251
|
Affiliation(s)
- J R Westphal
- Department of Pathology, University Hospital Nijmegen St. Radboud, Nijmegen, The Netherlands
| | | | | |
Collapse
|
252
|
Lanza GM, Abendschein DR, Hall CS, Scott MJ, Scherrer DE, Houseman A, Miller JG, Wickline SA. In vivo molecular imaging of stretch-induced tissue factor in carotid arteries with ligand-targeted nanoparticles. J Am Soc Echocardiogr 2000; 13:608-14. [PMID: 10849515 DOI: 10.1067/mje.2000.105840] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Molecular imaging permits tissues to be functionally characterized by identification of specific cell-surface receptors with targeted contrast agents. In our study, a ligand-targeted acoustic nanoparticle system was used to identify the angioplasty-induced expression of tissue factor by smooth muscle cells within the tunica media. Pig carotid arteries were overstretched bilaterally with balloon catheters, treated with a tissue factor-targeted or a control nanoparticle system, and imaged with intravascular ultrasound (20 MHz) before and after treatment. Carotid wall acoustic reflectivities were unaffected by overstretch injury. Tissue factor-targeted nanoemulsion bound and increased the echogenicity of smooth muscle cells expressing tissue factor within the tunica media. The targeted emulsion increased the arterial wall gray scale (99.4+/-14.5; P<.05) relative to pretreatment (41.8+/-11.1, P<0.05) and the control gray scale (pre-emulsion: 49.3+/-9.5; post-emulsion: 43.7+/-6.4; P<.05). The area of acoustic enhancement appeared to coincide with expression of induced tissue factor in the tunica media confirmed by immunohistochemistry. We have demonstrated that this novel nanoemulsion can infiltrate into arterial walls after balloon injury and localize the expression of overstretch-induced tissue factor within pig carotid arteries. Molecular imaging and quantification of complex, biochemical change, such as tissue factor expression after angioplasty, may prove to be a prognostically important predictor of subsequent restenosis.
Collapse
Affiliation(s)
- G M Lanza
- Department of Medicine, Division of Cardiology, Barnes-Jewish Hospital, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
253
|
Hagedorn M, Bikfalvi A. Target molecules for anti-angiogenic therapy: from basic research to clinical trials. Crit Rev Oncol Hematol 2000; 34:89-110. [PMID: 10799835 DOI: 10.1016/s1040-8428(00)00056-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
There is growing evidence that anti-angiogenic drugs will improve future therapies of diseases like cancer, rheumatoid arthritis and ocular neovascularisation. However, it is still uncertain which kind of substance, out of the large number of angiogenesis inhibitors, will prove to be a suitable agent to treat these human diseases. There are currently more than 30 angiogenesis inhibitors in clinical trials and a multitude of promising new candidates are under investigation in vitro and in animal models. Important therapeutic strategies are: suppression of activity of the major angiogenic regulators like vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF); inhibition of function of alphav-integrins and matrix metalloproteinases (MMPs); the exploitation of endogenous anti-angiogenic molecules like angiostatin, endostatin or thrombospondin. Given the wide spectrum of diseases which could be treated by anti-angiogenic compounds, it is important for today's clinicians to understand their essential mode of action at a cellular and molecular level. Here we give an in-depth overview of the basic pathophysiological mechanisms underlying the different anti-angiogenic approaches used to date based on the most recent fundamental and clinical research data. The angiogenesis inhibitors in clinical trials are presented and promising future drug candidates are discussed.
Collapse
Affiliation(s)
- M Hagedorn
- Laboratoire des Facteurs de Croissance et de la Différenciation cellulaire (Growth Factor and Cell Differenciation Laboratory), Bâtiment de Recherche Biologie Animale, Avenue des Facultés, Université de Bordeaux I, Talence, France
| | | |
Collapse
|
254
|
Wang JL, Liu YH, Lee MC, Nguyen TM, Lee C, Kim A, Nguyen M. Identification of tumor angiogenesis-related genes by subtractive hybridization. Microvasc Res 2000; 59:394-7. [PMID: 10792972 DOI: 10.1006/mvre.2000.2242] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- J L Wang
- Division of Surgical Oncology, UCLA Medical Center, Los Angeles, California 90095, USA
| | | | | | | | | | | | | |
Collapse
|
255
|
Helfrich W, Haisma HJ, Magdolen V, Luther T, Bom VJ, Westra J, van der Hoeven R, Kroesen BJ, Molema G, de Leij L. A rapid and versatile method for harnessing scFv antibody fragments with various biological effector functions. J Immunol Methods 2000; 237:131-45. [PMID: 10725458 DOI: 10.1016/s0022-1759(99)00220-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A versatile expression vector is described for the rapid construction and evaluation of bispecific scFvs and scFv-based fusion proteins. An important feature of this vector is the presence of two multiple cloning sites (MCS) separated by an in frame linker sequence. The first MCS was specifically designed to contain unique SfiI and NotI restriction enzyme sites that can be used for directional and in frame insertion of scFvs (or potentially any molecule) selected from established phage-display systems. Using this new vector, a functional bs-(scFv)(2) (2C11-MOC31) was constructed for retargeted T-cell cytotoxicity towards EGP2 positive tumor cells. The vector was also used for grafting of a number of promising biological effector principles onto scFv MOC31, including the prodrug converting enzyme cytosine deaminase, the anti-angiogenic factor angiostatin, and the thrombogenic molecule tissue factor. We aimed at producing biologically active fusion proteins by directing them through the endoplasmic reticulum-based protein folding machinery of eukaryotic cells (COS-7) using a kappa light chain leader, thereby taking advantage of the associated quality control mechanisms that allow only fully folded and processed fusion proteins to be secreted into the medium. Supernatants derived from fusion protein transfected COS-7 cells, which were transiently transfected at low transfection rates, were directly assayed for the biological and/or targeting activity of the excreted fusion proteins without any prior purification steps. This procedure might help to identify those fusion proteins that have favourable characteristics like stability and biological activity in the presence of serum and at low protein concentrations. Targeted delivery of all effector principles was subsequently assessed in an in vitro model system. The method we devised is both rapid and versatile and can be useful to construct and identify series of new chimeric proteins with enhanced therapeutic potential in human cancer therapy.
Collapse
Affiliation(s)
- W Helfrich
- Groningen University Institute for Drug Exploration (GUIDE) at the University Hospital Groningen, Department of Pathology and Laboratory Medicine, Medical Biology Branch, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
256
|
Lanza GM, Abendschein DR, Hall CS, Marsh JN, Scott MJ, Scherrer DE, Wickline SA. Molecular imaging of stretch-induced tissue factor expression in carotid arteries with intravascular ultrasound. Invest Radiol 2000; 35:227-34. [PMID: 10764091 DOI: 10.1097/00004424-200004000-00003] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
RATIONALE AND OBJECTIVES Molecular imaging with targeted contrast agents enables tissues to be distinguished by detecting specific cell-surface receptors. In the present study, a ligand-targeted acoustic nanoparticle system is used to identify angioplasty-induced expression of tissue factor by smooth muscle cells within carotid arteries. METHODS Pig carotid arteries were overstretched with balloon catheters, treated with tissue factor-targeted or a control nanoparticle system, and imaged with intravascular ultrasound before and after treatment. RESULTS Tissue factor-targeted emulsions bound and increased the echogenicity and gray-scale levels of overstretched smooth muscle cells within the tunica media, versus no change in contralateral control arteries. Expression of stretch-induced tissue factor in carotid artery media was confirmed by immunohistochemistry. CONCLUSIONS The potential for abnormal thrombogenicity of balloon-injured arteries, as reflected by smooth muscle expression of tissue factor, was imaged using a novel, targeted, nanoparticulate ultrasonic contrast agent.
