1
|
Kareff SA, Corbett V, Hallenbeck P, Chauhan A. TEM8 in Oncogenesis: Protein Biology, Pre-Clinical Agents, and Clinical Rationale. Cells 2023; 12:2623. [PMID: 37998358 PMCID: PMC10670355 DOI: 10.3390/cells12222623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
The TEM8 protein represents an emerging biomarker in many solid tumor histologies. Given the various roles it plays in oncogenesis, including but not limited to angiogenesis, epithelial-to-mesenchymal transition, and cell migration, TEM8 has recently served and will continue to serve as the target of novel oncologic therapies. We review herein the role of TEM8 in oncogenesis. We review its normal function, highlight the additional roles it plays in the tumor microenvironment, and synthesize pre-clinical and clinical data currently available. We underline the protein's prognostic and predictive abilities in various solid tumors by (1) highlighting its association with more aggressive disease biology and poor clinical outcomes and (2) assessing its associated clinical trial landscape. Finally, we offer future directions for clinical studies involving TEM8, including incorporating pre-clinical agents into clinical trials and combining previously tested oncologic therapies with currently available treatments, such as immunotherapy.
Collapse
Affiliation(s)
- Samuel A. Kareff
- University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33136, USA
| | | | | | - Aman Chauhan
- Division of Medical Oncology, Department of Medicine, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| |
Collapse
|
2
|
Zahedipour F, Zamani P, Jamialahmadi K, Jaafari MR, Sahebkar A. Vaccines targeting angiogenesis in melanoma. Eur J Pharmacol 2021; 912:174565. [PMID: 34656608 DOI: 10.1016/j.ejphar.2021.174565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis has a significant role in metastasis and progression of melanoma. Even small tumors may be susceptible to metastasis and hence lead to a worse outcome in patients with melanoma. One of the anti-angiogenic treatment approaches that is undergoing comprehensive study is specific immunotherapy. While tumor cells are challenging targets for immunotherapy due to their genetic instability and heterogeneity, endothelial cells (ECs) are genetically stable. Therefore, vaccines targeting angiogenesis in melanoma are appropriate choices that target both tumor cells and ECs while capable of inducing strong, anti-tumor immune responses with limited toxicity. The main targets of angiogenesis are VEGFs and their receptors but other potential targets have also been investigated, especially in preclinical studies. Various types of vaccines that target angiogenesis in melanoma have been studied including DNA, peptide, protein, dendritic cell-based, and endothelial cell vaccines. This review outlines a number of target antigens that are important for potential progress in developing vaccines for targeting angiogenesis in melanoma. We also discuss different types of vaccines that have been investigated, delivery mechanisms and popular adjuvants, and suggest ways to improve future clinical outcomes.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Sun KR, Lv HF, Chen BB, Nie CY, Zhao J, Chen XB. Latest therapeutic target for gastric cancer: Anthrax toxin receptor 1. World J Gastrointest Oncol 2021; 13:216-222. [PMID: 33889273 PMCID: PMC8040068 DOI: 10.4251/wjgo.v13.i4.216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/23/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
Anthrax toxin receptor 1 (ANTXR1), also known as tumor endothelial marker 8, is a highly conserved cell surface protein overexpressed in tumor-infiltrating vessels. It was first found in vascular endothelial cells of human colorectal cancer. Although our understanding of its physiological function is limited, it has been found that ANTXR1 binds collagen and promotes migration of endothelial cells in vitro. ANTXR1 is upregulated in vessels of different tumor types in mice and humans, and is also expressed by tumor cells themselves in some tumors, such as gastric, lung, intestinal and breast cancer. Developmental angiogenesis and wound healing were not disturbed in ANTXR1 knockout mice, but compared with wild-type mice, growth of melanoma was impaired after ANTXR1 knockout, indicating that host-derived ANTXR1 can promote tumor growth on the basis of immune activity. Previous studies have shown that ANTXR1 vaccines or sublethal doses of anthrax toxin can inhibit angiogenesis, slow tumor growth and prolong survival. These studies suggest that ANTXR1 is necessary for tumor rather than physiological angiogenesis. It has been found that ANTXR1 plays an important role in tumor angiogenesisas well as in the growth and metastasis of many kinds of tumors. This article reviews the physiological function of ANTXR1 and its role in different kinds of cancer.
