1
|
Kuret T, Kreft ME, Veranič P, Čemažar M, Pavlin M, Jerman UD. Phases of tight junction barrier disruption during transurothelial migration of invasive urothelial cancer cells. Sci Rep 2025; 15:12975. [PMID: 40234478 PMCID: PMC12000480 DOI: 10.1038/s41598-025-96267-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
Bladder cancer is characterised by its multifocal nature and a high recurrence, yet the underlying mechanisms of these phenomena remain only partially understood. In the present study, we aimed to investigate transurothelial invasion of urothelial cancer cells as a potential mechanism for dissemination of bladder cancer and to identify the key molecules involved in urothelial barrier disruption. Using confocal and electron microscopy, we were able to show that within a 24-hour timeframe muscle-invasive urothelial cancer cells T24 adhere to the partially differentiated normal urothelial in vitro model and initially cause localised disruption of the tight junctions between urothelial cells. Subsequently, urothelial cells separate and individual T24 cells migrate paracellularly through the urothelium. qPCR analysis identified fibroblast activation protein (FAP)/seprase as the candidate most likely to be involved in urothelial barrier disruption. In addition, treatment of T24 cells with Pefabloc resulted in the inhibition of T24 cell invasion. Our results contribute to the understanding of the mechanisms underlying transurothelial invasion of urothelial cancer cells. Among the molecules tested, FAP/sepraseis likely involved in cancer cell-induced disruption of the urothelial barrier, suggesting its potential as a therapeutic target to prevent progression and recurrence of bladder cancer.
Collapse
Affiliation(s)
- Tadeja Kuret
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urška Dragin Jerman
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Fu H, Huang J, Zhao L, Chen Y, Xu W, Cai J, Yu L, Pang Y, Guo W, Su B, Sun L, Wu H, Zhang J, Chen X, Chen H. 177Lu-LNC1004 Radioligand Therapy in Patients with End-stage Metastatic Cancers: A Single-Center, Single-Arm, Phase II Study. Clin Cancer Res 2025; 31:1415-1426. [PMID: 40084923 PMCID: PMC11995002 DOI: 10.1158/1078-0432.ccr-24-3918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/28/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE Fibroblast activation protein (FAP) is highly expressed in cancer-associated fibroblasts and certain tumor cells, making it a promising therapeutic target for various malignancies. This study evaluated the efficacy and safety of 177Lu-Evans blue-FAP inhibitor (177Lu-LNC1004) radioligand therapy (RLT) for treating end-stage metastatic tumors. PATIENTS AND METHODS This single-arm, single-center, phase II trial included 28 patients with progressive metastatic malignancies (11 types) and high FAP expression (defined as a maximum standardized uptake value ≥10 in >50% of tumors) who had exhausted all approved therapies, screened between June 2022 and April 2024. Patients were scheduled to receive four 177Lu-LNC1004 RLT cycles at 3.33 GBq/cycle every 6 weeks. The primary endpoint was post-RLT radiologic response. The secondary endpoints were progression-free survival (PFS), overall survival (OS), dosimetry, and safety. RESULTS Eastern Cooperative Oncology Group scores >2 were observed in 68% of patients. Overall, 63 177Lu-LNC1004 RLT cycles were performed, with 19 (68%) patients undergoing ≥2 cycles. Disease control was achieved in 13 (13/28, 46%) patients, with 4 and 9 patients demonstrating partial response and stable disease, respectively, and associated with improved PFS and OS (P < 0.001). The mean absorbed dose in tumors was 4.69 ± 3.83 Gy/GBq (1.18-25.03 Gy/GBq). Treatment-related grade 3/4 hematotoxicity was observed in six (21%) patients, with thrombocytopenia, leukopenia, and neutropenia most prevalent. No grade 3/4 hepatotoxicity or nephrotoxicity was observed. CONCLUSIONS FAP-directed RLT using 177Lu-LNC1004 at 3.33 GBq/cycle was well tolerated with an acceptable toxicity profile. Nearly half of patients achieved disease control, which was associated with prolonged PFS and OS.
Collapse
Affiliation(s)
- Hao Fu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingxiong Huang
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Liang Zhao
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuhang Chen
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Weizhi Xu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiayu Cai
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lingyu Yu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yizhen Pang
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Guo
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bishan Su
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Long Sun
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hua Wu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingjing Zhang
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaoyuan Chen
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Haojun Chen
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
3
|
Saito K, Watanabe H, Nakashima K, Ono M. Preclinical Characterization of Novel FAP-2286-Based Radioligand with Albumin Binder for Improved Tumor Retention. ACS Med Chem Lett 2025; 16:596-601. [PMID: 40236533 PMCID: PMC11995215 DOI: 10.1021/acsmedchemlett.4c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Fibroblast activation protein (FAP) is an attractive biomarker for tumor-targeting agents in cancer diagnosis and therapy. FAP-2286 shows retention in FAP-expressing tumors and is known as a promising FAP-targeting radioligand. In this study, we aimed to develop a FAP-2286 derivative that demonstrates higher tumor retention than FAP-2286. We designed DOTAGA-FAP-2286 and DOTAGA-FAP-2286-ALB by replacing DO3A with 2-(4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)pentanedioic acid (DOTAGA) and introducing an albumin binder. Both compounds were successfully radiolabeled with 111In. Compared with [111In]In-DOTAGA-FAP-2286, [111In]In-DOTAGA-FAP-2286-ALB showed higher stability in murine plasma. In the cell competition binding study, In-DOTAGA-FAP-2286-ALB exhibited a higher FAP-binding affinity than In-DOTAGA-FAP-2286. In the albumin-binding assay, [111In]In-DOTAGA-FAP-2286-ALB showed a high binding rate in the solution with albumin. The biodistribution assay revealed marked tumor retention of [111In]In-DOTAGA-FAP-2286-ALB, resulting in the enhancement of predicted tumor AUC values of [225Ac]Ac-DOTAGA-FAP-2286-ALB. These results suggest advantages of the introduction of an albumin binder to FAP-2286.
Collapse
Affiliation(s)
- Kosuke Saito
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-Ku, Kyoto 606-8501, Japan
| |
Collapse
|
4
|
Meng L, Lin R, Zhang J, Li H, Xia D, Zhao Z, Zhuang R, Huang L, Zhang X, Fang J, Miao W, Guo Z. Modification of Asp-Peptide Adapters: Giving the FAP-Targeted Radioligand a "Squirrel Tail". J Med Chem 2025; 68:6576-6587. [PMID: 40102034 DOI: 10.1021/acs.jmedchem.4c03133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Fibroblast activation protein (FAP) is a promising target for cancer theranostics, but most FAP-targeted radioprobes showed relatively insufficient tumor uptake and retention, which seriously hampered their further application. Inspired by the squirrel tail, this study developed a novel FAP-targeted molecule, FSND3, which is modified with three Asp-peptide adapters to enable both 68Ga ([68Ga]Ga-FSND3) and 18F ([18F]AlF-FSND3) PET imaging. Compared to [68Ga]Ga-FAPI-04, [68Ga]Ga-FAPI-42, and [18F]AlF-FAPI-42, [18F]AlF-FSND3 and [68Ga]Ga-FSND3 showed enhanced tumor uptake and prolonged residence in HT-1080-FAP and pancreatic tumor models, demonstrating the effectiveness of Asp-peptide adapters in pharmacomodulating FAP-targeted radioligands. The first-in-human pilot study revealed that [18F]AlF- and [68Ga]Ga-FSND3 exhibited comparable uptake in the primary lesion, higher-contrast images, and higher uptake in some metastases like in bone and brain, to 2-[18F] FDG PET/CT imaging. As a proof of concept, these results offer a significant enhancement to the diversity of the FAP-targeted tracer arsenal.
Collapse
Affiliation(s)
- Lingxin Meng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Rong Lin
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, China
| | - Jingru Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Huifeng Li
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Dongsheng Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Zuoquan Zhao
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Rongqiang Zhuang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Lumei Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xianzhong Zhang
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Jianyang Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Weibing Miao
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, China
| | - Zhide Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| |
Collapse
|
5
|
Faucher FF, Lovell S, Bertolini M, Blažková K, Cosco ED, Bogyo M, Barniol-Xicota M. Macrocyclic phage display for identification of selective protease substrates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643185. [PMID: 40161698 PMCID: PMC11952493 DOI: 10.1101/2025.03.13.643185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Traditional methods for identifying selective protease substrates have primarily relied on synthetic libraries of linear peptides, which offer limited sequence and structural diversity. Here, we present an approach that leverages phage display technology to screen large libraries of chemically modified cyclic peptides, enabling the identification of highly selective substrates for a protease of interest. Our method uses a reactive chemical linker to cyclize peptides on the phage surface, while simultaneously incorporating an affinity tag and a fluorescent reporter. The affinity tag enables capture of the phage library and subsequent release of phages expressing optimal substrates upon incubation with a protease of interest. The addition of a turn-on fluorescent reporter allows direct quantification of cleavage efficiency throughout each selection round. The resulting identified substrates can then be chemically synthesized, optimized and validated using recombinant enzymes and cells. We demonstrate the utility of this approach using Fibroblast Activation Protein alpha (FAPα) and the related proline-specific protease, dipeptidyl peptidase-4 (DPP4), as targets. Phage selection and subsequent optimization identified substrates with selectivity for each target that have the potential to serve as valuable tools for applications in basic biology and fluorescence image-guided surgery (FIGS). Overall, our strategy provides a rapid and unbiased platform for effectively discovering highly selective, non-natural protease substrates, overcoming key limitations of existing methods.
Collapse
Affiliation(s)
- Franco F. Faucher
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Scott Lovell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Life Sciences, University of Bath, Bath, BA2 7AX, United Kingdom
| | - Matilde Bertolini
- Department of Genetics, School of Medicine, Stanford University, California 94305, United States
| | - Kristýna Blažková
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Emily D. Cosco
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Marta Barniol-Xicota
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Medicine and Life Sciences, Biomedical Research Park (PRBB), Universitat Pompeu Fabra, 08003 Barcelona
| |
Collapse
|
6
|
Rizzo R, Capozza M, Conti L, Avalle L, Poli V, Terreno E. Novel FAP-Targeted Heptamethine Cyanines for NIRF Imaging Applications. Mol Pharm 2025; 22:1518-1528. [PMID: 39954291 PMCID: PMC11881144 DOI: 10.1021/acs.molpharmaceut.4c01232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Fibroblast activation protein (FAP) is a pan-cancer target that is useful for imaging, ideally all epithelial cancers. This work aimed to develop, characterize, and validate two novel FAP-targeted probes for optical imaging, both in vitro and in vivo. IRDye800CW and FNIRTag heptamethine cyanines were conjugated to the NH precursor of the well-known FAP inhibitor FAPI-46, which is widely employed in nuclear medicine. In addition to synthesis, the dyes were characterized in terms of physicochemical properties, biodistribution, and imaging performances in a breast cancer tumor model. FAPI-FNIRTag showed a stronger fluorescence and higher photostability compared to FAPI-IRDye800CW. Notably, both compounds exhibited strong tumor accumulation in TUBO breast cancer-bearing mice 24 h postadministration, suggesting potential for further investigation as fluorescence-guided surgery (FGS) agents.
Collapse
Affiliation(s)
- Rebecca Rizzo
- Department
of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44/bis, Turin 10126, Italy
| | - Martina Capozza
- Department
of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44/bis, Turin 10126, Italy
| | - Laura Conti
- Department
of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44/bis, Turin 10126, Italy
| | - Lidia Avalle
- DISIT, University of Eastern Piedmont, Viale Teresa Michel 11, Alessandria 15121, Italy
| | - Valeria Poli
- Department
of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin 10126, Italy
| | - Enzo Terreno
- Department
of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44/bis, Turin 10126, Italy
| |
Collapse
|
7
|
Qin Y, Miyake T, Muramoto K, Maekawa T, Nishina Y, Wang Y, Shimizu T, Tani M. Fibroblast Activation Protein-α Expression in Cancer-Associated Fibroblasts Shows the Poor Survival of Colorectal Cancer via Immune-Mediated Pathways : Implications of FAP in Cancer-Associated Fibroblasts Link Immune Dysregulation to Adverse Survival in Colorectal Cancer. Ann Surg Oncol 2025; 32:1941-1952. [PMID: 39623187 DOI: 10.1245/s10434-024-16593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/12/2024] [Indexed: 02/12/2025]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) and immune cells, the key components of the tumor microenvironment (TME), play critical roles in oncogenesis. Despite the recognized function of fibroblast activation protein-α (FAP), a specific biomarker of CAFs in cancer progression, its role in the survival of patients with colorectal cancer (CRC) and tumor immune microenvironment (TIME) remains unclear. METHODS We investigated 180 pathological sections obtained from 178 consecutive patients with CRC who underwent surgical resection at Shiga University of Medical Science Hospital between January 2013 and December 2015. FAP expression levels and CD3 and CD8 densities at the invasive margin and center of tumor were assessed using immunohistochemical (IHC) staining. Furthermore, we used single-cell RNA sequencing (scRNA-seq) of CAFs in a separate cohort of 10 untreated patients with CRC derived from the Gene Expression Omnibus database. RESULTS According to IHC evaluation, high FAP expression in patients with CRC showed a correlation with reduced tumor-infiltrating lymphocyte (TIL) distribution and poor survival. Based on the FAP transcription levels obtained through scRNA-seq analysis, CAFs were grouped into high and low FAP expression groups. Elevated FAP expression was correlated with decreased expression of T- and B-cell biomarkers, suggesting an association with an immunosuppressive TME promotion. Several genes associated with cancer-related immune-mediated pathways (CXCL12, COL11A1, CCL11, and COL10A1) were significantly upregulated in FAP-positive CAFs. CONCLUSIONS This study highlights the effects of FAP expression on survival of patients with CRC, its interaction with TILs, and relevant signaling pathways, and underscores potential immunotherapeutic targets for future investigation.
