
,
J.
Li
et al, “
Targeting cancer-associated fibroblasts by dual-responsive lipid-albumin nanoparticles to enhance drug perfusion for pancreatic tumor therapy
,”
J. Control Release
321
,
564
575
(
2020
).
152.
Y.
Gregoriou
,
G.
Gregoriou
,
V.
Yilmaz
et al, “
Resveratrol loaded polymeric micelles for theranostic targeting of breast cancer cells
,”
Nanotheranostics
5
(
1
),
113
124
(
2021
).
153.
P.
Dana
,
N.
Thumrongsiri
,
P.
Tanyapanyachon
,
W.
Chonniyom
,
P.
Punnakitikashem
, and
N.
Saengkrit
, “
Resveratrol loaded liposomes disrupt cancer associated fibroblast communications within the tumor microenvironment to inhibit colorectal cancer aggressiveness
,”
Nanomaterials
13
(
1
),
107
(
2022
).
154.
P. T.
Yin
,
T.
Pongkulapa
,
H. Y.
Cho
et al, “
Overcoming chemoresistance in cancer via combined MicroRNA therapeutics with anticancer drugs using multifunctional magnetic core-shell nanoparticles
,”
ACS Appl. Mater. Interfaces
10
(
32
),
26954
26963
(
2018
).
155.
A.
Ahmad
,
A.
Gupta
,
M. M.
Ansari
,
A.
Vyawahare
,
G.
Jayamurugan
, and
R.
Khan
, “
Hyperbranched polymer-functionalized magnetic nanoparticle-mediated hyperthermia and niclosamide bimodal therapy of colorectal cancer cells
,”
ACS Biomater. Sci. Eng.
6
(
2
),
1102
1111
(
2020
).
156.
X.
Li
,
X.
Wu
,
H.
Yang
,
L.
Li
,
Z.
Ye
, and
Y.
Rao
, “
A nuclear targeted Dox-aptamer loaded liposome delivery platform for the circumvention of drug resistance in breast cancer
,”
Biomed. Pharmacother.
117
,
109072
(
2019
).
157.
X.
Zhao
,
F.
Li
,
Y.
Li
et al, “
Co-delivery of HIF1α siRNA and gemcitabine via biocompatible lipid-polymer hybrid nanoparticles for effective treatment of pancreatic cancer
,”
Biomaterials
46
,
13
25
(
2015
).
158.
B.
Kim
,
J.
Shin
,
J.
Wu
et al, “
Engineering peptide-targeted liposomal nanoparticles optimized for improved selectivity for HER2-positive breast cancer cells to achieve enhanced in vivo efficacy
,”
J. Control Release
322
,
530
541
(
2020
).
159.
S.
Zanganeh
,
G.
Hutter
,
R.
Spitler
et al, “
Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues
,”
Nat. Nanotechnol.
11
(
11
),
986
994
(
2016
).
160.
T.
Zuo
,
J.
Zhang
,
J.
Yang
et al, “
On-demand responsive nanoplatform mediated targeting of CAFs and down-regulating mtROS-PYK2 signaling for antitumor metastasis
,”
Biomater. Sci.
9
(
5
),
1872
1885
(
2021
).
161.
N.
Pang
,
Z.
Yang
,
W.
Zhang
et al, “
Cancer-associated fibroblasts barrier breaking via TGF-β blockade paved way for docetaxel micelles delivery to treat pancreatic cancer
,”
Int. J. Pharm.
665
,
124706
(
2024
).
162.
S.
Lin
,
Y.
Sun
,
C.
Cao
et al, “
Single-nucleus RNA sequencing reveals heterogenous microenvironments and specific drug response between cervical squamous cell carcinoma and adenocarcinoma
,”
eBioMedicine
97
,
104846
(
2023
).
163.
T.
Guo
and
J.
Xu
, “
Cancer-associated fibroblasts: A versatile mediator in tumor progression, metastasis, and targeted therapy
,”
Cancer Metastasis Rev.
43
,
1095
(
2024
).
164.
R.
Mhaidly
et al, “
Role of cancer‐associated fibroblast subpopulations in immune infiltration, as a new means of treatment in cancer
,”
Immunol. Rev.
302
(
1
),
259
272
(
2021
).
165.
S.
Yazdani
,
R.
Bansal
, and
J.
Prakash
, “
Drug targeting to myofibroblasts: Implications for fibrosis and cancer
,”
Adv. Drug Delivery Rev.
121
,
101
116
(
2017
).
166.
X.
Guo
,
L.
Yang
,
C.
Deng
et al, “
Nanoparticles traversing the extracellular matrix induce biophysical perturbation of fibronectin depicted by surface chemistry
,”
Nanoscale
16
(
12
),
6199
6214
(
2024
).
167.
D.
Cheng
,
Y.
Ji
,
B.
Wang
et al, “
Dual-responsive nanohybrid based on degradable silica-coated gold nanorods for triple-combination therapy for breast cancer
,”
Acta Biomater.
128
,
435
446
(
2021
).
168.
A.
Masjedi
,
A.
Ahmadi
,
F.
Atyabi
et al, “
Silencing of IL-6 and STAT3 by siRNA loaded hyaluronate-N,N,N-trimethyl chitosan nanoparticles potently reduces cancer cell progression
,”
Int. J. Biol. Macromol.
149
,
487
500
(
2020
).
169.
R.
Zhao
,
J.
Liu
,
Z.
Li
,
W.
Zhang
,
F.
Wang
, and
B.
Zhang
, “
Recent advances in CXCL12/CXCR4 antagonists and nano-based drug delivery systems for cancer therapy
,”
Pharmaceutics
14
(
8
),
1541
(
2022
).
170.
Y.
Zeng
et al, “
Dual blockade of CXCL12‐CXCR4 and PD‐1–PD‐L1 pathways prolongs survival of ovarian tumor–bearing mice by prevention of immunosuppression in the tumor microenvironment
,”
FASEB J.
33
(
5
),
6596
6608
(
2019
).
171.
D.
Kerdidani
et al, “
Lung tumor MHCII immunity depends on in situ antigen presentation by fibroblasts
,”
J. Exp. Med.
219
(
2
),
e20210815
(
2022
).
172.
D. H.
Schaffert
,
A. H.
Okholm
,
R. S.
Sørensen
et al, “
Intracellular delivery of a planar DNA origami structure by the transferrin-receptor internalization pathway
,”
Small
12
(
19
),
2634
2640
(
2016
).
173.
Y.
Sun
,
J.
Sun
,
M.
Xiao
et al, “
DNA origami-based artificial antigen-presenting cells for adoptive T cell therapy
,”
Sci. Adv.
8
(
48
),
eadd1106
(
2022
).
174.
Y.
Zhao
and
A. A.
Adjei
, “
Targeting angiogenesis in cancer therapy: Moving beyond vascular endothelial growth factor
,”
Oncologist
20
(
6
),
660
673
(
2015
).
175.
S.
