3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Fibroblast Activating Protein: Skimming the Surface of Molecular Imaging to Assess Fibrotic Disease Activity

      editorial
      1 , 2
      American Journal of Respiratory and Critical Care Medicine
      American Thoracic Society

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Noninvasive studies to assist with the diagnosis of pulmonary fibrosis and individual prognostication represent a “holy grail” for idiopathic pulmonary fibrosis (IPF) and other interstitial lung diseases (ILDs). Multiple studies have investigated the ability of physiologic measurements, molecular profiling, circulating cellular markers (i.e., monocytes), and imaging biomarkers to predict disease progression. However, there remains no clinically used disease activity marker for single–time point assessment to assist with treatment decisions. In this issue of the Journal, Yang and colleagues (pp. 160–172) evaluate “fibroblast activating protein” (FAP) as a molecular marker of disease activity with the potential to be leveraged for prognostic purposes (1). FAP is a transmembrane serine protease that was initially found to be expressed by cancer-associated stromal cells and subsequently shown to be expressed on other cell types, including macrophages (2). While associated with fibrosis, FAP has been shown to have antifibrotic actions linked to its role in the proteolytic degradation and processing of collagen. Indeed, FAP-deficient mice have increased collagen accumulation in lungs after radiation- or bleomycin-induced injury (3). However, FAP is also reported to enhance α2-antiplasmin function, which would be expected to stabilize fibrinogen and fibronectin and contribute to lung fibrosis (4, 5). FAP expression is increased by the profibrotic cytokine TGF-β (transforming growth factor-β), and expression is increased in murine models of lung fibrosis. Within the lungs of patients with IPF, FAP was upregulated within fibroblastic foci, but not in the adjacent alveolar epithelium or in normal lung tissues (6). However, increased expression may not be detrimental; a recent study showed that depletion of FAP+ cells or genetic deletion of FAP exacerbated fibrosis in a murine model of repetitive bleomycin injury and had no impact on fibrosis induced by adenoviral overexpression of TGF-β (7). Regardless of its role in pathogenesis, recent studies have used FAP to direct delivery of approved and emerging antifibrotic drugs, supporting its potential as an indicator of active wound repair and fibrosis (8, 9). Here Yang and colleagues (1) evaluate FAP expression using human lung fibroblasts, a murine model of pulmonary fibrosis, single-cell sequencing, and a radiolabeled FAP inhibitor administered to patients with ILD. This study has several major findings. Fibroblast expression of FAP is induced by TGF-β. Similar findings have been shown in fibroblasts from several sources and disease states (2, 6). In vivo, increased FAP expression was detected 1 week after bleomycin-induced lung injury. FAP expression was increased in fibrotic human lung tissue, with half of explanted IPF samples and almost all of silicosis lung tissue samples showing elevations, demonstrating that increased FAP expression is not specific for IPF. Single-cell transcriptomic analysis indicated that FAP was highly expressed in mesenchymal cells and that there was moderate correlation with other mesenchymal cell markers associated with fibrosis (α-SMA [α-smooth muscle actin], fibronectin, and collagen I). In a small group of healthy volunteers and a larger group of subjects with ILD (IPF and non-IPF), 68Ga-FAPI-04 positron emission technology (PET) uptake was increased in the lungs of subjects with ILD compared with healthy volunteers. Differences in total standardized uptake value (SUVtotal) but not mean standardized uptake value were seen between the IPF and non-IPF ILD groups, with the IPF group having a higher SUVtotal. As SUVtotal, a measurement defined by the authors, captures the extent of probe uptake above a threshold, this finding hints at underlying biologic heterogeneity among ILD subtypes. SUVtotal correlated moderately with subsequent changes in FVC and Dl CO; however, the follow-up times for pulmonary function testing were nonuniform, ranging from 4 to 24 months. A major strength of this study lies in the application of molecular imaging to establish a link between FAP expression as a biologic indicator of fibroblast activation and a noninvasive indicator of disease activity. PET probes targeting type I collagen, CCR2 (C-C chemokine receptor 2), αvβ6 integrin, and CXCR4 (C-X-C motif chemokine receptor 4) have been recently applied to patients with IPF to detect disease or to assess treatment effects (10–13). 68Ga-FAPI-04 PET/computed tomography has been performed in systemic sclerosis–associated ILD, and the degree of probe uptake was associated with disease progression (14). This study by Yang and colleagues (1) illustrates several important applications of molecular imaging to ILD. First, it can be used to obtain molecular information otherwise not available and thus advance our understanding of molecular heterogeneity within individuals and ILD subtypes. Second, imaging of molecular pathways involved in fibrogenesis could provide a window into disease activity at a single time point. This study also highlights important considerations in using molecular imaging. PET biomarkers that incorporate both magnitude of uptake and amount of lung involved may better predict disease progression for diffuse lung diseases compared with traditional PET measurements such as mean standardized uptake value. Several important gaps remain in establishing FAP as a reliable marker of disease activity. This study does not definitively link FAP expression with profibrotic fibroblast activity. Indeed, although FAP correlated with ACTA2 (actin alpha 2, smooth muscle), COL1A1 (collagen type I alpha 1 chain), and FN1 (fibronectin 1) in single-cell analysis, transcriptional expression does not always reflect protein concentration. Given its role in collagen turnover, FAP may have a functional role in controlling the wound-repair response to prevent the excessive collagen accumulation that characterizes fibrosis. Thus, studies will need to determine the ability of molecular imaging for FAP to distinguish between physiologic and fibrotic repair. More definitive studies are needed to determine if 68Ga-FAPI-04 PET can reliably predict disease progression in ILD and whether FAP activity can predict responsiveness to antifibrotic therapies. Although the premise of differentiating “active” from “stable” disease by noninvasive FAP assessment as a decision tool to guide pharmacotherapy is appealing, fibrosis progression is not linear and for some individuals may occur in a stepwise manner. In the liver, FAP expression in hepatic stellate cells correlates with fibrosis severity (15), and circulating FAP is increased in patients with increased liver stiffness, supporting a potential role for FAP in the risk stratification of patients for liver fibrosis (16). In the lung, one modality may not provide the sensitivity and specificity needed for risk prediction or guidance of therapy in an individual patient. Nevertheless, the combination of accessible serum or genetic markers with physiologic measurements and molecular imaging may improve our ability to risk stratify individuals with ILD.

