51
|
ChallaSivaKanaka S, Vickman RE, Kakarla M, Hayward SW, Franco OE. Fibroblast heterogeneity in prostate carcinogenesis. Cancer Lett 2022; 525:76-83. [PMID: 34715252 PMCID: PMC8788937 DOI: 10.1016/j.canlet.2021.10.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/17/2021] [Accepted: 10/19/2021] [Indexed: 01/30/2023]
Abstract
Our understanding of stromal components, specifically cancer-associated fibroblasts (CAF), in prostate cancer (PCa), has evolved from considering these cells as inert bystanders to acknowledging their significance as players in prostate tumorigenesis. CAF are multifaceted-they promote cancer cell growth, migration and remodel the tumor microenvironment. Although targeting CAF could be a promising strategy for PCa treatment, they incorporate a high but undefined degree of intrinsic cellular heterogeneity. The interaction between CAF subpopulations, with the normal and tumor epithelium and with other cell types is not yet characterized. Defining these interactions and the critical signaling nodes that support tumorigenesis will enable the development of novel strategies to control prostate cancer progression. Here we will discuss the origins, molecular and functional heterogeneity of CAF in PCa. We highlight the challenges associated with delineating CAF heterogeneity and discuss potential areas of research that would assist in expanding our knowledge of CAF and their role in PCa tumorigenesis.
Collapse
Affiliation(s)
- Sathyavathi ChallaSivaKanaka
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Renee E Vickman
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Mamatha Kakarla
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Simon W Hayward
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Omar E Franco
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA. http://
| |
Collapse
|
52
|
Wen X, Xu P, Shi M, Liu J, Zeng X, Zhang Y, Shi C, Li J, Guo Z, Zhang X, Khong PL, Chen X. Evans blue-modified radiolabeled fibroblast activation protein inhibitor as long-acting cancer therapeutics. Am J Cancer Res 2022; 12:422-433. [PMID: 34987657 PMCID: PMC8690933 DOI: 10.7150/thno.68182] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Fibroblast activation protein (FAP) targeted molecular imaging radiotracers have shown promising preclinical and clinical results in tumor diagnosis. However, rapid clearance and inadequate tumor retention of these molecules have hindered them for further clinical translation in cancer therapy. In this study, we aimed to develop a series of albumin binder-truncated Evans blue (EB) modified FAP targeted radiotracers, and optimize the pharmacokinetic (PK) characteristics to overcome the existing limitations in order to apply in the radionuclide therapy of cancer. Methods: A series of compounds with the general structure of EB-FAPI-Bn were synthesized based on a FAP inhibitor (FAPI) variant (FAPI-02) and radiolabeled with 177LuCl3. To verify the binding affinity and FAP targeting specificity of these tracers in vitro, U87MG cell uptake and competition assays were performed. Preclinical PK was evaluated in U87MG tumor-bearing mice using SPECT imaging and biodistribution studies. The lead compound EB-FAPI-B1 was selected and cancer therapeutic efficacy of 177Lu-EB-FAPI-B1 was assessed in U87MG tumor-bearing mice. Results:177Lu-EB-FAPI-B1, B2, B3, B4 were stable in PBS (pH 7.4) and saline for at least 24 h. EB-FAPI-B1 showed high binding affinity (IC50 = 16.5 nM) to FAP in vitro, which was comparable with that of FAPI-02 (IC50 = 10.9 nM). SPECT imaging and biodistribution studies of 177Lu-EB-FAPI-B1, B2, B3, B4 have proved their prominently improved tumor accumulation and retention at 96 h post-injection, especially for 177Lu-EB-FAPI-B1, high tumor uptake and low background signal make it the optimal compound. Compared to the saline group, noteworthy tumor growth inhibitions of 177Lu-EB-FAPI-B1 have been observed after administration of different dosages. Conclusion: In this study, several EB modified FAPI-02 related radiopharmaceuticals have been synthesized successfully and evaluated. High binding affinity and FAP targeting specificity were identified in vitro and in vivo. Remarkably enhanced tumor uptake and retention of EB-FAPI-B1 were found over the unmodified FAPI-02. 177Lu-EB-FAPI-B1 showed remarkable tumor growth suppression in U87MG tumor model with negligible side effects, indicating that 177Lu-EB-FAPI-B1 is promising for clinical application and transformation.
Collapse
|
53
|
Ruan Q, Feng J, Jiang Y, Zhang X, Duan X, Wang Q, Yin G, Xiao D, Zhang J. Preparation and Bioevaluation of 99mTc-Labeled FAP Inhibitors as Tumor Radiotracers to Target the Fibroblast Activation Protein. Mol Pharm 2022; 19:160-171. [PMID: 34904839 DOI: 10.1021/acs.molpharmaceut.1c00712] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fibroblast activation protein (FAP) is overexpressed in cancer-associated fibroblasts (CAFs) in a majority of human epithelial cancers. With low expression in normal organs, FAP has become a promising molecular target for tumor theranostics. To develop a lower cost and more widely available alternative to positron emission tomography (PET), two isocyanide-containing FAP inhibitors (CN-C5-FAPI and CN-PEG4-FAPI) were synthesized and radiolabeled with 99mTc to obtain [99mTc][Tc-(CN-C5-FAPI)6]+ and [99mTc][Tc-(CN-PEG4-FAPI)6]+ in high yields (>95%). They showed good stability in saline and mouse serum. The partition coefficient (log P) values of [99mTc][Tc-(CN-C5-FAPI)6]+ and [99mTc][Tc-(CN-PEG4-FAPI)6]+ were -0.86 ± 0.03 and -2.38 ± 0.07, respectively, indicating that they were good hydrophilic complexes. The low nanomolar IC50 values of CN-C5-FAPI and CN-PEG4-FAPI indicated that they had specificity to FAP. In vitro cellular uptake and blocking experiments implied a FAP-targeted uptake mechanism. The nanomolar Kd values from the saturation binding assay indicated that they had significantly high target affinity to FAP. The biodistribution and blocking study in BALB/c nude mice bearing U87MG tumors showed that both exhibited specific tumor uptake. [99mTc][Tc-(CN-PEG4-FAPI)6]+ showed a higher tumor uptake and a higher tumor/nontarget ratio than [99mTc][Tc-(CN-C5-FAPI)6]+. The results of micro-single-photon emission computed tomography (SPECT) imaging studies of [99mTc][Tc-(CN-C5-FAPI)6]+ and [99mTc][Tc-(CN-PEG4-FAPI)6]+ were in accordance with the biodistribution results, suggesting that [99mTc][Tc-(CN-PEG4-FAPI)6]+ is a promising tumor imaging agent for targeting FAP.
Collapse
Affiliation(s)
- Qing Ruan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Xuran Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Di Xiao
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| |
Collapse
|
54
|
Standing D, Dandawate P, Anant S. Prolactin receptor signaling: A novel target for cancer treatment - Exploring anti-PRLR signaling strategies. Front Endocrinol (Lausanne) 2022; 13:1112987. [PMID: 36714582 PMCID: PMC9880166 DOI: 10.3389/fendo.2022.1112987] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Prolactin (PRL) is a peptide hormone mainly secreted from the anterior pituitary gland. PRL is reported to play a role in pregnancy, mammary gland development, immune modulation, reproduction, and differentiation of islet cells. PRL binds to its receptor PRLR, which belongs to a superfamily of the class I cytokine receptor that has no intrinsic kinase activity. In canonical signaling, PRL binding to PRLR induces downstream signaling including JAK-STAT, AKT and MAPK pathways. This leads to increased cell proliferation, stemness, migration, apoptosis inhibition, and resistance to chemotherapy. PRL-signaling is upregulated in numerous hormone-dependent cancers including breast, prostate, ovarian, and endometrial cancer. However, more recently, the pathway has been reported to play a tumor-promoting role in other cancer types such as colon, pancreas, and hepatocellular cancers. Hence, the signaling pathway is an attractive target for drug development with blockade of the receptor being a potential therapeutic approach. Different strategies have been developed to target this receptor including modification of PRL peptides (Del1-9-G129R-hPRL, G129R-Prl), growth hormone receptor/prolactin receptor bispecific antibody antagonist, neutralizing antibody LFA102, an antibody-drug conjugate (ABBV-176) of the humanized antibody h16f (PR-1594804) and pyrrolobenzodiazepine dimer, a bispecific antibody targeting both PRLR and CD3, an in vivo half-life extended fusion protein containing PRLR antagonist PrlRA and albumin binding domain. There have also been attempts to discover and develop small molecular inhibitors targeting PRLR. Recently, using structure-based virtual screening, we identified a few antipsychotic drugs including penfluridol as a molecule that inhibits PRL-signaling to inhibit PDAC tumor progression. In this review, we will summarize the recent advances in the biology of this receptor in cancer and give an account of PRLR antagonist development for the treatment of cancer.
Collapse
|
55
|
Mukkamala R, Lindeman SD, Kragness KA, Shahriar I, Srinivasarao M, Low PS. Design and Characterization of Fibroblast Activation Protein Targeted Pan-Cancer Imaging Agent for Fluorescence-Guided Surgery of Solid Tumors. J Mater Chem B 2022; 10:2038-2046. [PMID: 35255116 DOI: 10.1039/d1tb02651h] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tumor-targeted fluorescent dyes have been shown to significantly improve a surgeon's ability to locate and resect occult malignant lesions, thereby enhancing a patient’s chances of long term survival. Although several...
Collapse
Affiliation(s)
- Ramesh Mukkamala
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Spencer D Lindeman
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Kate A Kragness
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Imrul Shahriar
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, USA.
| | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
56
|
Liu M, Yang J, Xu B, Zhang X. Tumor metastasis: Mechanistic insights and therapeutic interventions. MedComm (Beijing) 2021; 2:587-617. [PMID: 34977870 PMCID: PMC8706758 DOI: 10.1002/mco2.100] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022] Open
Abstract
Cancer metastasis is responsible for the vast majority of cancer-related deaths worldwide. In contrast to numerous discoveries that reveal the detailed mechanisms leading to the formation of the primary tumor, the biological underpinnings of the metastatic disease remain poorly understood. Cancer metastasis is a complex process in which cancer cells escape from the primary tumor, settle, and grow at other parts of the body. Epithelial-mesenchymal transition and anoikis resistance of tumor cells are the main forces to promote metastasis, and multiple components in the tumor microenvironment and their complicated crosstalk with cancer cells are closely involved in distant metastasis. In addition to the three cornerstones of tumor treatment, surgery, chemotherapy, and radiotherapy, novel treatment approaches including targeted therapy and immunotherapy have been established in patients with metastatic cancer. Although the cancer survival rate has been greatly improved over the years, it is still far from satisfactory. In this review, we provided an overview of the metastasis process, summarized the cellular and molecular mechanisms involved in the dissemination and distant metastasis of cancer cells, and reviewed the important advances in interventions for cancer metastasis.
