Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 272
Filter
1.
Cancer J ; 30(3): 210-217, 2024.
Article in English | MEDLINE | ID: mdl-38753756

ABSTRACT

ABSTRACT: Fibroblast activation protein inhibitor positron emission tomography (PET) has gained interest for its ability to demonstrate uptake in a diverse range of tumors. Its molecular target, fibroblast activation protein, is expressed in cancer-associated fibroblasts, a major cell type in tumor microenvironment that surrounds various types of cancers. Although existing literature on FAPI PET is largely from single-center studies and case reports, initial findings show promise for some cancer types demonstrating improved imaging when compared with the widely used 18F-fludeoxyglucose PET for oncologic imaging. As we expand our knowledge of the utility of FAPI PET, accurate understanding of noncancerous uptake seen on FAPI PET is crucial for accurate evaluation. In this review, we summarize potential diagnostic and therapeutic applications of radiolabeled FAP inhibitors in oncological and nononcological disease processes.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/diagnosis , Neoplasms/metabolism , Positron-Emission Tomography/methods , Endopeptidases , Gelatinases/antagonists & inhibitors , Gelatinases/metabolism , Tumor Microenvironment/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Radiopharmaceuticals , Serine Endopeptidases/metabolism , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects
2.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791392

ABSTRACT

Malignant pleural mesothelioma (MPM) remains an incurable disease. This is partly due to the lack of experimental models that fully recapitulate the complexity and heterogeneity of MPM, a major challenge for therapeutic management of the disease. In addition, the contribution of the MPM microenvironment is relevant for the adaptive response to therapy. We established mesothelioma patient-derived organoid (mPDO) cultures from MPM pleural effusions and tested their response to pemetrexed and cisplatin. We aimed to evaluate the contribution of mesothelioma-associated fibroblasts (MAFs) to the response to pemetrexed and cisplatin (P+C). Organoid cultures were obtained from eight MPM patients using specific growth media and conditions to expand pleural effusion-derived cells. Flow cytometry was used to verify the similarity of the organoid cultures to the original samples. MAFs were isolated and co-cultured with mPDOs, and the addition of MAFs reduced the sensitivity of mPDOs to P+C. Organoid formation and expression of cancer stem cell markers such as ABCG2, NANOG, and CD44 were altered by conditioned media from treated MAFs. We identified IL-6 as the major contributor to the attenuated response to chemotherapy. IL-6 secretion by MAFs is correlated with increased resistance of mPDOs to pemetrexed and cisplatin.


Subject(s)
Cancer-Associated Fibroblasts , Cisplatin , Interleukin-6 , Mesothelioma, Malignant , Mesothelioma , Organoids , Pemetrexed , Humans , Organoids/metabolism , Organoids/drug effects , Organoids/pathology , Interleukin-6/metabolism , Cisplatin/pharmacology , Pemetrexed/pharmacology , Mesothelioma, Malignant/drug therapy , Mesothelioma, Malignant/pathology , Mesothelioma, Malignant/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Mesothelioma/pathology , Mesothelioma/drug therapy , Mesothelioma/metabolism , Tumor Microenvironment/drug effects , Male , Female , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Middle Aged , Aged , Antineoplastic Agents/pharmacology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
3.
J Proteome Res ; 23(6): 2160-2168, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38767394

ABSTRACT

Resistance is a major problem with effective cancer treatment and the stroma forms a significant portion of the tumor mass but traditional drug screens involve cancer cells alone. Cancer-associated fibroblasts (CAFs) are a major tumor stroma component and its secreted proteins may influence the function of cancer cells. The majority of secretome studies compare different cancer or CAF cell lines exclusively. Here, we present the direct characterization of the secreted protein profiles between CAFs and KRAS mutant-cancer cell lines from colorectal, lung, and pancreatic tissues using multiplexed mass spectrometry. 2573 secreted proteins were annotated, and differential analysis highlighted understudied CAF-enriched secreted proteins, including Wnt family member 5B (WNT5B), in addition to established CAF markers, such as collagens. The functional role of CAF secreted proteins was explored by assessing its effect on the response to 97 anticancer drugs since stromal cells may cause a differing cancer drug response, which may be missed on routine drug screening using cancer cells alone. CAF secreted proteins caused specific effects on each of the cancer cell lines, which highlights the complexity and challenges in cancer treatment and so the importance to consider stromal elements.