Collapse
Affiliation(s)
- Yubo Qin
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
- Department of Emergency Center, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Toru Miyake
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan.
| | - Keiji Muramoto
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Takeru Maekawa
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Yusuke Nishina
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Ying Wang
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Tomoharu Shimizu
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Masaji Tani
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
8
|
Gao H, Qu L, Li M, Guan X, Zhang S, Deng X, Wang J, Xing F. Unlocking the potential of chimeric antigen receptor T cell engineering immunotherapy: Long road to achieve precise targeted therapy for hepatobiliary pancreatic cancers. Int J Biol Macromol 2025; 297:139829. [PMID: 39814310 DOI: 10.1016/j.ijbiomac.2025.139829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/03/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Innovative therapeutic strategies are urgently needed to address the ongoing global health concern of hepatobiliary pancreatic malignancies. This review summarizes the latest and most comprehensive research of chimeric antigen receptor (CAR-T) cell engineering immunotherapy for treating hepatobiliary pancreatic cancers. Commencing with an exploration of the distinct anatomical location and the immunosuppressive, hypoxic tumor microenvironment (TME), this review critically assesses the limitations of current CAR-T therapy in hepatobiliary pancreatic cancers and proposes corresponding solutions. Various studies aim at enhancing CAR-T cell efficacy in these cancers through improving T cell persistence, enhancing antigen specificity and reducing tumor heterogeneity, also modulating the immunosuppressive and hypoxic TME. Additionally, the review examines the application of emerging nanoparticles and biotechnologies utilized in CAR-T therapy for these cancers. The results suggest that constructing optimized CAR-T cells to overcome physical barrier, manipulating the TME to relieve immunosuppression and hypoxia, designing CAR-T combination therapies, and selecting the most suitable delivery strategies, all together could collectively enhance the safety of CAR-T engineering and advance the effectiveness of adaptive cell therapy for hepatobiliary pancreatic cancers.
Collapse
Affiliation(s)
- Hongli Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Lianyue Qu
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001, China
| | - Mu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xin Guan
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shuang Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xin Deng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jin Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Fei Xing
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
9
|
Kumar BV, Sachan R, Garad P, Srivastava N, Saraf SA, Meher N. Dual Targeting of Prostate-Specific Membrane Antigen and Fibroblast Activation Protein: Bridging Prostate Cancer Theranostics with Precision. ACS APPLIED BIO MATERIALS 2025; 8:962-979. [PMID: 39895503 DOI: 10.1021/acsabm.4c01914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Targeting Prostate Specific Membrane Antigen (PSMA) has proven highly useful and beneficial for prostate cancer (PCa) theranostics. However, patients with advanced metastatic castration-resistant prostate cancer (mCRPC) lack optimal PSMA expression resulting in poor specificity. To address this limitation, combination targeting is gaining popularity by synergistically boosting the theranostic efficacy. Herein, we thoroughly reviewed the most recent development of drug formulation for PCa theranostics by targeting both PSMA and Fibroblast Activation Protein (FAP). FAP is known to overexpress in cancer-associated fibroblasts (CAFs) within the tumor microenvironment (TME). It has been extensively studied as an effective target for the identification and treatment of a variety of cancer phenotypes. Along with the advantages and current updates on combination targeting of PSMA and FAP, this Review thoroughly discussed the expression patterns of PSMA and FAP in various cancer phenotypes, as well as their role in tumor growth, invasion, and metastasis, which is of great interest in the design and development of prostate cancer theranostics.
Collapse
Affiliation(s)
- Boga Vijay Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Riya Sachan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Prajakta Garad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Shubhini A Saraf
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Niranjan Meher
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| |
Collapse
|
10
|
Zhang X, Lee KC, Choi JY, Lee KH, Choe YS. Comparative Study of Dimeric Fibroblast Activation Protein-Targeting Radioligands Labeled with Fluorine-18, Copper-64, and Gallium-68. Mol Pharm 2025; 22:906-917. [PMID: 39736080 DOI: 10.1021/acs.molpharmaceut.4c01080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025]
Abstract
Fibroblast activation protein inhibitors (FAPIs) labeled with gallium-68 and lutetium-177 show potential for use in the diagnosis and treatment of various cancers expressing FAP. However, 177Lu-labeled FAPIs often exhibit short tumor retention time, limiting their therapeutic applications. To improve tumor retention, we synthesized three radiolabeled dimeric FAPIs, [18F]1, [64Cu]2, and [68Ga]3. These were prepared by chelating Al[18F]F to 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA)-l-glutamic acid (E)-(FAPI)2 and copper-64 or gallium-68 to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-E-(FAPI)2. NOTA-E-(FAPI)2 and DOTA-E-(FAPI)2 showed higher binding affinities for FAP compared with that of FAPI-04 (IC50 = 0.47 and 0.16 nM vs 0.89 nM, respectively). All radioligands were synthesized in high decay-corrected radiochemical yields (59-96%) and were stable in fetal bovine serum and phosphate-buffered saline. The more hydrophilic radioligand, [68Ga]3, was selected for cellular uptake studies, which confirmed FAP-specific uptake. Positron emission tomography imaging and ex vivo biodistribution studies in U87MG tumor-bearing mice revealed high tumor uptake of all three radioligands, with significant blocking observed after preinjection of FAPI-04. [64Cu]2 and [68Ga]3 exhibited favorable in vivo pharmacokinetics compared to those of [18F]1. Notably, [68Ga]3 showed lower normal organ uptake than did the other two radioligands, and moreover, it exhibited higher, more prolonged tumor uptake than its monomeric counterpart [68Ga]Ga-FAPI-04 over a 3 h period, suggesting its potential as a promising FAP-specific theranostic radioligand.
Collapse
Affiliation(s)
- Xuran Zhang
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Yearn Seong Choe
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| |
Collapse
|
11
|
Aksoy SA, Earl J, Grahovac J, Karakas D, Lencioni G, Sığırlı S, Bijlsma MF. Organoids, tissue slices and organotypic cultures: Advancing our understanding of pancreatic ductal adenocarcinoma through in vitro and ex vivo models. Semin Cancer Biol 2025; 109:10-24. [PMID: 39730107 DOI: 10.1016/j.semcancer.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst prognoses of all common solid cancers. For the large majority of PDAC patients, only systemic therapies with very limited efficacy are indicated. In addition, immunotherapies have not brought the advances seen in other cancer types. Several key characteristics of PDAC contribute to poor treatment outcomes, and in this review, we will discuss how these characteristics are best captured in currently available ex vivo or in vitro model systems. For instance, PDAC is hallmarked by a highly desmoplastic and immune-suppressed tumor microenvironment that impacts disease progression and therapy resistance. Also, large differences in tumor biology exist between and within tumors, complicating treatment decisions. Furthermore, PDAC has a very high propensity for locally invasive and metastatic growth. The use of animal models is often not desirable or feasible and several in vitro and ex vivo model systems have been developed, such as organotypic cocultures and tissue slices, among others. However, the absence of a full host organism impacts the ability of these models to accurately capture the characteristics that contribute to poor outcomes in PDAC. We will discuss the caveats and advantages of these model systems in the context of PDAC's key characteristics and provide recommendations on model choice and the possibilities for optimization. These considerations should be of use to researchers aiming to study PDAC in the in vitro setting.
Collapse
Affiliation(s)
- Secil Ak Aksoy
- Bursa Uludag University, Faculty of Medicine, Department of Medical Microbiology, Bursa, Turkey
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Biomodels and Biomodels Platform Hospital Ramón y Cajal-IRYCIS, Carretera Colmenar Km 9,100, Madrid 28034, Spain; The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid 28029, Spain
| | - Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Didem Karakas
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Giulia Lencioni
- Department of Biology, University of Pisa, Pisa, Italy; Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Sıla Sığırlı
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| |
Collapse
|
12
|
Gunaratne GS, Gallant JP, Ott KL, Broome PL, Celada S, West JL, Mixdorf JC, Aluicio-Sarduy E, Engle JW, Boros E, Meimetis L, Lang JM, Zhao SG, Hernandez R, Kosoff D, LeBeau AM. Development of FAP-targeted theranostics discovered by next-generation sequencing-augmented mining of a novel immunized VNAR library. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632555. [PMID: 39868181 PMCID: PMC11761682 DOI: 10.1101/2025.01.13.632555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Cancer-associated fibroblasts (CAFs) in the stroma of solid tumors promote an immunosuppressive tumor microenvironment (TME) that drives resistance to therapies. The expression of the protease fibroblast activation protein (FAP) on the surface of CAFs has made FAP a target for development of therapies to dampen immunosuppression. Relatively few biologics have been developed for FAP and none have been developed that exploit the unique engagement properties of Variable New Antigen Receptors (VNARs) from shark antibodies. As the smallest binding domain in nature, VNARs cleverage unique geometries and recognize epitopes conventional antibodies cannot. By directly immunizing a nurse shark with FAP, we created a large anti-FAP VNAR phage display library. This library allowed us to identify a suite of anti-FAP VNARs through traditional biopanning and also by an in silico approach that did not require any prior affinity-based enrichment in vitro. We investigated four VNAR-Fc fusion proteins for theranostic properties and found that all four recognized FAP with high affinity and were rapidly internalized by FAP-positive cells. As a result, the VNAR-Fc constructs were effective antibody-drug conjugates in vitro and were able to localize to FAP-positive xenografts in vivo. Our findings establish VNAR-Fc constructs as a versatile platform for theranostic development that could yield innovative cancer therapies targeting the TME.
Collapse
Affiliation(s)
- Gihan S. Gunaratne
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Joseph P. Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kendahl L. Ott
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Payson L. Broome
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Sasha Celada
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cellular and Molecular Pathology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Jayden L. West
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Jason C. Mixdorf
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Eszter Boros
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Labros Meimetis
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Shuang G. Zhao
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Reinier Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S Middleton Memorial Veterans’ Hospital, Madison, Wisconsin
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
13
|
Butuzova DA, Kulebyakina MA, Basalova NA, Efimenko AY. Fibroblast Activation Protein Alpha (FAPα) as a Promising Target in the Diagnostics and Treatment of Cancer and Fibrotic Diseases: Recent Approaches to Imaging and Assessment of Functional Activity. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S135-S145. [PMID: 40164156 DOI: 10.1134/s000629792460279x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/16/2024] [Accepted: 06/19/2024] [Indexed: 04/02/2025]
Abstract
Fibroblast activation protein alpha (FAPα) is a transmembrane serine peptidase and a well-known marker of activated fibroblasts that are formed during onco- and fibrogenesis and play an important role in the progression of cancer and fibrosis. Identification of FAPα-positive cells is widely used to visualize pathological changes in the stroma in the diagnosis and treatment of cancer diseases. Recent evidence suggests that FAPα itself contributes to the development of tumors and fibrosis-associated diseases through its enzymatic activity and other mechanisms. Various methods for visualization and evaluation of FAPα enzymatic activity are being developed, which are essential for deciphering the role of FAPα in the development of stromal pathologies. Here we discuss current approaches to visualization and regulation of FAPα enzymatic activity.
Collapse
Affiliation(s)
- Daria A Butuzova
- Medical Research and Educational Institute, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Maria A Kulebyakina
- Medical Research and Educational Institute, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nataliya A Basalova
- Medical Research and Educational Institute, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Anastasia Yu Efimenko
- Medical Research and Educational Institute, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
14
|
Tian H, Wang W, Liang S, Ding J, Hua D. From darkness to light: Targeting CAFs as a new potential strategy for cancer treatment. Int Immunopharmacol 2024; 143:113482. [PMID: 39476569 DOI: 10.1016/j.intimp.2024.113482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024]
Abstract
Cancer-associated fibroblasts (CAFs), which are the most frequent stromal cells in the tumor microenvironment (TME), play a key role in the metastasis of tumor cells. Generally speaking, CAFs in cooperation with tumor cells can secrete various cytokines, proteins, growth factors, and metabolites to promote angiogenesis, mediate immune escape of tumor cells, enhance endothelial-to-mesenchymal transition, stimulate extracellular matrix remodeling, and preserve tumor cell stemness. These activities of CAFs provide a favorable exogenous pathway for tumor progression and metastasis, and a microenvironment that allows rapid growth of tumor cells, which always lead to poor prognosis for patients. More importantly, it seems that targeting CAFs is also a potential precision therapeutic strategy in clinical practice. Hence, this review outlines the origin of CAFs, the relationship between CAFs and cancer metastasis, and targeting CAFs as a potential strategy for cancer patients, which could give some inspirations for cancer treatment in clinic.
