Feasibility of in vivo Imaging of Fibroblast Activation Protein in Human Arterial Walls

2021 ◽  
pp. jnumed.121.262863
Author(s):  
Meiqi Wu ◽  
Jing Ning ◽  
Jingle Li ◽  
Zhichao Lai ◽  
Ximin Shi ◽  
...  
2018 ◽  
Vol 6 (10) ◽  
pp. 1449-1451 ◽  
Author(s):  
Jie Xing ◽  
Qiuyu Gong ◽  
Ruifen Zou ◽  
Zihou Li ◽  
Yuanzhi Xia ◽  
...  

Design and synthesis of a novel fibroblast activation protein “off–on” near-infrared fluorescent probe for cell detection, in vitro and in vivo imaging.


2012 ◽  
Vol 23 (8) ◽  
pp. 1704-1711 ◽  
Author(s):  
Jinbo Li ◽  
Kai Chen ◽  
Hongguang Liu ◽  
Kai Cheng ◽  
Meng Yang ◽  
...  

2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Xueying Zhang ◽  
Daoyun Chen ◽  
John W. Babich ◽  
Samuel J.E. Green ◽  
Xiang-Hua Deng ◽  
...  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ryoichi Katsube ◽  
Kazuhiro Noma ◽  
Toshiaki Ohara ◽  
Noriyuki Nishiwaki ◽  
Teruki Kobayashi ◽  
...  

AbstractCancer-associated fibroblasts (CAFs) have an important role in the tumor microenvironment. CAFs have the multifunctionality which strongly support cancer progression and the acquisition of therapeutic resistance by cancer cells. Near-infrared photoimmunotherapy (NIR-PIT) is a novel cancer treatment that uses a highly selective monoclonal antibody (mAb)-photosensitizer conjugate. We developed fibroblast activation protein (FAP)-targeted NIR-PIT, in which IR700 was conjugated to a FAP-specific antibody to target CAFs (CAFs-targeted NIR-PIT: CAFs-PIT). Thus, we hypothesized that the control of CAFs could overcome the resistance to conventional chemotherapy in esophageal cancer (EC). In this study, we evaluated whether EC cell acquisition of stronger malignant characteristics and refractoriness to chemoradiotherapy are mediated by CAFs. Next, we assessed whether the resistance could be rescued by eliminating CAF stimulation by CAFs-PIT in vitro and in vivo. Cancer cells acquired chemoradiotherapy resistance via CAF stimulation in vitro and 5-fluorouracil (FU) resistance in CAF-coinoculated tumor models in vivo. CAF stimulation promoted the migration/invasion of cancer cells and a stem-like phenotype in vitro, which were rescued by elimination of CAF stimulation. CAFs-PIT had a highly selective effect on CAFs in vitro. Finally, CAF elimination by CAFs-PIT in vivo demonstrated that the combination of 5-FU and NIR-PIT succeeded in producing 70.9% tumor reduction, while 5-FU alone achieved only 13.3% reduction, suggesting the recovery of 5-FU sensitivity in CAF-rich tumors. In conclusion, CAFs-PIT could overcome therapeutic resistance via CAF elimination. The combined use of novel targeted CAFs-PIT with conventional anticancer treatments can be expected to provide a more effective and sensible treatment strategy.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 813-813
Author(s):  
Angela Pennisi ◽  
Xin Li ◽  
Dana Gaddy ◽  
Nisreen Akel ◽  
Nazneen Aziz ◽  
...  