Subject(s)
Cancer-Associated Fibroblasts , Secretome , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Secretome/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Mass Spectrometry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proteomics/methods , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics
4.
Mikrochim Acta ; 191(5): 279, 2024 04 22.
Article in English | MEDLINE | ID: mdl-38647729

ABSTRACT

The therapeutic effect of gefitinib on colorectal cancer (CRC) is unclear, but it has been reported that stromal cells in the tumor microenvironment may have an impact on drug sensitivity. Herein, we established a microfluidic co-culture system and explored the sensitivity of CRC cells co-cultured with cancer-associated fibroblasts (CAFs) to gefitinib. The system consisted of a multichannel chip and a Petri dish. The chambers in the chip and dish were designed to continuously supply nutrients for long-term cell survival and create chemokine gradients for driving cell invasion without any external equipment. Using this system, the proliferation and invasiveness of cells were simultaneously evaluated by quantifying the area of cells and the migration distance of cells. In addition, the system combined with live cell workstation could evaluate the dynamic drug response of co-cultured cells and track individual cell trajectories in real-time. When CRC cells were co-cultured with CAFs, CAFs promoted CRC cell proliferation and invasion and reduced the sensitivity of cells to gefitinib through the exosomes secreted by CAFs. Furthermore, the cells that migrated out of the chip were collected, and EMT-related markers were determined by immunofluorescent and western blot assays. The results demonstrated that CAFs affected the response of CRC cells to gefitinib by inducing EMT, providing new ideas for further research on the resistance mechanism of gefitinib. This suggests that targeting CAFs or exosomes might be a new approach to enhance CRC sensitivity to gefitinib, and our system could be a novel platform for investigating the crosstalk between tumor cells and CAFs and understanding multiple biological changes of the tumor cells in the tumor microenvironment.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Coculture Techniques , Colorectal Neoplasms , Gefitinib , Gefitinib/pharmacology , Humans , Coculture Techniques/instrumentation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Cell Line, Tumor , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Exosomes/metabolism , Exosomes/chemistry , Exosomes/drug effects , Tumor Microenvironment/drug effects , Drug Resistance, Neoplasm/drug effects
5.
Cancer Lett ; 592: 216905, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38677641

ABSTRACT

Craniopharyngiomas (CPs), particularly Adamantinomatous Craniopharyngiomas (ACPs), often exhibit a heightened risk of postoperative recurrence and severe complications of the endocrine and hypothalamic function. The primary objective of this study is to investigate potential novel targeted therapies within the microenvironment of ACP tumors. Cancer-Associated Fibroblasts (CAFs) were identified in the craniopharyngioma microenvironment, notably in regions characterized by cholesterol clefts, wet keratin, ghost cells, and fibrous stroma in ACPs. CAFs, alongside ghost cells, basaloid-like epithelium cells and calcifications, were found to secrete PROS1 and GAS6, which can activate AXL receptors on the surface of tumor epithelium cells, promoting immune suppression and tumor progression in ACPs. Additionally, the AXL inhibitor Bemcentinib effectively inhibited the proliferation organoids and enhanced the immunotherapeutic efficacy of Atezolizumab. Furthermore, neural crest-like cells were observed in the glial reactive tissue surrounding finger-like protrusions. Overall, our results revealed that the AXL might be a potentially effective therapeutic target for ACPs.


Subject(s)
Axl Receptor Tyrosine Kinase , Craniopharyngioma , Pituitary Neoplasms , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Tumor Microenvironment , Humans , Craniopharyngioma/genetics , Craniopharyngioma/drug therapy , Craniopharyngioma/pathology , Craniopharyngioma/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/metabolism , Tumor Microenvironment/drug effects , Female , Male , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Gene Expression Profiling/methods , RNA-Seq , Benzocycloheptenes/pharmacology , Animals , Gene Expression Regulation, Neoplastic/drug effects , Mice , Cell Proliferation/drug effects , Adult , Molecular Targeted Therapy , Middle Aged , Triazoles
6.
Int J Pharm ; 656: 124078, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38569978

ABSTRACT

The role of tumor stroma in solid tumors has been widely recognized in cancer progression, metastasis and chemoresistance. Cancer-associated fibroblasts (CAFs) play a crucial role in matrix remodeling and promoting cancer cell stemness and resistance via reciprocal crosstalk. Residual tumor tissue after surgical removal as well as unresectable tumors face therapeutic challenges to achieve curable outcome. In this study, we propose to develop a dual delivery approach by combining p21-activated kinase 1 (PAK1) inhibitor (FRAX597) to inhibit tumor stroma and chemotherapeutic agent paclitaxel (PTX) to kill cancer cells using electrospun nanofibers. First, the role of the PAK1 pathway was established in CAF differentiation, migration and contraction using relevant in vitro models. Second, polycaprolactone polymer-based nanofibers were fabricated using a uniaxial electrospinning technique to incorporate FRAX597 and/or PTX, which showed a uniform texture and a prolonged release of both drugs for 16 days. To test nanofibers, stroma-rich 3D heterospheroid models were set up which showed high resistance to PTX nanofibers compared to stroma-free homospheroids. Interestingly, nanofibers containing PTX and FRAX597 showed strong anti-tumor effects on heterospheroids by reducing the growth and viability by > 90 % compared to either of single drug-loaded nanofibers. These effects were reflected by reduced intra-spheroidal expression levels of collagen 1 and α-smooth muscle actin (α-SMA). Overall, this study provides a new therapeutic strategy to inhibit the tumor stroma using PAK1 inhibitor and thereby enhance the efficacy of chemotherapy using nanofibers as a local delivery system for unresectable or residual tumor. Use of 3D models to evaluate nanofibers highlights these models as advanced in vitro tools to study the effect of controlled release local drug delivery systems before animal studies.