Collapse
Affiliation(s)
- Haixia Tian
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Weijing Wang
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Shuai Liang
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Junli Ding
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| | - Dong Hua
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
15
|
van der Heide CD, Campeiro JD, Ruigrok EAM, van den Brink L, Ponnala S, Hillier SM, Dalm SU. In vitro and ex vivo evaluation of preclinical models for FAP-targeted theranostics: differences and relevance for radiotracer evaluation. EJNMMI Res 2024; 14:125. [PMID: 39718718 DOI: 10.1186/s13550-024-01191-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Fibroblast activation protein (FAP) is an attractive target for cancer theranostics. Although FAP-targeted nuclear imaging demonstrated promising clinical results, only sub-optimal results are reported for targeted radionuclide therapy (TRT). Preclinical research is crucial in selecting promising FAP-targeted radiopharmaceuticals and for obtaining an increased understanding of factors essential for FAP-TRT improvement. FAP is mainly expressed by cancer-associated fibroblasts in the tumor stroma and less on cancer cells themselves. Therefore, other (complex) factors impact FAP-TRT efficacy compared to currently clinically applied TRT strategies. For accurate evaluation of these aspects, selection of a representative preclinical model is important. Currently mainly human cancer cell lines transduced to (over)express FAP are applied, lacking clinical representation. It is unclear how these and more physiological FAP-expressing models compare to each other, and whether/how the model influences the study outcome. We aimed to address this by comparing FAP tracer behavior in FAP-transduced HT1080-huFAP and HEK293-huFAP cells, and endogenous FAP-expressing U-87 MG cancer cells and PS-1 pancreatic stellate cells. [111In]In-FAPI-46 and a fluorescent FAP-targeted tracer (RTX-1370S) were used to compare tracer binding/uptake and localization in vitro and ex vivo. Additionally, FAP expression was determined with RT-qPCR and anti-FAP IHC. RESULTS Although FAP expression was highest in HEK293-huFAP cells and cell line derived xenografts, this did not result in the highest tracer uptake. [111In]In-FAPI-46 uptake was highest in HT1080-huFAP, closely followed by HEK293-huFAP, and a 6-10-fold lower uptake for U-87 MG and PS-1 cells. However, ex vivo U-87 MG xenografts only showed a 2-fold lower binding compared to HT1080-huFAP and HEK293-huFAP xenografts, mainly because the cell line attracts murine fibroblasts as demonstrated in our RT-qPCR and IHC studies. CONCLUSIONS The interaction between FAP and FAP-targeted tracers differs between models, indicating the need for appropriate model selection and that comparing results across studies using different models is difficult.
Collapse
Affiliation(s)
- Circe D van der Heide
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | - Joana D Campeiro
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | - Eline A M Ruigrok
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | - Lilian van den Brink
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | | | | | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands.
| |
Collapse
|
16
|
Thapa P, Debnath S, Bedi A, Parashar M, Gonzalez P, Reus J, Hammers H, Sun X. Monovalent and Divalent Designs of Copper Radiotheranostics Targeting Fibroblast Activation Protein in Cancer. Cancers (Basel) 2024; 16:4180. [PMID: 39766079 PMCID: PMC11675001 DOI: 10.3390/cancers16244180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Fibroblast activation protein (FAP)-targeted theranostic radiopharmaceuticals have shown desired tumor-to-background organ selectivity due to the ubiquitous presence of FAP within the tumor microenvironment. However, suboptimal tumor retention and fast clearance have hindered their use to deliver effective cancer therapies. With well-documented FAP-targeting moieties and linkers appending them to optimal chelators, the development of copper radiopharmaceuticals has attracted considerable interest, given the fact that an ideal theranostic pair of copper radionuclides (64Cu: t1/2 = 12.7 h; 17.4% β+; Eβ+max = 653 keV and 67Cu: t1/2 = 2.58 d; 100% β-; Eβ-max = 562 keV) are available. Herein, we report our design, synthesis, and comparative evaluation of monovalent and divalent FAP-targeted theranostic conjugates constructed from our previously reported bifunctional chelator scaffold (BFS) based on 1,4,8,11-tetraaza-bicyclo [6.6.2]hexadecane-4,11-diacetic acid (CB-TE2A), which forms the most stable complex with Cu(II). Methods: After synthesis and characterization, the monovalent and divalent conjugates were radiolabeled with 64Cu for in vitro cell assays, followed by in vivo positron emission tomography (PET) imaging evaluation in relevant mouse models. Results: Both 64Cu-labeled conjugates showed high in vitro stability and anticipated FAP-mediated cell binding and internalization. The divalent one showed significantly higher FAP-specific tumor uptake than its monovalent counterpart. Conclusions: Our results demonstrate that the BFS-based multivalent approach can be practically used to generate FAP-targeted radiotheranostic agents for effective cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Pawan Thapa
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.T.); (S.D.); (A.B.); (P.G.)
| | - Sashi Debnath
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.T.); (S.D.); (A.B.); (P.G.)
| | - Anjan Bedi
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.T.); (S.D.); (A.B.); (P.G.)
| | - Madhuri Parashar
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.P.); (J.R.); (H.H.)
| | - Paulina Gonzalez
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.T.); (S.D.); (A.B.); (P.G.)
| | - Joshua Reus
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.P.); (J.R.); (H.H.)
| | - Hans Hammers
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.P.); (J.R.); (H.H.)
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.T.); (S.D.); (A.B.); (P.G.)
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
17
|
Chan HW, Kuo DY, Shueng PW, Chuang HY. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals (Basel) 2024; 17:1663. [PMID: 39770505 PMCID: PMC11676442 DOI: 10.3390/ph17121663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor microenvironment (TME) is a critical factor in cancer progression, driving tumor growth, immune evasion, therapeutic resistance, and metastasis. Understanding the dynamic interactions within the TME is essential for advancing cancer management. Molecular imaging provides a non-invasive, real-time, and longitudinal approach to studying the TME, with techniques such as positron emission tomography (PET), magnetic resonance imaging (MRI), and fluorescence imaging offering complementary strengths, including high sensitivity, spatial resolution, and intraoperative precision. Recent advances in imaging probe development have enhanced the ability to target and monitor specific components of the TME, facilitating early cancer diagnosis, therapeutic monitoring, and deeper insights into tumor biology. By integrating these innovations, molecular imaging offers transformative potential for precision oncology, improving diagnostic accuracy and treatment outcomes through a comprehensive assessment of TME dynamics.
Collapse
Affiliation(s)
- Hui-Wen Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| |
Collapse
|
18
|
Geils C, Kathrein K. Augmentation of Solid Tumor Immunotherapy With IL-12. J Gene Med 2024; 26:e70000. [PMID: 39618102 PMCID: PMC11609498 DOI: 10.1002/jgm.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/15/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024] Open
Abstract
Immunotherapy describes a class of therapies in which the immune system is manipulated for therapeutic benefit. These treatments include immune checkpoint inhibitors, adoptive cell therapy, and vaccines. For many hematological malignancies, immunotherapy has emerged as an essential treatment component. However, this success has yet to be replicated for solid tumors, which develop advanced physical and molecular mechanisms for suppressing and evading immune destruction. Nevertheless, cytokine immunotherapy presents a potential remedy to these barriers by delivering a proinflammatory immune signal to the tumor and thereby transforming it from immunologically "cold" to "hot." Interleukin-12 (IL-12), one of the most potent proinflammatory cytokines, was initially investigated for this purpose. However, initial murine and human studies in which IL-12 was administered systemically resulted in dangerous immunotoxicity associated with off-target immune activation. As a result, recent studies have employed advanced cell and molecular engineering approaches to reduce IL-12 toxicity while increasing or maintaining its efficacy such that its effective doses can be tolerated in humans. This review highlights such developments and identifies promising future directions.
Collapse
Affiliation(s)
- Christian Geils
- Department of Biological SciencesUniversity of South CarolinaColumbiaSouth CarolinaUSA
| | - Katie L. Kathrein
- Department of Biological SciencesUniversity of South CarolinaColumbiaSouth CarolinaUSA
| |
Collapse
|
19
|
Şahin ÖF, Alçın G, Ergül N, Çermik TF, Arslan E. Physiological Gallbladder Accumulation on 68 Ga-FAPI-46 PET/CT. Clin Nucl Med 2024; 49:e685-e686. [PMID: 39466623 DOI: 10.1097/rlu.0000000000005480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
ABSTRACT Radiolabeled fibroblast activation protein inhibitors (FAPIs) are a novel approach in cancer detection and treatment. Targeting fibroblast activation protein, extensively produced by cancer-associated fibroblasts, FAPI plays a key role in tumor development. Its advanced imaging capabilities offer clearer results in solid tumors compared with traditional methods, drawing significant interest in oncology. Additionally, FAPI's activity in conditions requiring tissue remodeling, such as atherosclerosis and arthritis, highlights its broader potential. This interesting image shows the physiological accumulation of 68 Ga-FAPI in the gallbladder, emphasizing the importance of accurate interpretation to prevent misdiagnoses and ensure effective patient management.
Collapse
Affiliation(s)
- Ömer Faruk Şahin
- From the Clinic of Nuclear Medicine, Istanbul Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
20
|
Rodponthukwaji K, Thongchot S, Deureh S, Thongkleang T, Thaweesuvannasak M, Srichan K, Srisawat C, Thuwajit P, Nguyen KT, Tadpetch K, Thuwajit C, Punnakitikashem P. Development of cancer-associated fibroblasts-targeting polymeric nanoparticles loaded with 8- O-methylfusarubin for breast cancer treatment. Int J Pharm X 2024; 8:100294. [PMID: 39507587 PMCID: PMC11539325 DOI: 10.1016/j.ijpx.2024.100294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are abundant stromal cells residing in a tumor microenvironment (TME) which are associated with the progression of tumor. Herein, we developed novel CAFs-targeting polymeric nanoparticles encapsulating a synthetic 8-O-methylfusarubin (OMF) compound (OMF@NPs-anti-FAP). Anti-FAP/fibroblast activation protein antibody was employed as a CAFs-targeting ligand. The physicochemical properties of the synthesized nanomaterials were firstly investigated with various techniques. The cytocompatibility of polymeric nanoparticles (NPs) was elicited through cell viability of CAFs and human breast epithelial cells, MCF-10A. Additionally, the anti-FAP-conjugated NPs displayed different degrees of cellular internalization regarding the FAP expression level on the CAFs' surface. However, CAFs exposed to NPs containing OMF demonstrated significant cell death which were associated with the apoptotic pathway as confirmed by caspase-3/7 activity. Upon OMF@NPs-anti-FAP treatment, an enhanced toxicity was clearly observed in 3D spheroid models. High FAP-expressed PC-B-132CAFs demonstrated a high percentage of cell death compared to other cells with a low level of FAP expression analyzed by flow cytometry (e.g. MCF-10A, HDFa, and PC-B-142CAFs). This result emphasized the importance of anti-FAP antibody as a targeting ligand. These findings suggest that the fabricated nanosystem of OMF-loaded polymeric NPs with CAFs' high specificity holds a potential NP-based platform for improvement in breast cancer treatment.
Collapse
Affiliation(s)
- Kamonlatth Rodponthukwaji
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suttikiat Deureh
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Tanva Thongkleang
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Mattika Thaweesuvannasak
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kornrawee Srichan
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Kwanruthai Tadpetch
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
21
|
Wenta T, Nastaly P, Lipinska B, Manninen A. Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol 2024; 134:197-219. [PMID: 39500383 DOI: 10.1016/j.matbio.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024]
Abstract
The extracellular matrix (ECM) serves as a physical scaffold for tissues that is composed of structural proteins such as laminins, collagens, proteoglycans and fibronectin, forming a three dimensional network, and a wide variety of other matrix proteins with ECM-remodeling and signaling functions. The activity of ECM-associated signaling proteins is tightly regulated. Thus, the ECM serves as a reservoir for water and growth regulatory signals. The ECM architecture is dynamically modulated by multiple serine proteases that process both structural and signaling proteins to regulate physiological processes such as organogenesis and tissue homeostasis but they also contribute to pathological events, especially cancer progression. Here, we review the current literature regarding the role of ECM remodeling by serine proteases (KLKs, uPA, furin, HtrAs, granzymes, matriptase, hepsin) in tumorigenesis.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| | - Paulina Nastaly
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
22
|
Fouillet J, Torchio J, Rubira L, Fersing C. Unveiling the Tumor Microenvironment Through Fibroblast Activation Protein Targeting in Diagnostic Nuclear Medicine: A Didactic Review on Biological Rationales and Key Imaging Agents. BIOLOGY 2024; 13:967. [PMID: 39765634 PMCID: PMC11673949 DOI: 10.3390/biology13120967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/03/2025]
Abstract
The tumor microenvironment (TME) is a dynamic and complex medium that plays a central role in cancer progression, metastasis, and treatment resistance. Among the key elements of the TME, cancer-associated fibroblasts (CAFs) are particularly important for their ability to remodel the extracellular matrix, promote angiogenesis, and suppress anti-tumor immune responses. Fibroblast activation protein (FAP), predominantly expressed by CAFs, has emerged as a promising target in both cancer diagnostics and therapeutics. In nuclear medicine, targeting FAP offers new opportunities for non-invasive imaging using radiolabeled fibroblast activation protein inhibitors (FAPIs). These FAP-specific radiotracers have demonstrated excellent tumor detection properties compared to traditional radiopharmaceuticals such as [18F]FDG, especially in cancers with low metabolic activity, like liver and biliary tract tumors. The most recent FAPI derivatives not only enhance the accuracy of positron emission tomography (PET) imaging but also hold potential for theranostic applications by delivering targeted radionuclide therapies. This review examines the biological underpinnings of FAP in the TME, the design of FAPI-based imaging agents, and their evolving role in cancer diagnostics, highlighting the potential of FAP as a target for precision oncology.
