Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.617
Filter
1.
Front Immunol ; 15: 1433679, 2024.
Article in English | MEDLINE | ID: mdl-39086477

ABSTRACT

Introduction: Fibroblast activation protein (FAP) overexpression on cancer-associated fibroblasts (CAFs) is associated with poor prognosis and worse clinical outcomes. Selective ablation of pro-tumorgenic FAP+ stromal cells with CAR-T cells may be a new therapeutic strategy. However, the clinical use of FAP-CAR T cells is suggested to proceed with caution for occasional poor efficacy and induction of on-target off-tumor toxicity (OTOT), including lethal osteotoxicity and cachexia. Hence, more investigations and preclinical trials are required to optimize the FAP-CAR T cells and to approve their safety and efficacy. Methods: In this study, we designed second-generation CAR T cells targeting FAP with 4-1BB as a co-stimulatory molecule, and tested their cytotoxicity against FAP-positive cells (hFAP-HT1080 cells and a variety of primary CAFs) in vitro and in Cell line-derived xenograft (CDX) and a patient-derived xenograft (PDX) model. Results: Results showed that our FAP-CAR T cells were powerfully potent in killing human and murine FAP-positive tumor cells and CAFs in multiple types of tumors in BALB/c and C57BL/6 mice and in patient-derived xenografts (PDX) model. And they were proved to be biologically safe and exhibit low-level OTOT. Discussion: Taken together, the human/murine cross-reactive FAP-CAR T cells were powerfully potent in killing human and murine FAP positive tumor cells and CAFs. They were biologically safe and exhibit low-level OTOT, warranting further clinical investigation into our FAP-CAR T cells.


Subject(s)
Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Xenograft Model Antitumor Assays , Animals , Humans , Mice , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Cell Line, Tumor , Cross Reactions/immunology , Serine Endopeptidases/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Endopeptidases , Membrane Proteins/immunology , Membrane Proteins/genetics , Mice, Inbred BALB C , T-Lymphocytes/immunology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Mice, Inbred C57BL , Gelatinases/immunology , Gelatinases/metabolism , Neoplasms/immunology , Neoplasms/therapy , Female
2.
Oncol Res ; 32(8): 1323-1334, 2024.
Article in English | MEDLINE | ID: mdl-39055892

ABSTRACT

Background: Fibroblast activation protein (FAP), a cell surface serine protease, plays roles in tumor invasion and immune regulation. However, there is currently no pan-cancer analysis of FAP. Objective: We aimed to assess the pan-cancer expression profile of FAP, its molecular function, and its potential role in head and neck squamous cell carcinoma (HNSC). Methods: We analyzed gene expression, survival status, immune infiltration, and molecular functional pathways of FAP in The Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx) tumors. Furthermore, to elucidate the role of FAP in HNSC, we performed proliferation, migration, and invasion assays post-FAP overexpression or knock-down. Results: FAP expression was elevated in nine tumor types and was associated with poor survival in eight of them. In the context of immune infiltration, FAP expression negatively correlated with CD8+ T-cell infiltration in five tumor types and positively with regulatory T-cell infiltration in four tumor types. Our enrichment analysis highlighted FAP's involvement in the PI3K-Akt signaling pathway. In HNSC cells, FAP overexpression activated the PI3K-Akt pathway, promoting tumor proliferation, migration, and invasion. Conversely, FAP knockdown showed inhibitory effects. Conclusion: Our study unveils the association of FAP with poor tumor prognosis across multiple cancers and highlights its potential as a therapeutic target in HNSC.


Subject(s)
Biomarkers, Tumor , Cell Movement , Cell Proliferation , Endopeptidases , Head and Neck Neoplasms , Membrane Proteins , Serine Endopeptidases , Squamous Cell Carcinoma of Head and Neck , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/immunology , Prognosis , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Endopeptidases/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/metabolism , Gelatinases/metabolism , Gelatinases/genetics , Cell Line, Tumor , Signal Transduction , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism
3.
J Nucl Med ; 65(8): 1217-1223, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38960714