Subject(s)
Nanofibers , Paclitaxel , p21-Activated Kinases , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Nanofibers/administration & dosage , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Humans , Cell Line, Tumor , Spheroids, Cellular/drug effects , Polyesters/chemistry , Polyesters/administration & dosage , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Drug Delivery Systems/methods , Cell Movement/drug effects , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Drug Liberation , Cell Differentiation/drug effects
7.
Cell Rep Med ; 5(5): 101532, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38670097

ABSTRACT

Ovarian clear cell carcinoma (OCCC) is a gynecological cancer with a dismal prognosis; however, the mechanism underlying OCCC chemoresistance is not well understood. To explore the intracellular networks associated with the chemoresistance, we analyze surgical specimens by performing integrative analyses that combine single-cell analyses and spatial transcriptomics. We find that a chemoresistant OCCC subpopulation with elevated HIF activity localizes mainly in areas populated by cancer-associated fibroblasts (CAFs) with a myofibroblastic phenotype, which is corroborated by quantitative immunostaining. CAF-enhanced chemoresistance and HIF-1α induction are recapitulated in co-culture assays, which show that cancer-derived platelet-derived growth factor (PDGF) contributes to the chemoresistance and HIF-1α induction via PDGF receptor signaling in CAFs. Ripretinib is identified as an effective receptor tyrosine kinase inhibitor against CAF survival. In the co-culture system and xenograft tumors, ripretinib prevents CAF survival and suppresses OCCC proliferation in the presence of carboplatin, indicating that combination of conventional chemotherapy and CAF-targeted agents is effective against OCCC.


Subject(s)
Cancer-Associated Fibroblasts , Hypoxia-Inducible Factor 1, alpha Subunit , Ovarian Neoplasms , Platelet-Derived Growth Factor , Signal Transduction , Female , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Animals , Mice , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Disease Progression , Coculture Techniques , Cell Proliferation/drug effects , Mice, Nude , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/genetics , Feedback, Physiological/drug effects , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 30(11): 2582-2597, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38578281

ABSTRACT

PURPOSE: To explore the cellular cross-talk of tumor-resident mast cells (MC) in controlling the activity of cancer-associated fibroblasts (CAF) to overcome tumor microenvironment (TME) abnormalities, enhancing the efficacy of immune-checkpoint inhibitors in sarcoma. EXPERIMENTAL DESIGN: We used a coculture system followed by further validation in mouse models of fibrosarcoma and osteosarcoma with or without administration of the MC stabilizer and antihistamine ketotifen. To evaluate the contribution of ketotifen in sensitizing tumors to therapy, we performed combination studies with doxorubicin chemotherapy and anti-PD-L1 (B7-H1, clone 10F.9G2) treatment. We investigated the ability of ketotifen to modulate the TME in human sarcomas in the context of a repurposed phase II clinical trial. RESULTS: Inhibition of MC activation with ketotifen successfully suppressed CAF proliferation and stiffness of the extracellular matrix accompanied by an increase in vessel perfusion in fibrosarcoma and osteosarcoma as indicated by ultrasound shear wave elastography imaging. The improved tissue oxygenation increased the efficacy of chemoimmunotherapy, supported by enhanced T-cell infiltration and acquisition of tumor antigen-specific memory. Importantly, the effect of ketotifen in reducing tumor stiffness was further validated in sarcoma patients, highlighting its translational potential. CONCLUSIONS: Our study suggests the targeting of MCs with clinically administered drugs, such as antihistamines, as a promising approach to overcome resistance to immunotherapy in sarcomas.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Mast Cells , Tumor Microenvironment , Humans , Mice , Animals , Mast Cells/drug effects , Mast Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Sarcoma/drug therapy , Sarcoma/pathology , Sarcoma/immunology , Ketotifen/pharmacology , Ketotifen/therapeutic use , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Female , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/immunology
9.
Cancer Lett ; 591: 216859, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38615928