Collapse
Affiliation(s)
- G M Lanza
- Department of Medicine, Washington University Medical School, Barnes-Jewish Hospital, St. Louis, Missouri 63110, USA.
| | | | | | | | | | | | | |
Collapse
|
257
|
Stoelcker B, Ruhland B, Hehlgans T, Bluethmann H, Luther T, Männel DN. Tumor necrosis factor induces tumor necrosis via tumor necrosis factor receptor type 1-expressing endothelial cells of the tumor vasculature. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 156:1171-6. [PMID: 10751341 PMCID: PMC1876893 DOI: 10.1016/s0002-9440(10)64986-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Activation of endothelial cells, fibrin deposition, and coagulation within the tumor vasculature has been shown in vivo to correlate with the occurrence of tumor necrosis factor (TNF)-induced tumor necrosis in mice. In the present study we investigated which target cells mediate the TNF-induced necrosis in fibrosarcomas grown in wild type (wt), TNF receptor type 1-deficient (TNFRp55-/-), and TNF receptor type 2-deficient (TNFRp75-/-) mice. TNF administration resulted in tumor necrosis exclusively in wt and TNFRp75-/-, but not in TNFRp55-/- mice, indicating a dependence of TNF-mediated tumor necrosis on the expression of TNF receptor type 1. However, using wt and TNFRp55-/- fibrosarcomas in wt mice, we found that TNF-mediated tumor necrosis was completely independent of TNF receptor type 1 expression in tumor cells. Thus we could exclude any direct tumoricidal effect of TNF in this model. Soluble TNF induced leukostasis in wt and TNFRp75-/- mice but not in TNFRp55-/- mice. TNF-induced leukostasis in TNFRp55-/- mice was restored by adoptive bone marrow transplantation of wt hematopoietic cells, but TNF failed to induce tumor necrosis in these chimeric mice. Because TNF administration resulted in both activation and focal damage of tumor endothelium, TNF receptor type 1-expressing cells of the tumor vasculature, likely to be endothelial cells, appear to be target cells for mediating TNF-induced tumor necrosis.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Bone Marrow Transplantation
- Chimera
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Fibrosarcoma/blood supply
- Fibrosarcoma/pathology
- Leukostasis/chemically induced
- Leukostasis/surgery
- Mice
- Mice, Inbred C57BL/genetics
- Necrosis
- Neoplasm Transplantation
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Recombinant Proteins/pharmacology
- Tumor Necrosis Factor-alpha/deficiency
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- B Stoelcker
- Institute of Pathology/Tumor Immunology, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
258
|
Christofidou-Solomidou M, Pietra GG, Solomides CC, Arguiris E, Harshaw D, Fitzgerald GA, Albelda SM, Muzykantov VR. Immunotargeting of glucose oxidase to endothelium in vivo causes oxidative vascular injury in the lungs. Am J Physiol Lung Cell Mol Physiol 2000; 278:L794-805. [PMID: 10749757 DOI: 10.1152/ajplung.2000.278.4.l794] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vascular immunotargeting is a novel approach for site-selective drug delivery to endothelium. To validate the strategy, we conjugated glucose oxidase (GOX) via streptavidin with antibodies to the endothelial cell surface antigen platelet endothelial cell adhesion molecule (PECAM). Previous work documented that 1) anti-PECAM-streptavidin carrier accumulates in the lungs after intravenous injection in animals and 2) anti-PECAM-GOX binds to, enters, and kills endothelium via intracellular H(2)O(2) generation in cell culture. In the present work, we studied the targeting and effect of anti-PECAM-GOX in animals. Anti-PECAM-GOX, but not IgG-GOX, accumulated in the isolated rat lungs, produced H(2)O(2,) and caused endothelial injury manifested by a fourfold elevation of angiotensin-converting enzyme activity in the perfusate. In intact mice, anti-PECAM-GOX accumulated in the lungs (27 +/- 9 vs. 2.4 +/- 0.3% injected dose/g for IgG-GOX) and caused severe lung injury and 95% lethality within hours after intravenous injection. Endothelial disruption and blebbing, elevated lung wet-to-dry ratio, and interstitial and alveolar edema indicated that anti-PECAM-GOX damaged pulmonary endothelium. The vascular injury in the lungs was associated with positive immunostaining for iPF(2alpha)-III isoprostane, a marker for oxidative stress. In contrast, IgG-GOX caused a minor lung injury and little (5%) lethality. Anti-PECAM conjugated with inert proteins induced no death or lung injury. None of the conjugates caused major injury to other internal organs. These results indicate that an immunotargeting strategy can deliver an active enzyme to selected target cells in intact animals. Anti-PECAM-GOX provides a novel model of oxidative injury to the pulmonary endothelium in vivo.
Collapse
Affiliation(s)
- M Christofidou-Solomidou
- Pulmonary Critical Care Division, Department of Medicine, University of Pennsylvania Medical Center, Philadelphia 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
259
|
Abstract
The concept of treating solid tumors by inhibiting tumor angiogenesis was first articulated almost 30 years ago. For the next 10 years it attracted little scientific interest. This situation changed, relatively slowly, over the succeeding decade with the discovery of the first pro-angiogenic molecules such as basic fibroblast growth factor and vascular endothelial growth factor (VEGF), and the development of methods of successfully growing vascular endothelial cells in culture as well as in vivo assays of angiogenesis. However, the 1990s have witnessed a striking change in both attitude and interest in tumor angiogenesis and anti-angiogenic drug development, to the point where a remarkably diverse group of over 24 such drugs is currently undergoing evaluation in phase I, II or III clinical trials. In this review I will discuss the many reasons for this. These features, together with other recent discoveries have created intense interest in initiating and expanding anti-angiogenic drug discovery programs in both academia and industry, and the testing of such newly developed drugs, either alone, or in various combinations with conventional cytotoxic therapeutics. However, significant problems remain in the clinical application of angiogenesis inhibitors such as the need for surrogate markers to monitor the effects of such drugs when they do not cause tumor regressions, and the design of clinical trials. Also of concern is that the expected need to use anti-angiogenic drugs chronically will lead to delayed toxic side effects in humans, which do not appear in rodents, especially in short-term studies.
Collapse
Affiliation(s)
- R S Kerbel
- Sunnybrook and Women's College Health Sciences Centre, Division of Cancer Biology Research, S-218 Research Building, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada.
| |
Collapse
|
260
|
Kamada H, Tsutsumi Y, Kihira T, Tsunoda S, Yamamoto Y, Mayumi T. In vitro remodeling of tumor vascular endothelial cells using conditioned medium from various tumor cells and their sensitivity to TNF-alpha. Biochem Biophys Res Commun 2000; 268:809-13. [PMID: 10679287 DOI: 10.1006/bbrc.2000.2202] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prevention of tumor-associated blood vessel formation (angiogenesis) is a potentially powerful strategy to treat cancer. We found that tumor vascular endothelial cells were rearranged in vitro with conditioned culture medium derived from tumor cells and compared the sensitivity to the effects of TNF-alpha between normal and tumor endothelial cells. Incubation with tumor (Meth-A, Colon26)-derived conditioned medium showed that no effect was observed on cell growth. Tumor cells (Meth-A, Colon26, and B16BL6) only showed no sensitivity to TNF-alpha. Normal and control endothelial cells in culture showed little cytotoxicity in response to TNF-alpha treatment, but marked cytotoxicity of TNF-alpha was observed in endothelial cells cultured with tumor-derived conditioned medium. Sensitivity to TNF-alpha was different depending on the type of tumor from which the conditioned medium was derived. This difference in sensitivity was assumed to be due to the in vivo sensitivity to TNF-alpha. The results of this study suggested that the sensitivity of tumors to TNF-alpha is controlled by the sensitivity of tumor vasculature.
Collapse
Affiliation(s)
- H Kamada
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
261
|
Zhu Z, Witte L. Inhibition of tumor growth and metastasis by targeting tumor-associated angiogenesis with antagonists to the receptors of vascular endothelial growth factor. Invest New Drugs 2000; 17:195-212. [PMID: 10665474 DOI: 10.1023/a:1006314501634] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiogenesis, the formation of new blood vessels, is essential for both tumor growth and metastasis. Recent advances in our understanding of the molecular mechanisms underlying the angiogenesis process and its regulation have led to the discovery of a variety of pharmaceutical agents with anti-angiogenic activity. The potential application of these angiogenesis inhibitors is currently under intense clinical and pre-clinical investigation. Compelling evidence suggests that vascular endothelial growth factor (VEGF) and its receptors play critical roles in tumor-associated angiogenesis, and that they represent good targets for therapeutic intervention. This has been demonstrated in a variety of animal tumor models in which disabling the function of VEGF and its receptors was shown to inhibit both tumor growth and metastasis. We have produced a panel of antibodies directed against the VEGF receptor 2, KDR/F1k-1. These antibodies potently block VEGF/KDR/F1k-1 interaction, and inhibit VEGF-stimulated activation of the receptor and proliferation of human endothelial cells. Further, the antibodies significantly inhibited tumor-associated angiogenesis in several animal models. Antagonists of VEGF and/or its receptors may offer higher specificity towards tumors with reduced side effects, and may be less likely to elicit drug resistance compared to conventional therapy. Anti-angiogenesis therapy represents a novel strategy for the treatment of cancer and other human disorders where pathological angiogenesis is involved.
