1
|
Shah R, Johnson KA, Lippert AEL, Kraus SG, Emmerich PB, Pasch CA, Zhang W, Matkowskyj KA, LeBeau AM, Deming DA. Cancer-Associated Fibroblast Proteins as Potential Targets against Colorectal Cancers. Cancers (Basel) 2024; 16:3158. [PMID: 39335130 PMCID: PMC11440114 DOI: 10.3390/cancers16183158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
In colorectal cancer (CRC), attempts to identify cancer cell-specific markers to guide antibody-mediated therapeutics have failed to uncover markers that are both exclusive to cancer tissues and abundant across CRCs. Alternatively, cancer-associated fibroblasts (CAFs), which are abundant in the tumor microenvironment and upregulate unique surface markers, are not found in healthy tissues. Here, we evaluated the expression patterns of CAF-associated proteins α-smooth muscle actin (αSMA), fibroblast activation protein (FAP), podoplanin (PDPN), matrix metalloproteinase-2 (MMP2), transgelin (TAGLN), and THY1. While αSMA and THY1 were abundant in cancer tissues, high abundance in normal tissues limited their targeting potential. FAP was present in 94.5% of primary and metastatic CRC tissues and absent in 93.7% of adjacent normal colon and liver tissues assessed. These results indicate that FAP is a promising target for antibody conjugates with potential for broad application in CRC. Co-expression analyses showed that CRCs simultaneously expressing high levels of PDPN, MMP2, and THY1 were enriched for immune-related signatures, indicating potential for antibody-mediated immune engagers. Overall, this work highlights the potential of CAF proteins to act as therapeutic targets for novel anticancer agents and become important therapeutic biomarkers.
Collapse
Affiliation(s)
- Ruchi Shah
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Katherine A Johnson
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Anna E L Lippert
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Sean G Kraus
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Philip B Emmerich
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
| | - Cheri A Pasch
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Wei Zhang
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kristina A Matkowskyj
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
| | - Dustin A Deming
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53726, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
2
|
Gallant JP, Hicks D, Shi K, Moeller NH, Hoppe B, Lake EW, Baehr C, Pravetoni M, Aihara H, LeBeau AM. Identification and biophysical characterization of a novel domain-swapped camelid antibody specific for fentanyl. J Biol Chem 2024; 300:107502. [PMID: 38945452 PMCID: PMC11321312 DOI: 10.1016/j.jbc.2024.107502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
Opioid use disorders (OUD) and overdoses are ever-evolving public health threats that continue to grow in incidence and prevalence in the United States and abroad. Current treatments consist of opioid receptor agonists and antagonists, which are safe and effective but still suffer from some limitations. Murine and humanized monoclonal antibodies (mAb) have emerged as an alternative and complementary strategy to reverse and prevent opioid-induced respiratory depression. To explore antibody applications beyond traditional heavy-light chain mAbs, we identified and biophysically characterized a novel single-domain antibody specific for fentanyl from a camelid variable-heavy-heavy (VHH) domain phage display library. Structural data suggested that VHH binding to fentanyl was facilitated by a unique domain-swapped dimerization mechanism, which accompanied a rearrangement of complementarity-determining region loops leading to the formation of a fentanyl-binding pocket. Structure-guided mutagenesis further identified an amino acid substitution that improved the affinity and relaxed the requirement for dimerization of the VHH in fentanyl binding. Our studies demonstrate VHH engagement of an opioid and inform on how to further engineer a VHH for enhanced stability and efficacy, laying the groundwork for exploring the in vivo applications of VHH-based biologics against OUD and overdose.
Collapse
Affiliation(s)
- Joseph P Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nicholas H Moeller
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brooke Hoppe
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| | - Eric W Lake
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Carly Baehr
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Marco Pravetoni
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA; Center for Medication Development for Substance Use Disorders, University of Washington, Seattle, Washington, USA.