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a solid organ malignancy with a high mortality rate. Statistics indicate that its incidence has been increasing as well as the associated deaths. Most patients with PDAC show poor response to therapies making the clinical management of this cancer difficult. Stromal cells in the tumor microenvironment (TME) contribute to the development of resistance to therapy in PDAC cancer cells. Cancer-associated fibroblasts (CAFs), the most prevalent stromal cells in the TME, promote a desmoplastic response, produce extracellular matrix proteins and cytokines, and directly influence the biological behavior of cancer cells. These multifaceted effects make it difficult to eradicate tumor cells from the body. As a result, CAF-targeting synergistic therapeutic strategies have gained increasing attention in recent years. However, due to the substantial heterogeneity in CAF origin, definition, and function, as well as high plasticity, majority of the available CAF-targeting therapeutic approaches are not effective, and in some cases, they exacerbate disease progression. This review primarily elucidates on the effect of CAFs on therapeutic efficiency of various treatment modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Strategies for CAF targeting therapies are also discussed.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Tumor Microenvironment , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/drug effects , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Immunotherapy/methods , Animals , Molecular Targeted Therapy
10.
Toxicology ; 504: 153782, 2024 May.
Article in English | MEDLINE | ID: mdl-38493947

ABSTRACT

Tobacco carcinogens are recognized as critical hazard factors for bladder tumorigenesis, affecting the prognosis of patients through aromatic amines components. However, the specific function of tobacco carcinogens and systematic assessment models in the prognosis of bladder cancer remains poorly elucidated. We retrieved bladder cancer specific tobacco carcinogens-related genes from Comparative Toxicogenomic Database, our Nanjing Bladder Cancer cohort and TCGA database. Gene×Gene interaction method was utilized to establish a prognostic signature. Integrative assessment of immunogenomics, tumor microenvironments and single-cell RNA-sequencing were performed to illustrate the internal relations of key events from different levels. Finally, we comprehensively identified 33 essential tobacco carcinogens-related genes to construct a novel prognostic signature, and found that high-risk patients were characterized by significantly worse overall survival (HR=2.25; Plog-rank < 0.01). Single-cell RNA-sequencing and multi-omics analysis demonstrated that cancer-associated fibroblasts mediated the crosstalk between epithelial-mesenchymal transition progression and immune evasion. Moreover, an adverse outcome pathway framework was established to facilitate our understanding to the tobacco carcinogens-triggered bladder tumorigenesis. Our study systematically provided immune microenvironmental alternations for smoking-induced adverse survival outcomes in bladder cancer. These findings facilitated the integrative multi-omics insights into risk assessment and toxic mechanisms of tobacco carcinogens.


Subject(s)
Cancer-Associated Fibroblasts , Epithelial-Mesenchymal Transition , Tumor Microenvironment , Urinary Bladder Neoplasms , Humans , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/drug effects , Carcinogens/toxicity , Gene Expression Regulation, Neoplastic , Immune Evasion , Multiomics , Prognosis , Single-Cell Analysis , Smoking/adverse effects , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
11.
Nanomedicine ; 58: 102743, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38484918

ABSTRACT

Cancer-associated fibroblasts (CAFs) play a crucial role in creating an immunosuppressive environment and remodeling the extracellular matrix within tumors, leading to chemotherapy resistance and limited immune cell infiltration. To address these challenges, integrating CAFs deactivation into immunogenic chemotherapy may represent a promising approach to the reversal of immune-excluded tumor. We developed a tumor-targeted nanomedicine called the glutathione-responsive nanocomplex (GNC). The GNC co-loaded dasatinib, a CAF inhibitor, and paclitaxel, a chemotherapeutic agent, to deactivate CAFs and enhance the effects of immunogenic chemotherapy. Due to the modification with hyaluronic acid, the GNC preferentially accumulated in the tumor periphery and responsively released cargos, mitigating the tumor stroma as well as overcoming chemoresistance. Moreover, GNC treatment exhibited remarkable immunostimulatory efficacy, including CD8+ T cell expansion and PD-L1 downregulation, facilitating immune checkpoint blockade therapy. In summary, the integration of CAF deactivation and immunogenic chemotherapy using the GNC nanoplatform holds promise for rebuilding immune-excluded tumors.


