1
|
Ur Rehman O, Fatima E. Novel Antiendosialin Therapy and Soft Tissue Sarcoma in Children. J Pediatr Hematol Oncol 2023; 45:e1037-e1038. [PMID: 37794573 DOI: 10.1097/mph.0000000000002763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Affiliation(s)
- Obaid Ur Rehman
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | | |
Collapse
|
2
|
Gnesin S, Chouin N, Cherel M, Dunn SM, Schaefer N, Faivre-Chauvet A, Prior JO, Delage JA. From bench to bedside: 64Cu/ 177Lu 1C1m-Fc anti TEM-1: mice-to-human dosimetry extrapolations for future theranostic applications. EJNMMI Res 2023; 13:59. [PMID: 37314509 DOI: 10.1186/s13550-023-01010-4] [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: 04/24/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023] Open
Abstract
The development of diagnostic and therapeutic radiopharmaceuticals is an hot topic in nuclear medicine. Several radiolabeled antibodies are under development necessitating both biokinetic and dosimetry extrapolations for effective human translation. The validation of different animal-to-human dosimetry extrapolation methods still is an open issue. This study reports the mice-to-human dosimetry extrapolation of 64Cu/177Lu 1C1m-Fc anti-TEM-1 for theranostic application in soft-tissue sarcomas. We adopt four methods; direct mice-to-human extrapolation (M1); dosimetry extrapolation considering a relative mass scaling factor (M2), application of a metabolic scaling factor (M3) and combination of M2 and M3 (M4). Predicted in-human dosimetry for the [64Cu]Cu-1C1m-Fc resulted in an effective dose of 0.05 mSv/MBq. Absorbed dose (AD) extrapolation for the [177Lu]Lu-1C1m-Fc indicated that the AD of 2 Gy and 4 Gy to the red-marrow and total-body can be reached with 5-10 GBq and 25-30 GBq of therapeutic activity administration respectively depending on applied dosimetry method. Dosimetry extrapolation methods provided significantly different absorbed doses in organs. Dosimetry properties for the [64Cu]Cu-1C1m-Fc are suitable for a diagnostic in-human use. The therapeutic application of [177Lu]Lu-1C1m-Fc presents challenges and would benefit from further assessments in animals' models such as dogs before moving into the clinic.
Collapse
Affiliation(s)
- Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Nicolas Chouin
- Inserm, CNRS, University of Angers, Oniris, CRCI2NA, University of Nantes, Nantes, France
| | - Michel Cherel
- CHU Nantes, CNRS, Inserm, CRCINA, University of Nantes, 44000, Nantes, France
| | - Steven Mark Dunn
- LAbCore, Ludwig Institute for Cancer Research, Lausanne University Hospital and University of Lausanne, 1066, Epalinges, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | | | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| | - Judith Anna Delage
- Radiopharmacy Unit, Department of Pharmacy, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| |
Collapse
|
3
|
Yusni R, Mariya S, Saepuloh U, Mariya SS, Darusman HS. Kidney cell culture Macaca fascicularis as a candidate for vaccine development and in vitro model. J Med Primatol 2023. [PMID: 37296521 DOI: 10.1111/jmp.12655] [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: 08/31/2022] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Cell culture is the proliferation of a cell population in vitro by isolating from the original tissue or growing from existing ones. One essential source is the monkey kidney cell cultures which have an essential role in biomedical study. This is due to the significant homology between the human and macaque genomes making these useful for cultivating human viruses, especially enteroviruses, and growing vaccines. METHODS This study developed cell cultures derived from the kidney of Macaca fascicularis (Mf) and validated its gene expression. RESULTS The primary cultures were successfully subcultured up to six passages, grew as monolayers, and exhibited epithelial-like morphology. The cultured cells remained heterogeneous in phenotype and they expressed CD155 and CD46 as viral receptors, cell morphology (CD24, endosialin, and vWF), proliferation, also apoptosis markers (Ki67 and p53). CONCLUSIONS These results indicated that the cell cultures can be used as in vitro model cells for vaccine development and bioactive compound.
Collapse
Affiliation(s)
- Rahmat Yusni
- Biotechnology Graduate School of Bogor Agricultural University, Bogor, Indonesia
| | - Silmi Mariya
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
| | - Uus Saepuloh
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
| | - Sela S Mariya
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
- Center for Biomedical Research, National Research and Innovation Agency of Indonesia, Cibinong Sciences center, Bogor, Indonesia
| | - Huda S Darusman
- Biotechnology Graduate School of Bogor Agricultural University, Bogor, Indonesia
- Primate Research Center Bogor Agricultural University, Bogor, Indonesia
- Primatology Graduate School of Bogor Agricultural University, Bogor, Indonesia
- Faculty of Veterinary Medicine Bogor Agricultural University, Bogor, Indonesia
| |
Collapse
|
4
|
Lu S, Lu T, Zhang J, Gan L, Wu X, Han D, Zhang K, Xu C, Liu S, Qin W, Yang F, Wen W. CD248 promotes migration and metastasis of osteosarcoma through ITGB1-mediated FAK-paxillin pathway activation. BMC Cancer 2023; 23:290. [PMID: 36997926 PMCID: PMC10061858 DOI: 10.1186/s12885-023-10731-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common malignant bone tumor with a high incidence in children and adolescents. Frequent tumor metastasis and high postoperative recurrence are the most common challenges in OS. However, detailed mechanism is largely unknown. METHODS We examined the expression of CD248 in OS tissue microarrays by immunohistochemistry (IHC) staining. We studied the biological function of CD248 in cell proliferation, invasion and migration of OS cells by CCK8 assay, transwell and wound healing assay. We also studied its function in the metastasis of OS in vivo. At last, we explored the potential mechanism how CD248 promotes OS metastasis by using RNA-seq, western blot, immunofluorescence staining and co-immunoprecipitation using CD248 knockdown OS cells. RESULTS CD248 was highly expressed in OS tissues and its high expression was correlated with pulmonary metastasis of OS. Knockdown of CD248 in OS cells significantly inhibited cell migration, invasion and metastasis, while had no obvious effect on cell proliferation. Lung metastasis in nude mice was significantly inhibited when CD248 was knocked down. Mechanistically, we found that CD248 could promote the interaction between ITGB1 and extracellular matrix (ECM) proteins like CYR61 and FN, which activated the FAK-paxillin pathway to promote the formation of focal adhesion and metastasis of OS. CONCLUSION Our data showed that high CD248 expression is correlated with the metastatic potential of OS. CD248 may promote migration and metastasis through enhancing the interaction between ITGB1 and certain ECM proteins. Therefore, CD248 is a potential marker for diagnosis and effective target for the treatment of metastatic OS.