Collapse
Affiliation(s)
- Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jing Yang
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Bushu Xu
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
57
|
Lindner T, Giesel FL, Kratochwil C, Serfling SE. Radioligands Targeting Fibroblast Activation Protein (FAP). Cancers (Basel) 2021; 13:cancers13225744. [PMID: 34830898 PMCID: PMC8616197 DOI: 10.3390/cancers13225744] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary FAP-targeted radiotracers, recently introduced in cancer treatment, accumulate in Cancer-Associated Fibroblasts (CAFs). CAFs are present in tumor lesions but do not correspond to genuine cancer cells, although they behave in an abnormal and disease-promoting manner. One of their characteristic features, the expression of the surface protein FAP, can be utilized to discriminate between cancerous and healthy tissues. By the choice of an appropriate radionuclide, FAP-targeted tracers can be used for imaging or therapy in many cancer types. Therefore, the first successful application of FAP-targeted imaging has led to an enormous and growing interest in nuclear medicine and radiopharmacy. Abstract Targeting fibroblast activation protein (FAP) in cancer-associated fibroblasts (CAFs) has attracted significant attention in nuclear medicine. Since these cells are present in most cancerous tissues and FAP is rarely expressed in healthy tissues, anti-FAP tracers have a potential as pan-tumor agents. Compared to the standard tumor tracer [18F]FDG, these tracers show better tumor-to-background ratios (TBR) in many indications. Unlike [18F]FDG, FAP-targeted tracers do not require exhausting preparations, such as dietary restrictions on the part of the patient, and offer the possibility of radioligand therapy (RLT) in a theragnostic approach. Although a radiolabeled antibody was clinically investigated as early as the 1990s, the breakthrough event for FAP-targeting in nuclear medicine was the introduction and clinical application of the so-called FAPI-tracers in 2018. From then, the development and application of FAP-targeted tracers became hot topics for the radiopharmaceutical and nuclear medicine community, and attracted the interest of pharmaceutical companies. The aim of this review is to provide a comprehensive overview of the development of FAP-targeted radiopharmaceuticals and their application in nuclear medicine.
Collapse
Affiliation(s)
- Thomas Lindner
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany;
- Correspondence:
| | - Frederik L. Giesel
- Department of Nuclear Medicine, University Hospital Düsseldorf, 40225 Düsseldorf, Germany;
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Sebastian E. Serfling
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany;
| |
Collapse
|
58
|
Kuyumcu S, Sanli Y, Subramaniam RM. Fibroblast-Activated Protein Inhibitor PET/CT: Cancer Diagnosis and Management. Front Oncol 2021; 11:758958. [PMID: 34858834 PMCID: PMC8632139 DOI: 10.3389/fonc.2021.758958] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/18/2021] [Indexed: 01/13/2023] Open
Abstract
Fibroblast activation protein (FAP), overexpressed on cancer-associated fibroblasts (CAFs), is a novel target for molecular imaging of various tumors. Recently, the development of several small-molecule FAP inhibitors for radiolabeling with 68Ga has resulted in the emergence of studies evaluating its clinical role in cancer imaging. Preliminary findings have demonstrated that, in contrast to radiotracers taking advantage of cancer-specific targets such as PSMA and DOTATATE, FAPs as a target are the most promising that can compete with 18FDG in terms of widespread indications. They also have the potential to overcome the shortcomings of 18FDG, particularly false-positive uptake due to inflammatory or infectious processes, low sensitivity in certain cancer types, and radiotherapy planning. In addition, the attractive theranostic properties may facilitate the treatment of many refractory cancers. This review summarizes the current FAP variants and related clinical studies, focusing on radiopharmacy, dosimetry, and diagnostic and theranostic applications.
Collapse
Affiliation(s)
- Serkan Kuyumcu
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yasemin Sanli
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Rathan M. Subramaniam
- Otago Medical School, University of Otago, Dunedin, New Zealand
- Department of Radiology, Duke University, Durham, NC, United States
| |
Collapse
|
59
|
Engineered exosome-like nanovesicles suppress tumor growth by reprogramming tumor microenvironment and promoting tumor ferroptosis. Acta Biomater 2021; 135:567-581. [PMID: 34506976 DOI: 10.1016/j.actbio.2021.09.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 02/05/2023]
Abstract
Tumor vaccines that induce effective and sustained antitumor immunity are highly promising for cancer therapy. However, the antitumor potential of these vaccines is weakened due to the immunosuppressive characteristics of the tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are the most abundant stromal cells within the TME; they play an important role in tumor growth, metastasis, immunosuppression, and drug resistance. Fibroblast activation protein-α (FAP) is overexpressed in CAFs in more than 90% of human tumor tissues. Further, FAP+CAFs are an ideal interstitial target for the immunotherapy of solid tumors. Exosomes derived from tumor cells contain many tumor antigens, which can be used as the basis of tumor vaccines that elicit strong antitumor immunity. Almost all exosome-based cancer vaccines have been designed and developed for tumor parenchymal cells. Moreover, the exosome production is very low and the purification is very difficult, limiting their clinical application as tumor vaccines. In this study, we developed FAP gene-engineered tumor cell-derived exosome-like nanovesicles (eNVs-FAP) as a tumor vaccine that can be prepared easily and in large quantities. The eNVs-FAP vaccine inhibited tumor growth by inducing strong and specific cytotoxic T lymphocyte (CTL) immune responses against tumor cells and FAP+CAFs and reprogramming the immunosuppressive TME in the colon, melanoma, lung, and breast cancer models. Moreover, eNVs-FAP vaccine-activated cellular immune responses could promote tumor ferroptosis by releasing interferon-gamma (IFN-γ) from CTLs and depleting FAP+CAFs. Thus, eNVs-FAP is a candidate tumor vaccine targeting both the tumor parenchyma and the stroma. STATEMENT OF SIGNIFICANCE: Nanovaccines can activate immune cells and promote an antitumor immune response. In this study, we developed the fibroblast activation protein-α (FAP) gene-engineered tumor cell-derived exosome-like vesicle vaccines (eNVs-FAP). A large number of eNVs-FAP were obtained by continuously squeezing FAP gene-engineered tumor cells. eNVs-FAP showed excellent antitumor effects in a variety of tumor-bearing mouse models. The mechanistic analysis showed that eNVs-FAP promoted the maturation of dendritic cells (DCs), increased the infiltration of effector T cells into target tumor cells and FAP-positive cancer-associated fibroblasts (FAP+CAFs), and reduced the proportion of immunosuppressive cells, including M2-like tumor-associated macrophages (M2-TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), in the tumor microenvironment (TME). Moreover, the clearance of FAP+CAFs helped enhance interferon-gamma-induced tumor cell ferroptosis.
Collapse
|
60
|
Juillerat-Jeanneret L, Tafelmeyer P, Golshayan D. Regulation of Fibroblast Activation Protein-α Expression: Focus on Intracellular Protein Interactions. J Med Chem 2021; 64:14028-14045. [PMID: 34523930 DOI: 10.1021/acs.jmedchem.1c01010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prolyl-specific peptidase fibroblast activation protein-α (FAP-α) is expressed at very low or undetectable levels in nondiseased human tissues but is selectively induced in activated (myo)fibroblasts at sites of tissue remodeling in fibrogenic processes. In normal regenerative processes involving transient fibrosis FAP-α+(myo)fibroblasts disappear from injured tissues, replaced by cells with a normal FAP-α- phenotype. In chronic uncontrolled pathological fibrosis FAP-α+(myo)fibroblasts permanently replace normal tissues. The mechanisms of regulation and elimination of FAP-α expression in(myo)fibroblasts are unknown. According to a yeast two-hybrid screen and protein databanks search, we propose that the intracellular (co)-chaperone BAG6/BAT3 can interact with FAP-α, mediated by the BAG6/BAT3 Pro-rich domain, inducing proteosomal degradation of FAP-α protein under tissue homeostasis. In this Perspective, we discuss our findings in the context of current knowledge on the regulation of FAP-α expression and comment potential therapeutic strategies for uncontrolled fibrosis, including small molecule degraders (PROTACs)-modified FAP-α targeted inhibitors.
Collapse
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland.,University Institute of Pathology, CHUV and UNIL, CH1011 Lausanne, Switzerland
| | - Petra Tafelmeyer
- Hybrigenics Services, Laboratories and Headquarters-Paris, 1 rue Pierre Fontaine, 91000 Evry, France.,Hybrigenics Corporation, Cambridge Innovation Center, 50 Milk Street, Cambridge, Massachusetts 02142, United States
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland
| |
Collapse
|
61
|
Imlimthan S, Moon ES, Rathke H, Afshar-Oromieh A, Rösch F, Rominger A, Gourni E. New Frontiers in Cancer Imaging and Therapy Based on Radiolabeled Fibroblast Activation Protein Inhibitors: A Rational Review and Current Progress. Pharmaceuticals (Basel) 2021; 14:1023. [PMID: 34681246 PMCID: PMC8540221 DOI: 10.3390/ph14101023] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022] Open
Abstract
Over the past decade, the tumor microenvironment (TME) has become a new paradigm of cancer diagnosis and therapy due to its unique biological features, mainly the interconnection between cancer and stromal cells. Within the TME, cancer-associated fibroblasts (CAFs) demonstrate as one of the most critical stromal cells that regulate tumor cell growth, progression, immunosuppression, and metastasis. CAFs are identified by various biomarkers that are expressed on their surfaces, such as fibroblast activation protein (FAP), which could be utilized as a useful target for diagnostic imaging and treatment. One of the advantages of targeting FAP-expressing CAFs is the absence of FAP expression in quiescent fibroblasts, leading to a controlled targetability of diagnostic and therapeutic compounds to the malignant tumor stromal area using radiolabeled FAP-based ligands. FAP-based radiopharmaceuticals have been investigated strenuously for the visualization of malignancies and delivery of theranostic radiopharmaceuticals to the TME. This review provides an overview of the state of the art in TME compositions, particularly CAFs and FAP, and their roles in cancer biology. Moreover, relevant reports on radiolabeled FAP inhibitors until the year 2021 are highlighted-as well as the current limitations, challenges, and requirements for those radiolabeled FAP inhibitors in clinical translation.