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
3
|
Hoffmann M, Gallant J, LeBeau A, Schmidt D. Unlocking precision gene therapy: harnessing AAV tropism with nanobody swapping at capsid hotspots. NAR MOLECULAR MEDICINE 2024; 1:ugae008. [PMID: 39022346 PMCID: PMC11250487 DOI: 10.1093/narmme/ugae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Adeno-associated virus (AAV) has been remarkably successful in the clinic, but its broad tropism is a practical limitation of precision gene therapy. A promising path to engineer AAV tropism is the addition of binding domains to the AAV capsid that recognize cell surface markers present on a targeted cell type. We have recently identified two previously unexplored capsid regions near the 2/5-fold wall and 5-fold pore of the AAV capsid that are amenable to insertion of larger protein domains, including nanobodies. Here, we demonstrate that these hotspots facilitate AAV tropism switching through simple nanobody replacement without extensive optimization in both VP1 and VP2. Our data suggest that engineering VP2 is the preferred path for maintaining both virus production yield and infectivity. We demonstrate highly specific targeting of human cancer cells expressing fibroblast activating protein (FAP). Furthermore, we found that the combination of FAP nanobody insertion plus ablation of the heparin binding domain can reduce off-target infection to a minimum, while maintaining a strong infection of FAP receptor-positive cells. Taken together, our study shows that nanobody swapping at multiple capsid locations is a viable strategy for nanobody-directed cell-specific AAV targeting.
Collapse
Affiliation(s)
- Mareike D Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph P Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
4
|
Gallant JP, Hintz HM, Gunaratne GS, Breneman MT, Recchia EE, West JL, Ott KL, Heninger E, Jackson AE, Luo NY, Rosenkrans ZT, Hernandez R, Zhao SG, Lang JM, Meimetis L, Kosoff D, LeBeau AM. Mechanistic Characterization of Cancer-associated Fibroblast Depletion via an Antibody-Drug Conjugate Targeting Fibroblast Activation Protein. CANCER RESEARCH COMMUNICATIONS 2024; 4:1481-1494. [PMID: 38747612 PMCID: PMC11168342 DOI: 10.1158/2767-9764.crc-24-0248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/14/2024]
Abstract
Cancer-associated fibroblasts (CAF) are a prominent cell type within the tumor microenvironment (TME) where they are known to promote cancer cell growth and survival, angiogenesis, drug resistance, and immunosuppression. The transmembrane prolyl protease fibroblast activation protein (FAP) is expressed on the surface of highly protumorigenic CAFs found in the stroma of nearly every cancer of epithelial origin. The widespread expression of FAP has made it an attractive therapeutic target based on the underlying hypothesis that eliminating protumorigenic CAFs will disrupt the cross-talk between components of TME resulting in cancer cell death and immune infiltration. This hypothesis, however, has never been directly proven. To eliminate FAP-expressing CAFs, we developed an antibody-drug conjugate using our anti-FAP antibody, huB12, coupled to a monomethyl auristatin E (huB12-MMAE) payload. After determining that huB12 was an effective targeting vector, we found that huB12-MMAE potently eliminated FAP-expressing cells as monocultures in vitro and significantly prolonged survival in vivo using a xenograft engineered to overexpress FAP. We investigated the effects of selectively eliminating CAFs using a layered, open microfluidic cell coculture platform, known as the Stacks. Analysis of mRNA and protein expression found that treatment with huB12-MMAE resulted in the increased secretion of the proinflammatory cytokines IL6 and IL8 by CAFs and an associated increase in expression of proinflammatory genes in cancer cells. We also detected increased secretion of CSF1, a cytokine involved in myeloid recruitment and differentiation. Our findings suggest that the mechanism of FAP-targeted therapies is through effects on the immune microenvironment and antitumor immune response. SIGNIFICANCE The direct elimination of FAP-expressing CAFs disrupts the cross-talk with cancer cells leading to a proinflammatory response and alterations in the immune microenvironment and antitumor immune response.
Collapse
Affiliation(s)
- Joseph P. Gallant
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Hallie M. Hintz
- Department of Pharmacology, University of Minnesota School of Medicine, Minneapolis, Minnesota
| | - Gihan S. Gunaratne
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Matthew T. Breneman
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Emma E. Recchia
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jayden L. West
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kendahl L. Ott
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Erika Heninger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Abigail E. Jackson
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Natalie Y. Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Zachary T. Rosenkrans
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Reinier Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Shuang G. Zhao
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Labros Meimetis
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - David Kosoff
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- William S Middleton Memorial Veterans’ Hospital, Madison, Wisconsin
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
5
|
Mori JO, Elhussin I, Brennen WN, Graham MK, Lotan TL, Yates CC, De Marzo AM, Denmeade SR, Yegnasubramanian S, Nelson WG, Denis GV, Platz EA, Meeker AK, Heaphy CM. Prognostic and therapeutic potential of senescent stromal fibroblasts in prostate cancer. Nat Rev Urol 2024; 21:258-273. [PMID: 37907729 PMCID: PMC11058122 DOI: 10.1038/s41585-023-00827-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
The stromal component of the tumour microenvironment in primary and metastatic prostate cancer can influence and promote disease progression. Within the prostatic stroma, fibroblasts are one of the most prevalent cell types associated with precancerous and cancerous lesions; they have a vital role in the structural composition, organization and integrity of the extracellular matrix. Fibroblasts within the tumour microenvironment can undergo cellular senescence, which is a stable arrest of cell growth and a phenomenon that is emerging as a recognized hallmark of cancer. Supporting the idea that cellular senescence has a pro-tumorigenic role, a subset of senescent cells exhibits a senescence-associated secretory phenotype (SASP), which, along with increased inflammation, can promote prostate cancer cell growth and survival. These cellular characteristics make targeting senescent cells and/or modulating SASP attractive as a potential preventive or therapeutic option for prostate cancer.