Collapse
Affiliation(s)
- Ke-Ran Sun
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Hui-Fang Lv
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Bei-Bei Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Cai-Yun Nie
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Jing Zhao
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Xiao-Bing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| |
Collapse
|
4
|
Sabbieti MG, Lacava G, Amaroli A, Marchetti L, Censi R, Di Martino P, Agas D. Molecular Adjuvants Based on Plasmids Encoding Protein Aggregation Domains Affect Bone Marrow Niche Homeostasis. Curr Gene Ther 2019; 17:391-397. [PMID: 29303078 PMCID: PMC6751345 DOI: 10.2174/1566523218666180105122626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/12/2017] [Accepted: 12/29/2017] [Indexed: 12/19/2022]
Abstract
Background: During last years, DNA vaccine immunogenicity has been optimized by the employment of co-stimulatory molecules and molecular adjuvants. It has been reported that plasmid (pATRex), encompassing the DNA sequence for the von Willebrand A (vWA/A) domain of the An-thrax Toxin Receptor-1 (ANTXR-1, alias TEM8, Tumor Endothelial Marker 8), acts as strong immune adjuvant by inducing formation of insoluble intracellular aggregates. Markedly, we faced with upsetting findings regarding the safety of pATRex as adjuvant since the aggregosome formation prompted to os-teopenia in mice. Objective: The present study provides additional evidences about the proteinaceous adjuvants action within bone marrow and questioned regarding the self-aggregation protein adjuvants immunotoxicity on marrow niches. Methods & Results: Using histological, biochemical and proteomic assays we shed light on pATRex effects within bone marrow niche and specifically we evidenced an aplastic-like bone marrow with dis-rupted cytokine/chemokine production. Conclusion: The above findings provide compelling support to the thesis that adjuvants based on plas-mids encoding protein aggregation domains disrupt the physiological features of the bone marrow ele-ments.
Collapse
Affiliation(s)
- Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino (MC), Italy
| | - Giovanna Lacava
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino (MC), Italy
| | - Andrea Amaroli
- Department of Surgical and Diagnostic Sciences, University of Genova, Genova, Italy
| | - Luigi Marchetti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino (MC), Italy
| | - Roberta Censi
- School of Pharmacy, University of Camerino, Camerino, (MC), Italy
| | - Piera Di Martino
- School of Pharmacy, University of Camerino, Camerino, (MC), Italy
| | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino (MC), Italy
| |
Collapse
|
5
|
Evans DJ, Wasinger AM, Brey RN, Dunleavey JM, St Croix B, Bann JG. Seneca Valley Virus Exploits TEM8, a Collagen Receptor Implicated in Tumor Growth. Front Oncol 2018; 8:506. [PMID: 30460197 PMCID: PMC6232524 DOI: 10.3389/fonc.2018.00506] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/16/2018] [Indexed: 12/25/2022] Open
Abstract
Recent studies reveal that Seneca Valley Virus (SVV) exploits tumor endothelial marker 8 (TEM8) for cellular entry, the same surface receptor pirated by bacterial-derived anthrax toxin. This observation is particularly significant as SVV is a known oncolytic virus which selectively infects and kills tumor cells, particularly those of neuroendocrine origin. TEM8 is a transmembrane glycoprotein that is preferentially upregulated in some tumor cell and tumor-associated stromal cell populations. Both TEM8 and SVV have been evaluated for targeting of tumors of multiple origins, but the connection between the two was previously unknown. Here, we review currently understood interactions between TEM8 and SVV, anthrax protective antigen (PA), and collagen VI, a native binding partner of TEM8, with an emphasis on potential therapeutic directions moving forward.