Collapse
Affiliation(s)
- Shiqi Lu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jiayu Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lunbiao Gan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Xinjie Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Fa Yang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
| |
Collapse
|
5
|
Rijs Z, Belt E, Kalisvaart GM, Sier CFM, Kuppen PJK, Cleven AHG, Vahrmeijer AL, van de Sande MAJ, van Driel PBAA. Immunohistochemical Evaluation of Candidate Biomarkers for Fluorescence-Guided Surgery of Myxofibrosarcoma Using an Objective Scoring Method. Biomedicines 2023; 11:biomedicines11030982. [PMID: 36979961 PMCID: PMC10046284 DOI: 10.3390/biomedicines11030982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/22/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
INTRODUCTION Myxofibrosarcoma (MFS) is the most common soft-tissue sarcoma subtype in elderly patients. Local recurrence (LR) remains a major concern as the lack of intraoperative guidance and an infiltrative growth pattern with long, slender tails hamper surgeons' ability to achieve adequate resection margins for MFS. Fluorescence-guided surgery (FGS) could overcome this concern by delineating tumor tissue during surgery. One of the most important steps to successful FGS is to define a tumor-specific biomarker that is highly overexpressed in tumor tissue while low or absent in adjacent healthy tissue. The aim of this study is to evaluate the expression of eight previously selected promising biomarkers for FGS in MFS tissue samples with adjacent healthy tissue using immunohistochemistry (IHC). METHODS The following eight biomarkers were stained in seventeen paraffin-embedded MFS samples: tumor endothelial marker-1 (TEM-1, also known as endosialin/CD248), vascular endothelial growth factor receptor-1 (VEGFR-1, also known as Flt-1), vascular endothelial growth factor receptor-2 (VEGFR-2, also known as Flk1), vascular endothelial growth factor-A (VEGF-A), epidermal growth factor receptor (EGFR), insulin-like growth factor-1 receptor (IGF-1R), platelet derived growth factor receptor-α (PDGFR-α), and cluster of differentiation 40 (CD40, also known as TNFRSF5). A pathologist specializing in sarcoma annotated the margin between the tumor and adjacent healthy tissue in each MFS tissue sample. Subsequently, we used an objective IHC scoring method to assess and compare the difference in staining intensity between the tumor and adjacent healthy tissue, which is crucial for the use of FGS. RESULTS TEM-1, VEGF-A, and PDGFR-α stained all MFS tumors, while the other biomarkers did not show expression in all MFS tumors. Ultimately, TEM-1 was identified as the most suitable biomarker for FGS in MFS based on higher tumor-to-background (TBR) staining intensity compared to VEGF-A and PDGFR-α, regardless of preoperative therapy. CONCLUSION TEM-1-targeted FGS tracers should be further investigated to optimize MFS treatment.
Collapse
Affiliation(s)
- Zeger Rijs
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Esther Belt
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gijsbert M Kalisvaart
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Percuros BV, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Pathology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michiel A J van de Sande
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | |
Collapse
|
6
|
Krishnan S, Manoharan J, Wang H, Gupta D, Fatima S, Yu Y, Mathew A, Li Z, Kohli S, Schwab C, Körner A, Mertens PR, Nawroth P, Shahzad K, Naumann M, Isermann B, Biemann R. CD248 induces a maladaptive unfolded protein response in diabetic kidney disease. Kidney Int 2023; 103:304-319. [PMID: 36309126 DOI: 10.1016/j.kint.2022.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/06/2022]
Abstract
Dysfunction of mesangial cells plays a major role in the pathogenesis of diabetic kidney disease (DKD), the leading cause of kidney failure. However, the underlying molecular mechanisms are incompletely understood. By unbiased gene expression analysis of glucose-exposed mesangial cells, we identified the transmembrane receptor CD248 as the most upregulated gene, and the maladaptive unfolded protein response (UPR) as one of the most stimulated pathways. Upregulation of CD248 was further confirmed in glucose-stressed mesangial cells in vitro, in kidney glomeruli isolated from diabetic mice (streptozotocin; STZ and db/db models, representing type 1 and type 2 diabetes mellitus, respectively) in vivo, and in glomerular kidney sections from patients with DKD. Time course analysis revealed that glomerular CD248 induction precedes the onset of albuminuria, mesangial matrix expansion and maladaptive UPR activation (hallmarked by transcription factor C/EBP homologous protein (CHOP) induction) but is paralleled by loss of the adaptive UPR regulator spliced X box binding protein (XBP1). Mechanistically, CD248 promoted maladaptive UPR signaling via inhibition of the inositol requiring enzyme 1α (IRE1α)-mediated transcription factor XBP1 splicing in vivo and in vitro. CD248 induced a multiprotein complex comprising heat shock protein 90, BH3 interacting domain death agonist (BID) and IRE1α, in which BID impedes IRE1α-mediated XBP1 splicing and induced CHOP mediated maladaptive UPR signaling. While CD248 knockout ameliorated DKD-associated glomerular dysfunction and reverses maladaptive unfolded protein response signaling, concomitant XBP1 deficiency abolished the protective effect in diabetic CD248 knockout mice, supporting a functional interaction of CD248 and XBP1 in vivo. Hence, CD248 is a novel mesangial cell receptor inducing maladaptive UPR signaling in DKD.