Collapse
Affiliation(s)
- Joakin O Mori
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Isra Elhussin
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mindy K Graham
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clayton C Yates
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel R Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William G Nelson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerald V Denis
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elizabeth A Platz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alan K Meeker
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher M Heaphy
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
6
|
Caramella-Pereira F, Zheng Q, Hicks JL, Roy S, Jones T, Pomper M, Antony L, Meeker AK, Yegnasubramanian S, De Marzo AM, Brennen WN. Overexpression of Fibroblast Activation Protein (FAP) in stroma of proliferative inflammatory atrophy (PIA) and primary adenocarcinoma of the prostate. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.04.24305338. [PMID: 38633791 PMCID: PMC11023661 DOI: 10.1101/2024.04.04.24305338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Fibroblast activation protein (FAP) is a serine protease upregulated at sites of tissue remodeling and cancer that represents a promising therapeutic and molecular imaging target. In prostate cancer, studies of FAP expression using tissue microarrays are conflicting, such that its clinical potential is unclear. Furthermore, little is known regarding FAP expression in benign prostatic tissues. Here we demonstrated, using a novel iterative multiplex IHC assay in standard tissue sections, that FAP was nearly absent in normal regions, but was increased consistently in regions of proliferative inflammatory atrophy (PIA). In carcinoma, FAP was expressed in all cases, but was highly heterogeneous. High FAP levels were associated with increased pathological stage and cribriform morphology. We verified that FAP levels in cancer correlated with CD163+ M2 macrophage density. In this first report to quantify FAP protein in benign prostate and primary tumors, using standard large tissue sections, we clarify that FAP is present in all primary prostatic carcinomas, supporting its potential clinical relevance. The finding of high levels of FAP within PIA supports the injury/regeneration model for its pathogenesis and suggests that it harbors a protumorigenic stroma. Yet, high levels of FAP in benign regions could lead to false positive FAP-based molecular imaging results in clinically localized prostate cancer.
Collapse
|
7
|
Hoffmann MD, Gallant JP, LeBeau AM, Schmidt D. Unlocking Precision Gene Therapy: Harnessing AAV Tropism with Nanobody Swapping at Capsid Hotspots. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587049. [PMID: 38585985 PMCID: PMC10996663 DOI: 10.1101/2024.03.27.587049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Adeno-associated virus has been remarkably successful in the clinic, but its broad tropism is a practical limitation of precision gene therapy. A promising path to engineer AAV tropism is the addition of binding domains to the AAV capsid that recognize cell surface markers present on a targeted cell type. We have recently identified two previously unexplored capsid regions near the 2-fold valley and 5-fold pore of the AAV capsid that are amenable to insertion of larger protein domains including nanobodies. Here, we demonstrate that these hotspots facilitate AAV tropism switching through simple nanobody replacement without extensive optimization in both VP1 and VP2. We demonstrate highly specific targeting of human cancer cells expressing fibroblast activating protein (FAP). Our data suggest that engineering VP2 is the preferred path for maintaining both virus production yield and infectivity. Our study shows that nanobody swapping at multiple capsid location is a viable strategy for nanobody-directed cell-specific AAV targeting.