Collapse
Affiliation(s)
- Z Zhu
- Department of Molecular and Cell Biology, ImClone Systems Incorporated, New York, NY 10014, USA.
| | | |
Collapse
|
262
|
Molema G, Kroesen BJ, Helfrich W, Meijer DK, de Leij LF. The use of bispecific antibodies in tumor cell and tumor vasculature directed immunotherapy. J Control Release 2000; 64:229-39. [PMID: 10640660 DOI: 10.1016/s0168-3659(99)00137-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To overcome dose limiting toxicities and to increase efficacy of immunotherapy of cancer, a number of strategies are under development for selectively redirecting effector cells/molecules towards tumor cells. Many of these strategies exploit the specificity of tumor associated antigen recognition by monoclonal antibodies. Using either hybridoma fusion, chemical derivatization or molecular biology technology, antibodies with dual specificity can be constructed. These so called biospecific antibodies (BsAbs) have been used to redirect the cytolytic activity of a variety of immune effector cells such as cytotoxic T lymphocytes, natural killer cells, neutrophils and monocytes/macrophages to tumor cells. Local administration of BsAbs, either alone or in combination with autologous effector cells, is highly effective in eradicating tumor cells. In contrast, systemic application of BsAb at present is only suitable for adjuvant treatment of minimal residual disease due to poor tumor cell accessibility. As an alternative, angiogenesis related determinants on tumor blood vessels can be exploited for the selective delivery of effector cells/molecules apart from being used to inhibit angiogenesis. Important advantages of this strategy is that the endothelial cell associated target epitope(s) are easy accessible. The dependence of tumor growth on the tumor's blood supply also renders tumor endothelial cells an attractive target for therapy. Although still in its infancy, attacking the tumor's blood supply for example by delivering coagulation factors or toxins, or by BsAb directed immunotherapies holds great promise for antineoplastic therapy.
Collapse
Affiliation(s)
- G Molema
- Dept. Clinical Immunology, Groningen University Institute for Drug Exploration (GUIDE), Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
263
|
Abstract
The endothelium plays a crucial dynamic role as a protective interface between blood and the underlying tissues during the haemostatic process, which maintains blood flow in the circulation and prevents life-threatening blood loss. Following vessel wall injury with initial platelet adhesion and aggregation to exposed subendothelial extracellular matrix, the initiation, amplification, and control of haemostasis depend on structurally unrelated membrane-associated receptors for blood coagulation proteases including tissue factor, G-protein-coupled protease-activatable receptors, thrombomodulin, and protein C receptor, respectively. In addition to their regulatory role in haemostasis, the respective (pro-)enzyme ligands such as Factors VIIa and Xa, thrombin or protein C mediate specific signalling pathways in vascular cells related to migration, proliferation or adhesion. The functional importance of these receptors beyond haemostasis has been manifested by various lethal and pathological phenotypes in knock-out mice. These protease receptors thereby provide important molecular links in the vascular system and serve to integrate haemostasis with endothelial cell functions which are relevant for the (patho-)physiological responses to injury or inflammatory challenges.
Collapse
Affiliation(s)
- K T Preissner
- Institut für Biochemie, Fachbereich Humanmedizin, Justus-Liebig-Universität, D-35392 Giessen, Germany.
| | | | | |
Collapse
|
264
|
Abstract
Medicine and pharmaceutics are encountering critical needs and opportunities for transvascular drug delivery that improves site targeting and tissue permeation by mimicking natural tissue addressing and transport mechanisms. This is driven by the accelerated development of genomic agents requiring targeted controlled release. Although rationally designed for in vitro activity, such agents are not highly effective in vivo, due to opsonization and degradation by plasma constituents, and failure to transport across the local vascular endothelium and tissue matrix. A growing knowledge of the addresses of the body can be applied to engineer "Bio-Logically" staged delivery systems with sequential bioaddressins complementary to the discontinuous compartments encountered--termed discontinuum pharmaceutics. Effective tissue targeting is accomplished by leukocytes, bacteria, and viruses. We are increasingly able to mimic their bioaddressins by genomic means. Approaches described in this commentary include: (a) endothelial-directed adhesion mediated by oligosaccharides and carbohydrates (e.g. dermatan sulfate as a mimic of sulfated CD44) and peptidomimetics interacting with adhesins, selectins, integrins, hyaluronans, and locally induced growth factors (e.g. vascular endothelial growth factor, VEGF) and coagulation factors (e.g. factor VIII antigen); (b) improved tissue permeation conferred by hydrophilically "cloaked" carrier systems; (c) "uncloaking" by matrix dilution or selective triggering near the target cells; and (d) target binding-internalization by terminally exposed hydrophobic moieties, cationic polymers, and receptor-binding lectins, peptides, or carbohydrates. This commentary also describes intermediate technology solutions (e.g. "hybrid drugs"), and highlights the high-resolution, dynamic magnetic resonance imaging and radiopharmaceutical imaging technologies plus the groups and organizations capable of accelerating these important initiatives.
Collapse
Affiliation(s)
- D F Ranney
- Department of Radiology, University of Texas Southwestern Medical Center at Dallas 75235, USA.
| |
Collapse
|
265
|
Abstract
Radiolabeled antibodies in nuclear medicine may be used for therapy or diagnosis. Diagnostic antibodies may be monoclonal antibodies (mAbs), directed against a specific disease, or polyclonal antibodies, which are used largely as intravascular reference markers or markers of vascular permeability in the imaging of inflammatory disease.
Collapse
Affiliation(s)
- A M Peters
- Department of Nuclear Medicine, New Addenbrookes Hospital, Cambridge, UK
| |
Collapse
|
266
|
Vajkoczy P, Ullrich A, Menger MD. Intravital fluorescence videomicroscopy to study tumor angiogenesis and microcirculation. Neoplasia 2000; 2:53-61. [PMID: 10933068 PMCID: PMC1531866 DOI: 10.1038/sj.neo.7900062] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Angiogenesis and microcirculation play a central role in growth and metastasis of human neoplasms, and, thus, represent a major target for novel treatment strategies. Mechanistic analysis of processes involved in tumor vascularization, however, requires sophisticated in vivo experimental models and techniques. Intravital microscopy allows direct assessment of tumor angiogenesis, microcirculation and overall perfusion. Its application to the study of tumor-induced neovascularization further provides information on molecular transport and delivery, intra- and extravascular cell-to-cell and cell-to-matrix interaction, as well as tumor oxygenation and metabolism. With the recent advances in the field of bioluminescence and fluorescent reporter genes, appropriate for in vivo imaging, the intravital fluorescent microscopic approach has to be considered a powerful tool to study microvascular, cellular and molecular mechanisms of tumor growth.
Collapse
Affiliation(s)
- P Vajkoczy
- Department of Neurosurgery, Klinikum Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | |
Collapse
|
267
|
Affiliation(s)
- M E Beckner
- Robert C. Byrd Health Science Center, West Virginia University, Morgantown, USA.
| |
Collapse
|
268
|
Birchler M, Neri G, Tarli L, Halin C, Viti F, Neri D. Infrared photodetection for the in vivo localisation of phage-derived antibodies directed against angiogenic markers. J Immunol Methods 1999; 231:239-48. [PMID: 10648941 DOI: 10.1016/s0022-1759(99)00160-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing ones, is a characteristic process which underlies many diseases, including cancer, rheumatoid arthritis and blinding ocular disorders. Antibodies capable of selective targeting and occlusion of neovasculature would open diagnostic and therapeutic opportunities. We have recently demonstrated that phage-derived human antibody fragments with high affinity for the extra-domain B (ED-B) of fibronectin, a marker of angiogenesis, selectively localise in new-forming blood vessels upon intravenous injection. Here, we show that infrared fluorescence methodologies nicely complement radioactive techniques for the study of the antibody-mediated targeting of angiogenesis in a variety of animal models. Methods are presented for the construction and use of infrared fluorescence imagers, as well as for the production and characterisation of recombinant antibodies labeled with infrared fluorophores.