Collapse
Affiliation(s)
- Surachet Imlimthan
- Department of Nuclear Medicine, the Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.I.); (H.R.); (A.A.-O.); (A.R.)
| | - Euy Sung Moon
- Department of Chemistry—TRIGA Site, Johannes Gutenberg—University Mainz, 55128 Mainz, Germany; (E.S.M.); (F.R.)
| | - Hendrik Rathke
- Department of Nuclear Medicine, the Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.I.); (H.R.); (A.A.-O.); (A.R.)
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, the Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.I.); (H.R.); (A.A.-O.); (A.R.)
| | - Frank Rösch
- Department of Chemistry—TRIGA Site, Johannes Gutenberg—University Mainz, 55128 Mainz, Germany; (E.S.M.); (F.R.)
| | - Axel Rominger
- Department of Nuclear Medicine, the Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.I.); (H.R.); (A.A.-O.); (A.R.)
| | - Eleni Gourni
- Department of Nuclear Medicine, the Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.I.); (H.R.); (A.A.-O.); (A.R.)
| |
Collapse
|
62
|
Xin L, Gao J, Zheng Z, Chen Y, Lv S, Zhao Z, Yu C, Yang X, Zhang R. Fibroblast Activation Protein-α as a Target in the Bench-to-Bedside Diagnosis and Treatment of Tumors: A Narrative Review. Front Oncol 2021; 11:648187. [PMID: 34490078 PMCID: PMC8416977 DOI: 10.3389/fonc.2021.648187] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Fibroblast activation protein-α (FAP) is a type II integral serine protease that is specifically expressed by activated fibroblasts. Cancer-associated fibroblasts (CAFs) in the tumor stroma have an abundant and stable expression of FAP, which plays an important role in promoting tumor growth, invasion, metastasis, and immunosuppression. For example, in females with a high incidence of breast cancer, CAFs account for 50–70% of the cells in the tumor’s microenvironment. CAF overexpression of FAP promotes tumor development and metastasis by influencing extracellular matrix remodeling, intracellular signaling, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. This review discusses the basic biological characteristics of FAP and its applications in the diagnosis and treatment of various cancers. We review the emerging basic and clinical research data regarding the use of nanomaterials that target FAP.
Collapse
Affiliation(s)
- Lei Xin
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Ziliang Zheng
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yiyou Chen
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuxin Lv
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Zhikai Zhao
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunhai Yu
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaotang Yang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiping Zhang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
63
|
Kuklik J, Michelfelder S, Schiele F, Kreuz S, Lamla T, Müller P, Park JE. Development of a Bispecific Antibody-Based Platform for Retargeting of Capsid Modified AAV Vectors. Int J Mol Sci 2021; 22:ijms22158355. [PMID: 34361120 PMCID: PMC8347852 DOI: 10.3390/ijms22158355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
A major limiting factor for systemically delivered gene therapies is the lack of novel tissue specific AAV (Adeno-associated virus) derived vectors. Bispecific antibodies can be used to redirect AAVs to specific target receptors. Here, we demonstrate that the insertion of a short linear epitope “2E3” derived from human proprotein-convertase subtilisin/kexin type 9 (PCSK9) into different surface loops of the VP capsid proteins can be used for AAV de-targeting from its natural receptor(s), combined with a bispecific antibody-mediated retargeting. We chose to target a set of distinct disease relevant membrane proteins—fibroblast activation protein (FAP), which is upregulated on activated fibroblasts within the tumor stroma and in fibrotic tissues, as well as programmed death-ligand 1 (PD-L1), which is strongly upregulated in many cancers. Upon incubation with a bispecific antibody recognizing the 2E3 epitope and FAP or PD-L1, the bispecific antibody/rAAV complex was able to selectively transduce receptor positive cells. In summary, we developed a novel, rationally designed vector retargeting platform that can target AAVs to a new set of cellular receptors in a modular fashion. This versatile platform may serve as a valuable tool to investigate the role of disease relevant cell types and basis for novel gene therapy approaches.
Collapse
Affiliation(s)
- Juliane Kuklik
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387 Biberach an der Riss, Germany;
| | - Stefan Michelfelder
- Division of Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387 Biberach an der Riss, Germany; (S.M.); (S.K.)
| | - Felix Schiele
- Division of Biotherapeutics Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387 Biberach an der Riss, Germany;
| | - Sebastian Kreuz
- Division of Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387 Biberach an der Riss, Germany; (S.M.); (S.K.)
- Boehringer Ingelheim Venture Fund GmbH, 55216 Ingelheim am Rhein, Germany;
| | - Thorsten Lamla
- Division of Drug Discovery Sciences Biberach, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387 Biberach an der Riss, Germany;
| | - Philipp Müller
- Boehringer Ingelheim Venture Fund GmbH, 55216 Ingelheim am Rhein, Germany;
| | - John E. Park
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387 Biberach an der Riss, Germany;
- Correspondence:
| |
Collapse
|
64
|
Ansardamavandi A, Tafazzoli-Shadpour M. The functional cross talk between cancer cells and cancer associated fibroblasts from a cancer mechanics perspective. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119103. [PMID: 34293346 DOI: 10.1016/j.bbamcr.2021.119103] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 12/12/2022]
Abstract
The function of biological tissues in health and disease is regulated at cellular level and is highly influenced by the physical microenvironment, through the interaction of forces between cells and ECM, which are perceived through mechanosensing pathways. In cancer, both chemical and physical signaling cascades and their interactions are involved during cell-cell and cell-ECM communications to meet requirements of tumor growth. Among stroma cells, cancer associated fibroblasts (CAFs) play key role in tumor growth and pave the way for cancer cells to initiate metastasis and invasion to other tissues, and without recruitment of CAFs, the process of cancer invasion is dysfunctional. This is through an intense chemical and physical cross talks with tumor cells, and interactive remodeling of ECM. During such interaction CAFs apply traction forces and depending on the mechanical properties, deform ECM and in return receive physical signals from the micromechanical environment. Such interaction leads to ECM remodeling by manipulating ECM structure and its mechanical properties. The results are in form of deposition of extra fibers, stiffening, rearrangement and reorganization of fibrous structure, and degradation which are due to a complex secretion and expression of different markers triggered by mechanosensing of tumor cells, specially CAFs. Such events define cancer progress and invasion of cancer cells. A systemic knowledge of chemical and physical factors provides a holistic view of how cancer process and enhances the current treatment methods to provide more diversity among targets that involves tumor cells and ECM structure.
Collapse
Affiliation(s)
- Arian Ansardamavandi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | |
Collapse
|
65
|
Maia A, Wiemann S. Cancer-Associated Fibroblasts: Implications for Cancer Therapy. Cancers (Basel) 2021; 13:3526. [PMID: 34298736 PMCID: PMC8307167 DOI: 10.3390/cancers13143526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Tumour cells do not exist as an isolated entity. Instead, they are surrounded by and closely interact with cells of the environment they are emerged in. The tumour microenvironment (TME) is not static and several factors, including cancer cells and therapies, have been described to modulate several of its components. Fibroblasts are key elements of the TME with the capacity to influence tumour progression, invasion and response to therapy, which makes them attractive targets in cancer treatment. In this review, we focus on fibroblasts and their numerous roles in the TME with a special attention to recent findings describing their heterogeneity and role in therapy response. Furthermore, we explore how different therapies can impact these cells and their communication with cancer cells. Finally, we highlight potential strategies targeting this cell type that can be employed for improving patient outcome.
Collapse
Affiliation(s)
- Ana Maia
- German Cancer Research Center (DKFZ), Division of Molecular Genome Analysis, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Stefan Wiemann
- German Cancer Research Center (DKFZ), Division of Molecular Genome Analysis, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
66
|
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S, Zhou H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther 2021; 6:218. [PMID: 34108441 PMCID: PMC8190181 DOI: 10.1038/s41392-021-00641-0] [Citation(s) in RCA: 384] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ye Wang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qingxiang Zeng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuzhi Deng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
67
|
Zhang X, Chen D, Babich JW, Green SJE, Deng XH, Rodeo SA. In Vivo Imaging of Fibroblast Activity Using a 68Ga-Labeled Fibroblast Activation Protein Alpha (FAP-α) Inhibitor: Study in a Mouse Rotator Cuff Repair Model. J Bone Joint Surg Am 2021; 103:e40. [PMID: 33587512 DOI: 10.2106/jbjs.20.00831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Rotator cuff repair site failure is a well-established clinical concern. Tendon-to-bone healing is initiated by inflammatory mediators followed by matrix synthesis by fibroblasts. The kinetics of fibroblast accumulation and activity are currently poorly understood. METHODS Ninety-six mice underwent supraspinatus tendon repair. Six were used for imaging using a novel 68Gallium (Ga)-labeled fibroblast activation protein alpha (FAP-α) inhibitor and positron emission tomography-computed tomography (PET/CT) at days 0 (before surgery), 3, 7, 14, and 28. Sixty-eight animals were divided into 4 groups to be evaluated at 3, 7, 14, or 28 days. Twenty-two native shoulders from mice without surgery were used as the control group (intact tendon). Six animals from each group were used for histological analysis; 6 from each group were used for evaluation of fibroblastic response-related gene expression; and 10 mice each from the intact, 14-day, and 28-day groups were used for biomechanical testing. RESULTS There was minimal localization of 68Ga-labeled FAP-α inhibitor in the shoulders at day 0 (before surgery). There was significantly increased uptake in the shoulders with surgery compared with the contralateral sides without surgery at 3, 7, and 14 days. 68Ga-labeled FAP-α inhibitor uptake in the surgically treated shoulders increased gradually and peaked at 14 days followed by a decrease at 28 days. Gene expression for smooth muscle alpha (α)-2 (acta2), FAP-α, and fibronectin increased postsurgery followed by a drop at 28 days. Immunohistochemical analysis showed that FAP-α-positive cell density followed a similar temporal trend, peaking at 14 days. All trends matched closely with the PET/CT results. Biomechanical testing demonstrated a gradual increase in failure load during the healing process. CONCLUSIONS 68Ga-labeled FAP-α inhibitor PET/CT allows facile, high-contrast in vivo 3-dimensional imaging of fibroblastic activity in a mouse rotator cuff repair model. CLINICAL RELEVANCE Noninvasive imaging of activated fibroblasts using labeled radiotracers may be a valuable tool to follow the progression of healing at the bone-tendon interface.