Abstract Fibroblast activation protein (FAP), a cell surface serine protease with both dipeptidyl peptidase and collagenase activity, is selectively expressed by tumor stroma and involved in tumor metastasis. We have reported that FAP is upregulated in myelomatous bone and is overexpressed in osteoclasts after coculture with myeloma (MM) cells. FAP is not expressed by MM cells and FAP siRNA reduced MM cell survival in cocultures (Ge et al., BJH 2006). The aim of the study was to investigate the effect of FAP inhibitors, PT-100 and PT-630 on MM cell growth and osteoclastogenesis using coculture system and the SCID-hu model for primary MM. PT-630 inhibits cell surface dipeptidyl peptidase activity while PT-100 also inhibits intracellular activity of these enzymes. MM cells from 6 patients were cocultured with osteoclasts and treated twice a day with PT-100 and PT-630 (0.1–100 μM) for 5–7 days. Whereas PT-100 effectively inhibited MM cell growth in all tested doses by 38%–62% (p<0.002 vs. 100 μM), PT-630 inhibited MM cell growth in a dose dependent manner reaching 45% growth inhibition with 100 μM (p<0.02). These compounds had no direct effect on MM cell survival. Moreover, recombinant FAP had no impact on MM cells cultured alone, suggesting that FAP-induced MM cell survival depends on close contact between MM cells and osteoclasts. The anti-MM effect of PT-100 in cocultures was mediated through downregulation of phosphorylated p38 in MM cells as detected by Phospho MAPK array and confirmed by Western blot. MMP-2 and MMP-9 have been associated with FAP activity. The level of MMP-2 but not MMP-9 was reduced in coculture conditioned media by 44±7% (p<0.04) following treatment with PT-100 while PT-630 had no significant effect on production of these matrix metalloproteinases. To test effect on osteoclastogenesis, osteoclast precursors were incubated with RANKL and M-CSF in the absence and presence of PT-100 (1 μM) and PT-630 (10 μM) for 5–7 days. PT-100 and PT-630 inhibited formation of multinucleated osteoclasts by 78±6% (p<0.001) and 56±6% (p<0.003), respectively. Culture of osteoclasts on dentine slices in the presence of PT-100 and PT-630 reduced resorption pit area by 92% (p<0.01) and 69% (p<0.04), respectively. The anti-osteoclastogenic effects were mediated through inhibition of phosphorylated p38 MAPK in osteoclastic cultures in a dose related manner. In vivo, SCID-hu mice engrafted with MM cells from 4 patients were orally treated for 4–5 weeks with PT-100 (20 mg/day) and PT-630 (200 mg/day). These agents inhibited MM growth in 2 experiments, delayed growth in one experiment and had no effect on MM in an additional experiment. Overall, final hIg levels in hosts treated with vehicle, PT-100 and PT-630 were 355±170, 183±78 and 76±27 mg/ml, respectively. Bone mineral density (BMD) of the myelomatous bone was increased in responding hosts (3% vs. -32% change from pretreatment level in control) and had reduced severity of bone loss in myelomatous bone of nonresponding hosts (−15% vs. −28% change from pretreatment level in control), suggesting that, as shown in vitro, these agents directly affect bone cell function in vivo. We conclude that FAP is critically involved in MM osteolysis and tumor growth and thus approaches to inhibit FAP activity in myelomatous bone may help control MM and its associated bone disease.


Author(s):  
Euy Sung Moon ◽  
Filipe Elvas ◽  
Gwendolyn Vliegen ◽  
Stef De Lombaerde ◽  
Christel Vangestel ◽  
...  

Abstract Background Fibroblast activation protein (FAP) is a proline selective serine protease that is overexpressed in tumor stroma and in lesions of many other diseases that are characterized by tissue remodeling. In 2014, a most potent FAP-inhibitor (referred to as UAMC1110) with low nanomolar FAP-affinity and high selectivity toward related enzymes such as prolyl oligopeptidase (PREP) and the dipeptidyl-peptidases (DPPs): DPP4, DPP8/9 and DPP2 were developed. This inhibitor has been adopted recently by other groups to create radiopharmaceuticals by coupling bifunctional chelator-linker systems. Here, we report squaric acid containing bifunctional DATA5m and DOTA chelators relied on UAMC1110. Results The radiopharmaceuticals DOTA.SA.FAPi and DATA5m.SA.FAPi were synthesized, labeled with gallium-68 and further characterized for in vitro stability, inhibitory efficiency, in vivo targeting properties and ex vivo biodistribution. [68Ga]Ga-DOTA.SA.FAPi and [68Ga]Ga-DATA5m.SA.FAPi showed high complexation after already 10 minutes and high stability over a period of 2 h. Affinity to FAP of DOTA.SA.FAPi and its natGa and natLu-labeled derivatives were in low nanomolar range. Comparable results were obtained for DATA5m.SA.FAPi and its natGa analogue. Additionally, all five compounds showed low affinity for the related protease PREP (high µM range). First proof-of-principle in vivo PET-imaging animal studies of the [68Ga]Ga-DOTA.SA.FAPi precursor in a HT-29 human colorectal cancer xenograft mouse model indicated promising results with high accumulation in tumor and low background signal. Ex vivo biodistribution showed high tumor uptake at 60 min post injection with overall low uptake in healthy tissues. Conclusion In this work, novel PET radiotracers targeting fibroblast activation protein (FAP) were synthesized and biochemically investigated. Critical substructures of the novel compounds are a squaramide linker unit derived from the basic motif of squaric acid, DOTA and DATA5m bifunctional chelators and a FAP-targeting moiety. In conclusion, these new FAP-ligands appear promising, both for further research and development as well as for first human application.


Molecules ◽  
2022 ◽  
Vol 27 (1) ◽  
pp. 264
Author(s):  
Diana Trujillo-Benítez ◽  
Myrna Luna-Gutiérrez ◽  
Guillermina Ferro-Flores ◽  
Blanca Ocampo-García ◽  
Clara Santos-Cuevas ◽  
...  