ABSTRACT

Despite their unique histologic features, gliosarcomas belong to the group of glioblastomas and are treated according to the same standards. Fibroblast activation protein (FAP) is a component of a tumor-specific subpopulation of fibroblasts that plays a critical role in tumor growth and invasion. Some case studies suggest an elevated expression of FAP in glioblastoma and a particularly strong expression in gliosarcoma attributed to traits of predominant mesenchymal differentiation. However, the prognostic impact of FAP and its diagnostic and therapeutic potential remain unclear. Here, we investigate the clinical relevance of FAP expression in gliosarcoma and glioblastoma and how it correlates with 68Ga-FAP inhibitor (FAPI)-46 PET uptake. Methods: Patients diagnosed with gliosarcoma or glioblastoma without sarcomatous differentiation with an overall survival of less than 2.5 y were enrolled. Histologic examination included immunohistochemistry and semiquantitative scoring of FAP (0-3, with higher values indicating stronger expression). Additionally, 68Ga-FAPI-46 PET scans were performed in a subset of glioblastomas without sarcomatous differentiation patients. The clinical SUVs were correlated with FAP expression levels in surgically derived tumor tissue and relevant prognostic factors. Results: Of the 61 patients who were enrolled, 13 of them had gliosarcoma. Immunohistochemistry revealed significantly more FAP in gliosarcomas than in glioblastomas without sarcomatous differentiation of tumor tissue (P < 0.0001). In the latter, FAP expression was confined to the perivascular space, whereas neoplastic cells additionally expressed FAP in gliosarcoma. A significant correlation of immunohistochemical FAP with SUVmean and SUVpeak of 68Ga-FAPI-46 PET indicates that clinical tracer uptake represents FAP expression of the tumor. Although gliosarcomas express higher levels of FAP than do glioblastomas without sarcomatous differentiation, overall survival does not significantly differ between the groups. Conclusion: The analysis reveals a significant correlation between SUVmean and SUVpeak in 68Ga-FAPI-46 PET and immunohistochemical FAP expression. This study indicates that FAP expression is much more abundant in the gliosarcoma subgroup of glioblastomas. This could open not only a diagnostic but also a therapeutic gap, since FAP could be explored as a theranostic target to enhance survival in a distinct subgroup of high-risk brain tumor patients with poor survival prognosis.


Subject(s)
Glioblastoma , Gliosarcoma , Positron-Emission Tomography , Humans , Gliosarcoma/diagnostic imaging , Gliosarcoma/metabolism , Gliosarcoma/pathology , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Glioblastoma/pathology , Male , Female , Middle Aged , Aged , Adult , Serine Endopeptidases/metabolism , Endopeptidases , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Gelatinases/metabolism , Survival Analysis , Membrane Proteins/metabolism , Prognosis , Quinolines
4.
J Nucl Med ; 65(8): 1188-1193, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38960716

ABSTRACT

The fibroblast activation protein (FAP) is highly expressed in tumor and stromal cells of mesothelioma and thus is an interesting imaging and therapeutic target. Previous data on PET imaging with radiolabeled FAP inhibitors (FAPIs) suggest high potential for superior tumor detection. Here, we report the data of a large malignant pleural mesothelioma cohort within a 68Ga-FAPI46 PET observational trial (NCT04571086). Methods: Of 43 eligible patients with suspected or proven malignant mesothelioma, 41 could be included in the data analysis of the 68Ga-FAPI46 PET observational trial. All patients underwent 68Ga-FAPI46 PET/CT, contrast-enhanced CT, and 18F-FDG PET/CT. The primary study endpoint was the association of 68Ga-FAPI46 PET uptake intensity and histopathologic FAP expression. Furthermore, secondary endpoints were detection rate and sensitivity, specificity, and positive and negative predictive values as compared with 18F-FDG PET/CT. Datasets were interpreted by 2 masked readers. Results: The primary endpoint was met, and the association between 68Ga-FAPI46 SUVmax or SUVpeak and histopathologic FAP expression was significant (SUVmax: r = 0.49, P = 0.037; SUVpeak: r = 0.51, P = 0.030).68Ga-FAPI46 and 18F-FDG showed similar sensitivity by histopathologic validation on a per-patient (100.0% vs. 97.3%) and per region (98.0% vs. 95.9%) basis. Per-region analysis revealed higher 68Ga-FAPI46 than 18F-FDG specificity (81.1% vs. 36.8%) and positive predictive value (87.5% vs. 66.2%). Conclusion: We confirm an association of 68Ga-FAPI46 uptake and histopathologic FAP expression in mesothelioma patients. Additionally, we report high sensitivity and superior specificity and positive predictive value for 68Ga-FAPI46 versus 18F-FDG.


Subject(s)
Endopeptidases , Fluorodeoxyglucose F18 , Gelatinases , Mesothelioma, Malignant , Mesothelioma , Positron Emission Tomography Computed Tomography , Humans , Positron Emission Tomography Computed Tomography/methods , Male , Female , Aged , Prospective Studies , Middle Aged , Mesothelioma/diagnostic imaging , Mesothelioma/metabolism , Mesothelioma, Malignant/diagnostic imaging , Mesothelioma, Malignant/metabolism , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Aged, 80 and over
5.
Front Immunol ; 15: 1352632, 2024.
Article in English | MEDLINE | ID: mdl-39035007