          Related collections

          Most cited references16

          • Record: found
          • Abstract: found
          • Article: not found

          The role of fibroblast activation protein in health and malignancy

          Fibroblast activation protein-α (FAP) is a type-II transmembrane serine protease expressed almost exclusively to pathological conditions including fibrosis, arthritis, and cancer. Across most cancer types, elevated FAP is associated with worse clinical outcomes. Despite the clear association between FAP and disease severity, the biological reasons underlying these clinical observations remain unclear. Here we review basic FAP biology and FAP's role in non-oncologic and oncologic disease. We further explore how FAP may worsen clinical outcomes via its effects on extracellular matrix remodeling, intracellular signaling regulation, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. Lastly, we discuss the potential to exploit FAP biology to improve clinical outcomes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Fibroblast activation protein: a serine protease expressed at the remodeling interface in idiopathic pulmonary fibrosis.

            Fibroblast activation protein (FAPalpha) is a member of the cell surface dipeptidyl peptidase (DPP) family of serine proteases. In its dimer form, FAPalpha exhibits gelatinase, collagenase, and DPP activity in vitro. Reactive fibroblasts in healing wounds and stromal fibroblasts associated with epithelial tumors express FAPalpha. Idiopathic pulmonary fibrosis (IPF) is a disease of the lung characterized by progressive fibrosis with no clear etiology or molecular marker for disease activity. Recently, it has been shown that fibroblast FAPalpha expression is induced in liver cirrhosis, with an expression pattern distinct from alpha-smooth muscle actin (alpha-SMA). In this study, we determine whether FAPalpha expression is selectively induced in areas of ongoing tissue remodeling characterized by fibroblast foci in IPF. Human lung tissue was obtained from patients with IPF, centrilobular emphysema, and normal lung. Immunohistochemical studies were performed using anti-FAPalpha antibody and antibodies against alpha-SMA and CD26 (DPPIV), another member of the DPP family. We found that FAPalpha was not expressed in normal human lung tissue or tissue with evidence of centriacinar emphysema, but was induced in all patients with IPF and With a pattern distinct from that of CD26 found primarily on hyperplastic alveolar epithelium. Specifically, FAPalpha was detected in fibroblast foci and in fibrotic interstitium and not in the interstitium of adjacent architecturally normal lung. Alveolar/airway epithelium and vascular smooth muscle did not express FAPalpha. This is the first report of FAPalpha expression in IPF and our results suggest that FAPalpha is selectively induced in fibrotic foci, but not in normal or emphysematous lung. Future studies will address whether FAPalpha may be used as a marker for disease activity in IPF.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Evaluation of integrin αvβ 6 cystine knot PET tracers to detect cancer and idiopathic pulmonary fibrosis

              Advances in precision molecular imaging promise to transform our ability to detect, diagnose and treat disease. Here, we describe the engineering and validation of a new cystine knot peptide (knottin) that selectively recognizes human integrin αvβ6 with single-digit nanomolar affinity. We solve its 3D structure by NMR and x-ray crystallography and validate leads with 3 different radiolabels in pre-clinical models of cancer. We evaluate the lead tracer’s safety, biodistribution and pharmacokinetics in healthy human volunteers, and show its ability to detect multiple cancers (pancreatic, cervical and lung) in patients at two study locations. Additionally, we demonstrate that the knottin PET tracers can also detect fibrotic lung disease in idiopathic pulmonary fibrosis patients. Our results indicate that these cystine knot PET tracers may have potential utility in multiple disease states that are associated with upregulation of integrin αvβ6.
                Bookmark

                Author and article information

                Journal
                Am J Respir Crit Care Med
                Am J Respir Crit Care Med
                ajrccm
                American Journal of Respiratory and Critical Care Medicine
                American Thoracic Society
                1073-449X
                1535-4970
                8 September 2022
                15 January 2023
                8 September 2022
                : 207
                : 2
                : 122-124
                Affiliations
                [ 1 ]Division of Pulmonary and Critical Care Medicine

                Massachusetts General Hospital

                Boston, Massachusetts
                [ 2 ]Division of Pulmonary, Critical Care and Sleep Medicine

                The Ohio State University

                Columbus, Ohio
                Author information
                https://orcid.org/0000-0002-5724-6808
                https://orcid.org/0000-0002-1505-2837
                Article
                202208-1638ED
                10.1164/rccm.202208-1638ED
                9893323
                36075072
                30383443-518b-4e8f-a8ca-d5fc53f3a092
                Copyright © 2023 by the American Thoracic Society

                This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0. For commercial usage and reprints, please e-mail Diane Gern ( dgern@ 123456thoracic.org ).

                History
                Page count
                Figures: 0, Tables: 0, References: 16, Pages: 3
                Funding
                Funded by: National Heart, Lung, and Blood Institute, doi 10.13039/100000050;
                Award ID: K23HL150331
                Award ID: R01HL141195
                Categories
                Editorials

                Comments

                Comment on this article