Subject(s)
Cancer-Associated Fibroblasts , Paclitaxel , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/metabolism , Animals , Humans , Mice , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Dasatinib/pharmacology , Dasatinib/therapeutic use , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/pathology , Cell Line, Tumor , Nanoparticles/chemistry , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Glutathione/metabolism
12.
Cancer Sci ; 115(5): 1505-1519, 2024 May.
Article in English | MEDLINE | ID: mdl-38476010

ABSTRACT

The fibrotic tumor microenvironment is a pivotal therapeutic target. Nintedanib, a clinically approved multikinase antifibrotic inhibitor, is effective against lung adenocarcinoma (ADC) but not squamous cell carcinoma (SCC). Previous studies have implicated the secretome of tumor-associated fibroblasts (TAFs) in the selective effects of nintedanib in ADC, but the driving factor(s) remained unidentified. Here we examined the role of tissue inhibitor of metalloproteinase-1 (TIMP-1), a tumor-promoting cytokine overproduced in ADC-TAFs. To this aim, we combined genetic approaches with in vitro and in vivo preclinical models based on patient-derived TAFs. Nintedanib reduced TIMP-1 production more efficiently in ADC-TAFs than SCC-TAFs through a SMAD3-dependent mechanism. Cell culture experiments indicated that silencing TIMP1 in ADC-TAFs abolished the therapeutic effects of nintedanib on cancer cell growth and invasion, which were otherwise enhanced by the TAF secretome. Consistently, co-injecting ADC cells with TIMP1-knockdown ADC-TAFs into immunocompromised mice elicited a less effective reduction of tumor growth and invasion under nintedanib treatment compared to tumors bearing unmodified fibroblasts. Our results unveil a key mechanism underlying the selective mode of action of nintedanib in ADC based on the excessive production of TIMP-1 in ADC-TAFs. We further pinpoint reduced SMAD3 expression and consequent limited TIMP-1 production in SCC-TAFs as key for the resistance of SCC to nintedanib. These observations strongly support the emerging role of TIMP-1 as a critical regulator of therapy response in solid tumors.


Subject(s)
Adenocarcinoma of Lung , Cancer-Associated Fibroblasts , Indoles , Lung Neoplasms , Smad3 Protein , Tissue Inhibitor of Metalloproteinase-1 , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Humans , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Mice , Indoles/pharmacology , Indoles/therapeutic use , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Smad3 Protein/metabolism , Cell Line, Tumor , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female
13.
Biomater Adv ; 160: 213831, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38552501

ABSTRACT

Nanoparticle (NP) use in cancer therapy is extensively studied in skin cancers. Cancer-associated fibroblasts (CAFs), a major tumor microenvironment (TME) component, promote cancer progression, making dual targeting of cancer cells and CAFs an effective therapy. However, dual NP-based targeting therapy on both tumor cells and CAFs is poorly investigated in skin cancers. Herein, we prepared and characterized doxorubicin-loaded PLGA NPs (DOX@PLGA NPs) and studied their anti-tumor effects on cutaneous melanoma (SKCM)(AN, M14) and cutaneous squamous cell carcinoma (cSCC) (MET1, MET2) cell lines in monolayer, as well as their impact on CAF deactivation. Then, we established 3D full thickness models (FTM) models of SKCM and cSCC using AN or MET2 cells on dermis matrix populated with CAFs respectively, and assessed the NPs' tumor penetration, tumor-killing ability, and CAF phenotype regulation through both topical administration and intradermal injection. The results show that, in monolayer, DOX@PLGA NPs inhibited cancer cell growth and induced apoptosis in a dose- and time-dependent manner, with a weaker effect on CAFs. DOX@PLGA NPs reduced CAF-marker expression and had successful anti-tumor effects in 3D skin cancer FTMs, with decreased tumor-load and invasion. DOX@PLGA NPs also showed great delivery potential in the FTMs and could be used as a platform for future functional study of NPs in skin cancers using human-derived skin equivalents. This study provides promising evidence for the potential of DOX@PLGA NPs in dual targeting therapy for SKCM and cSCC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Doxorubicin , Melanoma , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Skin Neoplasms , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Melanoma/drug therapy , Melanoma/pathology , Nanoparticles/chemistry , Cell Line, Tumor , Apoptosis/drug effects , Animals , Tumor Microenvironment/drug effects , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use
14.
Curr Cancer Drug Targets ; 24(6): 642-653, 2024.
Article in English | MEDLINE | ID: mdl-38310462