ABSTRACT

Introduction: This study investigates the role of Fibroblast Activation Protein (FAP)-positive cancer-associated fibroblasts (FAP+CAF) in shaping the tumor immune microenvironment, focusing on its association with immune cell functionality and cytokine expression patterns. Methods: Utilizing immunohistochemistry, we observed elevated FAP+CAF density in metastatic versus primary renal cell carcinoma (RCC) tumors, with higher FAP+CAF correlating with increased T cell infiltration in RCC, a unique phenomenon illustrating the complex interplay between tumor progression, FAP+CAF density, and immune response. Results: Analysis of immune cell subsets in FAP+CAF-rich stromal areas further revealed significant correlations between FAP+ stroma and various T cell types, particularly in RCC and non-small cell lung cancer (NSCLC). This was complemented by transcriptomic analyses, expanding the range of stromal and immune cell subsets interrogated, as well as to additional tumor types. This enabled evaluating the association of these subsets with tumor infiltration, tumor vascularization and other components of the tumor microenvironment. Our comprehensive study also encompassed cytokine, angiogenesis, and inflammation gene signatures across different cancer types, revealing heterogeneous cellular composition, cytokine expressions and angiogenic profiles. Through cytokine pathway profiling, we explored the relationship between FAP+CAF density and immune cell states, uncovering potential immunosuppressive circuits that limit anti-tumor activity in tumor-resident immune cells. Conclusions: These findings underscore the complexity of tumor biology and the necessity for personalized therapeutic and patient enrichment approaches. The insights gathered from FAP+CAF prevalence, immune infiltration, and gene signatures provide valuable perspectives on tumor microenvironments, aiding in future research and clinical strategy development.


Subject(s)
Cancer-Associated Fibroblasts , Immunotherapy , Serine Endopeptidases , Tumor Microenvironment , Tumor Microenvironment/immunology , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Immunotherapy/methods , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Cytokines/metabolism , Endopeptidases , Membrane Proteins/metabolism , Membrane Proteins/genetics , Gelatinases/metabolism , Gelatinases/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lung Neoplasms/metabolism
6.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000301

ABSTRACT

PET/CT using radiolabeled fibroblast activation protein inhibitors (FAPIs) is a promising diagnostic tool in oncology, especially when non-increased and/or physiologically high [18F]FDG uptake (as in liver parenchyma) is observed. We aimed to review the role of PET/CT using radiolabeled FAPIs in primary and/or metastatic liver lesions, and to compare their performances with more "conventional" radiopharmaceuticals. A search algorithm based on the terms "FAPI" AND ("hepatic" OR "liver") was applied, with the last update on 1st January 2024. Out of 177 articles retrieved, 76 studies reporting on the diagnostic application of radiolabeled FAPI PET/CT in at least one patient harboring primary or metastatic liver lesion(s) were fully analyzed. Although there was some heterogeneity in clinical conditions and/or study methodology, PET/CT with radiolabeled FAPIs showed an excellent performance in common primary liver malignancies (hepatocarcinoma, intrahepatic cholangiocarcinoma) and liver metastases (mostly from the gastrointestinal tract and lungs). A higher tumor-to-background ratio for FAPIs than for [18F]FDG was found in primary and metastatic liver lesions, due to lower background activity. Despite limited clinical evidence, radiolabeled FAPIs may be used to assess the suitability and effectiveness of FAPI-derived therapeutic agents such as [177Lu]Lu-FAPI. However, future prospective research on a wider population is needed to confirm the excellent performance.


Subject(s)
Fluorodeoxyglucose F18 , Liver Neoplasms , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Humans , Positron Emission Tomography Computed Tomography/methods , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Liver Neoplasms/metabolism , Radiopharmaceuticals/chemistry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Endopeptidases/metabolism , Gelatinases/metabolism , Gelatinases/antagonists & inhibitors
7.
Molecules ; 29(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999044