Collapse
Affiliation(s)
- Shruthi Krishnan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jayakumar Manoharan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Hongjie Wang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dheerendra Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Sameen Fatima
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yanfei Yu
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Zhen Li
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Constantin Schwab
- Tissue Bank of the National Center for Tumor Diseases, Heidelberg, Germany
| | - Antje Körner
- Leipzig University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Peter Nawroth
- Department of Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), University of Heidelberg, Heidelberg, Germany
| | - Khurrum Shahzad
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany.
| |
Collapse
|
7
|
Wu C, Sun W, Shen D, Li H, Tong X, Guo Y. TEM1 up-regulates MMP-2 and promotes ECM remodeling for facilitating invasion and migration of uterine sarcoma. Discov Oncol 2023; 14:5. [PMID: 36639546 PMCID: PMC9839929 DOI: 10.1007/s12672-023-00613-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 01/09/2023] [Indexed: 04/17/2023] Open
Abstract
OBJECTIVES To explore the correlation between tumor endothelial marker 1 (TEM1) and matrix metalloproteinase 2 (MMP-2) in uterine sarcoma and their roles in the progression of uterine sarcoma. METHODS Uterine leiomyosarcoma (uLMS, n = 25) and uterine leiomyoma (n = 25) specimens were collected from a total of 50 patients. Immunohistochemistry assay was conducted to determine the expression of TEM1, MMP-2 and MMP-9. TEM1 over expression (hTEM1) and low expression (shRNA-TEM1) MES-SA cell lines were established as in vitro uterine sarcoma models. MMP-2 mRNA, protein expression and enzymatic activity were verified using qPCR, Western blot and gelatin zymography respectively. MMP-2 expression was downregulated using MMP-2 siRNA in hTEM1 MES-SA cells to better study the role of MMP-2. The invasive and migratory capacities of hTEM1, shRNA-TEM1, and hTEM1 treated with MMP-2 siRNA MES-SA cells were determined using transwell assays. Extracellular matrix (ECM) remodeling mediated by TEM1 was examined using cell-ECM adhesion and fluorescent gelatin-ECM degradation assays. The immunofluorescence of F-actin was examined to analyze the formation of invadopodia. Subcutaneous and intraperitoneal xenografts were established to validate the role of TEM1 in promoting uterine sarcoma metastasis. RESULTS TEM1 and MMP-2 were expressed in 92% (n = 23) and 88% (n = 22) of uterine leiomyosarcoma specimens, respectively. Both TEM1 and MMP-2 were highly expressed in 100% (n = 17) of high stage (III-IV) uterine leiomyosarcoma specimens. In addition, TEM1 expression was positively correlated with MMP-2 expression in uterine leiomyosarcoma. The successful establishment of in vitro uterine sarcoma models was confirmed with qPCR and Western blotting tests. TEM1 promoted the invasion and metastasis of uterine sarcoma in vivo and in vitro. MMP-2 expression and activity were up-regulated in hTEM1 cells but down-regulated in shRNA-TEM1 cells. Importantly, MMP-2 knockdown impaired the invasive and migratory capacity of hTEM1 cells. TEM1 promoted ECM remodeling by increasing cell-ECM adhesion and ECM degradation. TEM1 overexpression also induced the formation of invadopodia. CONCLUSION TEM1 was co-expressed and positively correlated with MMP-2 in uterine leiomyosarcoma specimens. In addition, both TEM1 and MMP-2 were associated with tumor development. TEM1 promoted uterine sarcoma progression by regulating MMP-2 activity and ECM remodeling.
Collapse
Affiliation(s)
- Chenghao Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, People's Republic of China
| | - Wenhuizi Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, People's Republic of China
| | - Dongsheng Shen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, People's Republic of China
| | - Huaifang Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, People's Republic of China
| | - Xiaowen Tong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, People's Republic of China
| | - Yi Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, No.389 Xincun Road, Shanghai, 200065, People's Republic of China.
| |
Collapse
|
8
|
Xie D, Wang Z, Li J, Guo DA, Lu A, Liang C. Targeted Delivery of Chemotherapeutic Agents for Osteosarcoma Treatment. Front Oncol 2022; 12:843345. [PMID: 35311145 PMCID: PMC8931218 DOI: 10.3389/fonc.2022.843345] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
Since osteosarcoma (OS) is an aggressive bone cancer with unknown molecular pathways of etiology and pathophysiology, improving patient survival has long been a challenge. The conventional therapy is a complex multidisciplinary management that include radiotherapy, chemotherapy which followed by surgery and then post-operative adjuvant chemotherapy. However, they have severe side effects because the majority of the medicines used have just a minor selectivity for malignant tissue. As a result, treating tumor cells specifically without damaging healthy tissue is currently a primary goal in OS therapy. The coupling of chemotherapeutic drugs with targeting ligands is a unique therapy method for OS that, by active targeting, can overcome the aforementioned hurdles. This review focuses on advances in ligands and chemotherapeutic agents employed in targeted delivery to improve the capacity of active targeting and provide some insight into future therapeutic research for OS.
Collapse
Affiliation(s)
- Duoli Xie
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Zhuqian Wang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - De-an Guo
- National Engineering Laboratory for Standardization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica of the Chinese Academy of Sciences, Shanghai, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- *Correspondence: Chao Liang, ; Aiping Lu,
| | - Chao Liang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- *Correspondence: Chao Liang, ; Aiping Lu,
| |
Collapse
|
9
|
Fierle JK, Brioschi M, de Tiani M, Wetterwald L, Atsaves V, Abram-Saliba J, Petrova TV, Coukos G, Dunn SM. Soluble trivalent engagers redirect cytolytic T cell activity toward tumor endothelial marker 1. CELL REPORTS MEDICINE 2021; 2:100362. [PMID: 34467246 PMCID: PMC8385295 DOI: 10.1016/j.xcrm.2021.100362] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/17/2021] [Accepted: 07/08/2021] [Indexed: 01/07/2023]
Abstract
Tumor endothelial marker 1 (TEM1) is an emerging cancer target with a unique dual expression profile. First, TEM1 is expressed in the stroma and neo-vasculature of many human carcinomas but is largely absent from healthy adult tissues. Second, TEM1 is expressed by tumor cells of mesenchymal origin, notably sarcoma. Here, we present two fully human anti-TEM1 single-chain variable fragment (scFv) reagents, namely, 1C1m and 7G22, that recognize distinct regions of the extracellular domain and possess substantially different affinities. In contrast to other, well-described anti-TEM1 binders, these fragments confer cytolytic activity when expressed as 2nd generation chimeric antigen receptors (CARs). Moreover, both molecules selectively redirect human T cell effector functions toward TEM1+ tumor cells when incorporated into experimental soluble bispecific trivalent engagers that we term TriloBiTEs (tBs). Furthermore, systemic delivery of 1C1m-tB prevents the establishment of Ewing sarcoma tumors in a xenograft model. Our observations confirm TEM1 as a promising target for cancer immunotherapy and illustrate the prospective translational potential of certain scFv-based reagents.