Collapse
Affiliation(s)
- M Birchler
- Department of Applied Biosciences, Swiss Federal Institute of Technology, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
269
|
Mastrobattista E, Koning GA, Storm G. Immunoliposomes for the targeted delivery of antitumor drugs. Adv Drug Deliv Rev 1999; 40:103-127. [PMID: 10837783 DOI: 10.1016/s0169-409x(99)00043-5] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This review presents an overview of the field of immunoliposome-mediated targeting of anticancer agents. First, problems that are encountered when immunoliposomes are used for systemic anticancer drug delivery and potential solutions are discussed. Second, an update is given of the in vivo results obtained with immunoliposomes in tumor models. Finally, new developments on the utilization of immunoliposomes for the treatment of cancer are highlighted.
Collapse
Affiliation(s)
- E Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Pharmacy, Utrecht University, Sorbonnelaan 16, 3508 TB, Utrecht, The Netherlands
| | | | | |
Collapse
|
270
|
|
271
|
Verheul HM, Panigrahy D, Flynn E, Pinedo HM, D'Amato RJ. Treatment of the Kasabach-Merritt syndrome with pegylated recombinant human megakaryocyte growth and development factor in mice: elevated platelet counts, prolonged survival, and tumor growth inhibition. Pediatr Res 1999; 46:562-5. [PMID: 10541319 DOI: 10.1203/00006450-199911000-00012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Kasabach-Merritt Syndrome (KMS) is seen in children with large vascular tumors. KMS is characterized by very low platelet counts and a consumption of coagulation factors causing life-threatening complications. It has been proposed that thrombopenia in these patients is caused by intratumoral trapping of platelets. The truncated form of the cMpl-receptor ligand thrombopoietin, pegylated human megakaryocyte growth and development factor (Peg-rHuMGDF), is an agent that stimulates platelet production. We hypothesized that stimulation of the platelet production would prevent the life-threatening complications of patients with KMS owing to low platelet counts. In a mouse model of KMS, with tumors derived from a hemangioendothelioma cell line, we studied the effect of Peg-rHuMGDF. Treatment with Peg-rHuMGDF (10 microg/kg/day intraperitoneally) increased platelet counts by 7-8-fold compared with control tumor-bearing mice after 11 d of treatment (p < 0.001, n = 8). Survival was significantly increased, with 50% of treated animals alive at 1 mo versus 0% in untreated controls. Interestingly, we also observed an inhibition of tumor growth by 75% (p < 0.001, n = 8). Hematoxylin and eosin staining showed fresh fibrin clots in the treated tumors, suggesting that higher platelet counts caused intravascular thrombosis of tumor vessels. We conclude that increased platelet production in this model of KMS resulted in an antivascular tumor effect via platelet trapping. Further, we propose that thrombopoietin may be of critical value in preventing life-threatening complications from KMS.
Collapse
Affiliation(s)
- H M Verheul
- The Department of Medical Oncology, Academic Hospital Vrije Universiteit, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
272
|
Schiefer D, Gottstein C, Diehl V, Engert A. [Anti-angiogenesis: a new approach to tumor therapy?]. MEDIZINISCHE KLINIK (MUNICH, GERMANY : 1983) 1999; 94:570-9. [PMID: 10554516 DOI: 10.1007/bf03044955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
BACKGROUND The overall mortality due to metastatic cancer has not or only minimally been reduced in spite of intensive research and many innovations in the field of conventional antineoplastic therapy in the past decade. In the last years it has become a fact that tumor growth is angiogenesis-dependent. Therefore, inhibitors of angiogenesis are a new class of antineoplastic substances with a novel mechanism of action that might be a powerful complement to conventional cytostatic therapy. SUBSTANCES AND CLINICAL TRIALS: Inhibitors of tumor-angiogenesis which have entered clinical trials, with their results published until December 1998 are discussed here. Most results originate from phase-I or phase-II clinical trials. They are discussed and compared in respect to toxicity and response. Also some substances with high therapeutic potential which are still in preclinical testing are discussed. RESULTS Many of the investigated angiogenesis inhibitors demonstrated anti-tumor effects in phase-I or phase-II clinical trials. The commonest manifestation was stable disease, followed by partial remissions. In a few cases, complete remissions were observed. The toxicities of these substances differ both in type and degree of side effects. CONCLUSION Some antiangiogenic drugs appear to be promising candidates for a clinical use in the therapy of solid tumors. Further conclusions can only be drawn after evaluation of the results of ongoing phase-III clinical trials.
Collapse
Affiliation(s)
- D Schiefer
- Klinik I für Innere Medizin, Universität zu Köln
| | | | | | | |
Collapse
|
273
|
Nishi T, Goto T, Takeshima H, Hamada K, Tada K, Saito Y, Kochi M, Kuratsu JI, Ushio Y. Tissue factor expressed in pituitary adenoma cells contributes to the development of vascular events in pituitary adenomas. Cancer 1999; 86:1354-61. [PMID: 10506725 DOI: 10.1002/(sici)1097-0142(19991001)86:7<1354::aid-cncr35>3.0.co;2-r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Tissue factor (TF) was initially identified as an important factor in the initiation of coagulation. TF has recently been found to be expressed highly in certain types of malignant tumors. It has also been reported to be involved in systemic coagulopathy in cancer patients and in the proliferative and invasive activities of tumor cells. Tissue factor pathway inhibitor (TFPI) is a strong biologic inhibitor of TF. To the authors' knowledge, this is the first study of the expression of TF and TFPI in human pituitary adenoma. METHODS The expression of TF and TFPI were analyzed by immunohistochemical methods in human pituitary adenoma samples. To examine whether TF and TFPI expression influence the proliferative and/or invasive character of pituitary adenomas, the authors determined the MIB-1 labeling indices and invasiveness of all the pituitary adenomas they examined. Furthermore, to determine whether TF contributes to coagulation inside adenoma tissues, the incidence of cysts or hematomas in adenomas was analyzed. RESULTS In cells from 29 of 83 pituitary adenomas, overexpression of TF was observed. This was not the case for normal pituitary gland cells. TFPI was not expressed in either the adenomas or the normal pituitary glands from adenoma-bearing individuals. The expression of TF was significantly correlated with the formation of cysts or hematomas in pituitary adenomas. However, no such correlation with either the proliferative activity or the invasive character of the adenomas was observed. CONCLUSIONS Locally overexpressed TF in adenoma cells may contribute to the development of vascular events, such as infarction and/or hemorrhagic infarction, in pituitary adenomas.
Collapse
Affiliation(s)
- T Nishi
- Department of Neurosurgery, Kumamoto University School of Medicine, Kumamoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Birchler M, Viti F, Zardi L, Spiess B, Neri D. Selective targeting and photocoagulation of ocular angiogenesis mediated by a phage-derived human antibody fragment. Nat Biotechnol 1999; 17:984-8. [PMID: 10504699 DOI: 10.1038/13679] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Molecules that selectively target and occlude new blood vessels would be useful for diagnosis and treatment of pathologies associated with angiogenesis. We show that a phage-derived human antibody fragment (L19) with high affinity for the ED-B domain of fibronectin, a marker of angiogenesis, selectively localizes to newly formed blood vessels in a rabbit model of ocular angiogenesis. The L19 antibody, chemically coupled to a photosensitizer and irradiated with red light, mediates complete and selective occlusion of ocular neovasculature and promotes apoptosis of the corresponding endothelial cells. These results demonstrate that new ocular blood vessels can be distinguished immunochemically from preexisting ones and suggest that the targeted delivery of photosensitizers may be effective in treating angiogenesis-related pathologies.