ABSTRACT

BACKGROUND: FAP radiopharmaceuticals show promise for cancer diagnosis; however, their limited tumor residency hinders treatment. This study compared two FAPi derivatives, DOTA.SA.FAPi and DOTAGA.(SA.FAPi)2, labeled with gallium-68 and lutetium-177, aiming to determine an optimum combination for creating theranostic pairs. METHODS: The radiotracers were studied for lipophilicity, binding to human serum proteins, and binding to human cancer-associated fibroblasts (CAFs) in vitro, including saturation and internalization/externalization studies. PET/SPECT/CT and biodistribution studies were conducted in PC3 and U87MG xenografts for [68Ga]Ga-DOTA.SA.FAPi and [68Ga]Ga-DOTAGA.(SA.FAPi)2. [177Lu]Lu-DOTA.SA.FAPi and [177Lu]Lu-DOTAGA.(SA.FAPi)2, were evaluated in PC3 xenografts. Biodistribution studies of [68Ga]Ga-DOTA.SA.FAPi were performed in healthy male and female mice. RESULTS: All radiotracers exhibited strong binding to FAP. Their internalization rate was fast while only [177Lu]Lu-DOTAGA.(SA.FAPi)2 was retained longer in CAFs. [68Ga]Ga-DOTAGA.(SA.FAPi)2 and [177Lu]Lu-DOTAGA.(SA.FAPi)2 displayed elevated lipophilicity and affinity for human serum proteins compared to [68Ga]Ga-DOTA.SA.FAPi and [177Lu]Lu-DOTA.SA.FAPi. In vivo studies revealed slower washout of [68Ga]Ga-DOTAGA.(SA.FAPi)2 within 3 h compared to [68Ga]Ga-DOTA.SA.FAPi. The tumor-to-tissue ratios of [68Ga]Ga-DOTAGA.(SA.FAPi)2 versus [68Ga]Ga-DOTA.SA.FAPi did not exhibit any significant differences. [177Lu]Lu-DOTAGA.(SA.FAPi)2 maintained a significant tumor uptake even after 96 h p.i. compared to [177Lu]Lu-DOTA.SA.FAPi. CONCLUSIONS: Dimeric compounds hold promise for therapy, while monomers are better suited for diagnostics. Finding the right combination is essential for effective disease management.


Subject(s)
Endopeptidases , Gallium Radioisotopes , Lutetium , Radioisotopes , Radiopharmaceuticals , Lutetium/chemistry , Humans , Animals , Mice , Tissue Distribution , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Gallium Radioisotopes/chemistry , Cell Line, Tumor , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Gelatinases/antagonists & inhibitors , Gelatinases/metabolism , Heterocyclic Compounds, 1-Ring/chemistry , Female , Male , Theranostic Nanomedicine
8.
Z Rheumatol ; 83(6): 455-459, 2024 Aug.
Article in German | MEDLINE | ID: mdl-39031195

ABSTRACT

Fibroblast activation protein (FAP) is mainly found on the surface of activated fibroblasts but is not expressed on the surface of inactive fibroblasts. Selective FAP inhibitors (FAPI), which are coupled to a radioactive tracer, can be used to quantify profibrotic and proinflammatory fibroblasts in patients using FAPI positron emission tomography (PET) computed tomography (CT). Following initial applications in neoplastic diseases, FAPI-PET/CT is also increasingly being applied in rheumatological diseases. The first studies have shown that in patients with systemic sclerosis (SSc) FAPI accumulates in actively fibrotically remodeled pulmonary and myocardial areas, that a high FAPI accumulation is associated with the risk of short-term progression and that this accumulation in the lungs regresses after successful treatment. In cases of immunoglobulin 4 (IgG4)-associated diseases (IgG4 rheumatic disease, RD), the FAPI signal correlates with the histological accumulation of activated fibroblasts and a poorer response to treatment to inhibit inflammation. Fibroblasts in chronically inflamed tissue, such as patients with inflammatory joint diseases, vasculitis or myositis, also express FAP and can be quantified by FAPI-PET/CT. The treatment-induced change of the phenotype from a destructive IL-6+/MMP3+THY1+ fibroblast subtype to an inflammation inhibiting CD200+DKK3+ subtype can be mechanistically demonstrated using FAPI-PET/CT. These studies provide indications that FAPI-PET/CT enables quantification of the tissue response in patients with fibrosing and chronic inflammatory diseases and can be used for patient stratification; however, further studies are essential for validation of the use of FAPI-PET/CT as a molecular imaging marker.


Subject(s)
Endopeptidases , Positron Emission Tomography Computed Tomography , Rheumatic Diseases , Humans , Rheumatic Diseases/diagnostic imaging , Membrane Proteins/metabolism , Gelatinases/metabolism , Serine Endopeptidases/metabolism , Radiopharmaceuticals , Fibroblasts/pathology , Treatment Outcome , Sensitivity and Specificity
9.
J Med Chem ; 67(15): 13392-13408, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39079004

ABSTRACT

Fibroblast activation protein (FAP) is a protein biomarker widely expressed in most solid human malignancies of epithelial origin. In recent years, a number of FAP-targeted small organic radioligands, including OncoFAP, have been utilized in the clinic for the detection and diagnosis of cancer. Despite their selective accumulation, conventional FAP ligands present a relatively short half-life in tumors, corresponding to a few hours after systemic administration. In order to maximize their efficacy, FAP-targeted radioligand therapeutics must possess prolonged tumor retention, thus irradiating tumor cells for days. In this work, we describe the development of compact OncoFAP multimers with improved FAP affinity (low picomolar IC50s), aimed at increasing tumor-residence time for therapeutic applications. An in silico analysis of the interaction of the multimers with FAP revealed a wide and deep pocket and six additional secondary binding sites. TriOncoFAP-DOTAGA emerged for its favorable in vitro profile and superior in vivo biodistribution performance in tumor-bearing mice.