Collapse
Affiliation(s)
- Xueying Zhang
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY.,Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of, China
| | - Daoyun Chen
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY
| | - John W Babich
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY
| | - Samuel J E Green
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY
| | - Xiang-Hua Deng
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY
| | - Scott A Rodeo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY
| |
Collapse
|
68
|
|
69
|
An ultra-high-affinity small organic ligand of fibroblast activation protein for tumor-targeting applications. Proc Natl Acad Sci U S A 2021; 118:2101852118. [PMID: 33850024 DOI: 10.1073/pnas.2101852118] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We describe the development of OncoFAP, an ultra-high-affinity ligand of fibroblast activation protein (FAP) for targeting applications with pan-tumoral potential. OncoFAP binds to human FAP with affinity in the subnanomolar concentration range and cross-reacts with the murine isoform of the protein. We generated various fluorescent and radiolabeled derivatives of OncoFAP in order to study biodistribution properties and tumor-targeting performance in preclinical models. Fluorescent derivatives selectively localized in FAP-positive tumors implanted in nude mice with a rapid and homogeneous penetration within the neoplastic tissue. Quantitative in vivo biodistribution studies with a lutetium-177-labeled derivative of OncoFAP revealed a preferential localization in tumors at doses of up to 1,000 nmol/kg. More than 30% of the injected dose had already accumulated in 1 g of tumor 10 min after intravenous injection and persisted for at least 3 h with excellent tumor-to-organ ratios. OncoFAP also served as a modular component for the generation of nonradioactive therapeutic products. A fluorescein conjugate mediated a potent and FAP-dependent tumor cell killing activity in combination with chimeric antigen receptor (CAR) T cells specific to fluorescein. Similarly, a conjugate of OncoFAP with the monomethyl auristatin E-based Vedotin payload was well tolerated and cured tumor-bearing mice in combination with a clinical-stage antibody-interleukin-2 fusion. Collectively, these data support the development of OncoFAP-based products for tumor-targeting applications in patients with cancer.
Collapse
|
70
|
Slania SL, Das D, Lisok A, Du Y, Jiang Z, Mease RC, Rowe SP, Nimmagadda S, Yang X, Pomper MG. Imaging of Fibroblast Activation Protein in Cancer Xenografts Using Novel (4-Quinolinoyl)-glycyl-2-cyanopyrrolidine-Based Small Molecules. J Med Chem 2021; 64:4059-4070. [PMID: 33730493 PMCID: PMC8214312 DOI: 10.1021/acs.jmedchem.0c02171] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fibroblast activation protein (FAP) has become a favored target for imaging and therapy of malignancy. We have synthesized and characterized two new (4-quinolinoyl)-glycyl-2-cyanopyrrolidine-based small molecules for imaging of FAP, QCP01 and [111In]QCP02, using optical and single-photon computed tomography/CT, respectively. Binding of imaging agents to FAP was assessed in six human cancer cell lines of different cancer types: glioblastoma (U87), melanoma (SKMEL24), prostate (PC3), NSCLC (NCIH2228), colorectal carcinoma (HCT116), and lung squamous cell carcinoma (NCIH226). Mouse xenograft models were developed with FAP-positive U87 and FAP-negative PC3 cells to test pharmacokinetics and binding specificity in vivo. QCP01 and [111In]QCP02 demonstrated nanomolar inhibition of FAP at Ki values of 1.26 and 16.20 nM, respectively. Both were selective for FAP over DPP-IV, a related serine protease. Both enabled imaging of FAP-expressing tumors specifically in vivo. [111In]QCP02 showed high uptake at 18.2 percent injected dose per gram in the U87 tumor at 30 min post-administration.
Collapse
Affiliation(s)
- Stephanie L Slania
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Deepankar Das
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Ala Lisok
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Yong Du
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Zirui Jiang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Ronnie C Mease
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Xing Yang
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Martin G Pomper
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
71
|
Jung HJ, Nam EH, Park JY, Ghosh P, Kim IS. Identification of BR102910 as a selective fibroblast activation protein (FAP) inhibitor. Bioorg Med Chem Lett 2021; 37:127846. [PMID: 33571650 DOI: 10.1016/j.bmcl.2021.127846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/23/2021] [Accepted: 01/30/2021] [Indexed: 01/10/2023]
Abstract
Fibroblast activation protein (FAP) belongs to the family of prolyl-specific serine proteases and displays both exopeptidase and endopeptidase activities. FAP expression is undetectable in most normal adult tissues, but is greatly upregulated in sites of tissue remodeling, which include fibrosis, inflammation and cancer. Due to its restricted expression pattern and dual enzymatic activities, FAP inhibition is investigated as a therapeutic option for several diseases. In the present study, we described the structure-activity relationship of several synthesized compounds against DPPIV and prolyl oligopeptidase (PREP). In particular, BR102910 (compound 24) showed nanomolar potency and high selectivity. Moreover, the in vivo FAP inhibition study of BR102910 (compound 24) using C57BL/6J mice demonstrated exceptional profiles and satisfactory FAP inhibition efficacy. Based on excellent in vitro and in vivo profiles, the potential of BR102910 (compound 24) as a lead candidate for the treatment of type 2 diabetes is considered.
Collapse
Affiliation(s)
- Hui Jin Jung
- Research Center, Boryung Pharmaceuticals Co. Ltd., Ansan 15425, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Eun Hye Nam
- Research Center, Boryung Pharmaceuticals Co. Ltd., Ansan 15425, Republic of Korea
| | - Jin Young Park
- Research Center, Boryung Pharmaceuticals Co. Ltd., Ansan 15425, Republic of Korea
| | - Prithwish Ghosh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
72
|
Zhou S, Zhen Z, Paschall AV, Xue L, Yang X, Bebin-Blackwell AG, Cao Z, Zhang W, Wang M, Teng Y, Zhou G, Li Z, Avci FY, Tang W, Xie J. FAP-Targeted Photodynamic Therapy Mediated by Ferritin Nanoparticles Elicits an Immune Response against Cancer Cells and Cancer Associated Fibroblasts. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007017. [PMID: 35822179 PMCID: PMC9273013 DOI: 10.1002/adfm.202007017] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 06/15/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are present in many types of tumors and play a pivotal role in tumor progression and immunosuppression. Fibroblast-activation protein (FAP), which is overexpressed on CAFs, has been indicated as a universal tumor target. However, FAP expression is not restricted to tumors, and systemic treatment against FAP often causes severe side effects. To solve this problem, a photodynamic therapy (PDT) approach was developed based on ZnF16Pc (a photosensitizer)-loaded and FAP-specific single chain variable fragment (scFv)-conjugated apoferritin nanoparticles, or αFAP-Z@FRT. αFAP-Z@FRT PDT efficiently eradicates CAFs in tumors without inducing systemic toxicity. When tested in murine 4T1 models, the PDT treatment elicits anti-cancer immunity, causing suppression of both primary and distant tumors, i.e. abscopal effect. Treatment efficacy is enhanced when αFAP-Z@FRT PDT is used in combination with anti-PD1 antibodies. Interestingly, it is found that the PDT treatment not only elicits a cellular immunity against cancer cells, but also stimulates an anti-CAFs immunity. This is supported by an adoptive cell transfer study, where T cells taken from 4T1-tumor-bearing animals treated with αFAP PDT retard the growth of A549 tumors established on nude mice. Overall, our approach is unique for permitting site-specific eradication of CAFs and inducing a broad spectrum anti-cancer immunity.
Collapse
Affiliation(s)
- Shiyi Zhou
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Zipeng Zhen
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Amy V Paschall
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine and Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Lijun Xue
- Department of Medical Oncology, Jinling Hospital, Nanjing University Clinical School of Medicine, Nanjing, Jiangsu 210002, China
| | - Xueyuan Yang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | | | - Zhengwei Cao
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Mengzhe Wang
- Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yong Teng
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fikri Y Avci
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine and Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Wei Tang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
73
|
Abstract
Fibroblast activation protein-α (FAP) is a type-II transmembrane serine protease expressed almost exclusively to pathological conditions including fibrosis, arthritis, and cancer. Across most cancer types, elevated FAP is associated with worse clinical outcomes. Despite the clear association between FAP and disease severity, the biological reasons underlying these clinical observations remain unclear. Here we review basic FAP biology and FAP's role in non-oncologic and oncologic disease. We further explore how FAP may worsen clinical outcomes via its effects on extracellular matrix remodeling, intracellular signaling regulation, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. Lastly, we discuss the potential to exploit FAP biology to improve clinical outcomes.