Mukherjee
and
C. R.
Patra
, “
Therapeutic application of anti-angiogenic nanomaterials in cancers
,”
Nanoscale
8
(
25
),
12444
12470
(
2016
).
176.
N.
Hashemi Goradel
,
F.
Ghiyami-Hour
,
S.
Jahangiri
et al, “
Nanoparticles as new tools for inhibition of cancer angiogenesis
,”
J. Cell. Physiol.
233
(
4
),
2902
2910
(
2018
).
177.
Z.
Chen
,
K.
Zhang
,
J.
Fan
et al, “
In situ construction of ligand nano-network to integrin αvβ3 for angiogenesis inhibition
,”
Chin. Chem. Lett.
31
(
12
),
3107
3112
(
2020
).
178.
P.
Ma
,
H.
Xiao
,
C.
Yu
et al, “
Enhanced cisplatin chemotherapy by iron oxide nanocarrier-mediated generation of highly toxic reactive oxygen species
,”
Nano Lett.
17
(
2
),
928
937
(
2017
).
179.
K.
Louault
,
R. R.
Li
, and
Y. A.
DeClerck
, “
Cancer-associated fibroblasts: Understanding their heterogeneity
,”
Cancers
12
(
11
),
3108
(
2020
).
180.
R.
Lappano
,
D. C.
Rigiracciolo
,
A.
Belfiore
,
M.
Maggiolini
, and
E. M.
De Francesco
, “
Cancer associated fibroblasts: Role in breast cancer and potential as therapeutic targets
,”
Expert Opin. Ther. Targets
24
(
6
),
559
572
(
2020
).
181.
S.
Jahan
,
M. E.
Karim
, and
E. H.
Chowdhury
, “
Nanoparticles targeting receptors on breast cancer for efficient delivery of chemotherapeutics
,”
Biomedicines
9
(
2
),
114
(
2021
).
182.
A.
Bulle
and
K. H.
Lim
, “
Beyond just a tight fortress: Contribution of stroma to epithelial-mesenchymal transition in pancreatic cancer
,”
Sig. Transduct. Target. Ther.
5
(
1
),
249
(
2020
).
183.
A.
Zinger
,
L.
Koren
,
O.
Adir
et al, “
Collagenase nanoparticles enhance the penetration of drugs into pancreatic tumors
,”
ACS Nano
13
(
10
),
11008
11021
(
2019
).
184.
J.
Wang
,
Q.
Wu
,
Y.
Wang
et al, “
Collagenase-loaded pH-sensitive nanocarriers efficiently remodeled tumor stroma matrixes and improved the enrichment of nanomedicines
,”
Nanoscale
13
(
20
),
9402
9414
(
2021
).
185.
J. L.
Hu
,
W.
Wang
,
X. L.
Lan
et al, “
CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer
,”
Mol. Cancer
18
(
1
),
91
(
2019
).
186.
Z.
Su
,
C.
Lu
,
F.
Zhang
et al, “
Cancer-associated fibroblasts-secreted exosomal miR-92a-3p promotes tumor growth and stemness in hepatocellular carcinoma through activation of Wnt/β-catenin signaling pathway by suppressing AXIN1
,”
J. Cell. Physiol.
239
(
9
),
e31344
(
2024
).
187.
B. J.
Choi
,
D.
Lee
,
J. H.
Park
et al, “
Innovative therapeutic delivery of metastasis-associated in colon cancer 1-suppressing miRNA using high transmembrane 4 L6 family member 5-targeting exosomes in colorectal cancer mouse models
,”
Int. J. Mol. Sci.
25
(
17
),
9232
(
2024
).
188.
S.
Ünal
,
S.
Can Öztürk
,
E.
Bilgiç
et al, “
Therapeutic efficacy and gastrointestinal biodistribution of polycationic nanoparticles for oral camptothecin delivery in early and late-stage colorectal tumor-bearing animal model
,”
Eur. J. Pharm. Biopharm.
169
,
168
177
(
2021
).
189.
D. E.
Gerber
,
P.
Gupta
,
M. T.
Dellinger
et al, “
Stromal platelet-derived growth factor receptor α (PDGFRα) provides a therapeutic target independent of tumor cell PDGFRα expression in lung cancer xenografts
,”
Mol. Cancer Ther.
11
(
11
),
2473
2482
(
2012
).
190.
K.
Plourde
,
R. M.
Derbali
,
A.
Desrosiers
,
C.
Dubath
,
A.
Vallée-Bélisle
, and
J.
Leblond
, “
Aptamer-based liposomes improve specific drug loading and release
,”
J. Control Release
251
,
82
91
(
2017
).
191.
H.
Zhang
,
T.
Deng
,
R.
Liu
et al, “
CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer
,”
Mol. Cancer
19
(
1
),
43
(
2020
).
192.
J.
Cui
,
X.
Cai
,
R.
Qian
et al, “
Tween 80 micelles loaded with Fe3O4 nanoparticles and artemisinin for combined oxygen-independent ferroptosis therapy of cancer
,”
Pharmaceutics
16
(
5
),
639
(
2024
).
193.
C.
Hong
,
D.
Wang
,
J.
Liang
et al, “
Novel ginsenoside-based multifunctional liposomal delivery system for combination therapy of gastric cancer
,”
Theranostics
9
(
15
),
4437
4449
(
2019
).
194.
H.
Duan
,
C.
Liu
,
Y.
Hou
et al, “
Sequential delivery of quercetin and paclitaxel for the fibrotic tumor microenvironment remodeling and chemotherapy potentiation via a dual-targeting hybrid micelle-in-liposome system
,”
ACS Appl. Mater. Interfaces
14
(
8
),
10102
10116
(
2022
).
195.
J.
Keyvan Rad
,
A. R.
Mahdavian
,
S.
Khoei
, and
S.
Shirvalilou
, “
Enhanced photogeneration of reactive oxygen species and targeted photothermal therapy of C6 glioma brain cancer cells by folate-conjugated gold-photoactive polymer nanoparticles
,”
ACS Appl. Mater. Interfaces
10
(
23
),
19483
19493
(
2018
).
196.
J.
Zhang
,
T.
Zuo
,
J.
Yang
et al, “
Hierarchically releasing bio-responsive nanoparticles for complete tumor microenvironment modulation via TGF-β pathway inhibition and TAF reduction
,”
ACS Appl. Mater. Interfaces
13
(
2
),
2256
2268
(
2021
).
197.
M.
Tsioumpekou
et al, “
Specific targeting of PDGFRβ in the stroma inhibits growth and angiogenesis in tumors with high PDGF-BB expression
,”
Theranostics
10
(
3
),
1122
1135
(
2020
).
198.
M. G.
Kim
,
S. D.
Jo
,
J. Y.
Yhee
et al, “
Synergistic anti-tumor effects of bevacizumab and tumor targeted polymerized VEGF siRNA nanoparticles
,”
Biochem. Biophys. Res. Commun.
489
(
1
),
35
41
(
2017
).
199.
Y.
Song
,
C.
Tang
, and
C.