Collapse
Affiliation(s)
- David J Evans
- Department of Chemistry, Wichita State University, Wichita, KS, United States
| | - Alexa M Wasinger
- Department of Chemistry, Wichita State University, Wichita, KS, United States
| | | | - James M Dunleavey
- Tumor Angiogenesis Unit, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD, United States
| | - James G Bann
- Department of Chemistry, Wichita State University, Wichita, KS, United States
| |
Collapse
|
6
|
Batista-Duharte A, Martínez DT, Carlos IZ. Efficacy and safety of immunological adjuvants. Where is the cut-off? Biomed Pharmacother 2018; 105:616-624. [DOI: 10.1016/j.biopha.2018.06.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/05/2018] [Accepted: 06/05/2018] [Indexed: 12/21/2022] Open
|
7
|
Anthrax Toxin Receptor 1 Is Essential for Arteriogenesis in a Mouse Model of Hindlimb Ischemia. PLoS One 2016; 11:e0146586. [PMID: 26785120 PMCID: PMC4718698 DOI: 10.1371/journal.pone.0146586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/18/2015] [Indexed: 11/24/2022] Open
Abstract
Anthrax toxin receptor 1/tumor endothelial marker 8 (Antxr1 or TEM8) is up-regulated in tumor vasculature and serves as a receptor for anthrax toxin, but its physiologic function is unclear. The objective of this study was to evaluate the role of Antxr1 in arteriogenesis. The role of Antxr1 in arteriogenesis was tested by measuring gene expression and immunohistochemistry in a mouse model of hindlimb ischemia using wild-type and ANTXR1-/- mice. Additional tests were performed by measuring gene expression in in vitro models of fluid shear stress and hypoxia, as well as in human muscle tissues obtained from patients having peripheral artery disease. We observed that Antxr1 expression transiently increased in ischemic tissues following femoral artery ligation and that its expression was necessary for arteriogenesis. In the absence of Antxr1, the mean arterial lumen area in ischemic tissues decreased. Antxr1 mRNA and protein expression was positively regulated by fluid shear stress, but not by hypoxia. Furthermore, Antxr1 expression was elevated in human peripheral artery disease requiring lower extremity bypass surgery. These findings demonstrate an essential physiologic role for Antxr1 in arteriogenesis and peripheral artery disease, with important implications for managing ischemia and other arteriogenesis-dependent vascular diseases.
Collapse
|
8
|
Wagner SC, Ichim TE, Ma H, Szymanski J, Perez JA, Lopez J, Bogin V, Patel AN, Marincola FM, Kesari S. Cancer anti-angiogenesis vaccines: Is the tumor vasculature antigenically unique? J Transl Med 2015; 13:340. [PMID: 26510973 PMCID: PMC4625691 DOI: 10.1186/s12967-015-0688-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/03/2015] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is essential for the growth and metastasis of solid tumors. The tumor endothelium exists in a state of chronic activation and proliferation, fueled by the tumor milieu where angiogenic mediators are aberrantly over-expressed. Uncontrolled tumor growth, immune evasion, and therapeutic resistance are all driven by the dysregulated and constitutive angiogenesis occurring in the vasculature. Accordingly, great efforts have been dedicated toward identifying molecular signatures of this pathological angiogenesis in order to devise selective tumor endothelium targeting therapies while minimizing potential autoimmunity against physiologically normal endothelium. Vaccination with angiogenic antigens to generate cellular and/or humoral immunity against the tumor endothelium has proven to be a promising strategy for inhibiting or normalizing tumor angiogenesis and reducing cancer growth. Here we review tumor endothelium vaccines developed to date including active immunization strategies using specific tumor endothelium-associated antigens and whole endothelial cell-based vaccines designed to elicit immune responses against diverse target antigens. Among the novel therapeutic options, we describe a placenta-derived endothelial cell vaccine, ValloVax™, a polyvalent vaccine that is antigenically similar to proliferating tumor endothelium and is supported by pre-clinical studies to be safe and efficacious against several tumor types.
