AccScience Publishing / ARNM / Volume 1 / Issue 2 / DOI: 10.36922/arnm.1667
Cite this article
76
Download
1529
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
REVIEW

Fibroblast activation protein-targeted radioligand therapy in various types of cancer: Background, clinical studies, and future development

Yizhen Pang1,2† Liang Zhao1,2† Jianhao Chen2 Weizhi Xu3 Jiayu Cai3 Haojun Chen1,3* Qin Lin1,2*
Show Less
1 The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
2 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
3 Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Development and Translation of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
Submitted: 23 August 2023 | Accepted: 19 September 2023 | Published: 24 October 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Cancer is the leading cause of mortality worldwide. The proliferation and viability of cancer cells are intricately related to the complex tumor microenvironment in which they reside. Cancer-associated fibroblasts (CAFs) play a pivotal role in multiple stages of tumorigenesis, including tumor growth, invasion, metastasis, and resistance to treatment, through intricate crosstalk with tumor and immune cells. Targeted radionuclide therapy involves the systemic administration of small molecule drugs labeled with β-emitting or α-emitting radioisotopes, allowing precise targeting of tumor sites and delivering radiation directly to the tumor. Prostate-specific membrane antigen-targeted radioligand therapy (RLT) and somatostatin receptor-targeted peptide receptor radionuclide therapy (PRRT) have demonstrated favorable safety profiles and significant antitumor efficacy, leading to their approval by the Food and Drug Administration. Recently, the utilization of theranostic approaches that target overexpressed fibroblast activation proteins (FAPs) within the CAFs of the tumor stroma has demonstrated encouraging preliminary outcomes. This review aims to provide a concise summary of current clinical research outcomes and the applications of RLT based on FAP inhibitors in the context of solid tumors.

Keywords
Fibroblast activation proteins
Radioligand therapy
Immunotherapy
Synergistic effect
Funding
National Natural Science Foundation of China
Fujian Research and Training Grants for Young and Middle-aged Leaders in Healthcare, Key Scientific Research Program for Yong Scholars in Fujian
Fujian Natural Science Foundation for Distinguished Yong Scholars
Natural Science Foundation of Fujian Province
Key Medical and Health Projects in Xiamen
Xiamen Key Laboratory of Radiopharmaceuticals, Xiamen Key Laboratory of Radiation Oncology, Xiamen Clinical Research Center for Head and Neck Cancer, and 2021 National Clinical Key Specialty
References
  1. Kim KJ, Kim KJ, Choi J, et al., 2023, Linear association between radioactive iodine dose and second primary malignancy risk in thyroid cancer. J Natl Cancer Inst, 115: 695–702. https://doi.org/10.1093/jnci/djad040

 

  1. Paskeh MDA, Entezari M, Mirzaei S, et al., 2022, Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol, 15: 83. https://doi.org/10.1186/s13045-022-01305-4

 

  1. Morad G, Helmink BA, Sharma P, et al., 2021, Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell, 184: 5309–5337. https://doi.org/10.1016/j.cell.2021.09.020

 

  1. Morris ZS, Wang AZ, Knox SJ, 2021, The radiobiology of radiopharmaceuticals. Semin Radiat Oncol, 31: 20–27. https://doi.org/10.1016/j.semradonc.2020.07.002

 

  1. Langbein T, Weber WA, Eiber M, 2019, Future of theranostics: An outlook on precision oncology in nuclear medicine. J Nucl Med, 60: 13S–19S. https://doi.org/10.2967/jnumed.118.220566

 

  1. Fallah J, Agrawal S, Gittleman H, et al., 2023, FDA approval summary: Lutetium Lu 177 vipivotide tetraxetan for patients with metastatic castration-resistant prostate cancer. Clin Cancer Res, 29: 1651–1657. https://doi.org/10.1158/1078-0432.CCR-22-2875

 

  1. Chen X, Song E, 2019, Turning foes to friends: Targeting cancer-associated fibroblasts. Nat Rev Drug Discov, 18: 99–115. https://doi.org/10.1038/s41573-018-0004-1

 

  1. Hamson EJ, Keane FM, Tholen S, et al., 2014, Understanding fibroblast activation protein (FAP): Substrates, activities, expression and targeting for cancer therapy. Proteomics Clin Appl, 8: 454–463. https://doi.org/10.1002/prca.201300095

 

  1. Kratochwil C, Flechsig P, Lindner T, et al., 2019, 68Ga-FAPI PET/CT: Tracer uptake in 28 different kinds of cancer. J Nucl Med, 60: 801–805. https://doi.org/10.2967/jnumed.119.227967