Collapse
Affiliation(s)
- Juliette Fouillet
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
| | - Jade Torchio
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
| | - Léa Rubira
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
| | - Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| |
Collapse
|
23
|
Ashok G, AlAsmari AF, AlAsmari F, Livingstone P, Anbarasu A, Ramaiah S. Transcriptomic, mutational and structural bioinformatics approaches to explore the therapeutic role of FAP in predominant cancer types. Discov Oncol 2024; 15:699. [PMID: 39579201 PMCID: PMC11585531 DOI: 10.1007/s12672-024-01531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/05/2024] [Indexed: 11/25/2024] Open
Abstract
Fibroblast activating protein (FAP) is a cell surface marker of cancer-associated fibroblasts with a distinct pro-tumorigenic role. The present study analyzed the pan-cancer expression; and clinical and mutational profiles of the FAP coding gene. Molecular dynamics simulation (MDS) deciphered the backbone dynamics and energetics of FAP. Virtual screening and subsequent pharmacokinetic-profiling (PK) filtered lead molecules, which were subjected to molecular docking. MDS projected a stable trajectory for the protein, as dynamics evidenced by low residue-level fluctuations, stable backbone dynamics, and energetics. Around five stabilization and deleterious mutations in the catalytic domain were identified. The low binding energy (BE) profiles from molecular docking studies screened the top five lead molecules for site-specific intermolecular interaction studies. Lead-16 (ZINC000245289699) exhibited a significant BE and inhibition constant of -6.87 kcal/mol and 12.27 μM, respectively, across FAP and its mutants. Interestingly, the docked complexes of Lead-16 interacted with the catalytic triad residues (S624, D702, and H734). The docked complexes of Lead-16 with FAP showed lower average root-mean-square fluctuations compared to the unbound protein, suggesting a stable ligand-protein complex. The tumor-specific expression and its critical overall survival suggest the inhibitors of FAP for potential cancer therapeutic intervention and hindering tumor microenvironment-driven cancer progression.
Collapse
Affiliation(s)
- Gayathri Ashok
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
- Department of Bio-Sciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, 11451, 13 Riyadh, Saudi Arabia
| | - Fawaz AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, 11451, 13 Riyadh, Saudi Arabia
| | - Paul Livingstone
- School of Sports and Health Sciences, Cardiff Metropolitan University, Cardiff, CF5 2YB, UK
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
- Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India.
- Department of Bio-Sciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
24
|
Dharani S, Cho H, Fernandez JP, Juillerat A, Valton J, Duchateau P, Poirot L, Das S. TALEN-edited allogeneic inducible dual CAR T cells enable effective targeting of solid tumors while mitigating off-tumor toxicity. Mol Ther 2024; 32:3915-3931. [PMID: 39169622 PMCID: PMC11573618 DOI: 10.1016/j.ymthe.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/29/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
Adoptive cell therapy using chimeric antigen receptor (CAR) T cells has proven to be lifesaving for many cancer patients. However, its therapeutic efficacy has been limited in solid tumors. One key factor for this is cancer-associated fibroblasts (CAFs) that modulate the tumor microenvironment (TME) to inhibit T cell infiltration and induce "T cell dysfunction." Additionally, the sparsity of tumor-specific antigens (TSA) and expression of CAR-directed tumor-associated antigens (TAA) on normal tissues often results in "on-target off-tumor" cytotoxicity, raising safety concerns. Using TALEN-mediated gene editing, we present here an innovative CAR T cell engineering strategy to overcome these challenges. Our allogeneic "Smart CAR T cells" are designed to express a constitutive CAR, targeting FAP+ CAFs in solid tumors. Additionally, a second CAR targeting a TAA such as mesothelin is specifically integrated at a TCR signaling-inducible locus like PDCD1. FAPCAR-mediated CAF targeting induces expression of the mesothelin CAR, establishing an IF/THEN-gated circuit sensitive to dual antigen sensing. Using this approach, we observe enhanced anti-tumor cytotoxicity, while limiting "on-target off-tumor" toxicity. Our study thus demonstrates TALEN-mediated gene editing capabilities for design of allogeneic IF/THEN-gated dual CAR T cells that efficiently target immunotherapy-recalcitrant solid tumors while mitigating potential safety risks, encouraging clinical development of this strategy.
Collapse
MESH Headings
- Humans
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Mice
- Mesothelin
- Gene Editing
- Cell Line, Tumor
- Transcription Activator-Like Effector Nucleases
- Tumor Microenvironment/immunology
- Neoplasms/therapy
- Neoplasms/immunology
- Cancer-Associated Fibroblasts/metabolism
- Cancer-Associated Fibroblasts/immunology
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Xenograft Model Antitumor Assays
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- GPI-Linked Proteins/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Membrane Proteins
- Endopeptidases
Collapse
Affiliation(s)
| | - Hana Cho
- Cellectis Inc, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Lin Y, Gao H, Zheng J, Al-Ibraheem A, Hu P, Shi H. Clinical Explorations of [ 68Ga] Ga-FAPI-04 and [ 18F] FDG Dual-Tracer Total-body PET/CT and PET/MR Imaging. Semin Nucl Med 2024; 54:904-913. [PMID: 39489646 DOI: 10.1053/j.semnuclmed.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024]
Abstract
Fibroblast activation protein inhibitor (FAPI) and [18F]fluorodeoxyglucose ([18F]FDG) provide complementary biological information, and FAPI/FDG dual-tracer imaging clinical application is increasing recently. However, optimal protocols for FAPI/FDG dual-tracer positron emission tomography/computed tomography (PET/CT) and PET/magnetic resonance (PET/MR) imaging are still under investigation. Due to its high sensitivity, total-body PET/CT allows for imaging with minimal tracer activity and supports the creation of new dual-tracer PET/CT imaging protocols. PET/MR, with its multiparametric MR imaging, provides additional biological information for diagnosis. Studies have investigated the clinical feasibility of low-activity PET/MR imaging, yielding promising results. As there are still few institutions in the world that have experience with the advances provided by the use of total-body PET/CT and equipped with a PET/MR scanner, we have discussed the clinical explorations to reduce radiation exposure and optimize workflows for [68Ga]Ga-FAPI-04 and [18F]FDG dual-tracer PET/CT and PET/MR imaging. The review also provides potential new clinical explorations of [68Ga]Ga-FAPI-04 and [18F]FDG dual-tracer total-body PET/CT and PET/MR imaging, including dual-tracer dual-low-activity imaging.
Collapse
Affiliation(s)
- Yu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huaping Gao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiefu Zheng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA
| | - Akram Al-Ibraheem
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman Jordan; Department of Radiology and Nuclear Medicine, School of Medicine, University of Jordan, Amman, Jordan
| | - Pengcheng Hu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Amrute JM, Luo X, Penna V, Yang S, Yamawaki T, Hayat S, Bredemeyer A, Jung IH, Kadyrov FF, Heo GS, Venkatesan R, Shi SY, Parvathaneni A, Koenig AL, Kuppe C, Baker C, Luehmann H, Jones C, Kopecky B, Zeng X, Bleckwehl T, Ma P, Lee P, Terada Y, Fu A, Furtado M, Kreisel D, Kovacs A, Stitziel NO, Jackson S, Li CM, Liu Y, Rosenthal NA, Kramann R, Ason B, Lavine KJ. Targeting immune-fibroblast cell communication in heart failure. Nature 2024; 635:423-433. [PMID: 39443792 DOI: 10.1038/s41586-024-08008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/03/2024] [Indexed: 10/25/2024]
Abstract
Inflammation and tissue fibrosis co-exist and are causally linked to organ dysfunction1,2. However, the molecular mechanisms driving immune-fibroblast cell communication in human cardiac disease remain unexplored and there are at present no approved treatments that directly target cardiac fibrosis3,4. Here we performed multiomic single-cell gene expression, epitope mapping and chromatin accessibility profiling in 45 healthy donor, acutely infarcted and chronically failing human hearts. We identified a disease-associated fibroblast trajectory that diverged into distinct populations reminiscent of myofibroblasts and matrifibrocytes, the latter expressing fibroblast activator protein (FAP) and periostin (POSTN). Genetic lineage tracing of FAP+ fibroblasts in vivo showed that they contribute to the POSTN lineage but not the myofibroblast lineage. We assessed the applicability of experimental systems to model cardiac fibroblasts and demonstrated that three different in vivo mouse models of cardiac injury were superior compared with cultured human heart and dermal fibroblasts in recapitulating the human disease phenotype. Ligand-receptor analysis and spatial transcriptomics predicted that interactions between C-C chemokine receptor type 2 (CCR2) macrophages and fibroblasts mediated by interleukin-1β (IL-1β) signalling drove the emergence of FAP/POSTN fibroblasts within spatially defined niches. In vivo, we deleted the IL-1 receptor on fibroblasts and the IL-1β ligand in CCR2+ monocytes and macrophages, and inhibited IL-1β signalling using a monoclonal antibody, and showed reduced FAP/POSTN fibroblasts, diminished myocardial fibrosis and improved cardiac function. These findings highlight the broader therapeutic potential of targeting inflammation to treat tissue fibrosis and preserve organ function.
Collapse
Affiliation(s)
- Junedh M Amrute
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Xin Luo
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tracy Yamawaki
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Sikander Hayat
- Institute of Experimental Medicine and Systems Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Andrea Bredemeyer
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - In-Hyuk Jung
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Farid F Kadyrov
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rajiu Venkatesan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sally Yu Shi
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Alekhya Parvathaneni
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Andrew L Koenig
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Department of Nephrology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | | | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Cameran Jones
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Benjamin Kopecky
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Xue Zeng
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Tore Bleckwehl
- Institute of Experimental Medicine and Systems Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Pan Ma
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Paul Lee
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yuriko Terada
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Angela Fu
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Milena Furtado
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Atilla Kovacs
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Simon Jackson
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Chi-Ming Li
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Department of Nephrology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Brandon Ason
- Amgen Discovery Research, Amgen Inc., South San Francisco, CA, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
27
|
Meng S, Hara T, Miura Y, Ishii H. Fibroblast activation protein constitutes a novel target of chimeric antigen receptor T-cell therapy in solid tumors. Cancer Sci 2024; 115:3532-3542. [PMID: 39169645 PMCID: PMC11531970 DOI: 10.1111/cas.16285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/14/2024] [Accepted: 07/04/2024] [Indexed: 08/23/2024] Open
Abstract
With recent advances in tumor immunotherapy, chimeric antigen receptor T (CAR-T) cell therapy has achieved unprecedented success in several hematologic tumors, significantly improving patient prognosis. However, in solid tumors, the efficacy of CAR-T cell therapy is limited because of high antigen uncertainty and the extremely restrictive tumor microenvironment (TME). This challenge has led to the exploration of new targets, among which fibroblast activation protein (FAP) has gained attention for its relatively stable and specific expression in the TME of various solid tumors, making it a potential new target for CAR-T cell therapy. This study comprehensively analyzed the biological characteristics of FAP and discussed its potential application in CAR-T cell therapy, including the theoretical basis, and preclinical and clinical research progress of targeting FAP with CAR-T cell therapy for solid tumor treatment. The challenges and future optimization directions of this treatment strategy were also explored, providing new perspectives and strategies for CAR-T cell therapy in solid tumors.
Collapse
Grants
- 2024 Princess Takamatsu Cancer Research Fund
- JP23ym0126809 Ministry of Education, Culture, Sports, Science and Technology
- JP24ym0126809 Ministry of Education, Culture, Sports, Science and Technology
- A20H0054100 Ministry of Education, Culture, Sports, Science and Technology
- T23KK01530 Ministry of Education, Culture, Sports, Science and Technology
- T22K195590 Ministry of Education, Culture, Sports, Science and Technology
- A22H031460 Ministry of Education, Culture, Sports, Science and Technology
- T23K183130 Ministry of Education, Culture, Sports, Science and Technology
- T23K195050 Ministry of Education, Culture, Sports, Science and Technology
- T24K199920 Ministry of Education, Culture, Sports, Science and Technology
- IFO Research Communications (2024)
- Oceanic Wellness Foundation (2024)
- Princess Takamatsu Cancer Research Fund
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Yutaka Miura
- Laboratory for Chemistry and Life ScienceInstitute of Innovative Research, Tokyo Institute of TechnologyYokohamaKanagawaJapan
- Department of Life Science and Technology, School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaKanagawaJapan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
28
|
Kim SM, Jang YJ. Enzymatic activity of fibroblast activation protein-α is essential for TGF-β1-induced fibroblastic differentiation of human periodontal ligament cells. Exp Cell Res 2024; 442:114230. [PMID: 39222867 DOI: 10.1016/j.yexcr.2024.114230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Human periodontal ligament cells (hPDLCs) contain multipotent postnatal stem cells that can differentiate into PDL fibroblasts, osteoblasts, and cementoblasts. Interaction between the extracellular environment and stem cells is an important factor for differentiation into other progenitor cells. To identify cell surface molecules that induce PDL fibroblastic differentiation, we developed a series of monoclonal antibodies against membrane/ECM molecules. One of these antibodies, an anti-PDL25 antibody, recognizes approximately a 100 kDa protein, and this antigenic molecule accumulates in the periodontal ligament region of tooth roots. By mass spectrometric analysis, we found that the antigenic molecule recognized by the anti-PDL25 antibody is fibroblast activation protein α (FAPα). The expression level of FAPα/PDL25 increased in TGF-β1-induced PDL fibroblasts, and this protein was localized in the cell boundaries and elongated processes of the fibroblastic cells. Ectopic expression of FAPα induced fibroblastic differentiation. In contrast, expression of representative markers for PDL differentiation was decreased by knock down and antibody blocking of FAPα/PDL25. Inhibition of dipeptidyl peptidase activity by a potent FAPα inhibitor dramatically inhibited PDL fibroblastic marker expression but did not affect in cell proliferation and migration.