Fibroblast activation protein (FAP) is expressed in the microenvironment of most human epithelial tumors. 68Ga-labeled FAP inhibitors based on the cyanopyrrolidine structure (FAPI) are currently used for the detection of the tumor microenvironment by PET imaging. This research aimed to design, synthesize and preclinically evaluate a new FAP inhibitor radiopharmaceutical based on the 99mTc-((R)-1-((6-hydrazinylnicotinoyl)-D-alanyl) pyrrolidin-2-yl) boronic acid (99mTc-iFAP) structure for SPECT imaging. Molecular docking for affinity calculations was performed using the AutoDock software. The chemical synthesis was based on a series of coupling reactions of 6-hidrazinylnicotinic acid (HYNIC) and D-alanine to a boronic acid derivative. The iFAP was prepared as a lyophilized formulation based on EDDA/SnCl2 for labeling with 99mTc. The radiochemical purity (R.P.) was verified via ITLC-SG and reversed-phase radio-HPLC. The stability in human serum was evaluated by size-exclusion HPLC. In vitro cell uptake was assessed using N30 stromal endometrial cells (FAP positive) and human fibroblasts (FAP negative). Biodistribution and tumor uptake were determined in Hep-G2 tumor-bearing nude mice, from which images were acquired using a micro-SPECT/CT. The iFAP ligand (Ki = 0.536 nm, AutoDock affinity), characterized by UV-Vis, FT-IR, 1H–NMR and UPLC-mass spectroscopies, was synthesized with a chemical purity of 92%. The 99mTc-iFAP was obtained with a R.P. >98%. In vitro and in vivo studies indicated high radiotracer stability in human serum (>95% at 24 h), specific recognition for FAP, high tumor uptake (7.05 ± 1.13% ID/g at 30 min) and fast kidney elimination. The results found in this research justify additional dosimetric and clinical studies to establish the sensitivity and specificity of the 99mTc-iFAP.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Jae Min Cho ◽  
Eun Hee Yang ◽  
Wenying Quan ◽  
Eun Hye Nam ◽  
Hyae Gyeong Cheon

AbstractFibroblast growth factor (FGF) 21 is a class of hepatokines that plays a protective role against obesity, insulin resistance, and liver damage. Despite this, protective effects of FGF21 in human appear to be minimal, possibly due to its proteolytic cleavage by the fibroblast activation protein (FAP). Here, we presented a novel FAP inhibitor, BR103354, and described its pharmacological activities as a potential therapeutic agent for the treatment of metabolic disorders. BR103354 inhibited FAP with an IC50 value of 14 nM, showing high selectivity against dipeptidyl peptidase (DPP)-related enzymes and prolyl oligopeptidase (PREP). In differentiated 3T3/L1 adipocytes, the addition of FAP diminished hFGF21-induced Glut1 and phosphorylated levels of ERK, which were restored by BR103354. BR103354 exhibited good pharmacokinetic properties as evidenced by oral bioavailability of 48.4% and minimal hERG inhibition. Single co-administration of BR103354 with hFGF21 reduced nonfasting blood glucose concentrations, in association with increased intact form of hFGF21 in ob/ob mice. Additionally, chronic treatment of BR103354 for 4 weeks reduced nonfasting blood glucose concentrations with improved glucose tolerance and with reduced triglyceride (TG) content in liver of ob/ob mice. Consistently, BR103354 improved hepatic steatosis and fibrosis in a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD)-induced non-alcoholic steatohepatitis (NASH) mouse model. FAP inhibitory effects of BR103354 were confirmed in normal cynomolgus monkeys. Together, BR103354 acts as an effective FAP inhibitor in vitro and in vivo, thereby demonstrating its potential application as an anti-diabetic and anti-NASH agent.


2021 ◽  
Vol 118 (16) ◽  
pp. e2101852118
Author(s):  
Jacopo Millul ◽  
Gabriele Bassi ◽  
Jacqueline Mock ◽  
Abdullah Elsayed ◽  
Christian Pellegrino ◽  
...  

We describe the development of OncoFAP, an ultra-high-affinity ligand of fibroblast activation protein (FAP) for targeting applications with pan-tumoral potential. OncoFAP binds to human FAP with affinity in the subnanomolar concentration range and cross-reacts with the murine isoform of the protein. We generated various fluorescent and radiolabeled derivatives of OncoFAP in order to study biodistribution properties and tumor-targeting performance in preclinical models. Fluorescent derivatives selectively localized in FAP-positive tumors implanted in nude mice with a rapid and homogeneous penetration within the neoplastic tissue. Quantitative in vivo biodistribution studies with a lutetium-177–labeled derivative of OncoFAP revealed a preferential localization in tumors at doses of up to 1,000 nmol/kg. More than 30% of the injected dose had already accumulated in 1 g of tumor 10 min after intravenous injection and persisted for at least 3 h with excellent tumor-to-organ ratios. OncoFAP also served as a modular component for the generation of nonradioactive therapeutic products. A fluorescein conjugate mediated a potent and FAP-dependent tumor cell killing activity in combination with chimeric antigen receptor (CAR) T cells specific to fluorescein. Similarly, a conjugate of OncoFAP with the monomethyl auristatin E-based Vedotin payload was well tolerated and cured tumor-bearing mice in combination with a clinical-stage antibody-interleukin-2 fusion. Collectively, these data support the development of OncoFAP-based products for tumor-targeting applications in patients with cancer.


Sign in / Sign up

Export Citation Format

Share Document