Collapse
Affiliation(s)
- Mareike D. Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joseph P. Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
8
|
Li Z, Liu C, Li C, Wang F, Liu J, Zheng Z, Wu J, Zhang B. Irinotecan/scFv co-loaded liposomes coaction on tumor cells and CAFs for enhanced colorectal cancer therapy. J Nanobiotechnology 2021; 19:421. [PMID: 34906155 PMCID: PMC8670172 DOI: 10.1186/s12951-021-01172-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/30/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), as an important component of stroma, not only supply the "soils" to promote tumor invasion and metastasis, but also form a physical barrier to hinder the penetration of therapeutic agents. Based on this, the combinational strategy that action on both tumor cells and CAFs simultaneously would be a promising approach for improving the antitumor effect. RESULTS In this study, the novel multifunctional liposomes (IRI-RGD/R9-sLip) were designed, which integrated the advantages including IRI and scFv co-loading, different targets, RGD mediated active targeting, R9 promoting cell efficient permeation and lysosomal escape. As expected, IRI-RGD/R9-sLip showed enhanced cytotoxicity in different cell models, effectively increased the accumulation in tumor sites, as well as exhibited deep permeation ability both in vitro and in vivo. Notably, IRI-RGD/R9-sLip not only exhibited superior in vivo anti-tumor effect in both CAFs-free and CAFs-abundant bearing mice models, but also presented excellent anti-metastasis efficiency in lung metastasis model. CONCLUSION In a word, the novel combinational strategy by coaction on both "seeds" and "soils" of the tumor provides a new approach for cancer therapy, and the prepared liposomes could efficiently improve the antitumor effect with promising clinical application prospects.
Collapse
Affiliation(s)
- Zhaohuan Li
- School of Pharmacy, Weifang Medical University Weifang, Shandong, 261053, People's Republic of China
| | - Chunxi Liu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, People's Republic of China
| | - Chenglei Li
- School of Pharmacy, Weifang Medical University Weifang, Shandong, 261053, People's Republic of China
| | - Fangqing Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Jianhao Liu
- School of Pharmacy, Weifang Medical University Weifang, Shandong, 261053, People's Republic of China
| | - Zengjuan Zheng
- School of Pharmacy, Weifang Medical University Weifang, Shandong, 261053, People's Republic of China
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, Shandong, People's Republic of China.
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University Weifang, Shandong, 261053, People's Republic of China.
| |
Collapse
|
9
|
Hintz HM, Snyder KM, Wu J, Hullsiek R, Dahlvang JD, Hart GT, Walcheck B, LeBeau AM. Simultaneous Engagement of Tumor and Stroma Targeting Antibodies by Engineered NK-92 Cells Expressing CD64 Controls Prostate Cancer Growth. Cancer Immunol Res 2021; 9:1270-1282. [PMID: 34452926 PMCID: PMC9119026 DOI: 10.1158/2326-6066.cir-21-0178] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/20/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) has been largely resistant to immunotherapy. Natural killer (NK) cells are cytotoxic lymphocytes that detect and kill transformed cells without prior sensitization, and their infiltration into prostate tumors corresponds with an increased overall survival among patients with mCRPC. We sought to harness this knowledge to develop an approach to NK-cell based immunotherapy for mCRPC. We engineered an NK cell line (NK-92MI) to express CD64, the sole human high-affinity IgG Fcγ receptor (FcγR1), and bound these cells with antibodies to provide interchangeable tumor-targeting elements. NK-92MICD64 cells were evaluated for cell-activation mechanisms and antibody-dependent cell-mediated cytotoxicity (ADCC). A combination of mAbs was used to target the prostate tumor antigen tumor-associated calcium signal transducer 2 (TROP2) and the cancer-associated fibroblast marker fibroblast activation protein alpha (FAP). We found that CD64, which is normally expressed by myeloid cells and associates with the adaptor molecule FcRγ, can be expressed by NK-92MI cells and mediate ADCC through an association with CD3ζ. Cytotoxicity from the combination approach was two-fold higher compared to treatment with NK-92MICD64 cells and either mAb alone, and seven-fold higher than NK-92MICD64 cells alone at an effector-target cell ratio of 20:1. The cytotoxic effect was lost when using isotype control antibodies, indicating a selective targeting mechanism. The combination approach demonstrated efficacy in vivo as well and significantly reduced tumor growth compared with the saline control. This combination therapy presents a potential approach for treating mCRPC and could improve immunotherapy response.
Collapse
Affiliation(s)
- Hallie M Hintz
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Kristin M Snyder
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota
| | - Robert Hullsiek
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota
| | - James D Dahlvang
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Geoffrey T Hart
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota.