Yin
, “
Combination antitumor immunotherapy with VEGF and PIGF siRNA via systemic delivery of multi-functionalized nanoparticles to tumor-associated macrophages and breast cancer cells
,”
Biomaterials
185
,
117
132
(
2018
).
200.
R.
Mezzapelle
,
M.
Leo
,
F.
Caprioglio
et al, “
CXCR4/CXCL12 activities in the tumor microenvironment and implications for tumor immunotherapy
,”
Cancers
14
(
9
),
2314
(
2022
).
201.
W.
Zhou
,
S.
Guo
,
M.
Liu
,
M. E.
Burow
, and
G.
Wang
, “
Targeting CXCL12/CXCR4 axis in tumor immunotherapy
,”
Curr. Med. Chem.
26
(
17
),
3026
3041
(
2019
).
202.
R.
Saxena
,
Y.
Wang
, and
J. W.
Mier
, “
CXCR4 inhibition modulates the tumor microenvironment and retards the growth of B16-OVA melanoma and Renca tumors
,”
Melanoma Res.
30
(
1
),
14
25
(
2020
).
203.
Y.
Kittirat
,
M.
Suksawat
,
S.
Thongchot
et al, “
Interleukin-6-derived cancer-associated fibroblasts activate STAT3 pathway contributing to gemcitabine resistance in cholangiocarcinoma
,”
Front. Pharmacol.
13
,
897368
(
2022
).
204.
S. B.
Im
,
J. M.
Cho
,
H. B.
Kim
et al, “
FSP-1 expression in cancer cells is relevant to long-term oncological outcomes in nonmetastatic colorectal cancer
,”
Korean J. Clin. Oncol.
18
(
2
),
66
77
(
2022
).
205.
Y.
Zheng
,
J.
Zeng
,
D.
Lin
et al, “
Extracellular vesicles derived from cancer-associated fibroblast carries miR-224-5p targeting SLC4A4 to promote the proliferation, invasion and migration of colorectal cancer cells
,”
Carcinogenesis
42
(
9
),
1143
1153
(
2021
).
206.
B.
Chandra Jena
,
S.
Sarkar
,
L.
Rout
, and
M.
Mandal
, “
The transformation of cancer-associated fibroblasts: Current perspectives on the role of TGF-β in CAF mediated tumor progression and therapeutic resistance
,”
Cancer Lett.
520
,
222
232
(
2021
).
207.
Y.
Wang
,
Z.
Gao
,
X.
Du
et al, “
Co-inhibition of the TGF-β pathway and the PD-L1 checkpoint by pH-responsive clustered nanoparticles for pancreatic cancer microenvironment regulation and anti-tumor immunotherapy
,”
Biomater. Sci.
8
(
18
),
5121
5132
(
2020
).
208.
L.
Tang
,
Y.
Mei
,
Y.
Shen
et al, “
Nanoparticle-mediated targeted drug delivery to remodel tumor microenvironment for cancer therapy
,”
Int. J. Nanomed.
16
,
5811
5829
(
2021
).
209.
Y.
Zhu
,
L.
Wen
,
S.
Shao
et al, “
Inhibition of tumor-promoting stroma to enforce subsequently targeting AT1R on tumor cells by pathological inspired micelles
,”
Biomaterials
161
,
33
46
(
2018
).
210.
Q.
Yao
,
L.
Kou
,
Y.
Tu
, and
L.
Zhu
, “
MMP-responsive ‘smart’ drug delivery and tumor targeting
,”
Trends Pharmacol. Sci.
39
(
8
),
766
781
(
2018
).
211.
G.
Xiao
,
Y.
Zhao
,
X.
Wang
,
C.
Zeng
,
F.
Luo
, and
J.
Jing
, “
Photothermally sensitive gold nanocage augments the antitumor efficiency of immune checkpoint blockade in immune “cold” tumors
,”
Front. Immunol.
14
,
1279221
(
2023
).
212.
V.
Thakur
and
B.
Bedogni
, “
The membrane tethered matrix metalloproteinase MT1-MMP at the forefront of melanoma cell invasion and metastasis
,”
Pharmacol. Res.
111
,
17
22
(
2016
).
213.
P.
Wang
,
M. A.
Rahman
,
Z.
Zhao
et al, “
Visualization of the cellular uptake and trafficking of DNA origami nanostructures in cancer cells
,”
J. Am. Chem. Soc.
140
(
7
),
2478
2484
(
2018
).
214.
Z.
He
,
W.
Xiang
,
Q.
Fan
,
L.
Wang
, and
J.
Chao
, “
A DNA origami nanostructure embedded with NQO1-activated prodrugs for precision drug delivery
,”
Chem. Commun.
59
(
7
),
912
915
(
2023
).
215.
M.
Panagi
,
C.
Voutouri
,
F.
Mpekris
et al, “
TGF-β inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity
,”
Theranostics
10
(
4
),
1910
1922
(
2020
).
216.
P. M.
Galbo
,
X.
Zang
, and
D.
Zheng
, “
Molecular features of cancer-associated fibroblast subtypes and their implication on cancer pathogenesis, prognosis, and immunotherapy resistance
,”
Clin. Cancer Res.
27
(
9
),
2636
2647
(
2021
).
217.
L.
Gorchs
,
C.
Fernández Moro
,
P.
Bankhead
et al, “
Human pancreatic carcinoma-associated fibroblasts promote expression of co-inhibitory markers on CD4+ and CD8+ T-Cells
,”
Front. Immunol.
10
,
847
(
2019
).
218.
R.
Zhang
,
Z.
Zhu
,
H.
Lv
et al, “
Immune checkpoint blockade mediated by a small-molecule nanoinhibitor targeting the PD-1/PD-L1 pathway synergizes with photodynamic therapy to elicit antitumor immunity and antimetastatic effects on breast cancer
,”
Small
15
(
49
),
e1903881
(
2019
).
219.
S.
Hu
,
J.
Ma
,
C.
Su
et al, “
Engineered exosome-like nanovesicles suppress tumor growth by reprogramming tumor microenvironment and promoting tumor ferroptosis
,”
Acta Biomater.
135
,
567
581
(
2021
).
220.
Q.
Cheng
,
X.
Shi
,
M.
Han
,
G.
Smbatyan
,
H. J.
Lenz
, and
Y.
Zhang
, “
Reprogramming exosomes as nanoscale controllers of cellular immunity
,”
J. Am. Chem. Soc.
140
(
48
),
16413
16417
(
2018
).
221.
Y. N.
Tan
,
Y. P.
Li
,
J. D.
Huang
et al, “
Thermal-sensitive lipid nanoparticles potentiate anti-PD therapy through enhancing drug penetration and T lymphocytes infiltration in metastatic tumor
,”
Cancer Lett.
522
,
238
254
(
2021
).
222.
I.
Dubrovsky
,
A.
Dmitrenko
,
A.
Dmitrenko
,
N.
Serov
, and
V.
Vinogradov
, “
Unveiling the potential of AI for nanomaterial morphology prediction
,” arXiv:2406.02591 (
2024
).