ABSTRACT

BACKGROUND: Immune-checkpoint inhibitors (ICIs) against programmed death (PD)-1/PD-L1 pathway immunotherapy have been demonstrated to be effective in only a subset of patients with cancer, while the rest may exhibit low response or may develop drug resistance after initially responding. Previous studies have indicated that extensive collagen-rich stroma secreted by cancer-associated fibroblasts (CAFs) within the tumor microenvironment is one of the key obstructions of the immunotherapy for some tumors by decreasing the infiltrating cytotoxic T cells. However, there is still a lack of effective therapeutic strategies to control the extracellular matrix by targeting CAFs. METHODS: The enhanced uptake of IR-780 by CAFs was assessed by using in vivo or ex vivo nearinfrared fluorescence imaging, confocal NIR fluorescent imaging, and CAFs isolation testing. The fibrotic phenotype down-regulation effects and in vitro CAFs killing effect of IR-780 were tested by qPCR, western blot, and flow cytometry. The in vivo therapeutic enhancement of anti-PD-L1 by IR-780 was evaluated on EMT6 and MC38 subcutaneous xenograft mice models. RESULTS: IR-780 has been demonstrated to be preferentially taken up by CAFs and accumulate in the mitochondria. Further results identified low-dose IR-780 to downregulate the fibrotic phenotype, while high-dose IR-780 could directly kill both CAFs and EMT6 cells in vitro. Moreover, IR-780 significantly inhibited extracellular matrix (ECM) protein deposition in the peri-tumoral stroma on subcutaneous EMT6 and MC38 xenografts, which increased the proportion of tumor-infiltrating lymphocytes (TILs) in the deep tumor and further promoted anti-PD-L1 therapeutic efficacy. CONCLUSION: This work provides a unique strategy for the inhibition of ECM protein deposition in the tumor microenvironment by targeted regulating of CAFs, which destroys the T cell barrier and further promotes tumor response to PD-L1 monoclonal antibody. IR-780 has been proposed as a potential therapeutic small-molecule adjuvant to promote the effect of immunotherapy.


Subject(s)
Cancer-Associated Fibroblasts , Immunotherapy , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Mice , Humans , Immunotherapy/methods , Tumor Microenvironment/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Immune Checkpoint Inhibitors/pharmacology , Indoles/pharmacology , Female , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Cell Line, Tumor , Xenograft Model Antitumor Assays
15.
Adv Healthc Mater ; 13(11): e2303837, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38183408

ABSTRACT

Targeted reprogramming of cancer-associated fibroblasts (CAFs) is one of the most essential cancer therapies. However, how to reprogram active CAFs toward deactivated state still remains immense challenge. To tackle this challenge, herein, one perylene N, N'-bis(2-((dimethylammonium)ethylene)-2-(methoxylethyl))-1, 6, 7, 12-tetrachloroperylene-3, 4, 9, 10-tetracarboxylic diimide (PDIC-OC) is prepared, which can trigger endogenous reactive oxygen species (ROS) burst to result in cytoskeletal dysfunction and cell apoptosis so that suppress transforming growth factor ß (TGF-ß) production. As a result, PDIC-OC can reprogram the activated CAFs and relieve immunosuppressive tumor microenvironment by efficient polarization of M2-typed macrophages into M1-typed ones, downregulation of alpha-smooth muscle actin (α-SMA), alleviation of hypoxic state to promote infiltration of cytotoxic T lymphocytes, and ultimately realizes outstanding antitumor performance on B16F10 tumor-xenografted and lung-metastatic mouse model even at low concentration of 1 mg kg-1 body weight. This work thus presents a novel strategy that cytoskeleton dysfunction and cell apoptosis cooperatively suppress the secretion of TGF-ß to reprogram CAFs and meanwhile clarifies intrinsic mechanism for perylene-triggered chemo-immunotherapy against hypoxic tumors.


Subject(s)
Cancer-Associated Fibroblasts , Cytoskeleton , Immunotherapy , Perylene , Animals , Perylene/analogs & derivatives , Perylene/pharmacology , Perylene/chemistry , Mice , Cytoskeleton/metabolism , Cytoskeleton/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Immunotherapy/methods , Cell Line, Tumor , Tumor Microenvironment/drug effects , Transforming Growth Factor beta/metabolism , Apoptosis/drug effects , Humans , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL
16.
Int J Mol Sci ; 24(17)2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37686288

ABSTRACT

The tumor microenvironment comprises multiple cell types, like cancer cells, endothelial cells, fibroblasts, and immune cells. In recent years, there have been massive research efforts focusing not only on cancer cells, but also on other cell types of the tumor microenvironment, thereby aiming to expand and determine novel treatment options. Fibroblasts represent a heterogenous cell family consisting of numerous subtypes, which can alter immune cell fractions, facilitate or inhibit tumor growth, build pre-metastatic niches, or stabilize vessels. These effects can be achieved through cell-cell interactions, which form the extracellular matrix, or via the secretion of cytokines or chemokines. The pro- or antitumorigenic fibroblast phenotypes show variability not only among different cancer entities, but also among intraindividual sites, including primary tumors or metastatic lesions. Commonly prescribed for arterial hypertension, the inhibitors of the renin-angiotensin system have recently been described as having an inhibitory effect on fibroblasts. This inhibition leads to modified immune cell fractions and increased tissue stiffness, thereby contributing to overcoming therapy resistance and ultimately inhibiting tumor growth. However, it is important to note that the inhibition of fibroblasts can also have the opposite effect, potentially resulting in increased tumor growth. We aim to summarize the latest state of research regarding fibroblast heterogeneity and its intricate impact on the tumor microenvironment and extracellular matrix. Specifically, we focus on highlighting recent advancements in the comprehension of intraindividual heterogeneity and therapy options within this context.