Collapse
Affiliation(s)
- M Birchler
- Institut für Molekularbiologie und Biophysik, Eidgenössische Technische Hochschule Hönggerberg, Einsteinstrasse, CH-8093 Zürich, Switzerland
| | | | | | | | | |
Collapse
|
275
|
Abstract
Monoclonal antibodies directed to tumor-associated antigens have been chemically conjugated to drugs with different mechanisms of action and different levels of potency. Monoclonal-antibody-directed drug delivery has the potential to both improve efficacy and reduce systemic toxicity. Several immunoconjugates have demonstrated impressive antigen-specific antitumor activity in preclinical models. Phase I trials of a calicheamicin immunoconjugate for treatment of acute myeloid leukemia and a doxorubicin immunoconjugate for treatment of carcinoma have recently been completed.
Collapse
Affiliation(s)
- P A Trail
- Bristol-Myers Squibb Pharmaceutical Research Institute, P.O. Box 4000, Princeton, NJ 08543, USA.
| | | |
Collapse
|
276
|
Nihei Y, Suga Y, Morinaga Y, Ohishi K, Okano A, Ohsumi K, Hatanaka T, Nakagawa R, Tsuji T, Akiyama Y, Saito S, Hori K, Sato Y, Tsuruo T. A novel combretastatin A-4 derivative, AC-7700, shows marked antitumor activity against advanced solid tumors and orthotopically transplanted tumors. Jpn J Cancer Res 1999; 90:1016-25. [PMID: 10551333 PMCID: PMC5926154 DOI: 10.1111/j.1349-7006.1999.tb00850.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
AC-7700, a novel combretastatin A-4 derivative, suppresses the growth of solid tumors by inhibiting tumor perfusion. We evaluated the antitumor activity of AC-7700 on solid tumors in two experimental models, an advanced tumor model (murine colon 26 (c26) adenocarcinoma, colon 38 (c38) adenocarcinoma, MethA fibrosarcoma, Sarcoma 180 (S180), Lewis lung carcinoma (3LL), human LS180 adenocarcinoma) and an orthotopically transplanted tumor model (c26), compared with that of cisplatin (CDDP). The maximum tolerable dose (MTD) of CDDP suppressed early-stage c26 and c38 tumor growth when treatment was started after the tumor volume (TV) reached 0.2-0.5 cm3, but it showed reduced activity against the same tumors at an advanced growth stage when TV exceeded 2 cm3. At its MTD, AC-7700 was active against all tumors tested except 3LL in both early and advanced growth stages, reducing the tumor mass and having a curative effect in advanced c38 tumors. AC-7700 was also effective on orthotopically transplanted c26 tumors, showing a comparable activity to that on subcutaneous tumors. Unlike flavon acetic acid, which damages tumor vasculature by inducing endogenous tumor necrosis factor-alpha production, AC-7700 potently suppressed the growth of advanced c26 tumors in athymic as well as euthymic mice. These results suggest that AC-7700 is a novel antivascular agent that may have potent activity against advanced-stage cancer in the clinical setting.
Collapse
Affiliation(s)
- Y Nihei
- Pharmaceutical Research Laboratories, Ajinomoto Co., Inc., Kawasaki.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
277
|
Abstract
Angiogenesis is a critical step in the progression of tumours from dormancy to a clinical relevant cancer. Inhibition of this process is one of the most promising new anti-cancer strategies. To develop new drugs that interfere with the cascade of events required for the formation of new blood vessels, insight into this process is essential. Here, we discuss the molecular basis of angiogenesis and the concepts of vascular targeting. Furthermore new strategies will be discussed to discover surface markers on endothelial cells that confer sufficient specificity for targeted intervention in the tumour vasculature.
Collapse
Affiliation(s)
- H J Bloemendal
- Department of Internal Medicine, University Hospital Utrecht, The Netherlands
| | | | | |
Collapse
|
278
|
Gow AJ, Branco F, Christofidou-Solomidou M, Black-Schultz L, Albelda SM, Muzykantov VR. Immunotargeting of glucose oxidase: intracellular production of H(2)O(2) and endothelial oxidative stress. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:L271-81. [PMID: 10444521 DOI: 10.1152/ajplung.1999.277.2.l271] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Extracellular and intracellular reactive oxygen species attack different targets and may, therefore, result in different forms of oxidative stress. To specifically study an oxidative stress induced by a regulated intracellular flux of a defined reactive oxygen species in endothelium, we used immunotargeting of the H(2)O(2)-generating enzyme glucose oxidase (GOX) conjugated with an antibody to platelet-endothelial cell adhesion molecule (PECAM)-1, an endothelial surface antigen. Anti-PECAM-(125)I-GOX conjugates specifically bind to both endothelial and PECAM-transfected cells. Approximately 70% of cell-bound anti-PECAM-(125)I-GOX was internalized. The cell-bound conjugate was enzymatically active and generated H(2)O(2) from glucose. Use of the fluorescent dye dihydrorhodamine 123 revealed that 70% of H(2)O(2) was generated intracellularly, whereas 30% of H(2)O(2) was detected in the cell medium. Catalase added to the cells eliminated H(2)O(2) in the medium but had little effect on the intracellular generation of H(2)O(2) by anti-PECAM-GOX. Both H(2)O(2) added exogenously to the cell medium (extracellular H(2)O(2)) and that generated by anti-PECAM-GOX caused oxidative stress manifested by time- and dose-dependent irreversible plasma membrane damage. Inactivation of cellular catalase by aminotriazole treatment augmented damage caused by either extracellular H(2)O(2) or anti-PECAM-GOX. Catalase added to the medium protected either normal or aminotriazole-treated cells against extracellular H(2)O(2), yet failed to protect cells against injury induced by anti-PECAM-GOX. Therefore, treatment of PECAM-positive cells with anti-PECAM-GOX leads to conjugate internalization, predominantly intracellular H(2)O(2) generation and intracellular oxidative stress. These results indicate that anti-PECAM-GOX 1) provides cell-specific intracellular delivery of an active enzyme and 2) causes intracellular oxidative stress in PECAM-positive cells.
Collapse
Affiliation(s)
- A J Gow
- Institute for Environmental Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6068, USA
| | | | | | | | | | | |
Collapse
|
279
|
|
280
|
Dubowchik GM, Walker MA. Receptor-mediated and enzyme-dependent targeting of cytotoxic anticancer drugs. Pharmacol Ther 1999; 83:67-123. [PMID: 10511457 DOI: 10.1016/s0163-7258(99)00018-2] [Citation(s) in RCA: 198] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
This review is a survey of various approaches to targeting cytotoxic anticancer drugs to tumors primarily through biomolecules expressed by cancer cells or associated vasculature and stroma. These include monoclonal antibody immunoconjugates; enzyme prodrug therapies, such as antibody-directed enzyme prodrug therapy, gene-directed enzyme prodrug therapy, and bacterial-directed enzyme prodrug therapy; and metabolism-based therapies that seek to exploit increased tumor expression of, e.g., proteases, low-density lipoprotein receptors, hormones, and adhesion molecules. Following a discussion of factors that positively and negatively affect drug delivery to solid tumors, we concentrate on a mechanistic understanding of selective drug release or generation at the tumor site.
Collapse
Affiliation(s)
- G M Dubowchik
- Bristol-Myers Squibb Pharmaceutical Research Institute, Wallingford, CT 06492-7660, USA.
| | | |
Collapse
|
281
|
Abe K, Shoji M, Chen J, Bierhaus A, Danave I, Micko C, Casper K, Dillehay DL, Nawroth PP, Rickles FR. Regulation of vascular endothelial growth factor production and angiogenesis by the cytoplasmic tail of tissue factor. Proc Natl Acad Sci U S A 1999; 96:8663-8. [PMID: 10411932 PMCID: PMC17573 DOI: 10.1073/pnas.96.15.8663] [Citation(s) in RCA: 240] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tissue factor (TF), a transmembrane receptor for coagulation factor VII/VIIa, is aberrantly expressed in human cancers. We demonstrated a significant correlation between TF and vascular endothelial growth factor (VEGF) production in 13 human malignant melanoma cell lines (r(2) = 0.869, P < 0.0001). Two of these cell lines, RPMI-7951, a high TF and VEGF producer, and WM-115, a low TF and VEGF producer, were grown s.c. in severe combined immunodeficient mice. The high-producer cell line generated solid tumors characterized by intense vascularity, whereas the low producer generated relatively avascular tumors, as determined by immunohistologic staining of tumor vascular endothelial cells with anti-von Willebrand factor antibody. To investigate the structure-function relationship of TF and VEGF, a low-producer melanoma cell line (HT144) was transfected with a TF cDNA containing the full-length sequence, a cytoplasmic deletion mutant lacking the coding sequence for the distal three serine residues (potential substrates for protein kinase C), or an extracellular domain mutant, which has markedly diminished function for activation of factor X. Cells transfected with the full-length sequence produced increased levels of both TF and VEGF. Transfectants with the full-length sequence and the extracellular domain mutant produced approximately equal levels of VEGF mRNA. However, cells transfected with the cytoplasmic deletion mutant construct produced increased levels of TF, but little or no VEGF. Thus, the cytoplasmic tail of TF plays a role in the regulation of VEGF expression in some tumor cells.