223.
Y.
Sun
,
Y.
Wang
,
Y.
Liu
et al, “
Intelligent tumor microenvironment-activated multifunctional nanoplatform coupled with turn-on and always-on fluorescence probes for imaging-guided cancer treatment
,”
ACS Appl. Mater. Interfaces
13
(
45
),
53646
53658
(
2021
).
224.
J.
Cui
,
T. Y.
Huang
,
Z.
Luo
et al, “
Nanomagnetic encoding of shape-morphing micromachines
,”
Nature
575
(
7781
),
164
168
(
2019
).
225.
O. V.
Pham-Nguyen
,
J.
Shin
,
Y.
Park
et al, “
Fluorescence-shadowing nanoparticle clusters for real-time monitoring of tumor progression
,”
Biomacromolecules
23
(
8
),
3130
3141
(
2022
).
226.
N. C. W.
Ho
,
J. Y. Y.
Yap
,
Z.
Zhao
et al, “
Bioengineered hydrogels recapitulate fibroblast heterogeneity in cancer
,”
Adv. Sci.
11
(
20
),
e2307129
(
2024
).
227.
X.
Zhang
,
P.
Wang
, and
J.
Qin
, “
Editorial: Enhanced biological mechanism study, drug discovery and individualized medicine with single-cell multiomics data and integrative analysis
,”
Front Genet
14
,
1236126
(
2023
).
228.
J.
Li
,
Z.
Kong
,
Y.
Qi
et al, “
Single-cell and bulk RNA-sequence identified fibroblasts signature and CD8 + T-cell - fibroblast subtype predicting prognosis and immune therapeutic response of bladder cancer, based on machine learning: Bioinformatics multi-Omics study
,”
Int. J. Surg.
110
(
8
),
4911
4931
(
2024
).
229.
L.
Yan
,
F.
Zhao
,
S.
Li
,
Z.
Hu
, and
Y.
Zhao
, “
Low-toxic and safe nanomaterials by surface-chemical design, carbon nanotubes, fullerenes, metallofullerenes, and graphenes
,”
Nanoscale
3
(
2
),
362
382
(
2011
).
230.
D.
Zheng
,
C.
Wan
,
H.
Yang
et al, “
Her2-targeted multifunctional nano-theranostic platform mediates tumor microenvironment remodeling and immune activation for breast cancer treatment
,”
Int. J. Nanomed.
15
,
10007
10028
(
2020
).
231.
B.
Ouyang
,
W.
Poon
,
Y. N.
Zhang
et al, “
The dose threshold for nanoparticle tumour delivery
,”
Nat. Mater.
19
(
12
),
1362
1371
(
2020
).
232.
L. S. L.
Price
,
S. T.
Stern
,
A. M.
Deal
,
A. V.
Kabanov
, and
W. C.
Zamboni
, “
A reanalysis of nanoparticle tumor delivery using classical pharmacokinetic metrics
,”
Sci. Adv.
6
(
29
),
eaay9249
(
2020
).
233.
T.
Schluep
,
J.
Hwang
,
I. J.
Hildebrandt
et al, “
Pharmacokinetics and tumor dynamics of the nanoparticle IT-101 from PET imaging and tumor histological measurements
,”
Proc. Natl. Acad. Sci. U. S. A.
106
(
27
),
11394
11399
(
2009
).
234.
E.
Forte
et al, “
Radiolabeled PET/MRI nanoparticles for tumor imaging
,”
J. Clin. Med.
9
(
1
),
89
(
2020
).
235.
J.
Zhu
,
H.
Li
,
Z.
Xiong
et al, “
Polyethyleneimine-coated manganese oxide nanoparticles for targeted tumor PET/MR imaging
,”
ACS Appl. Mater. Interfaces
10
(
41
),
34954
34964
(
2018
).
236.
A. M.
Wróblewska
,
N.
Gos
,
J.
Zajda
,
L.
Ruzik
, and
M.
Matczuk
, “
Drawbacks in the efficient monitoring of gold nanoparticle-based cisplatin delivery systems formation by HPLC-ICP-MS
,”
Metallomics
15
(
1
),
mfad002
(
2023
).
237.
D. J.
Koo
,
J.
Choi
,
M.
Ahn
,
B. H.
Ahn
,
D. H.
Min
, and
S. Y.
Kim
, “
Large-scale 3D optical mapping and quantitative analysis of nanoparticle distribution in tumor vascular microenvironment
,”
Bioconjugate Chem.
31
(
7
),
1784
1794
(
2020
).
238.
S.
Healy
,
E. T.
Henderson
,
S.
Ke
et al, “
Spatial analysis of nanoparticle distribution in human breast xenografts reveals nanoparticles targeted to cancer cells localized with tumor-associated stromal cells
,”
Nanotheranostics
7
(
4
),
393
411
(
2023
).
239.
H.
Garcia Romeu
,
S.
Deville
, and
A.
Salvati
, “
Time- and space-resolved flow-cytometry of cell organelles to quantify nanoparticle uptake and intracellular trafficking by cells
,”
Small
17
(
34
),
e2100887
(
2021
).
240.
P.
Vincent
,
H.
Wang
,
M.
Nieskoski
et al, “
High-resolution ex vivo elastography to characterize tumor stromal heterogeneity in situ in pancreatic adenocarcinoma
,”
IEEE Trans. Biomed. Eng.
67
(
9
),
2490
2496
(
2020
).
241.
Z.
Zhang
,
R.
Wang
, and
L.
Chen
, “
Drug delivery system targeting cancer-associated fibroblast for improving immunotherapy
,”
Int. J. Nanomed.
20
,
483
503
(
2025
).
242.
M.
Liu
,
W.
Song
, and
L.
Huang
, “
Drug delivery systems targeting tumor-associated fibroblasts for cancer immunotherapy
,”
Cancer Lett.
448
,
31
39
(
2019
).
243.
S.
Gavas
,
S.
Quazi
, and
T. M.
Karpiński
, “
Nanoparticles for cancer therapy: Current progress and challenges
,”
Nanoscale Res. Lett.
16
(
1
),
173
(
2021
).
244.
D.
Kalyane
,
N.
Raval
,
R.
Maheshwari
,
V.
Tambe
,
K.
Kalia
, and
R. K.
Tekade
, “
Employment of enhanced permeability and retention effect (EPR): Nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer
,”
Mater. Sci. Eng. C
98
,
1252
1276
(
2019
).
245.
S.
Ryu
,
S.
Park
,
H. Y.
Lee
,
H.
Lee
,
C. W.
Cho
, and
J. S.
Baek
, “
Biodegradable nanoparticles-loaded PLGA microcapsule for the enhanced encapsulation efficiency and controlled release of hydrophilic drug
,”
Int. J. Mol. Sci.
22
(
6
),
2792
(
2021
).
246.
J.
Yoo
,
C.
Park
,
G.
Yi
,
D.
Lee
, and
H.