Subject(s)
Endopeptidases , Animals , Humans , Mice , Endopeptidases/metabolism , Tissue Distribution , Membrane Proteins/metabolism , Cell Line, Tumor , Serine Endopeptidases/metabolism , Gelatinases/metabolism , Female , Neoplasms/drug therapy
10.
J Med Chem ; 67(14): 11827-11840, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39013156

ABSTRACT

Fibroblast activation protein (FAP) has attracted considerable attention as a possible target for the radiotherapy of solid tumors. Unfortunately, initial efforts to treat solid tumors with FAP-targeted radionuclides have yielded only modest clinical responses, suggesting that further improvements in the molecular design of FAP-targeted radiopharmaceutical therapies (RPT) are warranted. In this study, we report several advances on the previously described FAP6 radioligand that increase tumor retention and accelerate healthy tissue clearance. Seven FAP6 derivatives with different linkers or albumin binders were synthesized, radiolabeled, and investigated for their effects on binding and cellular uptake. The radioligands were then characterized in 4T1 tumor-bearing Balb/c mice using both single-photon emission computed tomography (SPECT) and ex vivo biodistribution analyses to identify the conjugate with the best tumor retention and tumor-to-healthy organ ratios. The results reveal an optimized FAP6 radioligand that exhibits efficacy and safety properties that potentially justify its translation into the clinic.


Subject(s)
Endopeptidases , Gelatinases , Membrane Proteins , Mice, Inbred BALB C , Radiopharmaceuticals , Serine Endopeptidases , Tomography, Emission-Computed, Single-Photon , Animals , Endopeptidases/metabolism , Mice , Tissue Distribution , Membrane Proteins/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/therapeutic use , Gelatinases/metabolism , Female , Serine Endopeptidases/metabolism , Cell Line, Tumor , Humans , Ligands
11.
Semin Nucl Med ; 54(4): 537-556, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39019653

ABSTRACT

The advancement of theranostics, which combines therapeutic and diagnostic capabilities in oncology, has significantly impacted cancer management. This review explores fibroblast activation protein (FAP) expression in the tumor microenvironment (TME) and its association with various malignancies, highlighting its potential as a theranostic marker for PET/CT imaging using FAP-targeted tracers and for FAP-targeted radiopharmaceutical therapy. We examine the development and clinical applications of FAP inhibitors (FAPIs) and peptides, providing insights into their diagnostic accuracy, initial therapeutic efficacy, and clinical impact across diverse cancer types, as well as the synthesis of novel FAP-targeted ligands. This review aims to showcase the promising outcomes and challenges in integrating FAP-targeted approaches into cancer management.


Subject(s)
Membrane Proteins , Peptides , Humans , Membrane Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Serine Endopeptidases/metabolism , Endopeptidases/metabolism , Animals , Gelatinases/metabolism , Gelatinases/antagonists & inhibitors , Neoplasms/diagnostic imaging , Neoplasms/therapy , Neoplasms/metabolism , Theranostic Nanomedicine/methods , Precision Medicine/methods , Tumor Microenvironment , Radiopharmaceuticals/therapeutic use
12.
Microb Pathog ; 193: 106762, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936638

ABSTRACT

Enterococcus faecalis is a troublesome nosocomial pathogen that acquired resistance to most available antimicrobial agents. Antivirulence agents represent an unconventional treatment approach. Here, transcription factor decoy (TFD)-loaded cationic liposomes (TLL) were developed as an inhibitor of the Fsr quorum-sensing system and its associated virulence traits, in E. faecalis. The consensus sequence of the FsrA binding site was found conserved among 651 E. faecalis annotated genomes. The TFD was synthesized as an 82 bp DNA duplex, containing the conserved binding sequence, and loaded onto cationic liposomes. The optimum loading capacity, mean particle size, and zeta potential of the TLL were characterized. The developed TLL lacked any effect on E. faecalis growth and significantly inhibited the in vitro production of the proteolytic enzymes controlled by the Fsr system; gelatinase and serine protease, in a concentration-dependent manner. This inhibition was accompanied by a significant reduction in the transcription levels of FsrA-regulated genes (fsrB, gelE, and sprE). The developed TLL were safe as evidenced by the nonhemolytic effect on human RBCs and the negligible cytotoxicity on human skin fibroblast cells. Moreover, in the larvae infection model, TLL displayed a significant abolish in the mortality rates of Galleria mellonella larvae infected with E. faecalis. In conclusion, the developed TLL offer a new safe strategy for combating E. faecalis infection through the inhibition of quorum-sensing-mediated virulence; providing a platform for the development of similar agents to combat many other pathogens.