Collapse
Affiliation(s)
- K Abe
- Emory University School of Medicine, Atlanta, GA 30333, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
282
|
Abstract
Malignant tumours are angiogenesis-dependent diseases. Several experimental studies suggest that primary tumour growth, invasiveness and metastasis require neovascularisation. Tumour-associated angiogenesis is a complex multistep process under the control of positive and negative soluble factors. A mutual stimulation occurs between tumour and endothelial cells by paracrine mechanisms. Angiogenesis is necessary, but not sufficient, as the single event for tumour growth. There is, however, compelling evidence that acquisition of the angiogenic phenotype is a common pathway for tumour progression, and that active angiogenesis is associated with other molecular mechanisms leading to tumour progression. Experimental research suggests that it is possible to block angiogenesis by specific inhibitory agents, and that modulation of angiogenic activity is associated with tumour regression in animals with different types of neoplasia. The more promising angiosuppressive agents for clinical testing are: naturally occurring inhibitors of angiogenesis (angiostatin, endostatin, platelet factor-4 and others), specific inhibitors of endothelial cell growth (TNP-470, thalidomide, interleukin-12 and others), agents neutralising angiogenic peptides (antibodies to fibroblast growth factor or vascular endothelial growth factor, suramin and analogues, tecogalan and others) or their receptors, agents that interfere with vascular basement membrane and extracellular matrix [metalloprotease (MMP) inhibitors, angiostatic steroids and others], antiadhesion molecules antibodies such as antiintegrin alpha v beta 3, and miscellaneous drugs that modulate angiogenesis by diverse mechanisms of action. Antiangiogenic therapy is to be distinguished from vascular targeting. Gene therapy aimed to block neovascularisation is also a feasible anticancer strategy in animals bearing experimental tumours. Antiangiogenic therapy represents one of the more promising new approaches to anticancer therapy and it is already in early clinical trials. Because angiosuppressive therapy is aimed at blocking tumour growth indirectly, through modulation of neovascularisation, antiangiogenic agents need to be developed and evaluated as biological response modifiers. Therefore, adequate and well designed clinical trials should be performed for a proper evaluation of antiangiogenic agents, by determination and monitoring of surrogate markers of angiogenic activity.
Collapse
Affiliation(s)
- G Gasparini
- Division of Medical Oncology, Azienda Ospedali Riuniti Bianchi-Melacrino-Morelli, Reggio Calabria, Italy.
| |
Collapse
|
283
|
Abstract
Angiogenesis is a characteristic feature of many aggressive tumors and of other relevant disorders. Molecules capable of specifically binding to new-forming blood vessels, but not to mature vessels, could be used as selective vehicles and would, therefore, open diagnostic and therapeutic opportunities. We have studied the distribution of the ED-B oncofetal domain of fibronectin, a marker of angiogenesis, in four different tumor animal models: the F9 murine teratocarcinoma, SKMEL-28 human melanoma, N592 human small cell lung carcinoma, and C51 human colon carcinoma. In all of these experimental models we observed accumulation of the fibronectin isoform containing the ED-B domain around neovascular structures when the tumors were in the exponentially growing phase, but not in the slow-growing phase. Then we performed biodistribution studies in mice bearing a subcutaneously implanted F9 murine teratocarcinoma, using a high-affinity human antibody fragment (L19) directed against the ED-B domain of fibronectin. Radiolabeled L19, but not an irrelevant anti-lysozyme antibody fragment (D1.3), efficiently localizes in the tumoral vessels. The maximal dose of L19 accumulated in the tumor was observed 3 hours after injection (8.2% injected dose per gram). By virtue of the rapid clearance of the antibody fragment from the circulation, tumor-to-blood ratios of 1.9, 3.7, and 11.8 were obtained at 3, 5, and 24 hours, respectively. The tumor-targeting performance of L19 was not dose-dependent in the 0.7 to 10 μg range of injected antibody. The integral of the radioactivity localized in tumoral vessels over 24 hours was greater than 70-fold higher than the integral of the radioactivity in blood over the same time period, normalized per gram of tissue or fluid. These findings quantitatively show that new-forming blood vessels can selectively be targeted in vivo using specific antibodies, and suggest that L19 may be of clinical utility for the immunoscintigraphic detection of angiogenesis in patients.
Collapse
|
284
|
Fattorusso R, Pellecchia M, Viti F, Neri P, Neri D, Wüthrich K. NMR structure of the human oncofoetal fibronectin ED-B domain, a specific marker for angiogenesis. Structure 1999; 7:381-90. [PMID: 10196121 DOI: 10.1016/s0969-2126(99)80051-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The process of angiogenesis (i.e. the formation of new blood vessels from pre-existing ones) is fundamental to physiological processes such as reproduction, development and repair, as well as to pathological conditions such as tumor progression, rheumathoid arthritis and ocular disorders. The oncofoetal ED-B domain, a specific marker of angiogenesis, consists of 91 amino acid residues that are inserted by alternative splicing into the fibronectin (FN) molecule. RESULTS The NMR structure of the ED-B domain is reported and reveals important differences from other FN type III domains. A comparison of the ED-B domain with the crystal structure of a four-domain FN fragment shows the novel features of ED-B to be located in loop regions that are buried at interdomain interfaces, and which therefore largely determine the global shape of the FN molecule. The negatively charged amino acids in this highly acidic protein are uniformly distributed over the molecular surface, with the sole exception of a solvent-exposed hydrophobic patch that represents a potential specific recognition site. Epitope mapping with 82 decapeptides that span the ED-B sequence revealed that three ED-B-specific monoclonal antibodies, which selectively target newly forming blood vessels in tumor-bearing mice, bind to adjacent regions on the ED-B surface. CONCLUSIONS The NMR structure enables the identification of a large surface area of the ED-B domain that appears to be accessible in vivo, opening up new diagnostic and therapeutic opportunities. Furthermore, the mapping of specific monoclonal antibodies to the three-dimensional structure of the ED-B domain, and their use in angiogenesis inhibition experiments, provides a basis for further investigation of the role of the ED-B domain in the formation of new blood vessels.
Collapse
Affiliation(s)
- R Fattorusso
- Institut für Molekularbiologie und Biophysik, Eidgenössische Technische Hochschule Hönggerberg, CH-8093 Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
285
|
Kennel SJ, Boll R, Stabin M, Schuller HM, Mirzadeh S. Radioimmunotherapy of micrometastases in lung with vascular targeted 213Bi. Br J Cancer 1999; 80:175-184. [PMID: 10389994 PMCID: PMC2362995 DOI: 10.1038/sj.bjc.6690337] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/1997] [Revised: 05/13/1998] [Accepted: 05/21/1998] [Indexed: 11/08/2022] Open
Abstract
A model system has been used to test the efficacy of vascular targeting of alpha-particle emitter 213Bi for therapy of small, 'artificial' metastases in mouse lung. Specific monoclonal antibody (mAb) 201 B was used to deliver greater than 30% of the injected dose to lung where tumours had developed due to intravenous injection of cells. Specific 213Bi-mAb 201B treatment of BALB/c mammary carcinoma EMT-6 tumours in lung resulted in a dose-dependent destruction of tumours and an extended lifespan of treated animals relative to controls. Significant reduction of lung tumour burden was noted in animals treated with 0.93 MBq injected dose or as little as 14 Gy absorbed dose to the lung. Animals treated with higher doses (2.6-6.7 MBq) had nearly complete cure of lung tumours but eventually died of lung fibrosis induced by the treatment. Four other tumour cell types were studied: murine Line 1 lung carcinomas in syngeneic BALB/c mice, rat IC-12 tracheal carcinoma growing in severe combined immune deficient (SCID) mice, and two human tumours--epidermoid carcinoma A431 and lung carcinoma A549--growing in SCID mice. In all cases, the number of lung tumour colonies was reduced in animals treated with specific, labelled mAb relative to those in animals treated with control 213Bi MAb or EDTA complexed 213Bi. Tumours treated in immunodeficient SCID mice were partially destroyed or at least retarded in growth, but ultimately regrew and proved fatal, indicating that an intact immune function is necessary for complete cure. The data show that the short-lived alpha-particle emitter 213Bi can be effectively targeted to lung blood vessels and that tumour cells growing in the lung are killed. The mechanism may involve direct killing of tumour cells from alpha-particle irradiation, killing through destruction of blood supply to the tumour, or a combination of the two.