Collapse
Affiliation(s)
- Allison A Fitzgerald
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3870 Reservoir Road NW, Washington, DC, 20057, USA
| | - Louis M Weiner
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3870 Reservoir Road NW, Washington, DC, 20057, USA.
| |
Collapse
|
74
|
Hicks RJ, Roselt PJ, Kallur KG, Tothill RW, Mileshkin L. FAPI PET/CT: Will It End the Hegemony of 18F-FDG in Oncology? J Nucl Med 2020; 62:296-302. [PMID: 33277397 DOI: 10.2967/jnumed.120.256271] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
For over 40 years, 18F-FDG has been the dominant PET tracer in neurology, cardiology, inflammatory diseases, and, most particularly, oncology. Combined with the ability to perform whole-body scanning, 18F-FDG has revolutionized the evaluation of cancer and has stifled the adoption of other tracers, except in situations where low avidity or high background activity limits diagnostic performance. The strength of 18F-FDG has generally been its ability to detect disease in the absence of structural abnormality, thereby enhancing diagnostic sensitivity, but its simultaneous weakness has been a lack of specificity due to diverse pathologies with enhanced glycolysis. Radiotracers that leverage other hallmarks of cancer or specific cell-surface targets are gradually finding a niche in the diagnostic armamentarium. However, none have had sufficient sensitivity to realistically compete with 18F-FDG for evaluation of the broad spectrum of malignancies. Perhaps, this situation is about to change with development of a class of tracers targeting fibroblast activation protein that have low uptake in almost all normal tissues but high uptake in most cancer types. In this review, the development and exciting preliminary clinical data relating to various fibroblast activation protein-specific small-molecule inhibitor tracers in oncology will be discussed along with potential nononcologic applications.
Collapse
Affiliation(s)
- Rodney J Hicks
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Roselt
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Richard W Tothill
- Department of Clinical Pathology and Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Linda Mileshkin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
75
|
Meng Q, Luo X, Chen J, Wang D, Chen E, Zhang W, Zhang G, Zhou W, Xu J, Song Z. Unmasking carcinoma-associated fibroblasts: Key transformation player within the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2020; 1874:188443. [DOI: 10.1016/j.bbcan.2020.188443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022]
|
76
|
Abstract
Cancer-associated fibroblasts (CAFs) are the key component of tumor stromal. High heterogeneity of CAFs reflects in their origin, phenotype and function. Biological function which can be suggested by biomarkers of distinct CAF subgroups may be different, even opposite, just like water and fire. Identifying CAF subpopulations expressing different biomarkers and reconciling the relationship of the "water and fire" among distinct CAF subsets may be a breakthrough in tumor therapy. Herein, we briefly summarize the biomarkers commonly used or newly identified for distinct CAFs in terms of their features and potential clinical benefits.
Collapse
|
77
|
Hintz HM, Gallant JP, Vander Griend DJ, Coleman IM, Nelson PS, LeBeau AM. Imaging Fibroblast Activation Protein Alpha Improves Diagnosis of Metastatic Prostate Cancer with Positron Emission Tomography. Clin Cancer Res 2020; 26:4882-4891. [PMID: 32636317 PMCID: PMC7683011 DOI: 10.1158/1078-0432.ccr-20-1358] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Metastatic castration-resistant prostate cancer (mCRPC) is a lethal, heterogeneous disease with few therapeutic strategies that significantly prolong survival. Innovative therapies for mCRPC are needed; however, the development of new therapies relies on accurate imaging to assess metastasis and monitor response. Standard imaging modalities for prostate cancer require improvement and there remains a need for selective and sensitive imaging probes that can be widely used in patients with mCRPC. EXPERIMENTAL DESIGN We evaluated the transmembrane protease fibroblast activation protein alpha (FAP) as a targetable cell surface antigen for mCRPC. Genomic and IHC analyses were performed to investigate FAP expression in prostate cancer. Our FAP-targeted antibody imaging probe, [89Zr]Zr-B12 IgG, was evaluated by PET/CT imaging in preclinical prostate cancer models. RESULTS Analysis of patient data documented FAP overexpression in metastatic disease across tumor subtypes. PET imaging with [89Zr]Zr-B12 IgG demonstrated high tumor uptake and long-term retention of the probe in the preclinical models examined. FAP-positive stroma tumor uptake of [89Zr]Zr-B12 IgG was 5-fold higher than the isotype control with mean %ID/cc of 34.13 ± 1.99 versus 6.12 ± 2.03 (n = 3/group; P = 0.0006) at 72 hours. Ex vivo biodistribution corroborated these results documenting rapid blood clearance by 24 hours and high tumor uptake of [89Zr]Zr-B12 IgG by 72 hours. CONCLUSIONS Our study reveals FAP as a target for imaging the tumor microenvironment of prostate cancer. Validation of [89Zr]Zr-B12 IgG as a selective imaging probe for FAP-expressing tumors presents a new approach for noninvasive PET/CT imaging of mCRPC.
Collapse
Affiliation(s)
- Hallie M Hintz
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Joseph P Gallant
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Donald J Vander Griend
- Department of Pathology and Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Ilsa M Coleman
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Peter S Nelson
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aaron M LeBeau
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota.
| |
Collapse
|
78
|
Pandya DN, Sinha A, Yuan H, Mutkus L, Stumpf K, Marini FC, Wadas TJ. Imaging of Fibroblast Activation Protein Alpha Expression in a Preclinical Mouse Model of Glioma Using Positron Emission Tomography. Molecules 2020; 25:molecules25163672. [PMID: 32806623 PMCID: PMC7464128 DOI: 10.3390/molecules25163672] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive glioma of the primary central nervous system. Due to the lack of effective treatment options, the prognosis for patients remains bleak. Fibroblast activation protein alpha (FAP), a 170 kDa type II transmembrane serine protease was observed to be expressed on glioma cells and within the glioma tumor microenvironment. To understand the utility of targeting FAP in this tumor type, the immuno-PET radiopharmaceutical [89Zr]Zr-Df-Bz-F19 mAb was prepared and Lindmo analysis was used for its in vitro evaluation using the U87MG cell line, which expresses FAP endogenously. Lindmo analysis revealed an association constant (Ka) of 10-8 M-1 and an immunoreactivity of 52%. Biodistribution studies in U87MG tumor-bearing mice revealed increasing radiotracer retention in tumors over time, leading to average tumor-to-muscle ratios of 3.1, 7.3, 7.2, and 8.3 at 2, 24, 48 and 72 h, respectively. Small animal PET corroborated the biodistribution studies; tumor-to-muscle ratios at 2, 24, 48, and 72 h were 2.0, 5.0, 6.1 and 7.8, respectively. Autoradiography demonstrated accumulated activity throughout the interior of FAP+ tumors, while sequential tumor sections stained positively for FAP expression. Conversely, FAP- tissues retained minimal radioactivity and were negative for FAP expression by immunohistochemistry. These results demonstrate FAP as a promising biomarker that may be exploited to diagnose and potentially treat GBM and other neuroepithelial cancers.
Collapse
Affiliation(s)
- Darpan N. Pandya
- Department of Radiology, University of Iowa, Iowa City, IA 52242, USA; (D.N.P.); (A.S.)
| | - Akesh Sinha
- Department of Radiology, University of Iowa, Iowa City, IA 52242, USA; (D.N.P.); (A.S.)
| | - Hong Yuan
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Lysette Mutkus
- Department of Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (L.M.); (K.S.); (F.C.M.)
| | - Kristina Stumpf
- Department of Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (L.M.); (K.S.); (F.C.M.)
| | - Frank C. Marini
- Department of Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (L.M.); (K.S.); (F.C.M.)
| | - Thaddeus J. Wadas
- Department of Radiology, University of Iowa, Iowa City, IA 52242, USA; (D.N.P.); (A.S.)
- Correspondence: ; Tel.: +1-319-335-5009
| |
Collapse
|
79
|
Barnhoorn MC, Hakuno SK, Bruckner RS, Rogler G, Hawinkels LJAC, Scharl M. Stromal Cells in the Pathogenesis of Inflammatory Bowel Disease. J Crohns Colitis 2020; 14:995-1009. [PMID: 32160284 PMCID: PMC7392167 DOI: 10.1093/ecco-jcc/jjaa009] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Up till now, research on inflammatory bowel disease [IBD] has mainly been focused on the immune cells present in the gastrointestinal tract. However, recent insights indicate that stromal cells also play an important and significant role in IBD pathogenesis. Stromal cells in the intestines regulate both intestinal epithelial and immune cell homeostasis. Different subsets of stromal cells have been found to play a role in other inflammatory diseases [e.g. rheumatoid arthritis], and these various stromal subsets now appear to carry out also specific functions in the inflamed gut in IBD. Novel potential therapies for IBD utilize, as well as target, these pathogenic stromal cells. Injection of mesenchymal stromal cells [MSCs] into fistula tracts of Crohn's disease patients is already approved and used in clinical settings. In this review we discuss the current knowledge of the role of stromal cells in IBD pathogenesis. We further outline recent attempts to modify the stromal compartment in IBD with agents that target or replace the pathogenic stroma.
Collapse
Affiliation(s)
- M C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands,Corresponding author: Prof. Dr Michael Scharl, Department of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, Zurich 8091, Switzerland. Tel: 41 44 255 3419; Fax: 41 44 255 9497;
| | - S K Hakuno
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - R S Bruckner
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands,Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - G Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - L J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - M Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
80
|
Langbein T, Weber WA, Eiber M. Future of Theranostics: An Outlook on Precision Oncology in Nuclear Medicine. J Nucl Med 2020; 60:13S-19S. [PMID: 31481583 DOI: 10.2967/jnumed.118.220566] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/24/2019] [Indexed: 01/13/2023] Open
Abstract
Molecular alterations in malignant disease result in the expression or upregulations of various targets that can be used for imaging and treatment with radiopharmaceuticals. This theranostic principle has acquired greater importance in personalized medicine in recent years, particularly in oncology, where advanced tumors can be treated effectively with low side effects. Since the pioneering use of 131I in differentiated thyroid cancer in the 1940s, remarkable achievements in nuclear medicine endoradiotherapy have been demonstrated, mainly in the treatment of neuroendocrine neoplasms by using 177Lu-labeled somatostatin analogs or in the treatment of advanced prostate cancer using prostate-specific membrane antigen-directed radionuclide therapy. Besides that, this review focuses on promising novel radiopharmaceuticals and describes their preclinical and clinical status. Radiolabeled antibodies, such as 131I-omburtamab directed against the B7-H3 protein on the surface of neuroblastoma cells; HuMab-5B1, a 89Zr/177Lu-labeled antibody for the treatment of CA19-9-expressing malignancies; and 177Lu-lilotomab, a CD37 antibody for the treatment of B-cell lymphomas, are being highlighted. The neurotensin receptor ligand 111In/177Lu-3B-227 has demonstrated high potential in imaging and therapy for several malignancies (e.g., pancreatic adenocarcinomas). Targeting of the fibroblast activation protein is currently being explored for different tumor entities using PET imaging with the fibroblast activation protein inhibitor (FAPI) 68Ga-FAPI-04, and the first therapeutic applications of 90Y-FAPI-04 have been applied. After 2 decades of rapid development in theranostics, a variety of new targets are available for further clinical investigation.