Collapse
Affiliation(s)
- Seong-Min Kim
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea
| | - Young-Joo Jang
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea; Department of Oral Biochemistry, School of Dentistry, Dankook University, Cheonan, 31116, South Korea.
| |
Collapse
|
29
|
Koyama T, Yonemori K, Shimizu T, Sato J, Kondo S, Sudo K, Yoshida T, Katsuya Y, Imaizumi T, Enomoto M, Seki R, Yamamoto N. Phase I Study of Simlukafusp Alfa (FAP-IL2v) with or without Atezolizumab in Japanese Patients with Advanced Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:2349-2358. [PMID: 39140264 PMCID: PMC11377867 DOI: 10.1158/2767-9764.crc-24-0185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/21/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
PURPOSE The aim of the study was to evaluate the safety/tolerability and pharmacokinetics of simlukafusp alfa (FAP-IL2v), an immunocytokine containing an anti-fibroblast activation protein-α (FAP) antibody and an IL2 variant, administered alone or with the PDL1 inhibitor atezolizumab, in Japanese patients with advanced solid tumors. PATIENTS AND METHODS In this phase 1, open-label, dose-escalation study, patients received i.v. FAP-IL2v at 10 or 15/20 mg alone or 10 mg when combined with i.v. atezolizumab. The primary objectives were identification of dose-limiting toxicities (DLT), recommended dose, and maximum tolerated dose, and evaluation of the safety/tolerability and pharmacokinetics of FAP-IL2v alone and combined with atezolizumab. RESULTS All 11 patients experienced adverse events (AE) during FAP-IL2v treatment. Although most AEs were of mild severity, four treatment-related AEs led to study treatment discontinuation in two patients: one with infusion-related reaction, hypotension, and capillary leak syndrome, and the other with increased aspartate aminotransferase. No AE-related deaths occurred. One DLT (grade 3 hypotension) occurred in a patient receiving FAP-IL2v 15/20 mg alone. The recommended dose and maximum tolerated dose could not be determined. The pharmacokinetics of FAP-IL2v remained similar with or without atezolizumab. The study was terminated early as FAP-IL2v development was discontinued because of portfolio prioritization (not for efficacy/safety reasons). CONCLUSIONS This study describes the safety/tolerability of FAP-IL2v 10 mg alone and in combination with atezolizumab in Japanese patients with advanced solid tumors; one DLT (hypotension) occurred with FAP-IL2v 15/20 mg. However, dose escalation of FAP-IL2v was not conducted because of early study termination. SIGNIFICANCE This phase I study assessed the safety/tolerability and PK of simlukafusp alfa alone or combined with atezolizumab in Japanese patients with advanced solid tumors. No notable differences in PK were noted with the combination versus simlukafusp alfa alone; however, high-dose simlukafusp alfa treatment was associated with recombinant IL2-related toxicity, despite the drug's FAP targeting and IL2Rβγ-biased IL2 variant design.
Collapse
Affiliation(s)
- Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | - Kan Yonemori
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Toshio Shimizu
- Department of Pulmonary Medicine and Medical Oncology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Jun Sato
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Kazuki Sudo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Tatsuya Yoshida
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Yuki Katsuya
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | | | | | - Ryoko Seki
- Chugai Pharmaceutical Co., Ltd, Tokyo, Japan.
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| |
Collapse
|
30
|
Abbasi S, Khademi S, Montazerabadi A, Sahebkar A. FAP-Targeted Nanoparticle-based Imaging in Cancer: A Systematic Review. J Biomed Phys Eng 2024; 14:323-334. [PMID: 39175559 PMCID: PMC11336055 DOI: 10.31661/jbpe.v0i0.2404-1754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/12/2024] [Indexed: 08/24/2024]
Abstract
Background Fibroblast Activation Protein (FAP)-targeted nanoparticles (NPs) are designed to accumulate in cancerous stroma. These NPs hold promise for imaging applications in cancer therapy. Objective This systematic review aimed to comprehensively explore the use of FAP-targeting NPs for cancer diagnosis through different imaging modalities. Material and Methods This systematic review followed the framework proposed by O'Malley and Arksey. Peer-reviewed studies were searched in the Scopus, Science Direct, PubMed, and Google Scholar databases. Eligible studies were selected, and data were extracted to investigate the FAP-targeting NPs in imaging. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline was also utilized to present the results. Results Five studies met the specified inclusion criteria and were finally selected for analysis. The extracted data was classified into two categories: general and specific data. The general group indicated that most studies have been conducted in Mexico and have increased since 2022, and the specific group showed that colorectal cancer and Nude mice have received the most research attention. Furthermore, FAP-targeted NPs have demonstrated superior diagnostic imaging capabilities, even compared to specific methods for each cancer type. Also, they have been safe, with no toxicity. Conclusion FAP-targeted NPs using different ligands, such as Fibroblast Activation Protein Inhibitor (FAPI), can accurately detect tumors and metastases, and outperform specific cancer peptides like PSMA in cancer diagnosis. They are also non-toxic and do not cause radiation damage to tissues. Therefore, FAP-targeted NPs have the potential to serve as a viable alternative to FAP-targeted radionuclides for cancer diagnosis.
Collapse
Affiliation(s)
- Samaneh Abbasi
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Khademi
- Department of Radiology Technology, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Montazerabadi
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
31
|
Eskandari-Malayeri F, Rezeai M, Narimani T, Esmaeil N, Azizi M. Investigating the effect of Fusobacterium nucleatum on the aggressive behavior of cancer-associated fibroblasts in colorectal cancer. Discov Oncol 2024; 15:292. [PMID: 39030445 PMCID: PMC11264641 DOI: 10.1007/s12672-024-01156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/13/2024] [Indexed: 07/21/2024] Open
Abstract
Fusobacterium nucleatum, (F. nucleatum) as a known factor in inducing oncogenic, invasive, and inflammatory responses, can lead to an increase in the incidence and progression of colorectal cancer (CRC). Cancer-associated fibroblasts (CAF) are also one of the key components of the tumor microenvironment (TME), which lead to resistance to treatment, metastasis, and disease recurrence with their markers, secretions, and functions. This study aimed to investigate the effect of F. nucleatum on the invasive phenotype and function of fibroblast cells isolated from normal and cancerous colorectal tissue. F. nucleatum bacteria were isolated from deep periodontal pockets and confirmed by various tests. CAF cells from tumor tissue and normal fibroblasts (NF) from a distance of 10 cm of tumor tissue were isolated from 5 patients by the explant method and were exposed to secretions and ghosts of F. nucleatum. The expression level of two markers, fibroblast activation protein (FAP), and α-smooth muscle actin (α-SMA), and the amount of production of two cytokines TGF-β and IL-6 from fibroblast cells were measured by flow cytometry and ELISA test, respectively before and after exposure to different bacterial components. The expression of the FAP marker was significantly higher in CAF cells compared to NF cells (P < 0.05). Also, the expression of IL-6 in CAF cells was higher than that of NF cells. In investigating the effect of bacterial components on the function of fibroblastic cells, after comparing the amount of IL-6 produced between the normal tissue of each patient and his tumoral tissue under 4 treated conditions, it was found that the amount of IL-6 production from the CAF cells of patients in the control group, treated with heat-killed ghosts and treated with paraformaldehyde-fixed ghosts had a significant increase compared to NF cells (P < 0.05). Due to the significant increase in FAP marker expression in fibroblast cells of tumor tissue compared to normal tissue, it seems that FAP can be used as a very good therapeutic marker, especially in patients with high levels of CAF cells. Various components of F. nucleatum could affect fibroblast cells differentially and at least part of the effect of this bacterium in the TME is mediated by CAF cells.
Collapse
Affiliation(s)
| | - Marzieh Rezeai
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Tahmineh Narimani
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
32
|
LI RUIFANG, NAN XINRONG, LI MING, RAHHAL OMAR. Fibroblast activation protein (FAP) as a prognostic biomarker in multiple tumors and its therapeutic potential in head and neck squamous cell carcinoma. Oncol Res 2024; 32:1323-1334. [PMID: 39055892 PMCID: PMC11267059 DOI: 10.32604/or.2024.046965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/02/2024] [Indexed: 07/28/2024] Open
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.
Collapse
Affiliation(s)
- RUIFANG LI
- School of Stomatology, Shanxi Medical University, Taiyuan, 030001, China
| | - XINRONG NAN
- Department of Stomatology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - MING LI
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatology Hospital, Central South University, Changsha, 410005, China
| | - OMAR RAHHAL
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatology Hospital, Central South University, Changsha, 410005, China
| |
Collapse
|
33
|
Saadh MJ, Mustafa MA, Malathi H, Ahluwalia G, Kaur S, Al-Dulaimi MAAH, Alubiady MHS, Zain Al-Abdeen SH, Shakier HG, Ali MS, Ahmad I, Abosaoda MK. Targeting the pancreatic tumor microenvironment by plant-derived products and their nanoformulations. Med Oncol 2024; 41:201. [PMID: 39001987 DOI: 10.1007/s12032-024-02443-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
Pancreatic cancer remains a significant health issue with limited treatment options. The tumor stroma, a complex environment made up of different cells and proteins, plays a crucial role in tumor growth and chemoresistance. Targeting tumor stroma, consisting of diverse non-tumor cells such as fibroblasts, extracellular matrix (ECM), immune cells, and also pre-vascular cells is encouraging for remodeling solid cancers, such as pancreatic cancer. Remodeling the stroma of pancreas tumors can be suggested as a strategy for reducing resistance to chemo/immunotherapy. Several studies have shown that phytochemicals from plants can affect the tumor environment and have anti-cancer properties. By targeting key pathways involved in stromal activation, phytochemicals may disrupt communication between the tumor and stroma and make tumor cells more sensitive to different treatments. Additionally, phytochemicals have immunomodulatory and anti-angiogenic properties, all of which contribute to their potential in treating pancreatic cancer. This review will provide a detailed look at how phytochemicals impact the tumor stroma and their effects on pancreatic tumor growth, spread, and response to treatment. It will also explore the potential of combining phytochemicals with other treatment options like chemotherapy, immunotherapy, and radiation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Gunveen Ahluwalia
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, 303012, Rajasthan, India
| | - Sumeet Kaur
- Department of Applied Sciences, Chandigarh Engineering Colleges, Chandigarh Group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | | | | | | | | | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
34
|
Hoffmann M, Gallant J, LeBeau A, Schmidt D. Unlocking precision gene therapy: harnessing AAV tropism with nanobody swapping at capsid hotspots. NAR MOLECULAR MEDICINE 2024; 1:ugae008. [PMID: 39022346 PMCID: PMC11250487 DOI: 10.1093/narmme/ugae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Adeno-associated virus (AAV) has been remarkably successful in the clinic, but its broad tropism is a practical limitation of precision gene therapy. A promising path to engineer AAV tropism is the addition of binding domains to the AAV capsid that recognize cell surface markers present on a targeted cell type. We have recently identified two previously unexplored capsid regions near the 2/5-fold wall and 5-fold pore of the AAV capsid that are amenable to insertion of larger protein domains, including nanobodies. Here, we demonstrate that these hotspots facilitate AAV tropism switching through simple nanobody replacement without extensive optimization in both VP1 and VP2. Our data suggest that engineering VP2 is the preferred path for maintaining both virus production yield and infectivity. We demonstrate highly specific targeting of human cancer cells expressing fibroblast activating protein (FAP). Furthermore, we found that the combination of FAP nanobody insertion plus ablation of the heparin binding domain can reduce off-target infection to a minimum, while maintaining a strong infection of FAP receptor-positive cells. Taken together, our study shows that nanobody swapping at multiple capsid locations is a viable strategy for nanobody-directed cell-specific AAV targeting.