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Aaron M LeBeau
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota.
| |
Collapse
|
10
|
Ye G, Gallant J, Zheng J, Massey C, Shi K, Tai W, Odle A, Vickers M, Shang J, Wan Y, Du L, Aihara H, Perlman S, LeBeau A, Li F. The development of Nanosota- 1 as anti-SARS-CoV-2 nanobody drug candidates. eLife 2021; 10:e64815. [PMID: 34338634 PMCID: PMC8354634 DOI: 10.7554/elife.64815] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 07/28/2021] [Indexed: 12/29/2022] Open
Abstract
Combating the COVID-19 pandemic requires potent and low-cost therapeutics. We identified a series of single-domain antibodies (i.e., nanobody), Nanosota-1, from a camelid nanobody phage display library. Structural data showed that Nanosota-1 bound to the oft-hidden receptor-binding domain (RBD) of SARS-CoV-2 spike protein, blocking viral receptor angiotensin-converting enzyme 2 (ACE2). The lead drug candidate possessing an Fc tag (Nanosota-1C-Fc) bound to SARS-CoV-2 RBD ~3000 times more tightly than ACE2 did and inhibited SARS-CoV-2 pseudovirus ~160 times more efficiently than ACE2 did. Administered at a single dose, Nanosota-1C-Fc demonstrated preventive and therapeutic efficacy against live SARS-CoV-2 infection in both hamster and mouse models. Unlike conventional antibodies, Nanosota-1C-Fc was produced at high yields in bacteria and had exceptional thermostability. Pharmacokinetic analysis of Nanosota-1C-Fc documented an excellent in vivo stability and a high tissue bioavailability. As effective and inexpensive drug candidates, Nanosota-1 may contribute to the battle against COVID-19.
Collapse
Affiliation(s)
- Gang Ye
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSaint PaulUnited States
- Center for Coronavirus Research, University of MinnesotaSaint PaulUnited States
| | - Joseph Gallant
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
| | - Jian Zheng
- Department of Microbiology and Immunology, University of IowaIowa CityUnited States
| | - Christopher Massey
- Institutional Office of Regulated Nonclinical Studies, University of Texas Medical BranchGalvestonUnited States
| | - Ke Shi
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Wanbo Tai
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood CenterNew YorkUnited States
| | - Abby Odle
- Department of Microbiology and Immunology, University of IowaIowa CityUnited States
| | - Molly Vickers
- Department of Microbiology and Immunology, University of IowaIowa CityUnited States
| | - Jian Shang
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSaint PaulUnited States
- Center for Coronavirus Research, University of MinnesotaSaint PaulUnited States
| | - Yushun Wan
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSaint PaulUnited States
- Center for Coronavirus Research, University of MinnesotaSaint PaulUnited States
| | - Lanying Du
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood CenterNew YorkUnited States
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of IowaIowa CityUnited States
| | - Aaron LeBeau
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
| | - Fang Li
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSaint PaulUnited States
- Center for Coronavirus Research, University of MinnesotaSaint PaulUnited States
| |
Collapse
|
11
|
Structural Characterization of a Minimal Antibody against Human APOBEC3B. Viruses 2021; 13:v13040663. [PMID: 33921405 PMCID: PMC8070380 DOI: 10.3390/v13040663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 01/03/2023] Open
Abstract
APOBEC3B (A3B) is one of seven human APOBEC3 DNA cytosine deaminases that restrict viral infections as part of the overall innate immune response, but it also plays a major role in tumor evolution by mutating genomic DNA. Given the importance of A3B as a restriction factor of viral infections and as a driver of multiple human cancers, selective antibodies against A3B are highly desirable for its specific detection in various research and possibly diagnostic applications. Here, we describe a high-affinity minimal antibody, designated 5G7, obtained via a phage display screening against the C-terminal catalytic domain (ctd) of A3B. 5G7 also binds APOBEC3A that is highly homologous to A3Bctd but does not bind the catalytic domain of APOBEC3G, another Z1-type deaminase domain. The crystal structure of 5G7 shows a canonical arrangement of the heavy and light chain variable domains, with their complementarity-determining region (CDR) loops lining an antigen-binding cleft that accommodates a pair of α-helices. To understand the mechanism of A3Bctd recognition by 5G7, we used the crystal structures of A3Bctd and 5G7 as templates and computationally predicted the A3B-5G7 complex structure. Stable binding poses obtained by the simulation were further tested by site-directed mutagenesis and in vitro binding analyses. These studies mapped the epitope for 5G7 to a portion of C-terminal α6 helix of A3Bctd, with Arg374 playing an essential role. The same region of A3Bctd was used previously as a peptide antigen for generating a rabbit monoclonal antibody (mAb 5210-87-13), suggesting that this region is particularly immunogenic and that these antibodies from very different origins may share similar binding modes. Our studies provide a platform for the development of selective antibodies against A3B and other APOBEC3 family enzymes.