Collapse
Affiliation(s)
- S J Kennel
- Life Sciences Division, Oak Ridge National Laboratory, TN 37831-6101, USA
| | | | | | | | | |
Collapse
|
286
|
Hayes AJ, Li LY, Lippman ME. Science, medicine, and the future. Antivascular therapy: a new approach to cancer treatment. BMJ (CLINICAL RESEARCH ED.) 1999; 318:853-6. [PMID: 10092266 PMCID: PMC1115280 DOI: 10.1136/bmj.318.7187.853] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- A J Hayes
- Lombardi Cancer Center, Georgetown University Medical Center, 3970 Reservoir Road, Washington DC, 20007, USA.
| | | | | |
Collapse
|
287
|
McMillan K, Perepelitsyn I, Wang Z, Shapshay SM. Tumor growth inhibition and regression induced by photothermal vascular targeting and angiogenesis inhibitor retinoic acid. Cancer Lett 1999; 137:35-44. [PMID: 10376792 DOI: 10.1016/s0304-3835(98)00339-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effect of photothermal vascular targeting, alone and in combination with antiangiogenic therapy, was evaluated using tumors produced in mice by transplantation of KB cells. Tumor growth inhibition and regression followed vascular damage produced by pulsed dye laser (PDL) radiation. Administration of the antiangiogenic agent all-trans-retinoic acid (RA) was associated with smaller average tumor volumes in the presence and absence of PDL irradiation, but this effect was not statistically significant. The ability of PDL photothermal vascular targeting to cause regression of tumors without harming normal tissue may be a consequence of preferential damage to supplying vessels at the tumor periphery.
Collapse
Affiliation(s)
- K McMillan
- Otolaryngology Research Center, Department of Otolaryngology, Head & Neck Surgery, New England Medical Center, Boston, MA 02111, USA.
| | | | | | | |
Collapse
|
288
|
Zhu Z, Lu D, Kotanides H, Santiago A, Jimenez X, Simcox T, Hicklin DJ, Bohlen P, Witte L. Inhibition of vascular endothelial growth factor induced mitogenesis of human endothelial cells by a chimeric anti-kinase insert domain-containing receptor antibody. Cancer Lett 1999; 136:203-13. [PMID: 10355750 DOI: 10.1016/s0304-3835(98)00324-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The kinase insert domain-containing receptor (KDR) is the human vascular endothelial growth factor (VEGF) receptor responsible for the mitogenic and angiogenic effects of VEGF. There is much experimental evidence to suggest that the VEGF/KDR pathway plays an important role in tumor angiogenesis, a process essential for tumor growth and metastasis. Here we produced a chimeric anti-KDR antibody (IgG1), c-p1C11, from a single chain (scFv) antibody isolated from a phage display library. C-p1C11 binds specifically to the extracellular domain of soluble as well as cell-surface expressed KDR. It effectively blocks VEGF-KDR interaction and inhibits VEGF-stimulated activation of KDR and MAP kinases p44/p42 of human endothelial cells. Furthermore, c-p1C11 efficiently neutralizes VEGF-induced mitogenesis of human endothelial cells. Our results suggest that antibodies against KDR have potential clinical applications in the treatment of cancer and other diseases where pathological angiogenesis is involved.
Collapse
Affiliation(s)
- Z Zhu
- Department of Molecular and Cell Biology, ImClone Systems Inc., New York, NY 10014, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
289
|
Jones A, Fujiyama C. Angiogenesis in urological malignancy: prognostic indicator and therapeutic target. BJU Int 1999; 83:535-55; quiz 555-6. [PMID: 10210606 DOI: 10.1046/j.1464-410x.1999.00018.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- A Jones
- Department of Urology, Churchill Hospital, Oxford, UK
| | | |
Collapse
|
290
|
Abstract
Abstract
Interleukin-12 (IL-12) inhibits angiogenesis in vivo by inducing interferon-γ (IFN-γ) and other downstream mediators. Here, we report that neutralization of natural killer (NK) cell function with antibodies to either asialo GM1 or NK 1.1 reversed IL-12 inhibition of basic fibroblast growth factor (bFGF)-induced angiogenesis in athymic mice. By immunohistochemistry, those sites where bFGF-induced neovascularization was inhibited by IL-12 displayed accumulation of NK cells and the presence of IP-10–positive cells. Based on expression of the cytolytic mediators perforin and granzyme B, the NK cells were locally activated. Experimental Burkitt lymphomas treated locally with IL-12 displayed tumor tissue necrosis, vascular damage, and NK-cell infiltration surrounding small vessels. After activation in vitro with IL-12, NK cells from nude mice became strongly cytotoxic for primary cultures of syngeneic aortic endothelial cells. Cytotoxicity was neutralized by antibodies to IFN-γ. These results document that NK cells are required mediators of angiogenesis inhibition by IL-12, and provide evidence that NK-cell cytotoxicity of endothelial cells is a potential mechanism by which IL-12 can suppress neovascularization.
Collapse
|
291
|
Abstract
Interleukin-12 (IL-12) inhibits angiogenesis in vivo by inducing interferon-γ (IFN-γ) and other downstream mediators. Here, we report that neutralization of natural killer (NK) cell function with antibodies to either asialo GM1 or NK 1.1 reversed IL-12 inhibition of basic fibroblast growth factor (bFGF)-induced angiogenesis in athymic mice. By immunohistochemistry, those sites where bFGF-induced neovascularization was inhibited by IL-12 displayed accumulation of NK cells and the presence of IP-10–positive cells. Based on expression of the cytolytic mediators perforin and granzyme B, the NK cells were locally activated. Experimental Burkitt lymphomas treated locally with IL-12 displayed tumor tissue necrosis, vascular damage, and NK-cell infiltration surrounding small vessels. After activation in vitro with IL-12, NK cells from nude mice became strongly cytotoxic for primary cultures of syngeneic aortic endothelial cells. Cytotoxicity was neutralized by antibodies to IFN-γ. These results document that NK cells are required mediators of angiogenesis inhibition by IL-12, and provide evidence that NK-cell cytotoxicity of endothelial cells is a potential mechanism by which IL-12 can suppress neovascularization.
Collapse
|
292
|
Harris AL. Anti-angiogenesis therapy and strategies for integrating it with adjuvant therapy. Recent Results Cancer Res 1999; 152:341-52. [PMID: 9928571 DOI: 10.1007/978-3-642-45769-2_33] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor angiogenesis is critical for the growth of primary cancers above 1-2 mm in diameter. A major vascular growth factor is VEGF, and approaches to inhibit VEGF have shown encouraging results in pre-clinical studies. The mechanisms involved in switching on angiogenesis involve activation of oncogenes and upregulation of the hypoxia-sensing pathway. These provide novel targets for therapy. Many anti-angiogenic drugs are in clinical trial currently and there are problems in assessing these types of drugs if they only cause disease stabilisation. It will be important to develop methods to assess inhibition of vascular growth in vivo. New generations of anti-angiogenesis drugs such as endostatin of angiostatin, which are more potent, may cause tumor regression, but this has not yet been studied in patients. These approaches for advanced disease should be more successful when applied early in an adjuvant situation. This will also require careful monitoring of long-term toxicity.