 

  1. Chen H, Pang Y, Wu J, et al., 2020, Comparison of [68Ga] Ga-DOTA-FAPI-04 and [18F] FDG PET/CT for the diagnosis of primary and metastatic lesions in patients with various types of cancer. Eur J Nucl Med Mol Imaging, 47: 1820–1832. https://doi.org/10.1007/s00259-020-04769-z

 

  1. Lindner T, Loktev A, Altmann A, et al., 2018, Development of quinoline-based theranostic ligands for the targeting of fibroblast activation protein. J Nucl Med, 59: 1415–1422. https://doi.org/10.2967/jnumed.118.210443

 

  1. Lindner T, Altmann A, Kramer S, et al., 2020, Design and development of 99mTc-labeled FAPI tracers for SPECT imaging and 188Re therapy. J Nucl Med, 61: 1507–1513. https://doi.org/10.2967/jnumed.119.239731

 

  1. Ballal S, Yadav MP, Kramer V, et al., 2021, A theranostic approach of [68Ga]Ga-DOTA.SA.FAPi PET/CT-guided [177Lu] Lu-DOTA.SA.FAPi radionuclide therapy in an end-stage breast cancer patient: New frontier in targeted radionuclide therapy. Eur J Nucl Med Mol Imaging, 48: 942–944. https://doi.org/10.1007/s00259-020-04990-w

 

  1. Ohlund D, Elyada E, Tuveson D, 2014, Fibroblast heterogeneity in the cancer wound. J Exp Med, 211: 1503–1523. https://doi.org/10.1084/jem.20140692

 

  1. Mer AS, Ba-Alawi W, Smirnov P, et al., 2019, Integrative pharmacogenomics analysis of patient-derived xenografts. Cancer Res, 79: 4539–4550. https://doi.org/10.1158/0008-5472.CAN-19-0349

 

  1. Ozdemir BC, Pentcheva-Hoang T, Carstens JL, et al., 2014, Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell, 25: 719–734. https://doi.org/10.1016/j.ccr.2014.04.005

 

  1. Rettig WJ, Su SL, Fortunato SR, et al., 1994, Fibroblast activation protein: Purification, epitope mapping and induction by growth factors. Int J Cancer, 58: 385–392. https://doi.org/10.1002/ijc.2910580314

 

  1. Zhao L, Chen J, Pang Y, et al., 2022, Fibroblast activation protein-based theranostics in cancer research: A state-of-the-art review. Theranostics, 12: 1557–1569. https://doi.org/10.7150/thno.69475

 

  1. Ora M, Soni N, Nazar AH, et al., 2023, Fibroblast activation protein inhibitor-based radionuclide therapies: Current status and future directions. J Nucl Med, 64: 1001–1008. https://doi.org/10.2967/jnumed.123.265594

 

  1. Ostermann E, Garin-Chesa P, Heider KH, et al., 2008, Effective immunoconjugate therapy in cancer models targeting a serine protease of tumor fibroblasts. Clin Cancer Res, 14: 4584–4592. https://doi.org/10.1158/1078-0432.CCR-07-5211

 

  1. Duperret EK, Trautz A, Ammons D, et al., 2018, Alteration of the tumor stroma using a consensus DNA vaccine targeting fibroblast activation protein (FAP) synergizes with antitumor vaccine therapy in mice. Clin Cancer Res, 24: 1190–11201. https://doi.org/10.1158/1078-0432.CCR-17-2033

 

  1. Gunderson AJ, Yamazaki T, McCarty K, et al., 2019, Blockade of fibroblast activation protein in combination with radiation treatment in murine models of pancreatic adenocarcinoma. PLoS One, 14: e0211117. https://doi.org/10.1371/journal.pone.0211117

 

  1. Wang LCS, Lo A, Scholler J, et al., 2014, Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res, 2: 154–166. https://doi.org/10.1158/2326-6066.CIR-13-0027

 

  1. Laklai H, Miroshnikova YA, Pickup MW, et al., 2016, Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat Med, 22: 497–505. https://doi.org/10.1038/nm.4082

 

  1. Valkenburg KC, de Groot AE, Pienta KJ, 2018, Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol, 15: 366–381. https://doi.org/10.1038/s41571-018-0007-1

 

  1. Kessler L, Ferdinandus J, Hirmas N, et al., 2022, Pitfalls and common findings in 68Ga-FAPI PET: A pictorial analysis. J Nucl Med, 63: 890–896. https://doi.org/10.2967/jnumed.121.262808