Koo
, “
Active targeting strategies using biological ligands for nanoparticle drug delivery systems
,”
Cancers
11
(
5
),
640
(
2019
).
247.
W.
Tao
,
X.
Zeng
,
J.
Wu
et al, “
Polydopamine-based surface modification of novel nanoparticle-aptamer bioconjugates for in vivo breast cancer targeting and enhanced therapeutic effects
,”
Theranostics
6
(
4
),
470
484
(
2016
).
248.
M.
Abri Aghdam
,
R.
Bagheri
,
J.
Mosafer
et al, “
Recent advances on thermosensitive and pH-sensitive liposomes employed in controlled release
,”
J. Control Release
315
,
1
22
(
2019
).
249.
V.
Makwana
,
J.
Karanjia
,
T.
Haselhorst
,
S.
Anoopkumar-Dukie
, and
S.
Rudrawar
, “
Liposomal doxorubicin as targeted delivery platform: Current trends in surface functionalization
,”
Int. J. Pharm.
593
,
120117
(
2021
).
250.
A.
Gabizon
,
R.
Isacson
,
E.
Libson
et al, “
Clinical studies of liposome-encapsulated doxorubicin
,”
Acta Oncol.
33
(
7
),
779
786
(
1994
).
251.
K.
Aloss
and
P.
Hamar
, “
Recent preclinical and clinical progress in liposomal doxorubicin
,”
Pharmaceutics
15
(
3
),
893
(
2023
).
252.
P. P.
Wibroe
,
D.
Ahmadvand
,
M. A.
Oghabian
,
A.
Yaghmur
, and
S. M.
Moghimi
, “
An integrated assessment of morphology, size, and complement activation of the PEGylated liposomal doxorubicin products Doxil®, Caelyx®, DOXOrubicin, and SinaDoxosome
,”
J. Control Release
221
,
1
8
(
2016
).
253.
Y.
Zhang
,
R.
Yu
,
C.
Zhao
et al, “
CAFs homologous biomimetic liposome bearing BET inhibitor and pirfenidone synergistically promoting antitumor efficacy in pancreatic ductal adenocarcinoma
,”
Adv. Sci.
11
(
1
),
e2305279
(
2024
).
254.
C. K.
Thakur
,
C.
Karthikeyan
,
C. R.
Ashby
et al, “
Ligand-conjugated multiwalled carbon nanotubes for cancer targeted drug delivery
,”
Front. Pharmacol.
15
,
1417399
(
2024
).
255.
L.
Meng
,
X.
Zhang
,
Q.
Lu
,
Z.
Fei
, and
P. J.
Dyson
, “
Single walled carbon nanotubes as drug delivery vehicles: Targeting doxorubicin to tumors
,”
Biomaterials
33
(
6
),
1689
1698
(
2012
).
256.
Z.
Liu
,
P.
Ji
,
H.
Liu
et al, “
FNIII14 peptide-enriched membrane nanocarrier to disrupt stromal barriers through reversing CAFs for augmenting drug penetration in tumors
,”
Nano Lett.
23
(
21
),
9963
9971
(
2023
).
257.
L.
Contreras
,
I.
Villarroel
,
C.
Torres
, and
R.
Rozas
, “
Doxorubicin encapsulation in carbon nanotubes having haeckelite or stone-wales defects as drug carriers: A molecular dynamics approach
,”
Molecules
26
(
6
),
1586
(
2021
).
258.
P.
Jurney
,
R.
Agarwal
,
V.
Singh
et al, “
Unique size and shape-dependent uptake behaviors of non-spherical nanoparticles by endothelial cells due to a shearing flow
,”
J. Control Release
245
,
170
176
(
2017
).
259.
K.
Nambara
,
K.
Niikura
,
H.
Mitomo
et al, “
Reverse size dependences of the cellular uptake of triangular and spherical gold nanoparticles
,”
Langmuir
32
(
47
),
12559
12567
(
2016
).
260.
Y.
Li
,
M.
Kröger
, and
W. K.
Liu
, “
Shape effect in cellular uptake of PEGylated nanoparticles: Comparison between sphere, rod, cube and disk
,”
Nanoscale
7
(
40
),
16631
16646
(
2015
).
261.
F.
Veider
,
E.
Sanchez Armengol
, and
A.
Bernkop-Schnürch
, “
Charge-reversible nanoparticles: Advanced delivery systems for therapy and diagnosis
,”
Small
20
(
3
),
e2304713
(
2024
).
262.
E.
Fröhlich
, “
The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles
,”
Int. J. Nanomed.
7
,
5577
5591
(
2012
).
263.
J.
Alcaraz
,
J. L.
Carrasco
,
L.
Millares
et al, “
Stromal markers of activated tumor associated fibroblasts predict poor survival and are associated with necrosis in non-small cell lung cancer
,”
Lung Cancer
135
,
151
160
(
2019
).
264.
A.
Fullár
,
J.
Dudás
,
L.
Oláh
et al, “
Remodeling of extracellular matrix by normal and tumor-associated fibroblasts promotes cervical cancer progression
,”
BMC Cancer
15
,
256
(
2015
).
265.
K. H.
Sun
,
Y.
Chang
,
N. I.
Reed
, and
D.
Sheppard
, “
α-Smooth muscle actin is an inconsistent marker of fibroblasts responsible for force-dependent TGFβ activation or collagen production across multiple models of organ fibrosis
,”
Am. J. Physiol.
310
(
9
),
L824
836
(
2016
).
266.
J.
Bolley
,
E.
Guenin
,
N.
Lievre
et al, “
Carbodiimide versus click chemistry for nanoparticle surface functionalization: A comparative study for the elaboration of multimodal superparamagnetic nanoparticles targeting αvβ3 integrins
,”
Langmuir
29
(
47
),
14639
14647
(
2013
).
267.
Q.
Chen
,
M.
Gao
,
Z.
Li
et al, “
Biodegradable nanoparticles decorated with different carbohydrates for efficient macrophage-targeted gene therapy
,”
J. Control Release
323
,
179
190
(
2020
).
268.
L.
Hou
,
D.
Chen
,
L.
Hao
et al, “
Transformable nanoparticles triggered by cancer-associated fibroblasts for improving drug permeability and efficacy in desmoplastic tumors
,”
Nanoscale
11
(
42
),
20030
20044
(
2019
).
269.
C.
Guo
,
W.
Zhang
,
Q.
Zhang
et al, “
Novel dual CAFs and tumour cell targeting pH and ROS dual sensitive micelles for targeting delivery of paclitaxel to liver cancer
,”
Artif. Cells Nanomed. Biotechnol.
51
(
1
),
170
179
(
2023
).
270.
H.
Javid
,
M. A.
Oryani
,
N.
Rezagholinejad
,
A.
Esparham
,
M.
Tajaldini
, and
M.
Karimi-Shahri
, “
RGD peptide in cancer targeting: Benefits, challenges, solutions, and possible integrin-RGD interactions
,”
Cancer Med.
13
(
2
),
e6800
(
2024
).
271.
S.
Fu
,
X.
Xu
,
Y.