Collapse
Affiliation(s)
- A L Harris
- ICRF Molecular Oncology Laboratory, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
293
|
Fenton BM, Paoni SF, Lee J, Koch CJ, Lord EM. Quantification of tumour vasculature and hypoxia by immunohistochemical staining and HbO2 saturation measurements. Br J Cancer 1999; 79:464-71. [PMID: 10027314 PMCID: PMC2362405 DOI: 10.1038/sj.bjc.6690072] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Despite the possibility that tumour hypoxia may limit radiotherapeutic response, the underlying mechanisms remain poorly understood. A new methodology has been developed in which information from several sophisticated techniques is combined and analysed at a microregional level. First, tumour oxygen availability is spatially defined by measuring intravascular blood oxygen saturations (HbO2) cryospectrophotometrically in frozen tumour blocks. Second, hypoxic development is quantified in adjacent sections using immunohistochemical detection of a fluorescently conjugated monoclonal antibody (ELK3-51) to a nitroheterocyclic hypoxia marker (EF5), thereby providing information relating to both the oxygen consumption rates and the effective oxygen diffusion distances. Third, a combination of fluorescent (Hoechst 33342 or DiOC7(3)) and immunohistological (PECAM-1/CD31) stains is used to define the anatomical vascular densities and the fraction of blood vessels containing flow. Using a computer-interfaced microscope stage, image analysis software and a 3-CCD colour video camera, multiple images are digitized, combined to form a photo-montage and revisited after each of the three staining protocols. By applying image registration techniques, the spatial distribution of HbO2 saturations is matched to corresponding hypoxic marker intensities in adjacent sections. This permits vascular configuration to be related to oxygen availability and allows the hypoxic marker intensities to be quantitated in situ.
Collapse
Affiliation(s)
- B M Fenton
- Department of Radiation Oncology, University of Rochester School of Medicine, NY, USA
| | | | | | | | | |
Collapse
|
294
|
Abstract
Inactivation of the tumour suppressors p53 and p16INK4a or activating mutations in the ras oncogene are the most common genetic alterations found in human cancers. In this review, novel approaches designed to evaluate the effect of targeting intracellular molecules are described and it is shown how information derived from small synthetic peptides can stimulate novel approaches for cancer drugs. This review also gives an example of how molecular, biochemical, and cell biology studies of cancer-associated gene products can, via organic chemistry, be translated into active drugs ready for testing in clinical trials. New cancer treatments are directly springing out of studies related to tumour physiology, where the prime target is not the tumour cells but the tumour blood vessels; some of the different approaches that are being tested will be highlighted here. Finally, some of the difficulties and promises using cancer-associated genes in gene therapy are discussed.
Collapse
Affiliation(s)
- R Fåhraeus
- CRC Laboratories, Department of Biochemistry, University of Dundee, Scotland, U.K
| | | | | | | |
Collapse
|
295
|
Kubitza M, Hickey L, Roberts WG. Influence of host microvascular environment on tumour vascular endothelium. Int J Exp Pathol 1999; 80:1-10. [PMID: 10365081 PMCID: PMC2517751 DOI: 10.1046/j.1365-2613.1999.00100.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This review will focus on the tumour microvascular endothelium; how it is derived, modulated by angiogenic factors, and how the structure and function is influenced by the host tissue microenvironment.
Collapse
Affiliation(s)
- M Kubitza
- University of California, San Diego, La Jolla 92093, USA
| | | | | |
Collapse
|
296
|
van Hinsbergh VW, Collen A, Koolwijk P. Angiogenesis and anti-angiogenesis: Perspectives for the treatment of solid tumors. Ann Oncol 1999. [DOI: 10.1093/annonc/10.suppl_4.s60] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
297
|
Abstract
Neuroblastoma is a malignant solid tumor of childhood with a poor prognosis. The growth of solid tumors has been shown to be dependent on new blood vessel formation, i.e. angiogenesis. Several steps in the metastatic process have also been found to be angiogenesis-dependent. Neuroblastomas grow quickly, are highly vascularized, and metastasize early, and hence inhibition of angiogenesis--angiostatic therapy--may be indicated in this disease. In order to investigate the effects of angiostatic agents in this disease, a new animal experimental model for human neuroblastoma was developed. Three angiostatic agents were tested in the model: TNP-470, the synthetic analogue of fumagillin, given subcutaneously, and the endogenous steroid 2-methoxyestradiol and its derivative 2-propynylestradiol, given orally. TNP-470 administration resulted in a significant reduction of the tumor growth rate and microvascular counts, and of the fraction of viable tumor cells, compared to controls. The fraction of apoptotic tumor cells increased threefold, while that of proliferative cells remained unaltered. This can explain the reduced net growth. Treatment with the angiostatic and chemotherapeutic steroids 2-methoxyestradiol and 2-propynylestradiol yielded similar results. However, the mechanism of action of these steroids was bimodal; the effect occurring both through inhibition of tumor angiogenesis and through induction of tumor cell apoptosis. It was shown for the first time that inhibition of angiogenesis regardless of agent induces striking chromaffin differentiation, observed as increased expression of insulin-like growth factor II gene, tyrosine hydroxylase, and chromogranin A, and increased formation of cellular processes. It is suggested that inhibition of angiogenesis induces metabolic stress, resulting in chromaffin differentiation and apoptosis. Such agonal differentiation may be the link between angiostatic therapy and tumor cell apoptosis. Angiostatic agents administered as single therapy have an objective tumoristatic effect in our neuroblastoma model. Angiostatic treatment of neuroblastoma is a new and promising treatment modality that merits clinical investigation.
Collapse
Affiliation(s)
- E Wassberg
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Sweden.
| |
Collapse
|
298
|
Wynendaele W, van Oosterom AT, Pawinski A, de Bruijn EA, Maes RA. Angiogenesis: possibilities for therapeutic interventions. PHARMACY WORLD & SCIENCE : PWS 1998; 20:225-35. [PMID: 9972522 DOI: 10.1023/a:1008600603059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Vascular proliferation normally occurs only during embryonic development, the female reproductive cycle and wound healing. Various pathological conditions such as diabetic retinopathy are characterized by persistent, uncontrolled angiogenesis. At the other hand, impaired development of new blood vessels has been found to be related with myocardial infarction. A series of anti-angiogenic drugs are currently included in experimental cancer treatment, whereas the failure of ulcers to heal may be limited by increased angiogenesis upon administration of growth factors. In the present review control mechanisms of the vasculature are summarized and therapeutic approaches discussed.
Collapse
Affiliation(s)
- W Wynendaele
- Laboratory of Experimental Oncology (LEO), Leuven, Belgium
| | | | | | | | | |
Collapse
|
299
|
Griffioen AW, Barendsz-Janson AF, Mayo KH, Hillen HF. Angiogenesis, a target for tumor therapy. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1998; 132:363-8. [PMID: 9823929 DOI: 10.1016/s0022-2143(98)90106-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- A W Griffioen
- Department of Internal Medicine, Tumor Angiogenesis Lab, University Hospital Maastricht, The Netherlands
| | | | | | | |
Collapse
|
300
|
Abstract
Engineering of recombinant coagulation factor X variants, which can be activated by tumor-associated proteinases may lead to the development of new therapeutic molecules. However, the evaluation of such variants requires an appropriate animal model. Therefore, we isolated the complete coding sequence of mouse coagulation factor X from mouse liver cDNA by polymerase chain reaction. The deduced amino acid sequence codes for a prepro protein of 481 amino acids homologous to factor X sequences from various species. Recombinant mouse factor X was expressed in human embryonic kidney cells and secreted into cell culture supernatant as zymogen, which could be converted to catalytically active factor Xa by Russell's viper venom. Purified recombinant mouse factor X restored coagulation in human factor X deficient plasma, demonstrating that mouse factor X is able to functionally interact with the human blood coagulation system. Recombinant mouse factor X opens the possibility to analyze therapeutically useful variants in the mouse system.
Collapse
Affiliation(s)
- H H Heidtmann
- Zentrum für Innere Medizin, Abt. Hämatologie und Onkologie, Philipps-Universität Marburg, Germany
| | | |
Collapse
|