Collapse
Affiliation(s)
- Mareike D Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph P Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
35
|
Al-Ibraheem A, Zimmermann R, Abdlkadir AS, Herrmann K. Radiotheranostics Global Market and Future Developments. Semin Nucl Med 2024; 54:622-633. [PMID: 38485583 DOI: 10.1053/j.semnuclmed.2024.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 08/05/2024]
Abstract
Radiotheranostics, a combination of diagnostic and therapeutic approaches, was first utilized in cancer management using radiopharmaceuticals to both image and selectively treat specific cancer subtypes nearly a century ago. Radiotheranostic strategies rooted in nuclear medicine have revolutionized the treatment landscape for individuals diagnosed with prostate cancer and neuroendocrine tumors in the past 10 years. In specific contexts, these approaches have emerged as the prevailing standard, yielding numerous positive results. The field of radiotheranostics shows great potential for future clinical applications. This article aims to examine the key factors that will contribute to the success of radiotheranostics in the future, as well as the current challenges and potential strategies to overcome them, with insight into the global radiotheranostic market.
Collapse
Affiliation(s)
- Akram Al-Ibraheem
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, 11942, Jordan; Division of Nuclear Medicine/Department of Radiology and Nuclear Medicine, University of Jordan, Amman, 11942, Jordan.
| | - Richard Zimmermann
- Chrysalium Consulting, Lalaye, France; MEDraysintell, Louvain-la-Neuve, Oncidium Foundation, Mont-Saint-Guibert, Belgium; Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, 11942, Jordan
| | - Ahmed S Abdlkadir
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, 11942, Jordan
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center (WTZ), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; National Center for Tumor Diseases (NCT), NCT West, Germany
| |
Collapse
|
36
|
Gallant JP, Hintz HM, Gunaratne GS, Breneman MT, Recchia EE, West JL, Ott KL, Heninger E, Jackson AE, Luo NY, Rosenkrans ZT, Hernandez R, Zhao SG, Lang JM, Meimetis L, Kosoff D, LeBeau AM. Mechanistic Characterization of Cancer-associated Fibroblast Depletion via an Antibody-Drug Conjugate Targeting Fibroblast Activation Protein. CANCER RESEARCH COMMUNICATIONS 2024; 4:1481-1494. [PMID: 38747612 PMCID: PMC11168342 DOI: 10.1158/2767-9764.crc-24-0248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/14/2024]
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.
Collapse
Affiliation(s)
- Joseph P. Gallant
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Hallie M. Hintz
- Department of Pharmacology, University of Minnesota School of Medicine, Minneapolis, Minnesota
| | - Gihan S. Gunaratne
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Matthew T. Breneman
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Emma E. Recchia
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jayden L. West
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kendahl L. Ott
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Erika Heninger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Abigail E. Jackson
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Natalie Y. Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Zachary T. Rosenkrans
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Reinier Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Shuang G. Zhao
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Labros Meimetis
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - David Kosoff
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- William S Middleton Memorial Veterans’ Hospital, Madison, Wisconsin
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
37
|
Li R, Zhang R, Shi X, Jiao X, Li Y, Zhao Y, Liu T, Zhang C. Expression of FAP in Oral Leukoplakia and Oral Squamous Cell Carcinoma. Int Dent J 2024; 74:581-588. [PMID: 38278714 PMCID: PMC11123527 DOI: 10.1016/j.identj.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/28/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the potential of fibroblast activation protein (FAP) as a biomarker in the progression of oral leukoplakia (OLK) carcinogenesis. This was achieved by evaluating FAP expression at different levels of the organisation, namely oral normal mucosa (NM), OLK, and oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS Altogether, 88 paraffin-embedded tissue samples were examined, including 55 cases of OLK, 13 cases of OSCC, and 20 cases of NM (control group). An exhaustive investigation was performed to examine FAP expression in NM, OLK, and OSCC tissues via immunohistochemistry (IHC). The relationship between FAP expression and clinical pathologic characteristics was analysed. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB) also proved the expression of FAP in NM, OLK, and OSCC cells. Aberrant FAP expression in OLK and OSCC was explored using in vitro experiments. RESULTS Immunohistochemical results showed that high FAP expression was significantly correlated with histopathologic grade (P = .038) but not correlated with age, sex, or region (P = .953, .622, and .108, respectively). The expression level of FAP in NM tissues (0.15 ± 0.01) was minimal, whereas it was observed in OLK (0.28 ± 0.04) and OSCC (0.39 ± 0.02) tissues with a noticeable increase in expression levels (P < .001). The expression level of FAP in OLK with severe abnormal hyperplasia (S-OLK) tissues (0.33 ± 0.04) was significantly higher than in OLK with mild abnormal hyperplasia (MI-OLK, 0.26 ± 0.02) and OLK with moderate abnormal hyperplasia (MO-OLK, 0.28 ± 0.03) tissues (P < .001 and P = .039, respectively). The results of RT-PCR illustrated that the relative expression of FAP mRNA in OLK cells (2.63 ± 0.62) was higher than in NM cells (0.87 ± 0.14), but lower than in OSCC cells (5.63 ± 1.06; P = .027 and .012, respectively). FAP expression was minimal in NM cells (0.78 ± 0.06), modest in OLK cells (1.04 ± 0.06), and significantly elevated in OSCC cells (1.61 ± 0.09) based on the results of WB (P < .001). CONCLUSIONS Significant variations in FAP expression were observed in NM, OLK, and OSCC tissues and cells. These findings revealed that FAP may be a reliable biomarker for the early diagnosis and evaluation of OLK carcinogenesis.
Collapse
Affiliation(s)
- Ran Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China.
| | - Rongrong Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Xiaotong Shi
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Xiaofeng Jiao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Yanwei Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Yingjiao Zhao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Tiantian Liu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China; Department of Pediatric and Preventive Dentistry, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Chunye Zhang
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology.
| |
Collapse
|
38
|
Wehrli M, Guinn S, Birocchi F, Kuo A, Sun Y, Larson RC, Almazan AJ, Scarfò I, Bouffard AA, Bailey SR, Anekal PV, Llopis PM, Nieman LT, Song Y, Xu KH, Berger TR, Kann MC, Leick MB, Silva H, Salas-Benito D, Kienka T, Grauwet K, Armstrong TD, Zhang R, Zhu Q, Fu J, Schmidts A, Korell F, Jan M, Choi BD, Liss AS, Boland GM, Ting DT, Burkhart RA, Jenkins RW, Zheng L, Jaffee EM, Zimmerman JW, Maus MV. Mesothelin CAR T Cells Secreting Anti-FAP/Anti-CD3 Molecules Efficiently Target Pancreatic Adenocarcinoma and its Stroma. Clin Cancer Res 2024; 30:1859-1877. [PMID: 38393682 PMCID: PMC11062832 DOI: 10.1158/1078-0432.ccr-23-3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Marc Wehrli
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Samantha Guinn
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Filippo Birocchi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Adam Kuo
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yi Sun
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Antonio J. Almazan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Irene Scarfò
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Amanda A. Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Stefanie R. Bailey
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | | | | | - Linda T. Nieman
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yuhui Song
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Katherine H. Xu
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Trisha R. Berger
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Michael C. Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Mark B. Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Harrison Silva
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Diego Salas-Benito
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Tamina Kienka
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Korneel Grauwet
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Todd D. Armstrong
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Rui Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Qingfeng Zhu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Juan Fu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Max Jan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Bryan D. Choi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Andrew S. Liss
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Genevieve M. Boland
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - David T. Ting
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Richard A. Burkhart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Russell W. Jenkins
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Marcela V. Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| |
Collapse
|
39
|
Su J, Desmarais J, Chu CQ, Zhu J. Potential therapeutic targets of fibrosis in inflammatory rheumatic diseases. Best Pract Res Clin Rheumatol 2024; 38:101945. [PMID: 38627168 DOI: 10.1016/j.berh.2024.101945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 09/02/2024]
Abstract
Fibrosis is commonly associated with chronic rheumatic diseases, and causes substantial morbidity and mortality. Treatment of fibrosis is extremely challenging but is badly needed, as approved antifibrotic therapies fibrosis do not halt its progression, which will be discussed with a focus on pulmonary fibrosis. Findings from recent studies indicate several therapeutic targets for treating fibrosis. Interleukin-11 is emerging as a fibrogenic cytokine whose activity can be blocked with neutralizing monoclonal antibodies. Fibroblast activation protein (FAP) is highly expressed by activated fibroblasts in inflammatory and fibrotic tissues. Targeting FAP with different modalities has been extensively explored as adjunct treatment for cancer, which can also apply to treating fibrosis in rheumatic diseases.
Collapse
Affiliation(s)
- Jiang Su
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Julianna Desmarais
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA; Rheumatology Section, VA Portland Health Care System, Portland, OR, 97239, USA.
| | - Jing Zhu
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
40
|
Chintamaneni PK, Pindiprolu SKSS, Swain SS, Karri VVSR, Nesamony J, Chelliah S, Bhaskaran M. Conquering chemoresistance in pancreatic cancer: Exploring novel drug therapies and delivery approaches amidst desmoplasia and hypoxia. Cancer Lett 2024; 588:216782. [PMID: 38453046 DOI: 10.1016/j.canlet.2024.216782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Pancreatic cancer poses a significant challenge within the field of oncology due to its aggressive behaviour, limited treatment choices, and unfavourable outlook. With a mere 10% survival rate at the 5-year mark, finding effective interventions becomes even more pressing. The intricate relationship between desmoplasia and hypoxia in the tumor microenvironment further complicates matters by promoting resistance to chemotherapy and impeding treatment efficacy. The dense extracellular matrix and cancer-associated fibroblasts characteristic of desmoplasia create a physical and biochemical barrier that impedes drug penetration and fosters an immunosuppressive milieu. Concurrently, hypoxia nurtures aggressive tumor behaviour and resistance to conventional therapies. a comprehensive exploration of emerging medications and innovative drug delivery approaches. Notably, advancements in nanoparticle-based delivery systems, local drug delivery implants, and oxygen-carrying strategies are highlighted for their potential to enhance drug accessibility and therapeutic outcomes. The integration of these strategies with traditional chemotherapies and targeted agents reveals the potential for synergistic effects that amplify treatment responses. These emerging interventions can mitigate desmoplasia and hypoxia-induced barriers, leading to improved drug delivery, treatment efficacy, and patient outcomes in pancreatic cancer. This review article delves into the dynamic landscape of emerging anticancer medications and innovative drug delivery strategies poised to overcome the challenges imposed by desmoplasia and hypoxia in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Pavan Kumar Chintamaneni
- Department of Pharmaceutics, GITAM School of Pharmacy, GITAM (Deemed to be University), Rudraram, 502329 Telangana, India.
| | | | - Swati Swagatika Swain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | | | - Jerry Nesamony
- College of Pharmacy and Pharmaceutical Sciences, The University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Selvam Chelliah
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX-77004, USA
| | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, The University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA.
| |
Collapse
|
41
|
Schmidt P, Lindemeyer J, Raut P, Schütz M, Saniternik S, Jönsson J, Endepols H, Fischer T, Quaas A, Schlößer HA, Thelen M, Grüll H. Multiparametric Characterization of the DSL-6A/C1 Pancreatic Cancer Model in Rats. Cancers (Basel) 2024; 16:1535. [PMID: 38672617 PMCID: PMC11049193 DOI: 10.3390/cancers16081535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The DSL-6A/C1 murine pancreatic ductal adenocarcinoma (PDAC) tumor model was established in Lewis rats and characterized through a comprehensive multiparametric analysis to compare it to other preclinical tumor models and explore potential diagnostic and therapeutical targets. DSL-6A/C1 tumors were histologically analyzed to elucidate PDAC features. The tumor microenvironment was studied for immune cell prevalence. Multiparametric MRI and PET imaging were utilized to characterize tumors, and 68Ga-FAPI-46-targeting cancer-associated fibroblasts (CAFs), were used to validate the histological findings. The histology confirmed typical PDAC characteristics, such as malformed pancreatic ductal malignant cells and CAFs. Distinct immune landscapes were identified, revealing an increased presence of CD8+ T cells and a decreased CD4+ T cell fraction within the tumor microenvironment. PET imaging with 68Ga-FAPI tracers exhibited strong tracer uptake in tumor tissues. The MRI parameters indicated increasing intralesional necrosis over time and elevated contrast media uptake in vital tumor areas. We have demonstrated that the DSL-6A/C1 tumor model, particularly due to its high tumorigenicity, tumor size, and 68Ga-FAPI-46 sensitivity, is a suitable alternative to established small animal models for many forms of preclinical analyses and therapeutic studies of PDAC.
Collapse
Affiliation(s)
- Patrick Schmidt
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Johannes Lindemeyer
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Pranali Raut
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Markus Schütz
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| | - Sven Saniternik
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| | - Jannika Jönsson
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Heike Endepols
- Faculty of Medicine and University Hospital of Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, 50937 Cologne, Germany;
- Faculty of Medicine and University Hospital of Cologne, Department of Nuclear Medicine, University of Cologne, 50937 Cologne, Germany;
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Thomas Fischer
- Faculty of Medicine and University Hospital of Cologne, Department of Nuclear Medicine, University of Cologne, 50937 Cologne, Germany;
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, 50937 Cologne, Germany;
| | - Hans Anton Schlößer
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (H.A.S.); (M.T.)