Collapse
Affiliation(s)
- Thomas Langbein
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
81
|
Roy J, Hettiarachchi SU, Kaake M, Mukkamala R, Low PS. Design and validation of fibroblast activation protein alpha targeted imaging and therapeutic agents. Theranostics 2020; 10:5778-5789. [PMID: 32483418 PMCID: PMC7254991 DOI: 10.7150/thno.41409] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/03/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Cancer-associated fibroblasts (CAFs) comprise a major cell type in the tumor microenvironment where they support tumor growth and survival by producing extracellular matrix, secreting immunosuppressive cytokines, releasing growth factors, and facilitating metastases. Because tumors with elevated CAFs are characterized by poorer prognosis, considerable effort is focused on developing methods to quantitate, suppress and/or eliminate CAFs. We exploit the elevated expression of fibroblast activation protein (FAP) on CAFs to target imaging and therapeutic agents selectively to these fibroblasts in solid tumors. Methods: FAP-targeted optical imaging, radioimaging, and chemotherapeutic agents were synthesized by conjugating FAP ligand (FL) to either a fluorescent dye, technetium-99m, or tubulysin B hydrazide. In vitro and in vivo studies were performed to determine the specificity and selectivity of each conjugate for FAP in vitro and in vivo. Results: FAP-targeted imaging and therapeutic conjugates showed high binding specificity and affinity in the low nanomolar range. Injection of FAP-targeted 99mTc into tumor-bearing mice enabled facile detection of tumor xenografts with little off-target uptake. Optical imaging of malignant lesions was also readily achieved following intravenous injection of FAP-targeted near-infrared fluorescent dye. Finally, systemic administration of a tubulysin B conjugate of FL promoted complete eradication of solid tumors with no evidence of gross toxicity to the animals. Conclusion: In view of the near absence of FAP on healthy cells, we conclude that targeting of FAP on cancer-associated fibroblasts can enable highly specific imaging and therapy of solid tumors.
Collapse
Affiliation(s)
| | | | | | | | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
82
|
Toms J, Kogler J, Maschauer S, Daniel C, Schmidkonz C, Kuwert T, Prante O. Targeting Fibroblast Activation Protein: Radiosynthesis and Preclinical Evaluation of an 18F-Labeled FAP Inhibitor. J Nucl Med 2020; 61:1806-1813. [PMID: 32332144 DOI: 10.2967/jnumed.120.242958] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/05/2020] [Indexed: 12/30/2022] Open
Abstract
Fibroblast activation protein (FAP) has emerged as an interesting molecular target used in the imaging and therapy of various types of cancers. 68Ga-labeled chelator-linked FAP inhibitors (FAPIs) have been successfully applied to PET imaging of various tumor types. To broaden the spectrum of applicable PET tracers for extended imaging studies of FAP-dependent diseases, we herein report the radiosynthesis and preclinical evaluation of an 18F-labeled glycosylated FAPI ([18F]FGlc-FAPI). Methods: An alkyne-bearing precursor was synthesized and subjected to click chemistry-based radiosynthesis of [18F]FGlc-FAPI by 2-step 18F-fluoroglycosylation. FAP-expressing HT1080hFAP cells were used to study competitive binding to FAP, cellular uptake, internalization, and efflux of [18F]FGlc-FAPI in vitro. Biodistribution studies and in vivo small-animal PET studies of [18F]FGlc-FAPI compared with [68Ga]Ga-FAPI-04 were conducted in nude mice bearing HT1080hFAP tumors or U87MG xenografts. Results: [18F]FGlc-FAPI was synthesized with a 15% radioactivity yield and a high radiochemical purity of more than 99%. In HT1080hFAP cells, [18F]FGlc-FAPI showed specific uptake, a high internalized fraction, and low cellular efflux. Compared with FAPI-04 (half maximal inhibitory concentration [IC50] = 32 nM), the glycoconjugate, FGlc-FAPI (IC50 = 167 nM), showed slightly lower affinity for FAP in vitro, whereas plasma protein binding was higher for [18F]FGlc-FAPI. Biodistribution studies revealed significant hepatobiliary excretion of [18F]FGlc-FAPI; however, small-animal PET studies in HT1080hFAP xenografts showed higher specific tumor uptake of [18F]FGlc-FAPI (4.5 percentage injected dose per gram of tissue [%ID/g]) than of [68Ga]Ga-FAPI-04 (2 %ID/g). In U87MG tumor-bearing mice, both tracers showed similar tumor uptake, but [18F]FGlc-FAPI showed a higher tumor retention. Interestingly, [18F]FGlc-FAPI demonstrated high specific uptake in bone structures and joints. Conclusion: [18F]FGlc-FAPI is an interesting candidate for translation to the clinic, taking advantage of the longer half-life and physical imaging properties of 18F. The availability of [18F]FGlc-FAPI may allow extended PET studies of FAP-related diseases, such as cancer, but also arthritis, heart diseases, or pulmonary fibrosis.
Collapse
Affiliation(s)
- Johannes Toms
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Erlangen, Germany
| | - Jürgen Kogler
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Erlangen, Germany
| | - Simone Maschauer
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Erlangen, Germany
| | - Christoph Daniel
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nephropathology, Erlangen, Germany; and
| | - Christian Schmidkonz
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Erlangen, Germany
| | - Torsten Kuwert
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Erlangen, Germany
| | - Olaf Prante
- Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Erlangen, Germany .,Friedrich-Alexander University Erlangen-Nürnberg (FAU), Department of Nuclear Medicine, Erlangen, Germany
| |
Collapse
|
83
|
Šimková A, Bušek P, Šedo A, Konvalinka J. Molecular recognition of fibroblast activation protein for diagnostic and therapeutic applications. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140409. [PMID: 32171757 DOI: 10.1016/j.bbapap.2020.140409] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/24/2020] [Accepted: 03/05/2020] [Indexed: 01/09/2023]
Abstract
Fibroblast activation protein (FAP) is a non-classical serine protease expressed predominantly in conditions accompanied by tissue remodeling, particularly cancer. Due to its plasma membrane localization, FAP represents a promising molecular target for tumor imaging and treatment. The unique enzymatic activity of FAP facilitates development of diagnostic and therapeutic tools based on molecular recognition of FAP by substrates and small-molecule inhibitors, in addition to conventional antibody-based strategies. In this review, we provide background on the pathophysiological role of FAP and discuss its potential for diagnostic and therapeutic applications. Furthermore, we present a detailed analysis of the structural patterns crucial for substrate and inhibitor recognition by the FAP active site and determinants of selectivity over the related proteases dipeptidyl peptidase IV and prolyl endopeptidase. We also review published data on targeting of the tumor microenvironment with FAP antibodies, FAP-targeted prodrugs, activity-based probes and small-molecule inhibitors. We describe use of a recently developed, selective FAP inhibitor with low-nanomolar potency in inhibitor-based targeting strategies including synthetic antibody mimetics based on hydrophilic polymers and inhibitor conjugates for PET imaging. In conclusion, recent advances in understanding of the molecular structure and function of FAP have significantly contributed to the development of several tools with potential for translation into clinical practice.
Collapse
Affiliation(s)
- Adéla Šimková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, 166 10 Praha 6, Czech Republic; Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 12843 Praha 2, Czech Republic.
| | - Petr Bušek
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Praha 2, Czech Republic.
| | - Aleksi Šedo
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Praha 2, Czech Republic.
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, 166 10 Praha 6, Czech Republic; Department of Biochemistry, Faculty of Science, Charles University, Hlavova 8, 12843 Praha 2, Czech Republic.
| |
Collapse
|
84
|
Mhaidly R, Mechta-Grigoriou F. Fibroblast heterogeneity in tumor micro-environment: Role in immunosuppression and new therapies. Semin Immunol 2020; 48:101417. [PMID: 33077325 DOI: 10.1016/j.smim.2020.101417] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
|
85
|
Truffi M, Sorrentino L, Corsi F. Fibroblasts in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:15-29. [PMID: 32040852 DOI: 10.1007/978-3-030-37184-5_2] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The implications of a tumor microenvironment in cancer initiation and progression have drawn interest in recent years. Within the tumor stroma, fibroblasts represent a predominant cell type and are responsible for the majority of extracellular components within the tumor microenvironment, such as matrix and soluble factors. A switch from quiescent fibroblasts to cancer-associated fibroblasts triggers a large variety of pro-tumorigenic signals that support tumor progression and shape the surrounding pathological stroma, with the remodeling of tissue architecture and repression of the local immune response. The heterogeneous nature of cancer-associated fibroblasts and their multiple functions are subject of active research as they could represent promising targets for cutting-edge therapeutic approaches to cancer and the tumor microenvironment.
Collapse
Affiliation(s)
- Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli studi di Milano, Milano, Italy
| | - Luca Sorrentino
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli studi di Milano, Milano, Italy
| | - Fabio Corsi
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy. .,Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli studi di Milano, Milano, Italy.
| |
Collapse
|
86
|
Abstract
This review discusses nuclear imaging of inflammation using molecular probes beyond fluoro-d-glucose, is structured by cellular targets, and focuses on those tracers that have been successfully applied clinically.