Collapse
Affiliation(s)
- Julie K Fierle
- LAbCore Immunoglobulin Discovery Platform, Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - Matteo Brioschi
- LAbCore Immunoglobulin Discovery Platform, Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - Mariastella de Tiani
- LAbCore Immunoglobulin Discovery Platform, Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - Laureline Wetterwald
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - Vasileios Atsaves
- LAbCore Immunoglobulin Discovery Platform, Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - Johan Abram-Saliba
- LAbCore Immunoglobulin Discovery Platform, Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland.,Department of Oncology, Centre hospitalier universitaire vaudois (CHUV), 1011 Lausanne, Switzerland
| | - Steven M Dunn
- LAbCore Immunoglobulin Discovery Platform, Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066 Epalinges, Switzerland.,Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|
10
|
Antibody Conjugates for Sarcoma Therapy: How Far along Are We? Biomedicines 2021; 9:biomedicines9080978. [PMID: 34440182 PMCID: PMC8392509 DOI: 10.3390/biomedicines9080978] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 01/12/2023] Open
Abstract
Sarcomas are one of the most difficult type of cancer to manage and treat because of their extremely heterogeneous molecular and morphological features. Despite the progress made over the years in the establishment of standard protocols for high and low grading/staging sarcoma patients, mostly with chemotherapy and/or radiotherapy, 50% of treated patients experience relapse episodes. Because of this, in the last 20 years, new therapeutic approaches for sarcoma treatment have been evaluated in preclinical and clinical studies. Among them, antibody-based therapies have been the most studied. Immunoconjugates consist of a carrier portion, frequently represented by an antibody, linked to a toxic moiety, i.e., a drug, toxin, or radionuclide. While the efficacy of immunoconjugates is well demonstrated in the therapy of hematological tumors and more recently also of epithelial ones, their potential as therapeutic agents against sarcomas is still not completely explored. In this paper, we summarize the results obtained with immunoconjugates targeting sarcoma surface antigens, considering both preclinical and clinical studies. To date, the encouraging results obtained in preclinical studies allowed nine immunoconjugates to enter clinical trials, demonstrating the validity of immunotherapy as a promising pharmacological tool also for sarcoma therapy.
Collapse
|
11
|
Milone MC, Xu J, Chen SJ, Collins MA, Zhou J, Powell DJ, Melenhorst JJ. Engineering enhanced CAR T-cells for improved cancer therapy. NATURE CANCER 2021; 2:780-793. [PMID: 34485921 PMCID: PMC8412433 DOI: 10.1038/s43018-021-00241-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/28/2021] [Indexed: 12/19/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have evolved from a research tool to a paradigm-shifting therapy with impressive responses in B cell malignancies. This review summarizes the current state of the CAR T-cell field, focusing on CD19- and B cell maturation antigen-directed CAR T-cells, the most developed of the CAR T-cell therapies. We discuss the many challenges to CAR-T therapeutic success and innovations in CAR design and T-cell engineering aimed at extending this therapeutic platform beyond hematologic malignancies.
Collapse
Affiliation(s)
- Michael C. Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jie Xu
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Sai-Juan Chen
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - McKensie A. Collins
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiafeng Zhou
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, PR China
| | - Daniel J. Powell
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J. Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Katal S, Maldonado A, Carrascoso J, Assadi M, Gholamrezanezhad A. Theranostic Agents in Musculoskeletal Disorders. PET Clin 2021; 16:441-448. [PMID: 34053587 DOI: 10.1016/j.cpet.2021.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Theranostic-based strategies, combining therapeutic and diagnostic properties of a single agent, have gained enormous attention in the past few years. Today, various multifunctional theranostic modalities have been examined, using different bioactive targeting, for the detection, quantifying, and monitoring of therapy response in different pathologies. Herein we review the newly emerging approaches in theranostic nanomedicine for the detection and therapy for musculoskeletal disorders to provide valuable insights for developing more efficient agents for clinical use. Some potential preclinical applications of radionuclide nanotheranostic agents are described in rheumatoid arthritis, osteoarthrosis, multiple myeloma, and neoplastic diseases.
Collapse
Affiliation(s)
- Sanaz Katal
- Department of Nuclear Medicine, Kowsar Hospital, Shiraz, Iran
| | - Antonio Maldonado
- Department of Nuclear Medicine, Quironsalud Madrid University Hospital, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Javier Carrascoso
- Department of Radiology, Quironsalud Madrid University Hospital, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Majid Assadi
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Gholamrezanezhad
- Department of Diagnostic Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA.
| |
Collapse
|
13
|
Nakata E, Fujiwara T, Kunisada T, Ito T, Takihira S, Ozaki T. Immunotherapy for sarcomas. Jpn J Clin Oncol 2021; 51:523-537. [PMID: 33611603 DOI: 10.1093/jjco/hyab005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
Sarcomas are a heterogeneous group of malignancies of mesenchymal origin; their molecular and genomic mechanisms differ with regard to histology. These characteristics lead to the presentation of varied immunological profiles based on the tumor microenvironment. Various immunotherapies are considered for the treatment of sarcoma. These treatments are performed either in isolation or in combination with other methods such as cytotoxic chemotherapy or the use of molecular target agents. Among these, two recently emerging immunotherapies include T-cell receptor gene therapy and immune checkpoint inhibitor therapy, which are expected to be effective for many types of sarcoma. A sarcoma with a disease-specific translocation and a limited number of mutations, such as synovial sarcoma, expresses high levels of self-antigens, like the New York esophageal squamous cell carcinoma 1, which has been targeted in T-cell receptor gene therapy. On the other hand, sarcomas with a greater number of mutations, such as undifferentiated pleomorphic sarcomas, myxofibrosarcoma and dedifferentiated liposarcomas, can be good candidates for immune checkpoint inhibitors. Among immune checkpoint inhibitor therapies, programmed cell death-1 blockade (nivolumab and pembrolizumab) and cytotoxic T-lymphocyte-associated antigen 4 blockade (ipilimumab) have been investigated most often in sarcoma. Although the sole use of immune checkpoint inhibitors provides limited efficacy, combined immunotherapy with immune checkpoint inhibitors or molecular target agents, especially antiangiogenic agents, has shown moderate results against some types of sarcoma, such as the alveolar soft part sarcoma. Several clinical trials utilizing immunotherapy, including T-cell receptor gene therapy and immune checkpoint inhibitors, in sarcomas are under progress. By clarifying the tumor microenvironment and biomarker-predictive capacity of immunotherapy in sarcomas, better clinical trials can be designed; this could lead to improved outcomes for immunotherapy in sarcoma.