Collapse
Affiliation(s)
- Samuel C Wagner
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Thomas E Ichim
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Hong Ma
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Julia Szymanski
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | | | - Javier Lopez
- Pan Am Cancer Treatment Center, Tijuana, Mexico.
| | - Vladimir Bogin
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Amit N Patel
- Department of Surgery, University of Utah, Salt Lake City, UT, USA.
| | | | | |
Collapse
|
9
|
Kuo F, Histed S, Xu B, Bhadrasetty V, Szajek LP, Williams MR, Wong K, Wu H, Lane K, Coble V, Vasalatiy O, Griffiths G, Paik CH, Elbuluk O, Szot C, Chaudhary A, St. Croix B, Choyke P, Jagoda EM. Immuno-PET imaging of tumor endothelial marker 8 (TEM8). Mol Pharm 2014; 11:3996-4006. [PMID: 24984190 PMCID: PMC4224515 DOI: 10.1021/mp500056d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 05/30/2014] [Accepted: 07/01/2014] [Indexed: 01/26/2023]
Abstract
Tumor endothelial marker 8 (TEM8) is a cell surface receptor that is highly expressed in a variety of human tumors and promotes tumor angiogenesis and cell growth. Antibodies targeting TEM8 block tumor angiogenesis in a manner distinct from the VEGF receptor pathway. Development of a TEM8 imaging agent could aid in patient selection for specific antiangiogenic therapies and for response monitoring. In these studies, L2, a therapeutic anti-TEM8 monoclonal IgG antibody (L2mAb), was labeled with (89)Zr and evaluated in vitro and in vivo in TEM8 expressing cells and mouse xenografts (NCI-H460, DLD-1) as a potential TEM8 immuno-PET imaging agent. (89)Zr-df-L2mAb was synthesized using a desferioxamine-L2mAb conjugate (df-L2mAb); (125)I-L2mAb was labeled directly. In vitro binding studies were performed using human derived cell lines with high, moderate, and low/undetectable TEM8 expression. (89)Zr-df-L2mAb in vitro autoradiography studies and CD31 IHC staining were performed with cryosections from human tumor xenografts (NCI-H460, DLD-1, MKN-45, U87-MG, T-47D, and A-431). Confirmatory TEM8 Western blots were performed with the same tumor types and cells. (89)Zr-df-L2mAb biodistribution and PET imaging studies were performed in NCI-H460 and DLD-1 xenografts in nude mice. (125)I-L2mAb and (89)Zr-df-L2mAb exhibited specific and high affinity binding to TEM8 that was consistent with TEM8 expression levels. In NCI-H460 and DLD-1 mouse xenografts nontarget tissue uptake of (89)Zr-df-L2mAb was similar; the liver and spleen exhibited the highest uptake at all time points. (89)Zr-L2mAb was highly retained in NCI-H460 tumors with <10% losses from day 1 to day 3 with the highest tumor to muscle ratios (T:M) occurring at day 3. DLD-1 tumors exhibited similar pharmacokinetics, but tumor uptake and T:M ratios were reduced ∼2-fold in comparison to NCI-H460 at all time points. NCI-H460 and DLD-1 tumors were easily visualized in PET imaging studies despite low in vitro TEM8 expression in DLD-1 cells indicating that in vivo expression might be higher in DLD-1 tumors. From in vitro autoradiography studies (89)Zr-df-L2mAb specific binding was found in 6 tumor types (U87-MG, NCI-H460, T-47D MKN-45, A-431, and DLD-1) which highly correlated to vessel density (CD31 IHC). Westerns blots confirmed the presence of TEM8 in the 6 tumor types but found undetectable TEM8 levels in DLD-1 and MKN-45 cells. This data would indicate that TEM8 is associated with the tumor vasculature rather than the tumor tissue, thus explaining the increased TEM8 expression in DLD-1 tumors compared to DLD-1 cell cultures. (89)Zr-df-L2mAb specifically targeted TEM8 in vitro and in vivo although the in vitro expression was not necessarily predictive of in vivo expression which seemed to be associated with the tumor vasculature. In mouse models, (89)Zr-df-L2mAb tumor uptakes and T:M ratios were sufficient for visualization during PET imaging. These results would suggest that a TEM8 targeted PET imaging agent, such as (89)Zr-df-L2mAb, may have potential clinical, diagnostic, and prognostic applications by providing a quantitative measure of tumor angiogenesis and patient selection for future TEM8 directed therapies.