Ma
,
S.
Zhang
, and
S.
Zhang
, “
RGD peptide-based non-viral gene delivery vectors targeting integrin αvβ3 for cancer therapy
,”
J. Drug Target
27
(
1
),
1
11
(
2019
).
272.
M.
Li
,
Y.
Wang
,
M.
Li
,
X.
Wu
,
S.
Setrerrahmane
, and
H.
Xu
, “
Integrins as attractive targets for cancer therapeutics
,”
Acta Pharm. Sin. B
11
(
9
),
2726
2737
(
2021
).
273.
S.
Affo
,
A.
Nair
,
F.
Brundu
et al, “
Promotion of cholangiocarcinoma growth by diverse cancer-associated fibroblast subpopulations
,”
Cancer Cell
39
(
6
),
883
(
2021
).
274.
Y.
Zhu
,
J.
Feijen
, and
Z.
Zhong
, “
Dual-targeted nanomedicines for enhanced tumor treatment
,”
Nano Today
18
,
65
85
(
2018
).
275.
C.
Martins
and
B.
Sarmento
, “
Multi-ligand functionalized blood-to-tumor sequential targeting strategies in the field of glioblastoma nanomedicine
,”
WIREs Nanomed. Nanobiotechnol.
15
(
5
),
e1893
(
2023
).
276.
H.
Yan
,
Y.
You
,
X.
Li
et al, “
Preparation of RGD peptide/folate acid double-targeted mesoporous silica nanoparticles and its application in human breast cancer MCF-7 cells
,”
Front. Pharmacol.
11
,
898
(
2020
).
277.
Z.
Gong
et al, “
Tumor acidic microenvironment-induced drug release of RGD peptide nanoparticles for cellular uptake and cancer therapy
,”
Colloids Surf. B
202
,
111673
(
2021
).
278.
B.
Chen
,
W.
Dai
,
B.
He
et al, “
Current multistage drug delivery systems based on the tumor microenvironment
,”
Theranostics
7
(
3
),
538
558
(
2017
).
279.
G.
Wang
,
M.
Zhang
,
W.
Lai
et al, “
Tumor microenvironment responsive RNA drug delivery systems: Intelligent platforms for sophisticated release
,”
Mol. Pharm.
21
(
9
),
4217
4237
(
2024
).
280.
F.
Danhier
,
O.
Feron
, and
V.
Préat
, “
To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery
,”
J. Control Release
148
(
2
),
135
146
(
2010
).
281.
Q.
Zhang
,
N.
Liu
,
J.
Wang
et al, “
The recent advance of cell-penetrating and tumor-targeting peptides as drug delivery systems based on tumor microenvironment
,”
Mol. Pharm.
20
(
2
),
789
809
(
2023
).
282.
H. S.
El-Sawy
,
A. M.
Al-Abd
,
T. A.
Ahmed
,
K. M.
El-Say
, and
V. P.
Torchilin
, “
Stimuli-responsive nano-architecture drug-delivery systems to solid tumor micromilieu: Past, present, and future perspectives
,”
ACS Nano
12
(
11
),
10636
10664
(
2018
).
283.
J.
Zheng
and
P.
Gao
, “
Toward normalization of the tumor microenvironment for cancer therapy
,”
Integr. Cancer Ther.
18
,
1534735419862352
(
2019
).
284.
L.
Feng
,
Z.
Dong
,
D.
Tao
,
Y.
Zhang
, and
Z.
Liu
, “
The acidic tumor microenvironment: A target for smart cancer nano-theranostics
,”
Natl. Sci. Rev.
5
,
269
286
(
2018
).
285.
E.
Boedtkjer
and
S. F.
Pedersen
, “
The acidic tumor microenvironment as a driver of cancer
,”
Annu. Rev. Physiol.
82
,
103
126
(
2020
).
286.
Z.
Li
,
L.
Qiu
,
Q.
Chen
et al, “
pH-sensitive nanoparticles of poly(L-histidine)-poly(lactide-co-glycolide)-tocopheryl polyethylene glycol succinate for anti-tumor drug delivery
,”
Acta Biomater.
11
,
137
150
(
2015
).
287.
F.
Hu
,
R.
Zhang
,
W.
Guo
et al, “
PEGylated-PLGA nanoparticles coated with pH-responsive tannic acid-Fe(III) complexes for reduced premature doxorubicin release and enhanced targeting in breast cancer
,”
Mol. Pharm.
18
(
6
),
2161
2173
(
2021
).
288.
A. D.
Tripathi
,
Y.
Labh
,
S.
Katiyar
,
A. K.
Singh
,
V. K.
Chaturvedi
, and
A.
Mishra
, “
Folate-mediated targeting and controlled release: PLGA-encapsulated mesoporous silica nanoparticles delivering capecitabine to pancreatic tumor
,”
ACS Appl. Bio Mater.
7
(
12
),
7838
7851
(
2024
).
289.
A.
Narmani
,
R.
Jahedi
,
E.
Bakhshian-Dehkordi
et al, “
Biomedical applications of PLGA nanoparticles in nanomedicine: Advances in drug delivery systems and cancer therapy
,”
Expert Opin. Drug Delivery
20
(
7
),
937
954
(
2023
).
290.
M.
Karimi
,
M.
Eslami
,
P.
Sahandi-Zangabad
et al, “
pH-Sensitive stimulus-responsive nanocarriers for targeted delivery of therapeutic agents
,”
Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol.
8
(
5
),
696
716
(
2016
).
291.
Z.
Chen
,
X.
Wang
,
N.
Zhao
,
H.
Chen
, and
G.
Guo
, “
Advancements in pH-responsive nanocarriers: Enhancing drug delivery for tumor therapy
,”
Expert Opin. Drug Delivery
20
(
11
),
1623
1642
(
2023
).
292.
B.
Grünwald
,
J.
Vandooren
,
E.
Locatelli
et al, “
Matrix metalloproteinase-9 (MMP-9) as an activator of nanosystems for targeted drug delivery in pancreatic cancer
,”
J. Control Release
239
,
39
48
(
2016
).
293.
K.
Vaghasiya
,
E.
Ray
,
A.
Sharma
,
O. P.
Katare
, and
R. K.
Verma
, “
Matrix metalloproteinase-responsive mesoporous silica nanoparticles cloaked with cleavable protein for “self-actuating” on-demand controlled drug delivery for cancer therapy
,”
ACS Appl. Bio Mater.
3
(
8
),
4987
4999
(
2020
).
294.
L.
Kou
,
R.
Sun
,
X.
Jiang
et al, “
Tumor microenvironment-responsive, multistaged liposome induces apoptosis and ferroptosis by amplifying oxidative stress for enhanced cancer therapy
,”
ACS Appl. Mater. Interfaces
12
(
27
),
30031
30043
(
2020
).
295.
E.
Timosenko
,
A. V.
Hadjinicolaou
, and
V.
Cerundolo
, “
Modulation of cancer-specific immune responses by amino acid degrading enzymes
,”
Immunotherapy
9
(
1
),
83
97
(
2017
).