Collapse
Affiliation(s)
- Eiji Nakata
- Department of Orthopedic Surgery, Okayama University Hospital, Okayama City, Okayama, Japan
| | - Tomohiro Fujiwara
- Department of Orthopedic Surgery, Okayama University Hospital, Okayama City, Okayama, Japan
| | - Toshiyuki Kunisada
- Department of Orthopedic Surgery, Okayama University Hospital, Okayama City, Okayama, Japan
| | - Tastuo Ito
- Department of Hygiene, Kawasaki Medical University, Kurashiki City, Okayama, Japan
| | - Shota Takihira
- Department of Orthopedic Surgery, Okayama University Hospital, Okayama City, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Hospital, Okayama City, Okayama, Japan
| |
Collapse
|
14
|
Candidate Biomarkers for Specific Intraoperative Near-Infrared Imaging of Soft Tissue Sarcomas: A Systematic Review. Cancers (Basel) 2021; 13:cancers13030557. [PMID: 33535618 PMCID: PMC7867119 DOI: 10.3390/cancers13030557] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Near-infrared imaging of tumors during surgery facilitates the oncologic surgeon to distinguish malignant from healthy tissue. The technique is based on fluorescent tracers binding to tumor biomarkers on malignant cells. Currently, there are no clinically available fluorescent tracers that specifically target soft tissue sarcomas. This review searched the literature to find candidate biomarkers for soft tissue sarcomas, based on clinically used therapeutic antibodies. The search revealed 7 biomarkers: TEM1, VEGFR-1, EGFR, VEGFR-2, IGF-1R, PDGFRα, and CD40. These biomarkers are abundantly present on soft tissue sarcoma tumor cells and are already being targeted with humanized monoclonal antibodies. The conjugation of these antibodies with a fluorescent dye will yield in specific tracers for image-guided surgery of soft tissue sarcomas to improve the success rates of tumor resections. Abstract Surgery is the mainstay of treatment for localized soft tissue sarcomas (STS). The curative treatment highly depends on complete tumor resection, as positive margins are associated with local recurrence (LR) and prognosis. However, determining the tumor margin during surgery is challenging. Real-time tumor-specific imaging can facilitate complete resection by visualizing tumor tissue during surgery. Unfortunately, STS specific tracers are presently not clinically available. In this review, STS-associated cell surface-expressed biomarkers, which are currently already clinically targeted with monoclonal antibodies for therapeutic purposes, are evaluated for their use in near-infrared fluorescence (NIRF) imaging of STS. Clinically targeted biomarkers in STS were extracted from clinical trial registers and a PubMed search was performed. Data on biomarker characteristics, sample size, percentage of biomarker-positive STS samples, pattern of biomarker expression, biomarker internalization features, and previous applications of the biomarker in imaging were extracted. The biomarkers were ranked utilizing a previously described scoring system. Eleven cell surface-expressed biomarkers were identified from which 7 were selected as potential biomarkers for NIRF imaging: TEM1, VEGFR-1, EGFR, VEGFR-2, IGF-1R, PDGFRα, and CD40. Promising biomarkers in common and aggressive STS subtypes are TEM1 for myxofibrosarcoma, TEM1, and PDGFRα for undifferentiated soft tissue sarcoma and EGFR for synovial sarcoma.
Collapse
|
15
|
Smalley H, Rowe JM, Nieto F, Zeledon J, Pollard K, Tomich JM, Fleming SD. Beta2 glycoprotein I-derived therapeutic peptides induce sFlt-1 secretion to reduce melanoma vascularity and growth. Cancer Lett 2020; 495:66-75. [PMID: 32891714 PMCID: PMC7899169 DOI: 10.1016/j.canlet.2020.08.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/11/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022]
Abstract
Melanoma, a form of skin cancer, is one of the most common cancers in young men and women. Tumors require angiogenesis to provide oxygen and nutrients for growth. Pro-angiogenic molecules such as VEGF and anti-angiogenic molecules such as sFlt-1 control angiogenesis. In addition, the serum protein, Beta2 Glycoprotein I (β2-GPI) induces or inhibits angiogenesis depending on conformation and concentration. β2-GPI binds to proteins and negatively charged phospholipids on hypoxic endothelial cells present in the tumor microenvironment. We hypothesized that peptides derived from the binding domain of β2-GPI would regulate angiogenesis and melanoma growth. In vitro analyses determined the peptides reduced endothelial cell migration and sFlt-1 secretion. In a syngeneic, immunocompetent mouse melanoma model, β2-GPI-derived peptides also reduced melanoma growth in a dose-dependent response with increased sFlt-1 and attenuated vascular markers compared to negative controls. Importantly, administration of peptide with sFlt-1 antibody resulted in tumor growth. These data demonstrate the therapeutic potential of novel β2-GPI-derived peptides to attenuate tumor growth and endothelial migration is sFlt-1 dependent.