Collapse
Affiliation(s)
- Malte Kircher
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany.
| |
Collapse
|
87
|
Oatmen KE, Cull E, Spinale FG. Heart failure as interstitial cancer: emergence of a malignant fibroblast phenotype. Nat Rev Cardiol 2019; 17:523-531. [DOI: 10.1038/s41569-019-0286-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 12/15/2022]
|
88
|
Pereira BA, Vennin C, Papanicolaou M, Chambers CR, Herrmann D, Morton JP, Cox TR, Timpson P. CAF Subpopulations: A New Reservoir of Stromal Targets in Pancreatic Cancer. Trends Cancer 2019; 5:724-741. [PMID: 31735290 DOI: 10.1016/j.trecan.2019.09.010] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/16/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are one of the most significant components in the tumour microenvironment (TME), where they can perform several protumourigenic functions. Several studies have recently reported that CAFs are more heterogenous and plastic than was previously thought. As such, there has been a shift in the field to study CAF subpopulations and the emergent functions of these subsets in tumourigenesis. In this review, we explore how different aspects of CAF heterogeneity are defined and how these manifest in multiple cancers, with a focus on pancreatic ductal adenocarcinoma (PDAC). We also discuss therapeutic approaches to selectively target protumourigenic CAF functions, while avoiding normal fibroblasts, providing insight into the future of stromal targeting for the treatment of PDAC and other solid tumours.
Collapse
Affiliation(s)
- Brooke A Pereira
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Claire Vennin
- Division of Molecular Pathology, Netherlands Cancer Institute (NKI), 1066 CX Amsterdam, The Netherlands
| | - Michael Papanicolaou
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Cecilia R Chambers
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - David Herrmann
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Jennifer P Morton
- Cancer Department, Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Thomas R Cox
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia.
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia.
| |
Collapse
|
89
|
Lindner T, Loktev A, Giesel F, Kratochwil C, Altmann A, Haberkorn U. Targeting of activated fibroblasts for imaging and therapy. EJNMMI Radiopharm Chem 2019; 4:16. [PMID: 31659499 PMCID: PMC6658625 DOI: 10.1186/s41181-019-0069-0] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
Tumors form a complex environment consisting of a variety of non-malignant cells. Especially cancer-associated fibroblasts have been shown to have an important role for different aspects of malignant tumors such as migration, metastasis, resistance to chemotherapy and immunosuppression. Therefore, a targeting of these cells may be useful for both imaging and therapy. In this respect, an interesting target is the fibroblast activation protein (FAP) which is expressed in activated fibroblasts, but not in quiescent fibroblasts, giving the opportunity to use this membrane-anchored enzyme as a target for radionuclide-based approaches for diagnosis and treatment of tumors and for the diagnosis of non-malignant disease associated with a remodelling of the extracellular matrix.
Collapse
Affiliation(s)
- Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Anastasia Loktev
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
90
|
Abstract
Cancers are not composed merely of cancer cells alone; instead, they are complex 'ecosystems' comprising many different cell types and noncellular factors. The tumour stroma is a critical component of the tumour microenvironment, where it has crucial roles in tumour initiation, progression, and metastasis. Most anticancer therapies target cancer cells specifically, but the tumour stroma can promote the resistance of cancer cells to such therapies, eventually resulting in fatal disease. Therefore, novel treatment strategies should combine anticancer and antistromal agents. Herein, we provide an overview of the advances in understanding the complex cancer cell-tumour stroma interactions and discuss how this knowledge can result in more effective therapeutic strategies, which might ultimately improve patient outcomes.
Collapse
|
91
|
Hintz HM, Cowan AE, Shapovalova M, LeBeau AM. Development of a Cross-Reactive Monoclonal Antibody for Detecting the Tumor Stroma. Bioconjug Chem 2019; 30:1466-1476. [PMID: 30966746 DOI: 10.1021/acs.bioconjchem.9b00206] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Here, we document the discovery of a monoclonal antibody that selectively binds to both human and murine fibroblast activation protein alpha (FAP), a serine protease that is overexpressed on cancer-associated fibroblasts (CAFs), making it an attractive therapeutic target for the aiding and abetting tumor microenvironment. The lead antibody, B12, was identified from a naïve murine single-chain variable fragment antibody phage display library screened against recombinant human FAP on magnetic beads. The heavy and light chains of B12 were cloned into full-length human immunoglobulin 1 (IgG) vectors and expressed as a chimeric monoclonal antibody (B12 IgG). We engineered a drug-resistant prostate cancer cell line, CWR-R1-EnzR, to express human FAP for antibody characterization and validation (R1-EnzRFAP). B12 IgG selectively bound to the R1-EnzRFAP cells by flow cytometry and was internalized in vitro by confocal microscopy. B12 IgG was further evaluated as a near-infrared (NIR) optical imaging probe in R1-EnzRFAP and parental xenograft models. High tumor uptake and retention of the NIR probe was observed in the R1-EnzRFAP xenografts, and endogenous expression of murine stromal origin FAP was detected in the parental xenografts. Ex vivo evaluation of these models by immunohistochemistry documented B12 IgG localization to both human and murine FAP-expressing cells.
Collapse
Affiliation(s)
- Hallie M Hintz
- Department of Pharmacology , University of Minnesota Medical School , Minneapolis , Minnesota 55455 , United States
| | - Aidan E Cowan
- Department of Pharmacology , University of Minnesota Medical School , Minneapolis , Minnesota 55455 , United States
| | - Mariya Shapovalova
- Department of Pharmacology , University of Minnesota Medical School , Minneapolis , Minnesota 55455 , United States
| | - Aaron M LeBeau
- Department of Pharmacology , University of Minnesota Medical School , Minneapolis , Minnesota 55455 , United States
| |
Collapse
|
92
|
Kratochwil C, Flechsig P, Lindner T, Abderrahim L, Altmann A, Mier W, Adeberg S, Rathke H, Röhrich M, Winter H, Plinkert PK, Marme F, Lang M, Kauczor HU, Jäger D, Debus J, Haberkorn U, Giesel FL. 68Ga-FAPI PET/CT: Tracer Uptake in 28 Different Kinds of Cancer. J Nucl Med 2019; 60:801-805. [PMID: 30954939 DOI: 10.2967/jnumed.119.227967] [Citation(s) in RCA: 981] [Impact Index Per Article: 163.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/19/2019] [Indexed: 12/31/2022] Open
Abstract
The recent development of quinoline-based PET tracers that act as fibroblast-activation-protein inhibitors (FAPIs) demonstrated promising preclinical and clinical results. FAP is overexpressed by cancer-associated fibroblasts of several tumor entities. Here, we quantify the tumor uptake on 68Ga-FAPI PET/CT of various primary and metastatic tumors to identify the most promising indications for future application. Methods: 68Ga-FAPI PET/CT scans were requested by various referring physicians according to individual clinical indications that were considered insufficiently covered by 18F-FDG PET/CT or other imaging modalities. All PET/CT was performed 1 h after injection of 122-312 MBq of 68Ga-FAPI-04. We retrospectively identified 80 patients with histopathologically proven primary tumors or metastases or radiologically unequivocal metastatic lesions of histologically proven primary tumors. Tumor uptake was quantified by SUVmax and SUVmean (60% isocontour). Results: Eighty patients with 28 different tumor entities (54 primary tumors and 229 metastases) were evaluated. The highest average SUVmax (>12) was found in sarcoma, esophageal, breast, cholangiocarcinoma, and lung cancer. The lowest 68Ga-FAPI uptake (average SUVmax < 6) was observed in pheochromocytoma, renal cell, differentiated thyroid, adenoid cystic, and gastric cancer. The average SUVmax of hepatocellular, colorectal, head-neck, ovarian, pancreatic, and prostate cancer was intermediate (SUV 6-12). SUV varied across and within all tumor entities. Because of low background in muscle and blood pool (SUVmax < 2), the tumor-to-background contrast ratios were more than 3-fold in the intermediate and more than 6-fold in the high-intensity uptake group. Conclusion: Several highly prevalent cancers presented with remarkably high uptake and image contrast on 68Ga-FAPI PET/CT. The high and rather selective tumor uptake may open up new applications for noninvasive tumor characterization, staging examinations, or radioligand therapy.
Collapse
Affiliation(s)
- Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Paul Flechsig
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Labidi Abderrahim
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Adeberg
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology, Heidelberg, Germany
| | - Hendrik Rathke
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Manuel Röhrich
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany.,Department of Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Peter K Plinkert
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Frederik Marme
- Department Obstetrics and Gynecology, University Hospital Heidelberg, Heidelberg, Germany.,Department Obstetrics and Gynecology, University Hospital Mannheim, Mannheim, Germany
| | - Matthias Lang
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Germany.,Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany; and
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
93
|
Loktev A, Lindner T, Burger EM, Altmann A, Giesel F, Kratochwil C, Debus J, Marmé F, Jäger D, Mier W, Haberkorn U. Development of Fibroblast Activation Protein-Targeted Radiotracers with Improved Tumor Retention. J Nucl Med 2019; 60:1421-1429. [PMID: 30850501 PMCID: PMC6785792 DOI: 10.2967/jnumed.118.224469] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/04/2019] [Indexed: 12/23/2022] Open
Abstract
Cancer-associated fibroblasts constitute a vital subpopulation of the tumor stroma and are present in more than 90% of epithelial carcinomas. The overexpression of the serine protease fibroblast activation protein (FAP) allows a selective targeting of a variety of tumors by inhibitor-based radiopharmaceuticals (FAPIs). Of these compounds, FAPI-04 has been recently introduced as a theranostic radiotracer and demonstrated high uptake into different FAP-positive tumors in cancer patients. To enable the delivery of higher doses, thereby improving the outcome of a therapeutic application, several FAPI variants were designed to further increase tumor uptake and retention of these tracers. Methods: Novel quinoline-based radiotracers were synthesized by organic chemistry and evaluated in radioligand binding assays using FAP-expressing HT-1080 cells. Depending on their in vitro performance, small-animal PET imaging and biodistribution studies were performed on HT-1080-FAP tumor–bearing mice. The most promising compounds were used for clinical PET imaging in 8 cancer patients. Results: Compared with FAPI-04, 11 of 15 FAPI derivatives showed improved FAP binding in vitro. Of these, 7 compounds demonstrated increased tumor uptake in tumor-bearing mice. Moreover, tumor–to–normal-organ ratios were improved for most of the compounds, resulting in images with higher contrast. Notably two of the radiotracers, FAPI-21 and -46, displayed substantially improved ratios of tumor to blood, liver, muscle, and intestinal uptake. A first diagnostic application in cancer patients revealed high intratumoral uptake of both radiotracers already 10 min after administration but a higher uptake in oral mucosa, salivary glands, and thyroid for FAPI-21. Conclusion: Chemical modification of the FAPI framework enabled enhanced FAP binding and improved pharmacokinetics in most of the derivatives, resulting in high-contrast images. Moreover, higher doses of radioactivity can be delivered while minimizing damage to healthy tissue, which may improve therapeutic outcome.