Collapse
|
12
|
Ye G, Gallant JP, Massey C, Shi K, Tai W, Zheng J, Odle AE, Vickers MA, Shang J, Wan Y, Drelich A, Kempaiah KR, Tat V, Perlman S, Du L, Tseng CT, Aihara H, LeBeau AM, Li F. The Development of a Novel Nanobody Therapeutic for SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.11.17.386532. [PMID: 33236012 PMCID: PMC7685322 DOI: 10.1101/2020.11.17.386532] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Combating the COVID-19 pandemic requires potent and low-cost therapeutics. We identified a novel series of single-domain antibodies (i.e., nanobody), Nanosota-1, from a camelid nanobody phage display library. Structural data showed that Nanosota-1 bound to the oft-hidden receptor-binding domain (RBD) of SARS-CoV-2 spike protein, blocking out viral receptor ACE2. The lead drug possessing an Fc tag ( Nanosota-1C-Fc ) bound to SARS-CoV-2 RBD with a K d of 15.7picomolar (∼3000 times more tightly than ACE2 did) and inhibited SARS-CoV-2 infection with an ND 50 of 0.16microgram/milliliter (∼6000 times more potently than ACE2 did). Administered at a single dose, Nanosota-1C-Fc demonstrated preventive and therapeutic efficacy in hamsters subjected to SARS-CoV-2 infection. Unlike conventional antibody drugs, Nanosota-1C-Fc was produced at high yields in bacteria and had exceptional thermostability. Pharmacokinetic analysis of Nanosota-1C-F c documented a greater than 10-day in vivo half-life efficacy and high tissue bioavailability. Nanosota-1C-Fc is a potentially effective and realistic solution to the COVID-19 pandemic. IMPACT STATEMENT Potent and low-cost Nanosota-1 drugs block SARS-CoV-2 infections both in vitro and in vivo and act both preventively and therapeutically.
Collapse
Affiliation(s)
- Gang Ye
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, USA
| | - Joseph P. Gallant
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Christopher Massey
- Institutional Office of Regulated Nonclinical Studies, University of Texas Medical Branch, Galveston, TX, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Wanbo Tai
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Jian Zheng
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Abby E. Odle
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Molly A. Vickers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Jian Shang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, USA
| | - Yushun Wan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, USA
| | - Aleksandra Drelich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kempaiah R. Kempaiah
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Vivian Tat
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lanying Du
- Laboratory of Viral Immunology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Chien-Te Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Center of Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston, TX, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Aaron M. LeBeau
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Fang Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, USA
| |
Collapse
|
13
|
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: 20] [Impact Index Per Article: 5.0] [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
|
14
|
Li C, Li J, Xu Y, Zhan Y, Li Y, Song T, Zheng J, Yang H. Application of Phage-Displayed Peptides in Tumor Imaging Diagnosis and Targeting Therapy. Int J Pept Res Ther 2020; 27:587-595. [PMID: 32901205 PMCID: PMC7471523 DOI: 10.1007/s10989-020-10108-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022]
Abstract
Phage display is an effective and powerful technique that provides a route to discovery unique peptides targeting to tumor cells. Specifically binding peptides are considered as the valuable target directing molecule fragments with potential efficiency to improve the current tumor clinic, and offer new approaches for tumor prevention, diagnosis and treatment. We focus on the recent advances in the isolation of tumor-targeting peptides by biopanning methods, with particular emphasis on molecular imaging, and pharmaceutical targeting therapy.
Collapse
Affiliation(s)
- Chunyan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| | - Jia Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| | - Ying Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| | - Ying Zhan
- 518 Hospital of PLA, Xi'an, 710043 Shaanxi China
| | - Yu Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| | - Tingting Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| | - Jiao Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| | - Hong Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Air Force Medical University, 127 West ChangLe Road, Xi'an, 710032 Shaanxi China
| |
Collapse
|
15
|
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: 25] [Impact Index Per Article: 6.3] [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
|
16
|
Li Z, Zheng Z, Li C, Li Z, Wu J, Zhang B. Therapeutic drugs and drug delivery systems targeting stromal cells for cancer therapy: a review. J Drug Target 2020; 28:714-726. [DOI: 10.1080/1061186x.2020.1744157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Zhaohuan Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zengjuan Zheng
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zhipeng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|