Collapse
Affiliation(s)
- Haley Smalley
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jennifer M Rowe
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Fernando Nieto
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jazmin Zeledon
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Kellyn Pollard
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - John M Tomich
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
16
|
Biological evaluation of new TEM1 targeting recombinant antibodies for radioimmunotherapy: In vitro, in vivo and in silico studies. Eur J Pharm Biopharm 2020; 158:233-244. [PMID: 33271301 DOI: 10.1016/j.ejpb.2020.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 12/27/2022]
Abstract
The tumour endothelial marker 1 (TEM1/endosialin/CD248) is a receptor overexpressed in several human solid tumours and silenced in normal adult tissues, representing a suitable and potentially safe target for radioimmunotherapy of sarcoma. To develop new tools with improved TEM1 targeting properties, a new panel of antibody fragments was for the first time evaluated preclinically following 125I radiolabelling. The antibody fragment 1C1m-Fc, with the highest human/murine TEM1 binding affinity, was extensively characterized in vitro and in vivo in a Ewing's sarcoma human xenograft mouse model. In silico studies were also performed to elucidate the influence of a single amino acid mutation in the complementarity-determining region (CDR3) of the heavy chain, upon affinity maturation of the parental clone 1C1-Fc. From this study, 1C1m-Fc emerged as a promising candidate for the development of TEM1-targeted radioimmunoconjugates, namely to be further explored for theranostic applications with other suitable medical radionuclides.
Collapse
|
17
|
Pietrzyk Ł, Wdowiak P. Endosialin (TEM1) as a Diagnostic, Progression, and Prognostic Serum Marker for Patients With Colorectal Cancer-A Preliminary Study. Cancer Control 2020; 27:1073274820903351. [PMID: 32107922 PMCID: PMC7053787 DOI: 10.1177/1073274820903351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide usually diagnosed in the advanced stage. In this study, the serum concentration of tumor endothelial marker 1 (TEM1) was measured and correlated with clinicopathological features to evaluate whether TEM1 might serve as a biomarker for early CRC diagnosis, progression, and prognosis. The concentration of TEM1 was measured in the serum samples of 45 patients with CRC and 35 healthy individuals using enzyme-linked immunosorbent assay test. The mean serum concentration of TEM1 was significantly higher in the patients with CRC compared to the healthy individuals (1.31 ± 0.16 vs 0.92 ± 0.90 ng/mL; P < .001). The mean concentration of TEM1 significantly increased in the patients having CRC with early stage (stage I + II) compared to noncancer control individuals (stage I + II vs control 1.21 ± 0.13 ng/mL: 0.92 ± 0.90 ng/mL; P < .001). The TEM1 concentration in blood serum also showed a significant association with the development of T stages (P < .001), N stages (P < .001), and M stages (P = .006). The TEM1 sensitivity and specificity in CRC detection are higher than routinely used blood markers (carcinoembryonic antigen [CEA] and carbohydrate antigen [Ca 19-9]). Patients with high TEM1 concentration (≥1.055 ng/mL) had a worse overall survival rate compared to the patients having CRC with low TEM1 concentration (<1.055 ng/mL). In conclusion, TEM1 can act as a potential diagnostic, progression, and prognostic serum biomarker for patients with CRC; TEM1 might be a good supplement for commonly used markers CEA and Ca 19-9.
Collapse
Affiliation(s)
- Łukasz Pietrzyk
- Department of Didactics and Medical Simulation, Medical University of Lublin, Lublin, Poland.,Department of General, Oncological and Minimally Invasive Surgery, 1st Military Clinical Hospital with the Outpatient Clinic, Lublin, Poland
| | - Paulina Wdowiak
- Chair and Department of Human Anatomy, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
18
|
Delage JA, Faivre-Chauvet A, Fierle JK, Gnesin S, Schaefer N, Coukos G, Dunn SM, Viertl D, Prior JO. 177Lu radiolabeling and preclinical theranostic study of 1C1m-Fc: an anti-TEM-1 scFv-Fc fusion protein in soft tissue sarcoma. EJNMMI Res 2020; 10:98. [PMID: 32804276 PMCID: PMC7431510 DOI: 10.1186/s13550-020-00685-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022] Open
Abstract
PURPOSE TEM-1 (tumor endothelial marker-1) is a single-pass transmembrane cell surface glycoprotein expressed at high levels by tumor vasculature and malignant cells. We aimed to perform a preclinical investigation of a novel anti-TEM-1 scFv-Fc fusion antibody, 1C1m-Fc, which was radiolabeled with 177Lu for use in soft tissue sarcomas models. METHODS 1C1m-Fc was first conjugated to p-SCN-Bn-DOTA using different excess molar ratios and labeled with 177Lu. To determine radiolabeled antibody immunoreactivity, Lindmo assays were performed. The in vivo behavior of [177Lu]Lu-1C1m-Fc was characterized in mice bearing TEM-1 positive (SK-N-AS) and negative (HT-1080) tumors by biodistribution and single-photon emission SPECT/CT imaging studies. Estimated organ absorbed doses were obtained based on biodistribution results. RESULTS The DOTA conjugation and the labeling with 177Lu were successful with a radiochemical purity of up to 95%. Immunoreactivity after radiolabeling was 86% ± 4%. Biodistribution showed a specific uptake in TEM-1 positive tumor versus liver as critical non-specific healthy organ, and this specificity is correlated to the number of chelates per antibody. A 1.9-fold higher signal at 72 h was observed in SPECT/CT imaging in TEM-1 positive tumors versus control tumors. CONCLUSION TEM-1 is a promising target that could allow a theranostic approach to soft-tissue sarcoma, and 1C1m-Fc appears to be a suitable targeting candidate. In this study, we observed the influence of the ratio DOTA/antibody on the biodistribution. The next step will be to investigate the best conjugation to achieve an optimal tumor-to-organ radioactivity ratio and to perform therapy in murine xenograft models as a prelude to future translation in patients.