 

  1. van den Hoven AF, Keijsers RGM, Lam M, et al., 2023, Current research topics in FAPI theranostics: A bibliometric analysis. Eur J Nucl Med Mol Imaging, 50: 1014–1027. https://doi.org/10.1007/s00259-022-06052-9

 

  1. Pang Y, Zhao L, Meng T, et al., 2023, PET imaging of fibroblast activation protein in various types of cancer using 68Ga-FAP-2286: Comparison with 18F-FDG and 68Ga-FAPI-46 in a single-center, prospective study. J Nucl Med, 64: 386–394. https://doi.org/10.2967/jnumed.122.264544

 

  1. Polson AG, Fuji RN, 2012, The successes and limitations of preclinical studies in predicting the pharmacodynamics and safety of cell-surface-targeted biological agents in patients. Br J Pharmacol, 166: 1600–1602. https://doi.org/10.1111/j.1476-5381.2012.01916.x

 

  1. Watabe T, Liu Y, Kaneda-Nakashima K, et al., 2020, Theranostics targeting fibroblast activation protein in the tumor stroma: 64Cu- and 225Ac-labeled FAPI-04 in pancreatic cancer xenograft mouse models. J Nucl Med, 61: 563–569. https://doi.org/10.2967/jnumed.119.233122

 

  1. Loktev A, Lindner T, Burger EM, et al., 2019, Development of fibroblast activation protein-targeted radiotracers with improved tumor retention. J Nucl Med, 60: 1421–1429. https://doi.org/10.2967/jnumed.118.224469

 

  1. Rathke H, Fuxius S, Giesel FL, et al., 2021, Two tumors, one target: Preliminary experience with 90Y-FAPI therapy in a patient with metastasized breast and colorectal cancer. Clin Nucl Med, 46: 842–844. https://doi.org/10.1097/RLU.0000000000003842.

 

  1. Kratochwil C, Giesel FL, Rathke H, et al., 2021, [153Sm] Samarium-labeled FAPI-46 radioligand therapy in a patient with lung metastases of a sarcoma. Eur J Nucl Med Mol Imaging, 48: 3011–3013. https://doi.org/10.1007/s00259-021-05273-8

 

  1. Fu K, Pang Y, Zhao L, et al., 2022, FAP-targeted radionuclide therapy with [177Lu]Lu-FAPI-46 in metastatic nasopharyngeal carcinoma. Eur J Nucl Med Mol Imaging, 49: 1767–1769. https://doi.org/10.1007/s00259-021-05634-3

 

  1. Zhao L, Niu B, Fang J, et al., 2022, Synthesis, preclinical evaluation, and a pilot clinical PET imaging study of 68Ga-Labeled FAPI dimer. J Nucl Med, 63: 862–868. https://doi.org/10.2967/jnumed.121.263016

 

  1. Pang Y, Zhao L, Fang J, et al., 2023, Development of FAPI tetramers to improve tumor uptake and efficacy of FAPI radioligand therapy. J Nucl Med, 64: 1449–1455. https://doi.org/10.2967/jnumed.123.265599

 

  1. Zhao L, Chen J, Pang Y, et al., 2022, Development of fibroblast activation protein inhibitor-based dimeric radiotracers with improved tumor retention and antitumor efficacy. Mol Pharm, 19: 364036-51. https://doi.org/10.1021/acs.molpharmaceut.2c00424

 

  1. Moon ES, Ballal S, Yadav MP, et al., 2021, Fibroblast activation protein (FAP) targeting homodimeric FAP inhibitor radiotheranostics: A step to improve tumor uptake and retention time. Am J Nucl Med Mol Imaging, 11: 476–491.

 

  1. Zboralski D, Hoehne A, Bredenbeck A, et al., 2022, Preclinical evaluation of FAP-2286 for fibroblast activation protein targeted radionuclide imaging and therapy. Eur J Nucl Med Mol Imaging, 49: 3651–3667. https://doi.org/10.1007/s00259-022-05842-5

 

  1. Wen X, Xu P, Shi M, et al., 2022, Evans blue-modified radiolabeled fibroblast activation protein inhibitor as long-acting cancer therapeutics. Theranostics, 12: 422–433. https://doi.org/10.7150/thno.68182

 

  1. Xu M, Zhang P, Ding J, et al., 2022, Albumin binder-conjugated fibroblast activation protein inhibitor radiopharmaceuticals for cancer therapy. J Nucl Med, 63: 952–958. https://doi.org/10.2967/jnumed.121.262533