Subject(s)
Bacterial Proteins , Enterococcus faecalis , Gram-Positive Bacterial Infections , Quorum Sensing , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Animals , Virulence/drug effects , Humans , Quorum Sensing/drug effects , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/drug therapy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Liposomes , Larva/microbiology , Transcription Factors/metabolism , Transcription Factors/genetics , Virulence Factors/genetics , Gelatinases/metabolism , Gelatinases/antagonists & inhibitors , Moths/microbiology , Erythrocytes/drug effects , Disease Models, Animal , Serine Proteases/metabolism , Serine Proteases/genetics , Gene Expression Regulation, Bacterial/drug effects , Nanoparticles/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
13.
Anal Cell Pathol (Amst) ; 2024: 8810804, 2024.
Article in English | MEDLINE | ID: mdl-38826849

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) poses significant challenges with poor survival rates and limited therapeutic strategies. Our study, using The Cancer Genome Atlas (TCGA) data, assesses cancer-associated fibroblast (CAF) gene signatures' clinical relevance. In our analysis across TCGA tumor types, differential gene expression analysis revealed that fibroblast activation protein (FAP) is upregulated in tumor tissues and associated with poorer survival rates in HNSCC. Furthermore, mechanistic studies employing gene-silencing techniques substantiated that FAP knockout led to a significant decrease in cellular proliferation, invasion, and migration in HNSCC cell lines. Through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses, we established that high FAP expression correlates with vital biological processes such as extracellular matrix organization, angiogenesis, and cellular motility. Importantly, FAP was found to regulate these processes by promoting the expression of key proteins involved in epithelial-mesenchymal transition-related pathways. Additionally, our analysis revealed a significant correlation between FAP expression and the expression profiles of immune checkpoint molecules, underscoring its potential role in immune modulation. Collectively, our findings illuminate FAP's pivotal role in HNSCC pathogenesis and its potential as a prognostic biomarker and therapeutic target. This research lays the groundwork for understanding the multifaceted roles and regulatory mechanisms of CAFs in HNSCC, thereby offering valuable perspectives for the development of targeted therapeutic strategies aimed at improving patient outcomes.


Subject(s)
Biomarkers, Tumor , Endopeptidases , Gelatinases , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms , Membrane Proteins , Serine Endopeptidases , Squamous Cell Carcinoma of Head and Neck , Humans , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Endopeptidases/metabolism , Endopeptidases/genetics , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line, Tumor , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Gelatinases/metabolism , Gelatinases/genetics , Epithelial-Mesenchymal Transition/genetics , Cell Proliferation/genetics , Cell Movement/genetics
14.
Signal Transduct Target Ther ; 9(1): 142, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38825657

ABSTRACT

Radiotherapy combined with immune checkpoint blockade holds great promise for synergistic antitumor efficacy. Targeted radionuclide therapy delivers radiation directly to tumor sites. LNC1004 is a fibroblast activation protein (FAP)-targeting radiopharmaceutical, conjugated with the albumin binder Evans Blue, which has demonstrated enhanced tumor uptake and retention in previous preclinical and clinical studies. Herein, we demonstrate that 68Ga/177Lu-labeled LNC1004 exhibits increased uptake and prolonged retention in MC38/NIH3T3-FAP and CT26/NIH3T3-FAP tumor xenografts. Radionuclide therapy with 177Lu-LNC1004 induced a transient upregulation of PD-L1 expression in tumor cells. The combination of 177Lu-LNC1004 and anti-PD-L1 immunotherapy led to complete eradication of all tumors in MC38/NIH3T3-FAP tumor-bearing mice, with mice showing 100% tumor rejection upon rechallenge. Immunohistochemistry, single-cell RNA sequencing (scRNA-seq), and TCR sequencing revealed that combination therapy reprogrammed the tumor microenvironment in mice to foster antitumor immunity by suppressing malignant progression and increasing cell-to-cell communication, CD8+ T-cell activation and expansion, M1 macrophage counts, antitumor activity of neutrophils, and T-cell receptor diversity. A preliminary clinical study demonstrated that 177Lu-LNC1004 was well-tolerated and effective in patients with refractory cancers. Further, scRNA-seq of peripheral blood mononuclear cells underscored the importance of addressing immune evasion through immune checkpoint blockade treatment. This was emphasized by the observed increase in antigen processing and presentation juxtaposed with T cell inactivation. In conclusion, our data supported the efficacy of immunotherapy combined with 177Lu-LNC1004 for cancer patients with FAP-positive tumors.