Collapse
Affiliation(s)
- J A Delage
- Radiopharmacy Unit, Department of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - J K Fierle
- LAbCore, Ludwig Institute for Cancer Research, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - S Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - N Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - G Coukos
- Ludwig Institute for Cancer Research and Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - S M Dunn
- LAbCore, Ludwig Institute for Cancer Research, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - D Viertl
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - J O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
19
|
Cicone F, Denoël T, Gnesin S, Riggi N, Irving M, Jakka G, Schaefer N, Viertl D, Coukos G, Prior JO. Preclinical Evaluation and Dosimetry of [ 111In]CHX-DTPA-scFv78-Fc Targeting Endosialin/Tumor Endothelial Marker 1 (TEM1). Mol Imaging Biol 2020; 22:979-991. [PMID: 31993928 PMCID: PMC7343747 DOI: 10.1007/s11307-020-01479-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Endosialin/tumor endothelial marker-1 (TEM1) is an attractive theranostic target expressed by the microenvironment of a wide range of tumors, as well as by sarcoma and neuroblastoma cells. We report on the radiolabeling and preclinical evaluation of the scFv78-Fc, a fully human TEM1-targeting antibody fragment cross-reactive with mouse TEM1. PROCEDURES The scFv78-Fc was conjugated with the chelator p-SCN-Bn-CHX-A"-DTPA, followed by labeling with indium-111. The number of chelators per molecule was estimated by mass spectrometry. A conventional saturation assay, extrapolated to infinite antigen concentration, was used to determine the immunoreactive fraction of the radioimmunoconjugate. The radiopharmaceutical biodistribution was assessed in immunodeficient mice grafted with Ewing's sarcoma RD-ES and neuroblastoma SK-N-AS human TEM1-positive tumors. The full biodistribution studies were preceded by a dose-escalation experiment based on the simultaneous administration of the radiopharmaceutical with increasing amounts of unlabeled scFv78-Fc. Radiation dosimetry extrapolations to human adults were obtained from mouse biodistribution data according to established methodologies and additional assumptions concerning the impact of the tumor antigenic sink in the cross-species translation. RESULTS [111In]CHX-DTPA-scFv78-Fc was obtained with a radiochemical purity > 98 % after 1 h incubation at 42 °C and ultrafiltration. It showed good stability in human serum and > 70 % immunoreactive fraction. Biodistribution data acquired in tumor-bearing mice confirmed fast blood clearance and specific tumor targeting in both xenograft models. The radiopharmaceutical off-target uptake was predominantly abdominal. After a theoretical injection of [111In]CHX-DTPA-scFv78-Fc to the reference person, the organs receiving the highest absorbed dose would be the spleen (0.876 mGy/MBq), the liver (0.570 mGy/MBq) and the kidneys (0.298 mGy/MBq). The total body dose and the effective dose would be 0.058 mGy/MBq and 0.116 mSv/MBq, respectively. CONCLUSIONS [111In]CHX-DTPA-scFv78-Fc binds specifically to endosialin/TEM1 in vitro and in vivo. Dosimetry estimates are in the range of other monoclonal antibodies radiolabeled with indium-111. [111In]CHX-DTPA-scFv78-Fc could be potentially translated into clinic.
Collapse
Affiliation(s)
- Francesco Cicone
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Department of Experimental and Clinical Medicine, Unit of Nuclear Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy.
| | - Thibaut Denoël
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nicolo Riggi
- Experimental Pathology Service, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066, Epalinges, Switzerland
| | - Gopinadh Jakka
- Department of Oncology, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066, Epalinges, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - David Viertl
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066, Epalinges, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
20
|
de Gooyer JM, Versleijen-Jonkers YMH, Hillebrandt-Roeffen MHS, Frielink C, Desar IME, de Wilt JHW, Flucke U, Rijpkema M. Immunohistochemical selection of biomarkers for tumor-targeted image-guided surgery of myxofibrosarcoma. Sci Rep 2020; 10:2915. [PMID: 32076024 PMCID: PMC7031512 DOI: 10.1038/s41598-020-59735-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/30/2020] [Indexed: 01/04/2023] Open
Abstract
Myxofibrosarcoma(MFS) is the most common soft tissue sarcoma(STS) in elderly patients. Surgical resection remains the main treatment modality but tumor borders can be difficult to delineate with conventional clinical methods. Incomplete resections are a common problem and local recurrence remains a clinical issue. A technique that has shown great potential in improving surgical treatment of solid tumors is tumor targeted imaging and image-guided surgery with near-infrared fluorescence. To facilitate this technique, it is essential to identify a biomarker that is highly and homogenously expressed on tumor cells, while being absent on healthy non-malignant tissue. The purpose of this study was to identify suitable molecular targets for tumor-targeted imaging of myxofibrosarcoma. Ten potential molecular targets for tumor targeted imaging were investigated with immunohistochemical analysis in myxofibrosarcoma tissue (n = 34). Results were quantified according to the immunoreactive score(IRS). Moderate expression rates were found for uPAR, PDGFRa and EMA/MUC1. High expression rates of VEGF and TEM1 were seen. Strong expression was most common for TEM1 (88.2%). These results confirms that TEM1 is a suitable target for tumor-targeted imaging of myxofibrosarcoma. Keywords Image-guided surgery; Immunohistochemistry; Molecular imaging; Myxofibrosarcoma; Soft tissue sarcoma; Tumor endothelial marker 1(TEM1), Vascular endothelial growth factor (VEGF).
Collapse
Affiliation(s)
- Jan Marie de Gooyer
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, the Netherlands. .,Department of Surgery, Radboud university medical center, Nijmegen, the Netherlands.
| | | | | | - Cathelijne Frielink
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud university medical center, Nijmegen, the Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud university medical center, Nijmegen, the Netherlands
| | - Uta Flucke
- Department of Pathology, Radboud university medical center, Nijmegen, the Netherlands
| | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
21
|
Dyson KA, Stover BD, Grippin A, Mendez-Gomez HR, Lagmay J, Mitchell DA, Sayour EJ. Emerging trends in immunotherapy for pediatric sarcomas. J Hematol Oncol 2019; 12:78. [PMID: 31311607 PMCID: PMC6636007 DOI: 10.1186/s13045-019-0756-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022] Open
Abstract
While promising, immunotherapy has yet to be fully unlocked for the preponderance of cancers where conventional chemoradiation reigns. This remains particularly evident in pediatric sarcomas where standard of care has not appreciably changed in decades. Importantly, pediatric bone sarcomas, like osteosarcoma and Ewing’s sarcoma, possess unique tumor microenvironments driven by distinct molecular features, as do rhabdomyosarcomas and soft tissue sarcomas. A better understanding of each malignancy’s biology, heterogeneity, and tumor microenvironment may lend new insights toward immunotherapeutic targets in novel platform technologies for cancer vaccines and adoptive cellular therapy. These advances may pave the way toward new treatments requisite for pediatric sarcomas and patients in need of new therapies.