Collapse
Affiliation(s)
- Frank Kuo
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Stephanie Histed
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Biying Xu
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Veerendra Bhadrasetty
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Lawrence P. Szajek
- PET Department and Nuclear Medicine Division,
Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mark R. Williams
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Karen Wong
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Haitao Wu
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Kelly Lane
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Vincent Coble
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Olga Vasalatiy
- Imaging Probe Development Center, National
Heart, Lung, and Blood Institute, National
Institutes of Health, Rockville, Maryland 20892-3372, United States
| | - Gary
L. Griffiths
- Clinical Research Directorate/CMRP, Leidos
Biomedical Research Inc. (formerly SAIC-Frederick, Inc.), Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Chang H. Paik
- PET Department and Nuclear Medicine Division,
Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Osama Elbuluk
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Christopher Szot
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Amit Chaudhary
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Brad St. Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Peter Choyke
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| | - Elaine M. Jagoda
- Molecular Imaging Program, National Cancer Institute, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
10
|
Chaudhary A, Hilton MB, Seaman S, Haines DC, Stevenson S, Lemotte PK, Tschantz WR, Zhang XM, Saha S, Fleming T, Croix BS. TEM8/ANTXR1 blockade inhibits pathological angiogenesis and potentiates tumoricidal responses against multiple cancer types. Cancer Cell 2012; 21:212-26. [PMID: 22340594 PMCID: PMC3289547 DOI: 10.1016/j.ccr.2012.01.004] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 09/19/2011] [Accepted: 01/05/2012] [Indexed: 01/19/2023]
Abstract
Current antiangiogenic agents used to treat cancer only partially inhibit neovascularization and cause normal tissue toxicities, fueling the need to identify therapeutic agents that are more selective for pathological angiogenesis. Tumor endothelial marker 8 (TEM8), also known as anthrax toxin receptor 1 (ANTXR1), is a highly conserved cell-surface protein overexpressed on tumor-infiltrating vasculature. Here we show that genetic disruption of Tem8 results in impaired growth of human tumor xenografts of diverse origin including melanoma, breast, colon, and lung cancer. Furthermore, antibodies developed against the TEM8 extracellular domain blocked anthrax intoxication, inhibited tumor-induced angiogenesis, displayed broad antitumor activity, and augmented the activity of clinically approved anticancer agents without added toxicity. Thus, TEM8 targeting may allow selective inhibition of pathological angiogenesis.
Collapse
Affiliation(s)
- Amit Chaudhary
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Mary Beth Hilton
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
- Basic Research Program, SAIC, NCI, NIH, Frederick, MD 21702, USA
| | - Steven Seaman
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Diana C. Haines
- Veterinary Pathology Section, Pathology/Histotechnology Laboratory, SAIC, NCI, Frederick, MD, 21702, USA
| | - Susan Stevenson
- Novartis Institutes for BioMedical Research, Inc, Cambridge, MA, 02139, USA
| | - Peter K. Lemotte
- Novartis Institutes for BioMedical Research, Inc, Cambridge, MA, 02139, USA
| | | | - Xiaoyan M. Zhang
- Novartis Institutes for BioMedical Research, Inc, Cambridge, MA, 02139, USA
| | - Saurabh Saha
- Novartis Institutes for BioMedical Research, Inc, Cambridge, MA, 02139, USA
| | - Tony Fleming
- Novartis Institutes for BioMedical Research, Inc, Cambridge, MA, 02139, USA
| | - Brad St. Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| |
Collapse
|
11
|
Matejuk A, Leng Q, Chou ST, Mixson AJ. Vaccines targeting the neovasculature of tumors. Vasc Cell 2011; 3:7. [PMID: 21385454 PMCID: PMC3061948 DOI: 10.1186/2045-824x-3-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 03/08/2011] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis has a critical role in physiologic and disease processes. For the growth of tumors, angiogenesis must occur to carry sufficient nutrients to the tumor. In addition to growth, development of new blood vessels is necessary for invasion and metastases of the tumor. A number of strategies have been developed to inhibit tumor angiogenesis and further understanding of the interplay between tumors and angiogenesis should allow new approaches and advances in angiogenic therapy. One such promising angiogenic approach is to target and inhibit angiogenesis with vaccines. This review will discuss recent advances and future prospects in vaccines targeting aberrant angiogenesis of tumors. The strategies utilized by investigators have included whole endothelial cell vaccines as well as vaccines with defined targets on endothelial cells and pericytes of the developing tumor endothelium. To date, several promising anti-angiogenic vaccine strategies have demonstrated marked inhibition of tumor growth in pre-clinical trials with some showing no observed interference with physiologic angiogenic processes such as wound healing and fertility.