Subject(s)
Cancer-Associated Fibroblasts , Carcinogenesis , Neoplasms , Cancer-Associated Fibroblasts/classification , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/physiology , Humans , Tumor Microenvironment , Neoplasms/drug therapy , Neoplasms/pathology , Antihypertensive Agents/pharmacology , Extracellular Matrix Proteins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology
17.
Biochem Biophys Res Commun ; 628: 133-140, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36084551

ABSTRACT

The response to medroxyprogesterone acetate (MPA) decreases as endometrial disease progresses from the benign to malignancy. In a mouse model, progesterone receptor (PR) expression in normal fibroblasts is accountable for the MPA's inhibitory effects in cancer cells. However, it is still unclear, if and how, fibroblasts from human tumors respond to MPA. In this study, three benign-associated fibroblasts (BAFs) and four cancer-associated fibroblasts (CAFs) were isolated from human benign and cancerous endometrial tissues, respectively, to examine MPA activation on PR signaling. PR-B protein expression were heterogeneously expressed in both CAFs and BAFs, despite a lower mRNA expression in the former. In a luciferase reporter assay, MPA treatment stimulated some PR DNA-binding activity in BAFs but not in CAFs. Yet, activation of PR target gene was generally more pronounced in MPA-treated CAFs compared to BAFs. Cyclin-dependent kinase 1 (CDK1) was exclusively upregulated by 10 nM MPA in CAFs (5.1-fold vs. 1.1-fold in BAFs, P < 0.05), leading to a higher CDK1 protein expression. Subsequently in a dose-response study, CAFs showed an average of ∼20% higher cell viability when compared to BAFs, indicative of drug resistance to MPA. MPA resistance was also observed in EC-CAFs co-culture, when MPA-treated cells showed greater tumor spheroid formation than in EC-BAFs co-culture (2-fold, P < 0.01). The increased cell viability observed in CAFs was reversed with mifepristone (RU486), a PR antagonist which suppressed MPA-induced CDK1 expression. This indicates that MPA-induced abnormal upregulation of CDK1 may contribute to the enhanced CAFs cell proliferation, suggesting a new mechanism of MPA resistance within endometrial cancer microenvironment.


Subject(s)
CDC2 Protein Kinase , Cancer-Associated Fibroblasts , Drug Resistance, Neoplasm , Medroxyprogesterone Acetate , Neoplasms , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/drug therapy , Endometrium/pathology , Female , Humans , Luciferases/metabolism , Medroxyprogesterone Acetate/pharmacology , Medroxyprogesterone Acetate/therapeutic use , Mifepristone/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , RNA, Messenger/genetics , Receptors, Progesterone/metabolism , Up-Regulation
18.
Int J Mol Sci ; 23(14)2022 Jul 09.
Article in English | MEDLINE | ID: mdl-35886951

ABSTRACT

Vasopressin type-2 receptor (V2R) is ectopically expressed and plays a pathogenic role in clear cell renal cell carcinoma (ccRCC) tumor cells. Here we examined how V2R signaling within human ccRCC tumor cells (Caki1 cells) stimulates stromal cancer-associated fibroblasts (CAFs). We found that cell culture conditioned media from Caki1 cells increased activation, migration, and proliferation of fibroblasts in vitro, which was inhibited by V2R gene silencing in Caki1 cells. Analysis of the conditioned media and mRNA of the V2R gene silenced and control Caki1 cells showed that V2R regulates the production of CAF-activating factors. Some of these factors were also found to be regulated by YAP in these Caki1 cells. YAP expression colocalized and correlated with V2R expression in ccRCC tumor tissue. V2R gene silencing or V2R antagonist significantly reduced YAP in Caki1 cells. Moreover, the V2R antagonist reduced YAP expression and myofibroblasts in mouse xenograft tumors. These results suggest that V2R plays an important role in secreting pro-fibrotic factors that stimulate fibroblast activation by a YAP-dependent mechanism in ccRCC tumors. Our results demonstrate a novel role for the V2R-YAP axis in the regulation of myofibroblasts in ccRCC and a potential therapeutic target.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Renal Cell , Kidney Neoplasms , Receptors, Vasopressin , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Culture Media, Conditioned , Fibroblasts/metabolism , Humans , Kidney/drug effects , Kidney/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Vasopressins/genetics , Vasopressins/metabolism , Xenograft Model Antitumor Assays
19.
Theranostics ; 12(10): 4564-4580, 2022.
Article in English | MEDLINE | ID: mdl-35832090