296.
P.
Wang
,
W.
Yang
,
S.
Shen
et al, “
Differential diagnosis and precision therapy of two typical malignant cutaneous tumors leveraging their tumor microenvironment: A photomedicine strategy
,”
ACS Nano
13
(
10
),
11168
11180
(
2019
).
297.
M.
Chen
,
D.
Liu
,
F.
Liu
,
Y.
Wu
,
X.
Peng
, and
F.
Song
, “
Recent advances of redox-responsive nanoplatforms for tumor theranostics
,”
J. Control Release
332
,
269
284
(
2021
).
298.
D.
Chen
,
G.
Zhang
,
R.
Li
et al, “
Biodegradable, hydrogen peroxide, and glutathione dual responsive nanoparticles for potential programmable paclitaxel release
,”
J. Am. Chem. Soc.
140
(
24
),
7373
7376
(
2018
).
299.
H.
Deng
,
X.
Zhao
,
J.
Liu
et al, “
Reactive oxygen species (ROS) responsive PEG-PCL nanoparticles with pH-controlled negative-to-positive charge reversal for intracellular delivery of doxorubicin
,”
J. Mater. Chem. B
3
(
48
),
9397
9408
(
2015
).
300.
I.
Azcune
and
I.
Odriozola
, “
Aromatic disulfide crosslinks in polymer systems: Self-healing, reprocessability, recyclability and more
,”
Eur. Polym. J.
84
,
147
160
(
2016
).
301.
M.
Novoselova
,
V. S.
Chernyshev
,
A.
Schulga
et al, “
Effect of surface modification of multifunctional nanocomposite drug delivery carriers with DARPin on their biodistribution in vitro and in vivo
,”
ACS Appl. Bio Mater.
5
(
6
),
2976
2989
(
2022
).
302.
W.
Liu
,
Y.
Pan
,
Y.
Zhong
et al, “
A multifunctional aminated UiO-67 metal-organic framework for enhancing antitumor cytotoxicity through bimodal drug delivery
,”
Chem. Eng. J.
412
,
127899
(
2021
).
303.
Q.
Yang
,
L.
Wu
,
L.
Li
,
Z.
Zhou
, and
Y.
Huang
, “
Subcellular co-delivery of two different site-oriented payloads for tumor therapy
,”
Nanoscale
9
(
4
),
1547
1558
(
2017
).
304.
N.
Li
,
L.
Zhao
,
L.
Qi
,
Z.
Li
, and
Y.
Luan
, “
Polymer assembly: Promising carriers as co-delivery systems for cancer therapy
,”
Prog. Polym. Sci.
58
,
1
26
(
2016
).
305.
D. M.
Silva
,
R.
Liu
,
A. F.
Gonçalves
et al, “
Design of polymeric core-shell carriers for combination therapies
,”
J. Colloid Interface Sci.
587
,
499
509
(
2021
).
306.
Y.
Yu
,
R.
Feng
,
S.
Yu
et al, “
Nanostructured lipid carrier-based pH and temperature dual-responsive hydrogel composed of carboxymethyl chitosan and poloxamer for drug delivery
,”
Int. J. Biol. Macromol.
114
,
462
469
(
2018
).
307.
W. T.
Al-Jamal
and
K.
Kostarelos
, “
Liposomes: From a clinically established drug delivery system to a nanoparticle platform for theranostic nanomedicine
,”
Acc. Chem. Res.
44
(
10
),
1094
1104
(
2011
).
308.
H.
Wang
,
L.
Gao
,
T.
Fan
et al, “
Strategic design of intelligent-responsive nanogel carriers for cancer therapy
,”
ACS Appl. Mater. Interfaces
13
(
46
),
54621
54647
(
2021
).
309.
R.
Bayat Mokhtari
,
T. S.
Homayouni
,
N.
Baluch
et al, “
Combination therapy in combating cancer
,”
Oncotarget
8
(
23
),
38022
38043
(
2017
).
310.
X.
Qian
,
W.
Hu
, and
J.
Yan
, “
Nano-chemotherapy synergize with immune checkpoint inhibitor- A better option?
Front. Immunol.
13
,
963533
(
2022
).
311.
Y.
Yan
,
A. B.
Kumar
,
H.
Finnes
et al, “
Combining immune checkpoint inhibitors with conventional cancer therapy
,”
Front. Immunol.
9
,
1739
(
2018
).
312.
L.
Sitia
,
A.
Bonizzi
,
S.
Mazzucchelli
et al, “
Selective targeting of cancer-associated fibroblasts by engineered H-ferritin nanocages loaded with navitoclax
,”
Cells
10
(
2
),
328
(
2021
).
313.
C.
Hopkins
,
K.
Javius-Jones
,
Y.
Wang
,
H.
Hong
,
Q.
Hu
, and
S.
Hong
, “
Combinations of chemo-, immuno-, and gene therapies using nanocarriers as a multifunctional drug platform
,”
Expert Opin. Drug Delivery
19
(
10
),
1337
1349
(
2022
).
314.
M. H.
Akhter
,
M.
Rizwanullah
,
J.
Ahmad
,
M. J.
Ahsan
,
M. A.
Mujtaba
, and
S.
Amin
, “
Nanocarriers in advanced drug targeting: Setting novel paradigm in cancer therapeutics
,”
Artif. Cells Nanomed. Biotechnol.
46
(
5
),
873
884
(
2018
).
315.
B. M.
Dicheva
and
G. A.
Koning
, “
Targeted thermosensitive liposomes: An attractive novel approach for increased drug delivery to solid tumors
,”
Expert Opin. Drug Delivery
11
(
1
),
83
100
(
2014
).
316.
W.
Yan
,
S. S.
Leung
, and
K. K.
To
, “
Updates on the use of liposomes for active tumor targeting in cancer therapy
,”
Nanomedicine
15
(
3
),
303
318
(
2020
).
317.
S. A.
Moosavian
and
A.
Sahebkar
, “
Aptamer-functionalized liposomes for targeted cancer therapy
,”
Cancer Lett.
448
,
144
154
(
2019
).
318.
M.
Gupta
,
G.
Chashoo
,
P. R.
Sharma
et al, “
Dual targeted polymeric nanoparticles based on tumor endothelium and tumor cells for enhanced antitumor drug delivery
,”
Mol. Pharm.
11
(
3
),
697
715
(
2014
).
319.
F.
Ordikhani
,
M.
Uehara
,
V.
Kasinath
et al, “
Targeting antigen-presenting cells by anti-PD-1 nanoparticles augments antitumor immunity
,”
JCI Insight
3
(
20
),
e122700
(
2018
).
320.
S.
Song
,
Y.
Hao
,
X.
Yang
,
P.
Patra
, and
J.
Chen
, “
Using gold nanoparticles as delivery vehicles for targeted delivery of chemotherapy drug fludarabine phosphate to treat hematological cancers
,”
J. Nanosci. Nanotechnol.
16
(
3
),
2582
2586
(
2016
).