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Martin Thelen
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (H.A.S.); (M.T.)
| | - Holger Grüll
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
42
|
Trujillo-Benítez D, Luna-Gutiérrez M, Aguirre-De Paz JG, Cruz-Nova P, Bravo-Villegas G, Vargas-Ahumada JE, Vallejo-Armenta P, Morales-Avila E, Jiménez-Mancilla N, Oros-Pantoja R, Santos-Cuevas C, Azorín-Vega E, Ocampo-García B, Ferro-Flores G. 68Ga-DOTA-D-Alanine-BoroPro Radiotracer for Imaging of the Fibroblast Activation Protein in Malignant and Non-Malignant Diseases. Pharmaceutics 2024; 16:532. [PMID: 38675193 PMCID: PMC11054143 DOI: 10.3390/pharmaceutics16040532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 04/28/2024] Open
Abstract
Recently, we reported a new fibroblast activation protein (FAP) inhibitor radiopharmaceutical based on the 99mTc-((R)-1-((6-hydrazinylnicotinoyl)-D-alanyl) pyrrolidin-2-yl) boronic acid (99mTc-HYNIC-D-Alanine-BoroPro)(99mTc-HYNIC-iFAP) structure for tumor microenvironment SPECT imaging. This research aimed to synthesize 68Ga-[2,2',2″,2‴-(2-(4-(2-(5-(((S)-1-((S)-2-boronopyrrolidin-1-yl)-1-oxopropan-2-yl)carbamoyl)pyridin-2-yl)hydrazine-1-carbothioamido)benzyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrayl)tetraacetic acid] (68Ga-DOTA-D-Alanine-BoroPro)(68Ga-iFAP) as a novel radiotracer for PET imaging and evaluate its usefulness for FAP expression in malignant and non-malignant tissues. The coupling of p-SCN-benzene DOTA with HYNIC-iFAP was used for the chemical synthesis and further labeling with 68Ga. Radiochemical purity was verified by radio-HPLC. The specificity of 68Ga-iFAP was evaluated in HCT116 cells, in which FAP expression was verified by immunofluorescence and Western blot. Biodistribution and biokinetic studies were performed in murine models. 68Ga-iFAP uptake at the myocardial level was assessed in mice with induced infarction. First-in-human images of 68Ga-iFAP in healthy subjects and patients with myocardial infarction, glioblastoma, prostate cancer, and breast cancer were also obtained. DOTA-D-Alanine BoroPro was prepared with a chemical purity of 98% and was characterized by UPLC mass spectroscopy, FT-IR, and UV-vis. The 68Ga-iFAP was obtained with a radiochemical purity of >95%. In vitro and in vivo studies demonstrated 68Ga-iFAP-specific recognition for FAP, rapid renal elimination, and adequate visualization of the glioblastoma, breast tumor, prostate cancer, and myocardial infarction sites. The results of this research justify further dosimetry and clinical trials to establish the specificity and sensitivity of 68Ga-iFAP PET for FAP expression imaging.
Collapse
Affiliation(s)
- Diana Trujillo-Benítez
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50180, Mexico
| | - Myrna Luna-Gutiérrez
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
| | - José G. Aguirre-De Paz
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50180, Mexico
| | - Pedro Cruz-Nova
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
| | | | - Joel E. Vargas-Ahumada
- Nuclear Medicine Department, Instituto Nacional de Cardiología, Mexico City 14000, Mexico
| | - Paola Vallejo-Armenta
- Nuclear Medicine Department, Instituto Nacional de Cancerología, Mexico City 14000, Mexico
| | - Enrique Morales-Avila
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50180, Mexico
| | | | | | - Clara Santos-Cuevas
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
| | - Erika Azorín-Vega
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
| | - Blanca Ocampo-García
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
| | - Guillermina Ferro-Flores
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (D.T.-B.); (C.S.-C.)
| |
Collapse
|
43
|
Hoffmann MD, Gallant JP, LeBeau AM, Schmidt D. Unlocking Precision Gene Therapy: Harnessing AAV Tropism with Nanobody Swapping at Capsid Hotspots. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587049. [PMID: 38585985 PMCID: PMC10996663 DOI: 10.1101/2024.03.27.587049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Adeno-associated virus has been remarkably successful in the clinic, but its broad tropism is a practical limitation of precision gene therapy. A promising path to engineer AAV tropism is the addition of binding domains to the AAV capsid that recognize cell surface markers present on a targeted cell type. We have recently identified two previously unexplored capsid regions near the 2-fold valley and 5-fold pore of the AAV capsid that are amenable to insertion of larger protein domains including nanobodies. Here, we demonstrate that these hotspots facilitate AAV tropism switching through simple nanobody replacement without extensive optimization in both VP1 and VP2. We demonstrate highly specific targeting of human cancer cells expressing fibroblast activating protein (FAP). Our data suggest that engineering VP2 is the preferred path for maintaining both virus production yield and infectivity. Our study shows that nanobody swapping at multiple capsid location is a viable strategy for nanobody-directed cell-specific AAV targeting.
Collapse
Affiliation(s)
- Mareike D. Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joseph P. Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
44
|
Song N, Cui K, Zeng L, Li M, Fan Y, Shi P, Wang Z, Su W, Wang H. Advance in the role of chemokines/chemokine receptors in carcinogenesis: Focus on pancreatic cancer. Eur J Pharmacol 2024; 967:176357. [PMID: 38309677 DOI: 10.1016/j.ejphar.2024.176357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
The chemokines/chemokine receptors pathway significantly influences cell migration, particularly in recruiting immune cells to the tumor microenvironment (TME), impacting tumor progression and treatment outcomes. Emerging research emphasizes the involvement of chemokines in drug resistance across various tumor therapies, including immunotherapy, chemotherapy, and targeted therapy. This review focuses on the role of chemokines/chemokine receptors in pancreatic cancer (PC) development, highlighting their impact on TME remodeling, immunotherapy, and relevant signaling pathways. The unique immunosuppressive microenvironment formed by the interaction of tumor cells, stromal cells and immune cells plays an important role in the tumor proliferation, invasion, migration and therapeutic resistance. Chemokines/chemokine receptors, such as chemokine ligand (CCL) 2, CCL3, CCL5, CCL20, CCL21, C-X-C motif chemokine ligand (CXCL) 1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CXCL16, CXCL17, and C-X3-C motif chemokine ligand (CX3CL)1, derived mainly from leukocyte cells, cancer-related fibroblasts (CAFs), pancreatic stellate cells (PSCs), and tumor-associated macrophages (TAMs), contribute to PC progression and treatment resistance. Chemokines recruit myeloid-derived suppressor cells (MDSC), regulatory T cells (Tregs), and M2 macrophages, inhibiting the anti-tumor activity of immune cells. Simultaneously, they enhance pathways like epithelial-mesenchymal transition (EMT), Akt serine/threonine kinase (AKT), extracellular regulated protein kinases (ERK) 1/2, and nuclear factor kappa-B (NF-κB), etc., elevating the risk of PC metastasis and compromising the efficacy of radiotherapy, chemotherapy, and anti-PD-1/PD-L1 immunotherapy. Notably, the CCLx-CCR2 and CXCLx-CXCR2/4 axis emerge as potential therapeutic targets in PC. This review integrates recent findings on chemokines and receptors in PC treatment, offering valuable insights for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Na Song
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Kai Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Liqun Zeng
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Mengxiao Li
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China
| | - Yanwu Fan
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Pingyu Shi
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Ziwei Wang
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Wei Su
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China.
| | - Haijun Wang
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China.
| |
Collapse
|
45
|
Beckabir W, Wobker SE, Damrauer JS, Midkiff B, De la Cruz G, Makarov V, Flick L, Woodcock MG, Grivas P, Bjurlin MA, Harrison MR, Vincent BG, Rose TL, Gupta S, Kim WY, Milowsky MI. Spatial Relationships in the Tumor Microenvironment Demonstrate Association with Pathologic Response to Neoadjuvant Chemoimmunotherapy in Muscle-invasive Bladder Cancer. Eur Urol 2024; 85:242-253. [PMID: 38092611 PMCID: PMC11022933 DOI: 10.1016/j.eururo.2023.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/11/2023] [Accepted: 11/09/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Platinum-based neoadjuvant chemotherapy (NAC) is standard for patients with muscle-invasive bladder cancer (MIBC). Pathologic response (complete: ypT0N0 and partial: OBJECTIVE Using the NanoString GeoMx platform, we performed proteomic digital spatial profiling (DSP) on transurethral resections of bladder tumors from 18 responders ( DESIGN, SETTING, AND PARTICIPANTS Pretreatment tumor samples were stained by hematoxylin and eosin and immunofluorescence (panCK and CD45) to select four regions of interest (ROIs): tumor enriched (TE), immune enriched (IE), tumor/immune interface (tumor interface = TX and immune interface = IX). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS DSP was performed with 52 protein markers from immune cell profiling, immunotherapy drug target, immune activation status, immune cell typing, and pan-tumor panels. RESULTS AND LIMITATIONS Protein marker expression patterns were analyzed to determine their association with pathologic response, incorporating or agnostic of their ROI designation (TE/IE/TX/IX). Overall, DSP-based marker expression showed high intratumoral heterogeneity; however, response was associated with markers including PD-L1 (ROI agnostic), Ki-67 (ROI agnostic, TE, IE, and TX), HLA-DR (TX), and HER2 (TE). An elastic net model of response with ROI-inclusive markers demonstrated better validation set performance (area under the curve [AUC] = 0.827) than an ROI-agnostic model (AUC = 0.432). A model including DSP, tumor mutational burden, and clinical data performed no better (AUC = 0.821) than the DSP-only model. CONCLUSIONS Despite high intratumoral heterogeneity of DSP-based marker expression, we observed associations between pathologic response and specific DSP-based markers in a spatially dependent context. Further exploration of tumor region-specific biomarkers may help predict response to neoadjuvant chemoimmunotherapy in MIBC. PATIENT SUMMARY In this study, we used the GeoMx platform to perform proteomic digital spatial profiling on transurethral resections of bladder tumors from 18 responders and 18 nonresponders from two studies of neoadjuvant chemotherapy (gemcitabine and cisplatin) plus immune checkpoint inhibitor therapy (LCCC1520 [pembrolizumab] and BLASST-1 [nivolumab]). We found that assessing protein marker expression in the context of tumor architecture improved response prediction.
Collapse
Affiliation(s)
- Wolfgang Beckabir
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Sara E Wobker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeffrey S Damrauer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bentley Midkiff
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Vladmir Makarov
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Leah Flick
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Petros Grivas
- Department of Medicine, Division of Medical Oncology, University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Marc A Bjurlin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael R Harrison
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA; Division of Hematology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA; Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA; Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Tracy L Rose
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shilpa Gupta
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Matthew I Milowsky
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
46
|
Guglielmo P, Alongi P, Baratto L, Conte M, Abenavoli EM, Buschiazzo A, Celesti G, Dondi F, Filice R, Gorica J, Jonghi-Lavarini L, Laudicella R, Librando M, Linguanti F, Mattana F, Miceli A, Olivari L, Piscopo L, Santo G, Volpe F, Evangelista L. FAPi-Based Agents in Thyroid Cancer: A New Step towards Diagnosis and Therapy? A Systematic Review of the Literature. Cancers (Basel) 2024; 16:839. [PMID: 38398230 PMCID: PMC10887091 DOI: 10.3390/cancers16040839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/11/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Background: Thyroid cancer (TC) is often treated with surgery followed by iodine-131. Up to 50% of the instances of TC lose their avidity to 131I, becoming more aggressive. In this scenario, [18F]FDG PET/CT imaging is used for evaluating the widespread nature of the disease, despite its low sensitivity and a false negative rate of 8-21.1%. A novel class of PET agents targeting the fibroblast activation protein inhibitor (FAPi) has emerged, studied particularly for their potential application to theranostics. (2) Methods: A search of the literature was performed by two independent authors (P.G. and L.E.) using the PubMed, Scopus, Web of Science, Cochrane Library, and EMBASE databases. The following terms were used: "FAP" or "FAPi" or "Fibroblast activating protein" and "thyroid" or "thyroid cancer", in different combinations. The included papers were original articles, clinical studies, and case reports in the English language. No time limits were used. Editorials, conference papers, reviews, and preclinical studies were excluded. (3) Results: There were 31 papers that were selected. Some studies reported a low or absent FAPi uptake in TC lesions; others reported promising findings for the detection of metastases. (4) Conclusions: The preliminary results are encouraging. FAPI agents are an alternative to [18F]FDG and a promising theranostic tool. However, further studies with a larger population are needed.
Collapse
Affiliation(s)
| | - Pierpaolo Alongi
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, 90127 Palermo, Italy;
| | - Lucia Baratto
- Division of Pediatric Radiology, Department of Radiology, Lucile Packard Children’s Hospital, Stanford University, Stanford, CA 94304, USA;
| | - Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00185 Rome, Italy;
| | | | - Ambra Buschiazzo
- Nuclear Medicine Division, Santa Croce and Carle Hospital, 12100 Cuneo, Italy;
| | - Greta Celesti
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98122 Messina, Italy; (G.C.); (M.L.)
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Rossella Filice
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90133 Palermo, Italy; (R.F.); (R.L.)