 

  1. Tranel J, Palm S, Graves SA, et al., 2022, Impact of radiopharmaceutical therapy (177Lu, 225Ac) microdistribution in a cancer-associated fibroblasts model. EJNMMI Phys, 9: 67. https://doi.org/10.1186/s40658-022-00497-5

 

  1. Jokar N, Velikyan I, Ahmadzadehfar H, et al., 2021, Theranostic approach in breast cancer: A treasured tailor for future oncology. Clin Nucl Med, 46: e410–e420. https://doi.org/10.1097/RLU.0000000000003678

 

  1. Assadi M, Rekabpour SJ, Jafari E, et al., 2021, Feasibility and therapeutic potential of 177Lu-fibroblast activation protein inhibitor-46 for patients with relapsed or refractory cancers: A preliminary study. Clin Nucl Med, 46: e523–e530. https://doi.org/10.1097/RLU.0000000000003810

 

  1. Ballal S, Yadav MP, Moon ES, et al., 2021, First-in-human results on the biodistribution, pharmacokinetics, and dosimetry of [177Lu]Lu-DOTA.SA.FAPi and [177Lu]Lu-DOTAGA.(SA.FAPi) (2). Pharmaceuticals (Basel), 14: 1212. https://doi.org/10.3390/ph14121212

 

  1. Kuyumcu S, Kovan B, Sanli Y, et al., 2021, Safety of fibroblast activation protein-targeted radionuclide therapy by a low-dose dosimetric approach using 177Lu-FAPI04. Clin Nucl Med, 46: 641–646. https://doi.org/10.1097/RLU.0000000000003667

 

  1. Baum RP, Schuchardt C, Singh A, et al., 2022, Feasibility, biodistribution, and preliminary dosimetry in peptide-targeted radionuclide therapy of diverse adenocarcinomas using 177Lu-FAP-2286: First-in-humans results. J Nucl Med, 63: 415–423. https://doi.org/10.2967/jnumed.120.259192

 

  1. Ferdinandus J, Costa PF, Kessler L, et al., 2022, Initial clinical experience with 90Y-FAPI-46 radioligand therapy for advanced-stage solid tumors: A case series of 9 patients. J Nucl Med, 63: 727–734. https://doi.org/10.2967/jnumed.121.262468

 

  1. Fendler WP, Pabst KM, Kessler L, et al., 2022, Safety and efficacy of 90Y-FAPI-46 radioligand therapy in patients with advanced sarcoma and other cancer entities. Clin Cancer Res, 28: 4346–4353. https://doi.org/10.1158/1078-0432.CCR-22-1432

 

  1. Kaghazchi F, Aghdam RA, Haghighi S, et al., 2022, 177Lu-FAPI therapy in a patient with end-stage metastatic pancreatic adenocarcinoma. Clin Nucl Med, 47: e243–e245. https://doi.org/10.1097/RLU.0000000000004021

 

  1. Ballal S, Yadav MP, Moon ES, et al., 2022, Novel fibroblast activation protein inhibitor-based targeted theranostics for radioiodine-refractory differentiated thyroid cancer patients: A pilot study. Thyroid, 32: 65–77. https://doi.org/10.1089/thy.2021.0412

 

  1. Darvin P, Toor SM, Sasidharan Nair V, et al., 2018, Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp Mol Med, 50: 1–11. https://doi.org/10.1038/s12276-018-0191-1

 

  1. Borghaei H, Paz-Ares L, Horn L, et al., 2015, Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med, 373: 1627–1639. https://doi.org/10.1056/NEJMoa1507643

 

  1. Schoenfeld AJ, Hellmann MD, 2020, Acquired resistance to immune checkpoint inhibitors. Cancer Cell, 37: 443–455. https://doi.org/10.1016/j.ccell.2020.03.017

 

  1. Joyce JA, Fearon DT, 2015, T cell exclusion, immune privilege, and the tumor microenvironment. Science, 348: 74–80. https://doi.org/10.1126/science.aaa6204

 

  1. Sahai E, Astsaturov I, Cukierman E, et al., 2020, A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer, 20: 174–186. https://doi.org/10.1038/s41568-019-0238-1

 

  1. Grout JA, Sirven P, Leader AM, et al., 2022, Spatial positioning and matrix programs of cancer-associated fibroblasts promote T-cell exclusion in human lung tumors. Cancer Discov, 12: 2606–2625. https://doi.org/10.1158/2159-8290.CD-21-1714

 