321.
M.
Yafout
,
A.
Ousaid
,
Y.
Khayati
, and
I. S.
El Otmani
, “
Gold nanoparticles as a drug delivery system for standard chemotherapeutics: A new lead for targeted pharmacological cancer treatments
,”
Sci. Afr.
11
,
e00685
(
2021
).
322.
Y.
Zhang
,
C.
Zhang
,
J.
Chen
et al, “
Trackable mitochondria-targeting nanomicellar loaded with doxorubicin for overcoming drug resistance
,”
ACS Appl. Mater. Interfaces
9
(
30
),
25152
25163
(
2017
).
323.
C.
Liu
,
M.
Li
,
P.
Li
et al, “
Platinum-containing supramolecular drug self-delivery nanomicelles for efficient synergistic combination chemotherapy
,”
Biomacromolecules
22
(
6
),
2382
2392
(
2021
).
324.
E.
Sánchez-López
,
M.
Guerra
,
J.
Dias-Ferreira
et al, “
Current applications of nanoemulsions in cancer therapeutics
,”
Nanomaterials
9
(
6
),
821
(
2019
).
325.
B.
Gorain
,
H.
Choudhury
,
A. B.
Nair
,
S. K.
Dubey
, and
P.
Kesharwani
, “
Theranostic application of nanoemulsions in chemotherapy
,”
Drug Discovery Today
25
(
7
),
1174
1188
(
2020
).
326.
J.
Pardo
,
Z.
Peng
, and
R. M.
Leblanc
, “
Cancer targeting and drug delivery using carbon-based quantum dots and nanotubes
,”
Molecules
23
(
2
),
378
(
2018
).
327.
F.
Wu
,
J.
Yang
,
J.
Liu
et al, “
Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer
,”
Sig. Transduct. Target. Ther.
6
(
1
),
1
35
(
2021
).
328.
D. T.
Peters
,
H.
Waller
,
M. A.
Birch
, and
J. H.
Lakey
, “
Engineered mosaic protein polymers; a simple route to multifunctional biomaterials
,”
J. Biol. Eng.
13
,
54
(
2019
).
329.
H.
Shigemitsu
and
I.
Hamachi
, “
Design strategies of stimuli-responsive supramolecular hydrogels relying on structural analyses and cell-mimicking approaches
,”
Acc. Chem. Res.
50
(
4
),
740
750
(
2017
).
330.
A.
Zielińska
,
P.
Eder
,
L.
Rannier
et al, “
Hydrogels for modified-release drug delivery systems
,”
Curr. Pharm. Des.
28
(
8
),
609
618
(
2022
).
331.
J. A.
Serna
,
L.
Rueda-Gensini
,
D. N.
Céspedes-Valenzuela
,
J.
Cifuentes
,
J. C.
Cruz
, and
C.
Muñoz-Camargo
, “
Recent advances on stimuli-responsive hydrogels based on tissue-derived ECMs and their components: Towards improving functionality for tissue engineering and controlled drug delivery
,”
Polymers
13
(
19
),
3263
(
2021
).
332.
S. Y.
Park
,
J. H.
Kang
,
H. S.
Kim
,
J. Y.
Hwang
, and
U. S.
Shin
, “
Electrical and thermal stimulus-responsive nanocarbon-based 3D hydrogel sponge for switchable drug delivery
,”
Nanoscale
14
(
6
),
2367
2382
(
2022
).
333.
H.
Chen
,
Y.
Cheng
,
X.
Wang
et al, “
3D printed in vitro tumor tissue model of colorectal cancer
,”
Theranostics
10
(
26
),
12127
12143
(
2020
).
334.
H.
Cao
,
H. S.
Cheng
,
J. K.
Wang
,
N. S.
Tan
, and
C. Y.
Tay
, “
A 3D physio-mimetic interpenetrating network-based platform to decode the pro and anti-tumorigenic properties of cancer-associated fibroblasts
,”
Acta Biomater.
132
,
448
460
(
2021
).
335.
P.
Shukla
,
S.
Yeleswarapu
,
M. A.
Heinrich
,
J.
Prakash
, and
F.
Pati
, “
Mimicking tumor microenvironment by 3D bioprinting: 3D cancer modeling
,”
Biofabrication
14
(
3
),
032002
(
2022
).
336.
J.
Wang
,
W.
Li
,
L.
Zhang
et al, “
Chemically edited exosomes with dual ligand purified by microfluidic device for active targeted drug delivery to tumor cells
,”
ACS Appl. Mater. Interfaces
9
(
33
),
27441
27452
(
2017
).
337.
L.
Mathur
,
M.
Ballinger
,
R.
Utharala
, and
C. A.
Merten
, “
Microfluidics as an enabling technology for personalized cancer therapy
,”
Small
16
(
9
),
e1904321
(
2020
).
338.
S. Y.
Jeong
,
J. H.
Lee
,
Y.
Shin
,
S.
Chung
, and
H. J.
Kuh
, “
Co-culture of tumor spheroids and fibroblasts in a collagen matrix-incorporated microfluidic chip mimics reciprocal activation in solid tumor microenvironment
,”
PLoS One
11
(
7
),
e0159013
(
2016
).
339.
M.
Martinez-Lage
,
R.
Torres-Ruiz
,
P.
Puig-Serra
et al, “
In vivo CRISPR/Cas9 targeting of fusion oncogenes for selective elimination of cancer cells
,”
Nat. Commun.
11
(
1
),
5060
(
2020
).
340.
L.
Gelsomino
,
A.
Caruso
,
E.
Tasan
et al, “
Evidence that CRISPR-Cas9 Y537S-mutant expressing breast cancer cells activate Yes-associated protein 1 to driving the conversion of normal fibroblasts into cancer-associated fibroblasts
,”
Cell Commun. Signal.
22
(
1
),
545
(
2024
).
341.
K.
Scholten
and
E.
Meng
, “
A review of implantable biosensors for closed-loop glucose control and other drug delivery applications
,”
Int. J. Pharm.
544
(
2
),
319
334
(
2018
).
342.
H. K. A.
Tsai
et al, “
Integrating biosensors and drug delivery: A step closer toward scalable responsive drug-delivery systems
,”
Adv. Mater.
21
(
6
),
656
660
(
2009
).
343.
R.
Kanzaki
and
K.
Pietras
, “
Heterogeneity of cancer-associated fibroblasts: Opportunities for precision medicine
,”
Cancer Sci.
111
(
8
),
2708
2717
(
2020
).
344.
E. V.
Nguyen
,
B. A.
Pereira
,
M. G.
Lawrence
et al, “
Proteomic profiling of human prostate cancer-associated Fibroblasts (CAF) Reveals LOXL2-dependent regulation of the tumor microenvironment
,”
Mol. Cell Proteomics
18
(
7
),
1410
1427
(
2019
).
345.
H.
Li
,
E. T.
Courtois
,
D.
Sengupta
et al, “
Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors
,”
Nat. Genet.
49
(
5
),
708
718
(
2017
).