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00185 Rome, Italy;
| | - Lorenzo Jonghi-Lavarini
- Department of Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy;
| | - Riccardo Laudicella
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90133 Palermo, Italy; (R.F.); (R.L.)
| | - Maria Librando
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98122 Messina, Italy; (G.C.); (M.L.)
| | - Flavia Linguanti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Francesco Mattana
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCSS, 20141 Milan, Italy;
| | - Alberto Miceli
- Nuclear Medicine Unit, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy;
| | - Laura Olivari
- Nuclear Medicine Unit, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy;
| | - Leandra Piscopo
- Department of Advanced Biomedical Sciences, University Federico II, 80138 Naples, Italy; (L.P.); (F.V.)
| | - Giulia Santo
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy;
| | - Fabio Volpe
- Department of Advanced Biomedical Sciences, University Federico II, 80138 Naples, Italy; (L.P.); (F.V.)
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy;
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| |
Collapse
|
47
|
Hamacher R, Pabst KM, Cheung PF, Heilig CE, Hüllein J, Liffers ST, Borchert S, Costa PF, Schaarschmidt BM, Kessler L, Miera MA, Droste M, Akbulut M, Falkenhorst J, Zarrad F, Kostbade K, Mavroeidi IA, Glimm H, Umutlu L, Schuler M, Hübschmann D, Bauer S, Fröhling S, Herrmann K, Siveke JT, Schildhaus HU, Fendler WP. Fibroblast Activation Protein α-Directed Imaging and Therapy of Solitary Fibrous Tumor. J Nucl Med 2024; 65:252-257. [PMID: 38176718 DOI: 10.2967/jnumed.123.266411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/07/2023] [Indexed: 01/06/2024] Open
Abstract
Fibroblast activation protein α (FAPα) is expressed at high levels in several types of tumors. Here, we report the expression pattern of FAPα in solitary fibrous tumor (SFT) and its potential use as a radiotheranostic target. Methods: We analyzed FAPα messenger RNA and protein expression in biopsy samples from SFT patients using immunohistochemistry and multiplexed immunofluorescence. Tracer uptake and detection efficacy were assessed in patients undergoing clinical 68Ga-FAPα inhibitor (FAPI)-46 PET,18F-FDG PET, and contrast-enhanced CT. 90Y-FAPI-46 radioligand therapy was offered to eligible patients with progressive SFT. Results: Among 813 patients and 126 tumor entities analyzed from the prospective observational MASTER program of the German Cancer Consortium, SFT (n = 34) had the highest median FAPα messenger RNA expression. Protein expression was confirmed in tumor biopsies from 29 of 38 SFT patients (76%) in an independent cohort. Most cases showed intermediate to high FAPα expression by immunohistochemistry (24/38 samples, 63%), which was located primarily on the tumor cell surface. Nineteen patients who underwent 68Ga-FAPI-46 PET imaging demonstrated significantly increased tumor uptake, with an SUVmax of 13.2 (interquartile range [IQR], 10.2), and an improved mean detection efficacy of 94.5% (SEM, 4.2%), as compared with 18F-FDG PET (SUVmax, 3.2 [IQR, 3.1]; detection efficacy, 77.3% [SEM, 5.5%]). Eleven patients received a total of 34 cycles (median, 3 cycles [IQR, 2 cycles]) of 90Y-FAPI-46 radioligand therapy, which resulted in disease control in 9 patients (82%). Median progression-free survival was 227 d (IQR, 220 d). Conclusion: FAPα is highly expressed by SFT and may serve as a target for imaging and therapy. Further studies are warranted to define the role of FAPα-directed theranostics in the care of SFT patients.
Collapse
Affiliation(s)
- Rainer Hamacher
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany;
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
| | - Kim M Pabst
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Phyllis F Cheung
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (Partner Site Essen) and German Cancer Research Center, Heidelberg, Germany
| | - Christoph E Heilig
- Department of Translational Medical Oncology, National Center for Tumor Diseases, Heidelberg and German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
| | - Jennifer Hüllein
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Sven-Thorsten Liffers
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (Partner Site Essen) and German Cancer Research Center, Heidelberg, Germany
| | - Sabrina Borchert
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Institute of Pathology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Pedro Fragoso Costa
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Benedikt M Schaarschmidt
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Lukas Kessler
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Monika A Miera
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Margret Droste
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (Partner Site Essen) and German Cancer Research Center, Heidelberg, Germany
| | - Merve Akbulut
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Johanna Falkenhorst
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
| | - Fadi Zarrad
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Karina Kostbade
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
| | - Ilektra A Mavroeidi
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
| | - Hanno Glimm
- Department for Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center, Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium, Dresden, Germany; and
| | - Lale Umutlu
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
| | - Daniel Hübschmann
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Sebastian Bauer
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases, Heidelberg and German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
| | - Ken Herrmann
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Jens T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (Partner Site Essen) and German Cancer Research Center, Heidelberg, Germany
| | - Hans-Ulrich Schildhaus
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Institute of Pathology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Wolfgang P Fendler
- German Cancer Consortium, Partner site University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| |
Collapse
|
48
|
Azari F, Kennedy GT, Chang A, Low P, Basil M, Planer J, Katzen J, Eruslanov E, Albelda S, Singhal S. Molecular Imaging in Precision-Cut Non-Small Cell Lung Cancer Slices. Ann Thorac Surg 2024; 117:458-465. [PMID: 37572959 PMCID: PMC10841063 DOI: 10.1016/j.athoracsur.2023.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Small animal models remain invaluable for the preclinical study of emerging molecular imaging agents. However, the data obtained in this setting are generated in genetically homogenous animals that do not mimic human pathophysiology. The purpose of this study was to prospectively validate precision-cut lung slices (PCLSs) obtained from patients with lung cancer as a translational tool for the development of targeted fluorophores. METHODS The lung tissue was gently inflated with 2% Low-Melt Agarose (Fisher, 16520050) to avoid lung damage and minimize inflation pressure. The slices were then loaded into specialized cylindrical cartridges and inserted into a compressotome, and slices 150 to 350 μm thick were cut. Samples were incubated with fluorophore conjugates for ex vivo validation and immunohistochemical staining for receptor expression. RESULTS A total of 184 unique 3-dimensional, architecturally preserved normal lung and non-small cell lung cancer samples were obtained between 2020 and 2022. The median nodule size was 1.1 ± 0.21 cm for benign lesions and 2.1 ± 0.19 cm for malignant nodules. A total of 101 of 135 (74.8%) malignant lesions were adenocarcinoma spectrum lung cancers. The median viability was 9.78 ± 1.86 days, and 1 μM of FAPL-S0456 (high-affinity fibroblast activation protein [FAP] targeting ligand linked to the near-infrared fluorophore S0456, On Target Laboratories)-targeted near-infrared fluorochrome localization demonstrated correlative labeling of FAP-positive tumor areas with a correlation coefficient of +0.94 (P < .01). There was no FAP fluorochrome uptake in normal lungs (r = -1; P < .001). CONCLUSIONS PCLSs comprise a novel human tissue-based translational model that can be used to validate the efficacy of molecular imaging fluorochromes. PCLSs preserve the tumor microenvironment and parenchymal architecture that closely resemble the interactions of the immune and stromal components in humans.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gregory T Kennedy
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ashley Chang
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Philip Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Maria Basil
- Department of Medicine, Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph Planer
- Department of Medicine, Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeremy Katzen
- Department of Medicine, Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evgeniy Eruslanov
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven Albelda
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
49
|
Yadav MP, Ballal S, Martin M, Roesch F, Satapathy S, Moon ES, Tripathi M, Gogia A, Bal C. Therapeutic potential of [ 177Lu]Lu-DOTAGA-FAPi dimers in metastatic breast cancer patients with limited treatment options: efficacy and safety assessment. Eur J Nucl Med Mol Imaging 2024; 51:805-819. [PMID: 37932560 DOI: 10.1007/s00259-023-06482-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/15/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE The upregulation of fibroblast activation protein (FAP) expression has been observed in various cancers, including metastatic breast carcinoma, prompting research into small molecule inhibitors for both diagnostic and therapeutic purposes. While the diagnostic value of PET/CT imaging using 68 Ga- or 18F-labelled FAPi-monomers in breast cancer diagnosis is well-established, there is a significant need for therapeutic analogs. This retrospective study aimed to assess the safety and effectiveness of [177Lu]Lu-DOTAGA.FAPi dimer radionuclide therapy in patients with advanced-stage breast cancer who had previously undergone [68 Ga]Ga-DOTA.SA.FAPi PET/CT scans to confirm the expression of FAP. MATERIALS AND METHODS Between November 2020 and March 2023, a compassionate treatment approach was utilized to administer [177Lu]Lu-DOTAGA.FAPi dimer radionuclide therapy to heavily pretreated patients with advanced breast cancer. Nineteen patients (18 females, 1 male) with metastatic breast cancer participated in the study, with an average age of 44.6 ± 10.7 years. The therapy was administered at intervals of 8 to 12 weeks, and the median follow-up duration was 14 months. The primary objective of the study was to assess molecular response using [68 Ga]Ga-DOTA.SA.FAPi PET/CT scans, with response evaluation based on the PERCIST criteria. Secondary endpoints included overall survival (OS), progression-free survival (PFS), clinical response assessment, and safety evaluation using CTCAE v5.0 guidelines. RESULTS A total of 65 cycles were administered, with a mean cumulative activity of 19 ± 5.7 GBq (510 ± 154 mCi) ranging from 11 to 33.3 GBq (300 to 900 mCi) of [177Lu]Lu-DOTAGA.FAPi dimer. The number of cycles ranged from 2 to 6, with a median of 3 cycles. The treatment protocol consisted of different numbers of cycles administered to the patients: specifically, two cycles were given to five patients, three cycles to nine patients, four cycles to one patient, and six cycles to four patients. Most patients had invasive/infiltrative ductal carcinoma (94.7%), while a small percentage had invasive lobular carcinoma (5.3%). All patients had bone metastases, and five of them also had liver involvement, while seven had brain metastases. Response assessment using [68 Ga]Ga-DOTA.SA.FAPi PET/CT scans showed that 25% of the 16 patients evaluated had partial remission, while 37.5% exhibited disease progression. According to the VAS response criteria, 26.3% achieved complete response, 15.7% had partial response, 42% showed minimal response, 11% had stable disease, and 5% had no response. The clinical disease control rate was promising, with 95% of patients achieving disease control. The clinical objective response rate was 84%. The median follow-up period was 14 months. At the time of analysis, the median overall survival was 12 months, and the median progression-free survival was 8.5 months. Notably, no severe hematological, renal, or hepatic toxicities, electrolyte imbalances, or adverse events of grade 3 or 4 were observed during the study. CONCLUSION The findings suggest that [177Lu]Lu-DOTAGA.FAPi dimer therapy is well-tolerated, safe, and effective for treating end-stage metastatic breast cancer patients. [177Lu]Lu-DOTAGA.FAPi dimer treatment demonstrated promising efficacy in patients with advanced breast cancer, as indicated by high disease control rates, favorable response outcomes, and acceptable safety profile. Further research and longer follow-up are warranted to assess long-term outcomes and validate these findings.
Collapse
Affiliation(s)
- Madhav P Yadav
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sanjana Ballal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Marcel Martin
- Department of Chemistry - TRIGA site, Johannes Gutenberg University, Mainz, Germany
| | - Frank Roesch
- Department of Chemistry - TRIGA site, Johannes Gutenberg University, Mainz, Germany
| | - Swayamjeet Satapathy
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Euy S Moon
- Department of Chemistry - TRIGA site, Johannes Gutenberg University, Mainz, Germany
| | - Madhavi Tripathi
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
50
|
Loureiro LR, Hoffmann L, Neuber C, Rupp L, Arndt C, Kegler A, Kubeil M, Hagemeyer CE, Stephan H, Schmitz M, Feldmann A, Bachmann M. Immunotheranostic target modules for imaging and navigation of UniCAR T-cells to strike FAP-expressing cells and the tumor microenvironment. J Exp Clin Cancer Res 2023; 42:341. [PMID: 38102692 PMCID: PMC10722841 DOI: 10.1186/s13046-023-02912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cells are a promising approach in cancer immunotherapy, particularly for treating hematologic malignancies. Yet, their effectiveness is limited when tackling solid tumors, where immune cell infiltration and immunosuppressive tumor microenvironments (TME) are major hurdles. Fibroblast activation protein (FAP) is highly expressed on cancer-associated fibroblasts (CAFs) and various tumor cells, playing an important role in tumor growth and immunosuppression. Aiming to modulate the TME with increased clinical safety and effectiveness, we developed novel small and size-extended immunotheranostic UniCAR target modules (TMs) targeting FAP. METHODS The specific binding and functionality of the αFAP-scFv TM and the size-extended αFAP-IgG4 TM were assessed using 2D and 3D in vitro models as well as in vivo. Their specific tumor accumulation and diagnostic potential were evaluated using PET studies after functionalization with a chelator and suitable radionuclide. RESULTS The αFAP-scFv and -IgG4 TMs effectively and specifically redirected UniCAR T-cells using 2D, 3D, and in vivo models. Moreover, a remarkably high and specific accumulation of radiolabeled FAP-targeting TMs at the tumor site of xenograft mouse models was observed. CONCLUSIONS These findings demonstrate that the novel αFAP TMs are promising immunotheranostic tools to foster cancer imaging and treatment, paving the way for a more convenient, individualized, and safer treatment of cancer patients.
Collapse
Affiliation(s)
- Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
| | - Lydia Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Christin Neuber
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Alexandra Kegler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Manja Kubeil
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|