  1. Dearnaley DP, Jovic G, Syndikus I, et al., 2014, Escalated-dose versus control-dose conformal radiotherapy for prostate cancer: Long-term results from the MRC RT01 randomised controlled trial. Lancet Oncol, 15: 464–473. https://doi.org/10.1016/S1470-2045(14)70040-3

 

  1. Hall S, Rudrawar S, Zunk M, et al., 2016, Protection against radiotherapy-induced toxicity. Antioxidants (Basel), 5: 22. https://doi.org/10.3390/antiox5030022

 

  1. Jagodinsky JC, Jin WJ, Bates AM, et al., 2021, Temporal analysis of Type 1 interferon activation in tumor cells following external beam radiotherapy or targeted radionuclide therapy. Theranostics, 11: 6120–6137. https://doi.org/10.7150/thno.54881

 

  1. Patel RB, Hernandez R, Carlson P, et al., 2021, Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade. Sci Transl Med, 13: eabb3631. https://doi.org/10.1126/scitranslmed.abb3631

 

  1. Hernandez R, Walker KL, Grudzinski JJ, et al., 2019, 90Y-NM600 targeted radionuclide therapy induces immunologic memory in syngeneic models of T-cell non-Hodgkin’s lymphoma. Commun Biol, 2: 79. https://doi.org/10.1038/s42003-019-0327-4

 

  1. Twyman-Saint Victor C, Rech AJ, Maity A, et al., 2015, Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature, 520: 373–377. https://doi.org/10.1038/nature14292

 

  1. Thierens HM, Monsieurs MA, Brans B, et al., 2001, Dosimetry from organ to cellular dimensions. Comput Med Imaging Graph, 25: 187–193. https://doi.org/10.1016/s0895-6111(00)00047-1

 

  1. Gill MR, Falzone N, Du Y, et al., 2017, Targeted radionuclide therapy in combined-modality regimens. Lancet Oncol, 18: e414–e423. https://doi.org/10.1016/S1470-2045(17)30379-0

 

  1. Chen H, Zhao L, Fu K, et al., 2019, Integrin alpha(v)beta(3)- targeted radionuclide therapy combined with immune checkpoint blockade immunotherapy synergistically enhances anti-tumor efficacy. Theranostics, 9: 7948–7960. https://doi.org/10.7150/thno.39203

 

  1. Choi J, Beaino W, Fecek RJ, et al., 2018, Combined VLA- 4-targeted radionuclide therapy and immunotherapy in a mouse model of melanoma. J Nucl Med, 59: 1843–1849. https://doi.org/10.2967/jnumed.118.209510

 

  1. Czernin J, Current K, Mona CE, et al., 2021, Immune-checkpoint blockade enhances 225Ac-PSMA617 efficacy in a mouse model of prostate cancer. J Nucl Med, 62: 228–231. https://doi.org/10.2967/jnumed.120.246041

 

  1. Esfahani SA, De Aguiar Ferreira C, Summer P, et al., 2023, Addition of peptide receptor radiotherapy to immune checkpoint inhibition therapy improves outcomes in neuroendocrine tumors. J Nucl Med, 64: 1056–1061. https://doi.org/10.2967/jnumed.123.265391

 

  1. Feig C, Jones JO, Kraman M, et al., 2013, Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A, 110: 20212–20217. https://doi.org/10.1073/pnas.1320318110

 

  1. Zboralski D, Osterkamp F, Christensen E, et al., 2023, Fibroblast activation protein targeted radiotherapy induces an immunogenic tumor microenvironment and enhances the efficacy of PD-1 immune checkpoint inhibition. Eur J Nucl Med Mol Imaging, 50: 2621–2635. https://doi.org/10.1007/s00259-023-06211-6

 

  1. Fu H, Huang J, Sun L, et al., 2022, FAP-targeted radionuclide therapy of advanced radioiodine-refractory differentiated thyroid cancer with multiple cycles of 177 Lu-FAPI-46. Clin Nucl Med, 47: 906–907. https://doi.org/10.1097/RLU.0000000000004260

 

  1. Rao Z, Zhang Y, Liu L, et al., 2023, 177LuLu-FAP-2286 therapy in a case of right lung squamous cell carcinoma with systemic metastases. Eur J Nucl Med Mol Imaging, 50: 1266–1267. https://doi.org/10.1007/s00259-022-06048-5
Conflict of interest
The authors declare that they have no conflicts of interest.
Share
Back to top
Advances in Radiotherapy & Nuclear Medicine, Electronic ISSN: 2972-4392 Published by AccScience Publishing