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Pathology, University of Maryland Baltimore, MSTF Building, 10 South Pine Street, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
12
|
Yang MY, Chaudhary A, Seaman S, Dunty J, Stevens J, Elzarrad MK, Frankel AE, St. Croix B. The cell surface structure of tumor endothelial marker 8 (TEM8) is regulated by the actin cytoskeleton. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1813:39-49. [PMID: 21129411 PMCID: PMC3014418 DOI: 10.1016/j.bbamcr.2010.11.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 11/17/2010] [Accepted: 11/19/2010] [Indexed: 01/01/2023]
Abstract
Tumor endothelial marker 8 (TEM8) is an integrin-like cell surface protein upregulated on tumor blood vessels and a potential vascular target for cancer therapy. Here, we found that the ability of an anti-TEM8 antibody, clone SB5, to recognize the extracellular domain of TEM8 on the cell surface depends on other host-cell factors. By taking advantage of SB5's ability to distinguish different forms of cell surface TEM8, we identified alpha-smooth muscle actin and transgelin, an actin binding protein, as intracellular factors able to alter TEM8 cell surface structure. Overexpression of either of these proteins in cells converted TEM8 from an SB5-exposed to an SB5-masked form and protected cells from SB5-saporin immunotoxins. Because the predominant form of TEM8 on the cell surface is not recognized by SB5, we also developed a new monoclonal antibody, called AF334, which is able to recognize both the SB5-exposed and the SB5-masked forms of TEM8. AF334-saporin selectively killed TEM8-positive cells independent of TEM8 cell surface structure. These studies reveal that TEM8 exists in different forms at the cell surface, a structure dependent on interactions with components of the actin cytoskeleton, and should aid in the rational design of the most effective diagnostic and therapeutic anti-TEM8 monoclonal antibodies.
Collapse
Affiliation(s)
- Mi Young Yang
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Amit Chaudhary
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Steven Seaman
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Jill Dunty
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Janine Stevens
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Mohammed K. Elzarrad
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| | | | - Brad St. Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program (MCGP), National Cancer Institute-Frederick, Frederick, Maryland, USA
| |
Collapse
|
13
|
Zuo SG, Chen Y, Wu ZP, Liu X, Liu C, Zhou YC, Wu CL, Jin CG, Gu YL, Li J, Chen XQ, Li Y, Wei HP, Li LH, Wang XC. Orally administered DNA vaccine delivery by attenuated Salmonella typhimurium targeting fetal liver kinase 1 inhibits murine Lewis lung carcinoma growth and metastasis. Biol Pharm Bull 2010; 33:174-82. [PMID: 20118536 DOI: 10.1248/bpb.33.174] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The vascular endothelial growth factor (VEGF) receptor 2 (VEGFR-2), also called fetal liver kinase 1 (FLK1) in mice and kinase insert domain receptor (KDR) in humans, is an endothelial cell specific receptor tyrosine kinase that mediates lung cancer angiogenesis. We hypothesized that an active immunotherapy approach targeting FLK1 may inhibit lung cancer growth and metastasis. To test this hypothesis, we evaluated whether immune responses to FLK1 could be elicited in mice by immunization with an orally administered DNA vaccine encoding the extracellular domain (ECD) of FLK1 (pcDNA3.1-FLK1(ECD)) carried by attenuated Salmonella typhimurium. We found that the vaccine was effective at protective antitumor immunity in Lewis lung carcinoma models in mice by breaking immune tolerance to FLK1 self-antigen. Both FLK1-specific humoral and cellular immune responses against endothelial cells can be induced in mice by immunization with pcDNA3.1-FLK1(ECD). Immunization with pcDNA3.1-FLK1(ECD) resulted in tumor suppression and prolonged survival in mice challenged with Lewis lung carcinomas cells. Experimental pulmonary metastases were strongly inhibited in pcDNA3.1-FLK1(ECD) immunized mice challenged with Lewis lung carcinoma cells. Thus, we conclude that the plasmid DNA vaccine encoding the extracellular domain of FLK1 could be an important component of FLK1 DNA vaccine to prevent lung carcinoma recurrence and metastasis after surgery.