Collapse
Affiliation(s)
- Anastasia Loktev
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Eva-Maria Burger
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Frederik Marmé
- Department of Gynecologic Oncology, National Center for Tumor Diseases and Department of Obstetrics and Gynecology, University Women's Clinic, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany; and
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany .,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| |
Collapse
|
94
|
Lee HJ, Ehlerding EB, Cai W. Antibody-Based Tracers for PET/SPECT Imaging of Chronic Inflammatory Diseases. Chembiochem 2019; 20:422-436. [PMID: 30240550 PMCID: PMC6377337 DOI: 10.1002/cbic.201800429] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Indexed: 12/18/2022]
Abstract
Chronic inflammatory diseases are often progressive, resulting not only in physical damage to patients but also social and economic burdens, making early diagnosis of them critical. Nuclear medicine techniques can enhance the detection of inflammation by providing functional as well as anatomical information when combined with other modalities such as magnetic resonance imaging, computed tomography or ultrasonography. Although small molecules and peptides were mainly used for the treatment and imaging of chronic inflammatory diseases in the past, antibodies and their fragments have also been emerging for chronic inflammatory diseases as they show high specificity to their targets and can have various biological half-lives depending on how they are engineered. In addition, imaging with antibodies or their fragments can visualize the in vivo biodistribution of the probes or help monitor therapeutic responses, thereby providing physicians with a greater understanding of drug behavior in vivo and another means of monitoring their patients. In this review, we introduce various targets and radiolabeled antibody-based probes for the molecular imaging of chronic inflammatory diseases in preclinical and clinical studies. Targets can be classified into three different categories: 1) cell-adhesion molecules, 2) surface markers on immune cells, and 3) cytokines or enzymes. The limitations and future directions of using radiolabeled antibodies for imaging inflammatory diseases are also discussed.
Collapse
Affiliation(s)
- Hye Jin Lee
- Pharmaceutical Sciences Department, University of Wisconsin – Madison, Madison WI 53705, USA
| | - Emily B. Ehlerding
- Medical Physics Department, University of Wisconsin – Madison, Madison WI 53705, USA
| | - Weibo Cai
- Pharmaceutical Sciences Department, University of Wisconsin – Madison, Madison WI 53705, USA
- Medical Physics Department, University of Wisconsin – Madison, Madison WI 53705, USA
- Department of Radiology and Carbone Cancer Center, University of Wisconsin – Madison, Madison WI 53705, USA
| |
Collapse
|
95
|
Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov 2018; 18:99-115. [DOI: 10.1038/s41573-018-0004-1] [Citation(s) in RCA: 633] [Impact Index Per Article: 90.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
96
|
Abstract
The concept that progression of cancer is regulated by interactions of cancer cells with their microenvironment was postulated by Stephen Paget over a century ago. Contemporary tumour microenvironment (TME) research focuses on the identification of tumour-interacting microenvironmental constituents, such as resident or infiltrating non-tumour cells, soluble factors and extracellular matrix components, and the large variety of mechanisms by which these constituents regulate and shape the malignant phenotype of tumour cells. In this Timeline article, we review the developmental phases of the TME paradigm since its initial description. While illuminating controversies, we discuss the importance of interactions between various microenvironmental components and tumour cells and provide an overview and assessment of therapeutic opportunities and modalities by which the TME can be targeted.
Collapse
Affiliation(s)
- Shelly Maman
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
97
|
Loktev A, Lindner T, Mier W, Debus J, Altmann A, Jäger D, Giesel F, Kratochwil C, Barthe P, Roumestand C, Haberkorn U. A Tumor-Imaging Method Targeting Cancer-Associated Fibroblasts. J Nucl Med 2018; 59:1423-1429. [PMID: 29626120 PMCID: PMC6126438 DOI: 10.2967/jnumed.118.210435] [Citation(s) in RCA: 509] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/21/2018] [Indexed: 11/16/2022] Open
Abstract
The tumor stroma, which accounts for a large part of the tumor mass, represents an attractive target for the delivery of diagnostic and therapeutic compounds. Here, the focus is notably on a subpopulation of stromal cells, known as cancer-associated fibroblasts, which are present in more than 90% of epithelial carcinomas, including pancreatic, colon, and breast cancer. Cancer-associated fibroblasts feature high expression of fibroblast activation protein (FAP), which is not detectable in adult normal tissue but is associated with a poor prognosis in cancer patients. Methods: We developed an iodinated and a DOTA-coupled radiotracer based on a FAP-specific enzyme inhibitor (FAPI) and evaluated them in vitro using uptake, competition, and efflux studies as well as confocal microscopy of a fluorescence-labeled variant. Furthermore, we performed imaging and biodistribution studies on tumor-bearing animals. Finally, proof of concept was realized by imaging patients with 68Ga-labeled FAPI. Results: Both FAPIs showed high specificity, affinity, and rapid internalization into FAP-expressing cells in vitro and in vivo. Biodistribution studies on tumor-bearing mice and on the first cancer patients demonstrated high intratumoral uptake of the tracer and fast body clearance, resulting in high-contrast images and negligible exposure of healthy tissue to radiation. A comparison with the commonly used radiotracer 18F-FDG in a patient with locally advanced lung adenocarcinoma revealed that the new FAP ligand was clearly superior. Conclusion: Radiolabeled FAPIs allow fast imaging with very high contrast in tumors having a high stromal content and may therefore serve as pantumor agents. Coupling of these molecules to DOTA or other chelators allows labeling not only with 68Ga but also with therapeutic isotopes such as 177Lu or 90Y.
Collapse
Affiliation(s)
- Anastasia Loktev
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Philippe Barthe
- Centre de Biochimie Structurale, Université de Montpellier, CNRS, INSERM, Montpellier, France; and
| | - Christian Roumestand
- Centre de Biochimie Structurale, Université de Montpellier, CNRS, INSERM, Montpellier, France; and
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany .,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
98
|
Lindner T, Loktev A, Altmann A, Giesel F, Kratochwil C, Debus J, Jäger D, Mier W, Haberkorn U. Development of Quinoline-Based Theranostic Ligands for the Targeting of Fibroblast Activation Protein. J Nucl Med 2018; 59:1415-1422. [PMID: 29626119 DOI: 10.2967/jnumed.118.210443] [Citation(s) in RCA: 589] [Impact Index Per Article: 84.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/22/2018] [Indexed: 01/04/2023] Open
Abstract
Fibroblast activation protein (FAP) is overexpressed in cancer-associated fibroblasts and is involved in a variety of tumor-promoting activities such as matrix remodeling, angiogenesis, chemotherapy resistance, and immunosuppression. Because FAP shows low expression in most normal organs, it presents an interesting target for imaging and endoradiotherapy. In this investigation, FAP inhibitors (FAPIs) were modified and optimized for use as theranostic tracers. Methods: FAPIs based on a quinoline structure were synthesized and characterized with respect to binding, internalization, and efflux in cells expressing human and murine FAP as well as CD26. Preclinical pharmacokinetics were determined in tumor-bearing animals with biodistribution experiments and small-animal PET. Finally, a proof-of-concept approach toward imaging and therapy was chosen for 2 patients with metastasized breast cancer. Results: Of 15 synthesized FAPIs, FAPI-04 was identified as the most promising tracer for clinical application. Compared with the previously published ligand, FAPI-02, FAPI-04 showed excellent stability in human serum, higher affinity for FAP as opposed to CD26, and slower excretion in vitro. In vivo, a higher SUV was reached in tumor-bearing animals, leading to larger areas under the curve as calculated from biodistribution experiments. Finally, PET/CT scans with 68Ga-FAPI-04 in 2 patients with metastasized breast cancer revealed high tracer uptake in metastases and a reduction in pain symptoms after therapy with a considerably low dose of 90Y-FAPI-04. Conclusion: FAPI-04 represents a promising tracer for both diagnostic imaging and, possibly, targeted therapy of malignant tumors with a high content of activated fibroblasts, such as breast cancer.
Collapse
Affiliation(s)
- Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Anastasia Loktev
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; and
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany .,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
99
|
Antibody-Drug Conjugates: Targeting the Tumor Microenvironment. CANCER DRUG DISCOVERY AND DEVELOPMENT 2018. [DOI: 10.1007/978-3-319-78154-9_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
100
|
Jiang GM, Xu W, Du J, Zhang KS, Zhang QG, Wang XW, Liu ZG, Liu SQ, Xie WY, Liu HF, Liu JS, Wu BP. The application of the fibroblast activation protein α-targeted immunotherapy strategy. Oncotarget 2017; 7:33472-82. [PMID: 26985769 PMCID: PMC5078111 DOI: 10.18632/oncotarget.8098] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/28/2016] [Indexed: 12/31/2022] Open
Abstract
Cancer immunotherapy has primarily been focused on attacking tumor cells. However, given the close interaction between tumor cells and cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME), CAF-targeted strategies could also contribute to an integrated cancer immunotherapy. Fibroblast activation protein α (FAP α) is not detectible in normal tissues, but is overexpressed by CAFs and is the predominant component of the stroma in most types of cancer. FAP α has both dipeptidyl peptidase and endopeptidase activities, cleaving substrates at a post-proline bond. When all FAP α-expressing cells (stromal and cancerous) are destroyed, tumors rapidly die. Furthermore, a FAP α antibody, FAP α vaccine, and modified vaccine all inhibit tumor growth and prolong survival in mouse models, suggesting FAP α is an adaptive tumor-associated antigen. This review highlights the role of FAP α in tumor development, explores the relationship between FAP α and immune suppression in the TME, and discusses FAP α as a potential immunotherapeutic target.
Collapse
Affiliation(s)
- Guan-Min Jiang
- Department of Clinical Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kun-Shui Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiu-Gui Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiao-Wei Wang
- Department of Clinical Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhi-Gang Liu
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shuang-Quan Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Wan-Ying Xie
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Hui-Fang Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jing-Shi Liu
- Department of Anesthesia, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bai-Ping Wu
- Department of Clinical Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|