Subject(s)
Immune Checkpoint Inhibitors , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Endopeptidases/genetics , NIH 3T3 Cells , Radiopharmaceuticals/therapeutic use , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Xenograft Model Antitumor Assays , Immunotherapy , Gelatinases/genetics , Gelatinases/immunology , Lutetium/pharmacology , Cell Line, Tumor
15.
Environ Pollut ; 356: 124311, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38838811

ABSTRACT

Prolonged exposure to free silica leads to the development of silicosis, wherein activated fibroblasts play a pivotal role in its pathogenesis and progression. Fibroblast Activation Protein (FAP), as a biomarker for activated fibroblasts, its expression pattern and role in key aspects of silicosis pathogenesis remain unclear. This study elucidated the expression pattern and function of FAP through population-based epidemiological investigations, establishment of mouse models of silicosis, and in vitro cellular models. Results indicated a significant elevation of FAP in plasma from silicosis patients and lung tissues from mouse models of silicosis. In the cellular model, we observed a sharp increase in FAP expression early in the differentiation process, which remained high expression. Inhibition of FAP suppressed fibroblast differentiation, while overexpression of FAP produced the opposite effect. Moreover, fibroblast-derived FAP can alter the phenotype and function of neighboring macrophages. In summary, we revealed a high expression pattern of FAP in silicosis and its potential mechanistic role in fibrosis, suggesting FAP as a potential therapeutic target for silicosis.


Subject(s)
Fibroblasts , Membrane Proteins , Silicosis , Silicosis/metabolism , Animals , Mice , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Fibroblasts/metabolism , Male , Serine Endopeptidases/metabolism , Gelatinases/metabolism , Disease Models, Animal , Endopeptidases/metabolism , Lung/metabolism , Middle Aged , Female , Mice, Inbred C57BL , Macrophages/metabolism
16.
J Nucl Med ; 65(8): 1257-1263, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38871387

ABSTRACT

Because of upregulated expression on cancer-associated fibroblasts, fibroblast activation protein (FAP) has emerged as an attractive biomarker for the imaging and therapy of solid tumors. Although many FAP ligands have already been developed for radiopharmaceutical therapies (RPTs), most suffer from inadequate tumor uptake, insufficient tumor residence times, or off-target accumulation in healthy tissues, suggesting a need for further improvements. Methods: A new FAP-targeted RPT with a novel ligand (FAP8-PEG3-IP-DOTA) was designed by combining the desirable features of several previous ligand-targeted RPTs. Uptake and retention of [111In]In or [177Lu]Lu-FAP8-PEG3-IP-DOTA were assessed in KB, HT29, MDA-MB-231, and 4T1 murine tumor models by radioimaging or ex vivo biodistribution analyses. Radiotherapeutic potencies and gross toxicities were also investigated by monitoring tumor growth, body weight, and tissue damage in tumor-bearing mice. Results: FAP8-PEG3-IP-DOTA exhibited high affinity (half-maximal inhibitory concentration, 1.6 nM) and good selectivity for FAP relative to its closest homologs, prolyl oligopeptidase (half-maximal inhibitory concentration, ∼14.0 nM) and dipeptidyl peptidase-IV (half-maximal inhibitory concentration, ∼860 nM). SPECT/CT scans exhibited high retention in 2 different solid tumor models and minimal uptake in healthy tissues. Quantitative biodistribution analyses revealed tumor-to-healthy-tissue ratios of more than 5 times for all major organs, and live animal studies demonstrated 65%-93% suppression of tumor growth in all 4 models tested, with minimal or no evidence of systemic toxicity. Conclusion: We conclude that [177Lu]Lu-FAP8-PEG3-IP-DOTA constitutes a promising and safe RPT candidate for FAPα-targeted radionuclide therapy of solid tumors.


Subject(s)
Endopeptidases , Gelatinases , Membrane Proteins , Radiopharmaceuticals , Serine Endopeptidases , Animals , Mice , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Gelatinases/metabolism , Humans , Cell Line, Tumor , Serine Endopeptidases/metabolism , Membrane Proteins/metabolism , Tissue Distribution , Female , Drug Design , Lutetium/therapeutic use , Neoplasms/radiotherapy , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/therapeutic use , Molecular Targeted Therapy , Radioisotopes
17.
Talanta ; 278: 126475, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38944939

ABSTRACT

Fibroblast activation protein-α (FAP) plays a crucial role in various physiological and pathological processes, making it a key target for cancer diagnostics and therapeutics. However, in vivo detection of FAP activity with fluorogenic probes remains challenging due to the rapid diffusion and clearance of fluorescent products from the target. Herein, we developed a self-immobilizing near-infrared (NIR) fluorogenic probe, Hcy-CF2H-PG, by introducing a difluoromethyl group to FAP substrate-caged NIR fluorophore. Upon selective activation by FAP, the fluorescence of Hcy-CF2H-PG was triggered, followed by the covalent labelling of FAP. Hcy-CF2H-PG demonstrated significantly improved sensitivity, selectivity, and long-lasting labelling capacity for FAP both in vitro and in vivo, compared to that of non-immobilized probes. This represents a noteworthy advancement in FAP detection and cancer diagnostics within complex physiological systems.