ABSTRACT

Background: Since T cell exclusion contributes to tumor immune evasion and immunotherapy resistance, how to improve T cell infiltration into solid tumors becomes an urgent challenge. Methods: We employed deep learning to profile the tumor immune microenvironment (TIME) in triple negative breast cancer (TNBC) samples from TCGA datasets and noticed that fibroblast growth factor receptor (FGFR) signaling pathways were enriched in the immune-excluded phenotype of TNBC. Erdafitinib, a selective FGFR inhibitor, was then used to investigate the effect of FGFR blockade on TIME landscape of TNBC syngeneic mouse models by flow cytometry, mass cytometry (CyTOF) and RNA sequencing. Cell Counting Kit-8 (CCK-8) assay and transwell migration assay were carried out to detect the effect of FGFR blockade on cell proliferation and migration, respectively. Cytokine array, western blot, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence (IF) were employed to investigate the potential mechanism by which FGFR inhibition enhanced T cell infiltration. Results: Blocking FGFR pathway by Erdafitinib markedly suppressed tumor growth with increased T cell infiltration in immunocompetent mouse models of TNBC. Mechanistically, FGFR blockade inhibited cancer-associated fibroblasts (CAFs) proliferation, migration and secretion of vascular cell adhesion molecule 1 (VCAM-1) by down-regulating MAPK/ERK pathway in CAFs, thus promoting T cell infiltration by breaking physical and chemical barriers built by CAFs in TIME. Furthermore, we observed that FGFR inhibition combined with immune checkpoint blockade therapy (ICT) greatly improved the therapeutic response of TNBC tumor models. Conclusions: FGFR blockade enhanced ICT response by turning immune "cold" tumor into "hot" tumor, providing remarkable implications of FGFR inhibitors as adjuvant agents for combinatorial immunotherapy.


Subject(s)
Cancer-Associated Fibroblasts , Receptors, Fibroblast Growth Factor , T-Lymphocytes , Triple Negative Breast Neoplasms , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/immunology , Cell Line, Tumor , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
20.
BMC Cancer ; 22(1): 205, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35209871

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) are an important component of the tumour microenvironment. Recent studies revealed CAFs are heterogeneous and CAF subset(s) that suppress cancer progression (cancer-restraining CAFs [rCAFs]) must exist in addition to well-characterised cancer-promoting CAFs (pCAFs). However, the identity and specific markers of rCAFs are not yet reported. We recently identified Meflin as a specific marker of rCAFs in pancreatic and colon cancers. Our studies revealed that rCAFs may represent proliferating resident fibroblasts. Interestingly, a lineage tracing experiment showed Meflin-positive rCAFs differentiate into α-smooth muscle actin-positive and Meflin-negative CAFs, which are generally hypothesised as pCAFs, during cancer progression. Using a pharmacological approach, we identified AM80, a synthetic unnatural retinoid, as a reagent that effectively converts Meflin-negative pCAFs to Meflin-positive rCAFs. We aimed to investigate the efficacy of a combination of AM80 and gemcitabine (GEM) and nab-paclitaxel (nab-PTX) in patients with advanced pancreatic cancer. METHODS: The phase I part is a 3 + 3 design, open-label, and dose-finding study. The dose-limiting toxicity (DLT) of these combination therapies would be evaluated for 4 weeks. After the DLT evaluation period, if no disease progression is noted based on the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 or if the patient has no intolerable toxicity, administration of AM80 with GEM and nab-PTX would be continued for up to 24 weeks. The phase II part is an open-label, single-arm study. The maximum tolerated dose (MTD) of AM80 with GEM and nab-PTX, determined in phase I, would be administered until intolerable toxicity or disease progression occurs, up to a maximum of 24 weeks, to confirm efficacy and safety. The primary endpoints are frequency of DLT and MTD of AM80 with GEM and nab-PTX in the phase I part and response rate based on the RECIST in the phase II part. Given the historical control data, we hope that the response rate will be over 23% in phase II. DISCUSSION: Strategies to convert pCAFs into rCAFs have been developed in recent years. We hypothesised that AM80 would be a promising enhancer of chemosensitivity and drug distribution through CAF conversion in the stroma. TRIAL REGISTRATION: Clinicaltrial.gov: NCT05064618 , registered on 1 October 2021. jRCT: jRCT2041210056 , registered on 27 August 2021.


Subject(s)
Albumins/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzoates/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Repositioning/methods , Paclitaxel/administration & dosage , Pancreatic Neoplasms/drug therapy , Tetrahydronaphthalenes/administration & dosage , Adult , Aged , Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/drug effects , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Deoxycytidine/administration & dosage , Female , Humans , Immunoglobulins/drug effects , Male , Maximum Tolerated Dose , Middle Aged , Stromal Cells/drug effects , Treatment Outcome , Tumor Microenvironment/drug effects , Young Adult , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...