Collapse
Affiliation(s)
- Shu Guang Zuo
- Tumor Institute of Yunnan Province, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), No. 519 Kunzhou Road, Kunming, Yunnan 650118, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Preise D, Oren R, Glinert I, Kalchenko V, Jung S, Scherz A, Salomon Y. Systemic antitumor protection by vascular-targeted photodynamic therapy involves cellular and humoral immunity. Cancer Immunol Immunother 2009; 58:71-84. [PMID: 18488222 PMCID: PMC11030999 DOI: 10.1007/s00262-008-0527-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Accepted: 04/17/2008] [Indexed: 12/13/2022]
Abstract
Vascular-targeted photodynamic therapy (VTP) takes advantage of intravascular excitation of a photosensitizer (PS) to produce cytotoxic reactive oxygen species (ROS). These ROS are potent mediators of vascular damage inducing rapid local thrombus formation, vascular occlusion, and tissue hypoxia. This light-controlled process is used for the eradication of solid tumors with Pd-bacteriochlorophyll derivatives (Bchl) as PS. Unlike classical photodynamic therapy (PDT), cancer cells are not the primary target for VTP but instead are destroyed by treatment-induced oxygen deprivation. VTP initiates acute local inflammation inside the illuminated area accompanied by massive tumor tissue death. Consequently, in the present study, we addressed the possibility of immune response induction by the treatment that may be considered as an integral part of the mechanism of VTP-mediated tumor eradication. The effect of VTP on the host immune system was investigated using WST11, which is now in phase II clinical trials for age-related macular degeneration and intended to be evaluated for cancer therapy. We found that a functional immune system is essential for successful VTP. Long-lasting systemic antitumor immunity was induced by VTP involving both cellular and humoral components. The antitumor effect was cross-protective against mismatched tumors, suggesting VTP-mediated production of overlapping tumor antigens, possibly from endothelial origin. Based on our findings we suggest that local VTP might be utilized in combination with other anticancer therapies (e.g., immunotherapy) for the enhancement of host antitumor immunity in the treatment of both local and disseminated disease.
Collapse
Affiliation(s)
- Dina Preise
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | - Roni Oren
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | - Itai Glinert
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | - Vyacheslav Kalchenko
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot, Israel
| | - Steffen Jung
- Department of Chemical Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Avigdor Scherz
- Department of Plant Science, The Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Salomon
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
15
|
Zhang HG, Mehta K, Cohen P, Guha C. Hyperthermia on immune regulation: a temperature's story. Cancer Lett 2008; 271:191-204. [PMID: 18597930 DOI: 10.1016/j.canlet.2008.05.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Revised: 02/11/2008] [Accepted: 05/06/2008] [Indexed: 01/12/2023]
Abstract
Over the last decade the linkage between hyperthermia, heat shock proteins and fever with the body's immune system has been well investigated. The immunomodulatory function of hyperthermia has been found to be quite sensitively regulated by temperature, as different levels of heating can bring different modulatory effect on different sensitive targets. Understanding these intrinsic mechanisms could bring new inspirations on the design of clinical trials combining local tumor hyperthermia with immunotherapy in cancer patients. This review will attempt to tell the story about the effect of temperature on immune regulation, with special emphasis on the clinical application of hyperthermia and the feasibility of combining it with immunotherapy in the clinic.
Collapse
Affiliation(s)
- Hua-Gang Zhang
- Department of Radiation Oncology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | | | | | | |
Collapse
|
16
|
Tumor Endothelial Markers. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Affiliation(s)
- B St Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| |
Collapse
|