Collapse
Affiliation(s)
- Kyle A Dyson
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Brian D Stover
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA.,Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, PO Box 100298, Gainesville, FL, 32610, USA
| | - Adam Grippin
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Hector R Mendez-Gomez
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Joanne Lagmay
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, PO Box 100298, Gainesville, FL, 32610, USA
| | - Duane A Mitchell
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Elias J Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA. .,Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, PO Box 100298, Gainesville, FL, 32610, USA.
| |
Collapse
|
22
|
Cicone F, Gnesin S, Denoël T, Stora T, van der Meulen NP, Müller C, Vermeulen C, Benešová M, Köster U, Johnston K, Amato E, Auditore L, Coukos G, Stabin M, Schaefer N, Viertl D, Prior JO. Internal radiation dosimetry of a 152Tb-labeled antibody in tumor-bearing mice. EJNMMI Res 2019; 9:53. [PMID: 31187358 PMCID: PMC6560118 DOI: 10.1186/s13550-019-0524-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/28/2019] [Indexed: 11/10/2022] Open
Abstract
Background Biodistribution studies based on organ harvesting represent the gold standard pre-clinical technique for dose extrapolations. However, sequential imaging is becoming increasingly popular as it allows the extraction of longitudinal data from single animals, and a direct correlation with deterministic radiation effects. We assessed the feasibility of mouse-specific, microPET-based dosimetry of an antibody fragment labeled with the positron emitter 152Tb [(T1/2 = 17.5 h, Eβ+mean = 1140 keV (20.3%)]. Image-based absorbed dose estimates were compared with those obtained from the extrapolation to 152Tb of a classical biodistribution experiment using the same antibody fragment labeled with 111In. 152Tb was produced by proton-induced spallation in a tantalum target, followed by mass separation and cation exchange chromatography. The endosialin-targeting scFv78-Fc fusion protein was conjugated with the chelator p-SCN-Bn-CHX-A”-DTPA, followed by labeling with either 152Tb or 111In. Micro-PET images of four immunodeficient female mice bearing RD-ES tumor xenografts were acquired 4, 24, and 48 h after the i.v. injection of 152Tb-CHX-DTPA-scFv78-Fc. After count/activity camera calibration, time-integrated activity coefficients (TIACs) were obtained for the following compartments: heart, lungs, liver, kidneys, intestines, tumor, and whole body, manually segmented on CT. For comparison, radiation dose estimates of 152Tb-CHX-DTPA-scFv78-Fc were extrapolated from mice dissected 4, 24, 48, and 96 h after the injection of 111In-CHX-DTPA-scFv78-Fc (3–5 mice per group). Imaging-derived and biodistribution-derived organ TIACs were used as input in the 25 g mouse model of OLINDA/EXM® 2.0, after appropriate mass rescaling. Tumor absorbed doses were obtained using the OLINDA2 sphere model. Finally, the relative percent difference (RD%) between absorbed doses obtained from imaging and biodistribution were calculated. Results RD% between microPET-based dosimetry and biodistribution-based dose extrapolations were + 12, − 14, and + 17 for the liver, the kidneys, and the tumors, respectively. Compared to biodistribution, the imaging method significantly overestimates the absorbed doses to the heart and the lungs (+ 89 and + 117% dose difference, respectively). Conclusions MicroPET-based dosimetry of 152Tb is feasible, and the comparison with organ harvesting resulted in acceptable dose discrepancies for body districts that can be segmented on CT. These encouraging results warrant additional validation using radiolabeled biomolecules with a different biodistribution pattern.
Collapse
Affiliation(s)
- Francesco Cicone
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, CH-1011, Lausanne, CH, Switzerland.
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne, CH, Switzerland
| | - Thibaut Denoël
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, CH-1011, Lausanne, CH, Switzerland
| | | | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute (PSI), Villigen, CH, Switzerland.,Laboratory of Radiochemistry, Paul Scherrer Institute (PSI), Villigen, CH, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute (PSI), Villigen, CH, Switzerland
| | - Christiaan Vermeulen
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute (PSI), Villigen, CH, Switzerland
| | - Martina Benešová
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute (PSI), Villigen, CH, Switzerland
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, FR, France
| | | | - Ernesto Amato
- Section of Radiological Sciences, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, IT, Italy
| | - Lucrezia Auditore
- Section of Radiological Sciences, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, IT, Italy
| | - George Coukos
- Department of Oncology and Ludwig Center for Cancer Research, Lausanne, CH, Switzerland
| | | | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, CH-1011, Lausanne, CH, Switzerland
| | - David Viertl
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, CH-1011, Lausanne, CH, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, CH-1011, Lausanne, CH, Switzerland
| |
Collapse
|
23
|
Teicher BA. CD248: A therapeutic target in cancer and fibrotic diseases. Oncotarget 2019; 10:993-1009. [PMID: 30847027 PMCID: PMC6398180 DOI: 10.18632/oncotarget.26590] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/22/2018] [Indexed: 01/07/2023] Open
Abstract
CD248/endosialin/TEM1 is a type 1 transmembrane glycoprotein found on the plasma membrane of activated mesenchymal cells. CD248 functions during embryo development and is either not expressed or found at very low levels in adult tissues. CD248 is expressed at high levels by malignant sarcoma cells, by the pericyte component of tumor vasculature and by mesenchymal cells in some fibrotic diseases. CD248 is being targeted by several experimental therapeutics including antibodies, antibody drug conjugates, as an antigen for CART cells and in therapeutic vaccines. Although the function of CD248 has yet to be fully elucidated, this protein is a potential broad scope therapeutic target.
Collapse
Affiliation(s)
- Beverly A Teicher
- Molecular Pharmacology Branch, Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda 20892, MD, USA
| |
Collapse
|