Subject(s)
Endopeptidases , Fluorescent Dyes , Gelatinases , Membrane Proteins , Serine Endopeptidases , Fluorescent Dyes/chemistry , Animals , Humans , Membrane Proteins/analysis , Membrane Proteins/metabolism , Gelatinases/metabolism , Endopeptidases/metabolism , Serine Endopeptidases/metabolism , Serine Endopeptidases/analysis , Infrared Rays , Mice , Optical Imaging
18.
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
19.
Cancer Res Commun ; 4(6): 1481-1494, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38747612

ABSTRACT

Cancer-associated fibroblasts (CAF) are a prominent cell type within the tumor microenvironment (TME) where they are known to promote cancer cell growth and survival, angiogenesis, drug resistance, and immunosuppression. The transmembrane prolyl protease fibroblast activation protein (FAP) is expressed on the surface of highly protumorigenic CAFs found in the stroma of nearly every cancer of epithelial origin. The widespread expression of FAP has made it an attractive therapeutic target based on the underlying hypothesis that eliminating protumorigenic CAFs will disrupt the cross-talk between components of TME resulting in cancer cell death and immune infiltration. This hypothesis, however, has never been directly proven. To eliminate FAP-expressing CAFs, we developed an antibody-drug conjugate using our anti-FAP antibody, huB12, coupled to a monomethyl auristatin E (huB12-MMAE) payload. After determining that huB12 was an effective targeting vector, we found that huB12-MMAE potently eliminated FAP-expressing cells as monocultures in vitro and significantly prolonged survival in vivo using a xenograft engineered to overexpress FAP. We investigated the effects of selectively eliminating CAFs using a layered, open microfluidic cell coculture platform, known as the Stacks. Analysis of mRNA and protein expression found that treatment with huB12-MMAE resulted in the increased secretion of the proinflammatory cytokines IL6 and IL8 by CAFs and an associated increase in expression of proinflammatory genes in cancer cells. We also detected increased secretion of CSF1, a cytokine involved in myeloid recruitment and differentiation. Our findings suggest that the mechanism of FAP-targeted therapies is through effects on the immune microenvironment and antitumor immune response. SIGNIFICANCE: The direct elimination of FAP-expressing CAFs disrupts the cross-talk with cancer cells leading to a proinflammatory response and alterations in the immune microenvironment and antitumor immune response.


Subject(s)
Cancer-Associated Fibroblasts , Endopeptidases , Immunoconjugates , Tumor Microenvironment , Humans , Animals , Immunoconjugates/pharmacology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/immunology , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Endopeptidases/genetics , Endopeptidases/metabolism , Cell Line, Tumor , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Xenograft Model Antitumor Assays , Gelatinases/metabolism , Gelatinases/genetics , Oligopeptides/pharmacology , Female
20.
Pathol Res Pract ; 259: 155380, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815508

ABSTRACT

PURPOSE: Fibroblast activation protein (FAP) is a serine integral membrane protease, the expression of which has been confirmed in various cancer types. Solitary fibrous tumors of the pleura (SFTP) are rare mesenchymal fibroblastic neoplasms. We present a case of 18F-labeled FAP inhibitor ([18F]FAPI-74) PET imaging and its correlation with histological FAP expression and review an SFTP series at our institution in relation to the extent of FAP expression. METHODS: This retrospective study included 13 patients who underwent surgery between March 2011 and December 2022 at our institute. One of the patients also underwent [18F]FAPI-74 PET imaging. We semi-quantitatively evaluated FAP expression in SFTPs using immunohistochemical staining and H-scores. RESULTS: Nine of the 13 patients were male, with a median age of 64 years (range, 28-79 years). The median tumor size was 6.6 cm (1.1, 16 cm). In the pathological findings, expression levels of Ki67 were 1-5% in 12 of 13 cases. Furthermore, FAP expression was observed in all patients, and the median H-score was 160 (range, 10-280). The H-score of FAP expression in two of the 13 patients was low (10 in both), and that in two of the 13 patients was high (240 and 280). The SUVmax value of [18F]FAPI-74 PET was 3.57 in a patient in whom the H-score of FAP expression was 180. CONCLUSIONS: SFTPs expressed FAP to varying degrees in different patients and the [18F]FAPI-74 PET results in one patient reflected FAP expression in the tumor tissue.


Subject(s)
Endopeptidases , Gelatinases , Membrane Proteins , Serine Endopeptidases , Humans , Male , Middle Aged , Adult , Endopeptidases/metabolism , Aged , Serine Endopeptidases/metabolism , Serine Endopeptidases/analysis , Female , Retrospective Studies , Membrane Proteins/metabolism , Membrane Proteins/analysis , Gelatinases/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Positron-Emission Tomography , Solitary Fibrous Tumors/pathology , Solitary Fibrous Tumors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL