151
|
Trachsel E, Neri D. Antibodies for angiogenesis inhibition, vascular targeting and endothelial cell transcytosis. Adv Drug Deliv Rev 2006; 58:735-54. [PMID: 16822576 DOI: 10.1016/j.addr.2005.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2005] [Accepted: 05/06/2006] [Indexed: 12/20/2022]
Abstract
The endothelium is increasingly recognized as a target for biomedical intervention, not only for its accessibility to molecular agents coming from the blood-stream, but also for the active role played by endothelial cell proliferation to support diseases such as cancer, blinding ocular disorders and chronic inflammatory conditions. The notion that solid tumors cannot grow beyond a size of few millimeters without inducing the proliferation of new blood vessels has stimulated the search for mediators of angiogenesis and for inhibitors of this process, culminating in the approval of a humanized monoclonal antibody to VEGF-A for oncology applications. In parallel, researchers have begun to consider imaging and therapeutic strategies based on the selective delivery of bioactive agents to the new blood vessels, mediated by monoclonal antibody derivatives. Recently, the field of vascular targeting research has been extended to the investigation of molecular agents that may mediate endothelial cell transcytosis, in the hope to overcome this body barrier for drug delivery. This article reviews some of the most significant advances in these areas, and outlines future challenges and opportunities.
Collapse
Affiliation(s)
- Eveline Trachsel
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str.10, ETH Hönggerberg, HCI G396, CH-8093 Zürich, Switzerland
| | | |
Collapse
|
152
|
Brack SS, Silacci M, Birchler M, Neri D. Tumor-targeting properties of novel antibodies specific to the large isoform of tenascin-C. Clin Cancer Res 2006; 12:3200-8. [PMID: 16707621 DOI: 10.1158/1078-0432.ccr-05-2804] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The targeted delivery of bioactive molecules with antibodies specific to tumor-associated antigens represents a promising strategy for improving the efficacy of tumor therapy. The large isoform of tenascin-C, an abundant glycoprotein of the tumor extracellular matrix, is strongly overexpressed in adult tissue undergoing tissue remodeling, including wound healing and neoplasia, and has been implicated in a variety of different cancers while being virtually undetectable in most normal adult tissues. EXPERIMENTAL DESIGN We have used antibody phage technology to generate good-quality human recombinant antibodies (F16 and P12) specific to the alternatively spliced domains A1 and D of the large isoform of tenascin-C. The tumor-targeting properties of F16 and P12 were assessed by biodistribution studies in tumor xenografts using the antibodies in small immunoprotein (SIP) format. RESULTS SIP(F16) selectively accumulated at the tumor site with 4.5%ID/g at 24 hours in the U87 glioblastoma model but was rapidly cleared from other organs (tumor-to-organ ratios, approximately 10:1). The accumulation of SIP(P12) in the tumor was lower compared with SIP(F16) and persistent levels of radioactivity were observed in the intestine. CONCLUSIONS These data suggest that the F16 antibody, specific to domain A1 of tenascin-C, is a promising building block for the development of antibody-based pharmaceuticals in view of its excellent tumor-targeting performance and the strong expression of the antigen in a variety of primary and metastatic tumors.
Collapse
Affiliation(s)
- Simon S Brack
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zürich, Switzerland
| | | | | | | |
Collapse
|
153
|
Shinozaki K, Suominen E, Carrick F, Sauter B, Kähäri VM, Lieber A, Woo SLC, Savontaus M. Efficient infection of tumor endothelial cells by a capsid-modified adenovirus. Gene Ther 2006; 13:52-9. [PMID: 16107861 DOI: 10.1038/sj.gt.3302598] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Targeted antiangiogenic gene therapy is an attractive approach to treat metastatic cancer. However, the relative paucity of the receptors of the commonly used adenovirus serotype 5 in endothelial cells as compared with liver cells undermines the use of this vector for targeting the endothelial cells in tumors. To overcome this problem, we analyzed the ability of a hybrid Ad5/35 virus, where the serotype 5 fiber has been replaced with the fiber from serotype 35, to target tumor vasculature. Infection of human umbilical vein endothelial cells (HUVECs) with Ad5/35 at MOI 120 infected 100% of cells. In contrast, infection with Ad5 at the same MOI infected only 10% HUVECs. Ad5/35 was even more effective in transducing human aortic endothelial cells (HAECs), as infection with Ad5/35 at MOI 3.6 was sufficient to transduce 95% of cells. Gene expression analyses demonstrated that infection of HUVECs and HAECs with Ad5/35 resulted in between 1 and 3 orders of magnitude higher gene expression than infection with Ad5. Furthermore, various liver-derived cells were less infectable with Ad5/35 than Ad5, indicating a favorable toxicity profile for this virus. In a rat colon carcinoma tumor model, Ad5 was located mainly in the liver parenchyma after hepatic artery administration. In contrast, Ad5/35 was found only in the angiogenesis-rich border region of the tumor. Double immunostaining revealed that Ad5/35 colocalized with CD31 and Flk-1 positive endothelial cells. These results indicate that Ad5/35 may be useful in anticancer strategies targeting tumor endothelial cells.
Collapse
Affiliation(s)
- K Shinozaki
- Carl C. Icahn Center for Gene Therapy and Molecular Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
154
|
Balza E, Mortara L, Sassi F, Monteghirfo S, Carnemolla B, Castellani P, Neri D, Accolla RS, Zardi L, Borsi L. Targeted Delivery of Tumor Necrosis Factor-α to Tumor Vessels Induces a Therapeutic T Cell–Mediated Immune Response that Protects the Host Against Syngeneic Tumors of Different Histologic Origin. Clin Cancer Res 2006; 12:2575-82. [PMID: 16638868 DOI: 10.1158/1078-0432.ccr-05-2448] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We sought to demonstrate that a single systemic administration of L19mTNFalpha (a fusion protein constituted by the scFv L19 specific for the oncofetal ED-B domain of fibronectin and tumor necrosis factor alpha, TNFalpha) in combination with melphalan induced complete and long-lasting tumor eradication in tumor-bearing mice and triggered the generation of a specific T cell-based immune response that protects the animals from a second tumor challenge, as well as from challenges with syngeneic tumor cells of different histologic origin. EXPERIMENTAL DESIGN AND RESULTS Treatment with L19mTNFalpha, in combination with melphalan, induced complete tumor regression in 83% of BALB/c mice with WEHI-164 fibrosarcoma and 33% of animals with C51 colon carcinoma. All cured mice rejected challenges with the same tumor cells and, in a very high percentage of animals, also rejected challenges with syngeneic tumor cells of different histologic origin. In adoptive immunity transfer experiments, the splenocytes from tumor-cured mice protected naive mice both from C51 colon carcinoma and from WEHI-164 fibrosarcoma. Similar results were also obtained in adoptive immunity transfer experiments using severely immunodepressed mice. Experiments using depleted splenocytes showed that T cells play a major role in tumor rejection. CONCLUSIONS The results show that the selective targeting of mTNFalpha to the tumor enhances its immunostimulatory properties to the point of generating a therapeutic immune response against different histologically unrelated syngeneic tumors. These findings predicate treatment approaches for cancer patients based on the targeted delivery of TNFalpha to the tumor vasculature.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Line, Tumor
- Cell Survival/drug effects
- Cytotoxicity, Immunologic/immunology
- Dose-Response Relationship, Drug
- Fibronectins/genetics
- Fibronectins/immunology
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunoglobulin Fragments/genetics
- Immunotherapy, Adoptive/methods
- Melphalan/administration & dosage
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/prevention & control
- Neoplasms, Experimental/therapy
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/therapeutic use
- Spleen/cytology
- Spleen/immunology
- Spleen/transplantation
- Survival Analysis
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- Time Factors
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/pharmacology
- Tumor Necrosis Factor-alpha/therapeutic use
Collapse
Affiliation(s)
- Enrica Balza
- Department of Translational Oncology, Istituto Nazionale per la Ricerca sul Cancro, Unit of Innovative Therapies, Istituto Giannina Gaslini, Centro Biotecnologie Avanzate, Genoa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Fabbrini M, Trachsel E, Soldani P, Bindi S, Alessi P, Bracci L, Kosmehl H, Zardi L, Neri D, Neri P. Selective occlusion of tumor blood vessels by targeted delivery of an antibody-photosensitizer conjugate. Int J Cancer 2006; 118:1805-13. [PMID: 16217760 DOI: 10.1002/ijc.21412] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The irregular vasculature and high interstitial pressure of solid tumors hinder the delivery of cytotoxic agents to cancer cells. As a consequence, the doses of chemotherapy necessary to achieve complete tumor eradication are associated with unacceptably high toxicities. The selective thrombosis of tumor blood vessels has been postulated as an alternative avenue for combating cancer, depriving tumors of nutrients and oxygen and causing an avalanche of tumor cell deaths. The human antibody L19, specific to the EDB domain of fibronectin, a marker of angiogenesis, is capable of selective in vivo localization around tumor blood vessels and is thus a suitable agent for delivering toxic payloads to the tumor neovasculature. Here we show that a chemical conjugate of the L19 antibody with the photosensitizer bis(triethanolamine)Sn(IV) chlorin e(6), after intravenous injection and irradiation with red light, caused an arrest of tumor growth in mice with subcutaneous tumors. By contrast, a photosensitizer conjugate obtained with an antibody of identical pharmacokinetic properties but irrelevant specificity did not exhibit a significant therapeutic effect. These results confirm that vascular targeting strategies, aimed at the selective occlusion/disruption of tumor blood vessels, have a significant anticancer therapeutic potential and encourage the use of antibody-photosensitizer conjugates for the therapy of superficial tumors and possibly other angiogenesis-related pathologies.
Collapse
Affiliation(s)
- Monica Fabbrini
- Department of Molecular Biology, University of Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Berndorff D, Borkowski S, Sieger S, Rother A, Friebe M, Viti F, Hilger CS, Cyr JE, Dinkelborg LM. Radioimmunotherapy of solid tumors by targeting extra domain B fibronectin: identification of the best-suited radioimmunoconjugate. Clin Cancer Res 2006; 11:7053s-7063s. [PMID: 16203802 DOI: 10.1158/1078-0432.ccr-1004-0015] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The expression of extra domain B (ED-B) fibronectin is always associated with angiogenic processes and can be exclusively observed in tissues undergoing growth and/or extensive remodeling. Due to this selective expression, ED-B fibronectin is an interesting target for radioimmunotherapy of malignant diseases. The aim of this study was to identify the most appropriate ED-B-targeting radioimmunoconjugate for the therapy of solid tumors. EXPERIMENTAL DESIGN Three ED-B fibronectin-binding human antibody formats of L19 were investigated: dimeric single-chain Fv (approximately 50 kDa), "small immunoprotein" (SIP, approximately 80 kDa), and immunoglobulin G1 (IgG1, approximately 150 kDa). These L19 derivatives were either labeled with I-125 or with In-111 (using MX-diethylenetriaminepentaacetic acid, MX-DTPA). Pharmacokinetics and tumor accumulation of the radiolabeled immunoconjugates were investigated in F9 (murine teratocarcinoma) tumor-bearing mice. Subsequently, dosimetry for the corresponding therapeutic isotopes I-13-1 and Y-90 was done. After testing the myelotoxicity of I-131-L19-SIP and I-131-L19-IgG1 in non-tumor-bearing mice, the therapeutic efficacy of these iodinated antibody formats was finally investigated in F9 tumor-bearing mice. RESULTS The most favorable therapeutic index was found for I-131-L19-SIP followed by I-131-L19-IgG1. The therapeutic index of all In-111-labeled derivatives was significantly inferior. Considering the bone marrow as the dose-limiting organ, it was calculated that activities of 74 MBq I-131-L19-SIP and 25 MBq I-131-L19-IgG1 could be injected per mouse without causing severe myelotoxicity. The best therapeutic efficacy was observed using I-131-L19-SIP, resulting in significant tumor growth delay and prolonged survival after a single injection. CONCLUSION Compared with other L19-based radioimmunoconjugates, I-131-L19-SIP is characterized by superior antitumor efficacy and toxicity profile in the F9 teratocarcinoma animal model. These results indicate that ED-B fibronectin-targeted radioimmunotherapy using I-131-L19-SIP has potential to be applied to treatment of solid cancers.
Collapse
|
157
|
Ou-Yang F, Lan KL, Chen CT, Liu JC, Weng CL, Chou CK, Xie X, Hung JY, Wei Y, Hortobagyi GN, Hung MC. Endostatin-cytosine deaminase fusion protein suppresses tumor growth by targeting neovascular endothelial cells. Cancer Res 2006; 66:378-84. [PMID: 16397252 DOI: 10.1158/0008-5472.can-05-1578] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endostatin, an angiogenesis inhibitor tested in multiple clinical trials, selectively targets neovascular endothelial cells, suppressing tumor growth. To enhance the therapeutic efficacy of endostatin, we fused endostatin with cytosine deaminase, which converts a prodrug 5-flucytosine into a cytotoxic 5-fluorouracil. This therapeutic strategy was developed based on the observation that the endostatin-green fluorescence protein gene and endostatin-luciferase gene selectively target to endothelial cells in vitro and to the tumor site in vivo, respectively. When we used the endostatin-cytosine deaminase fusion protein to treat s.c. grafted tumors or experimental metastasis tumors, our results showed that endostatin-cytosine deaminase treatment provided stronger tumor growth suppression and increased mean survival time of the mice compared with the treatments of endostatin alone, cytosine deaminase alone, or endostatin plus cytosine deaminase. The endostatin-cytosine deaminase protein significantly inhibited the growth of endothelial cells and preferentially induced tumor cell apoptosis. This endostatin-cytosine deaminase fusion approach opens an avenue for cancer-targeting therapy.
Collapse
Affiliation(s)
- Fu Ou-Yang
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Pilch J, Brown DM, Komatsu M, Järvinen TAH, Yang M, Peters D, Hoffman RM, Ruoslahti E. Peptides selected for binding to clotted plasma accumulate in tumor stroma and wounds. Proc Natl Acad Sci U S A 2006; 103:2800-4. [PMID: 16476999 PMCID: PMC1413849 DOI: 10.1073/pnas.0511219103] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Screening of a phage library for peptides that bind to clotted plasma in the presence of liquid plasma yielded two cyclic decapeptides, CGLIIQKNEC (CLT1) and CNAGESSKNC (CLT2). When injected intravenously into mice bearing various types of tumors, fluorescein-conjugated CLT peptides accumulated in a fibrillar meshwork in the extracellular compartment of the tumors, but were not detectable in other tissues of the tumor-bearing mice. The tumor homing of both peptides was strongly reduced after coinjection with unlabeled CLT2, indicating that the two peptides recognize the same binding site. The CLT peptide fluorescence colocalized with staining for fibrin(ogen) present in the extravascular compartment of tumors, but not in other tissues. The CLT peptides did not home to tumors grown in fibrinogen-null mice or in mice that lack plasma fibronectin. The CLT peptides also accumulated at the sites of injury in arteries, skeletal muscle, and skin. We conclude that the CLT peptides recognize fibrin-fibronectin complexes formed by clotting of plasma proteins that have leaked into the extravascular space in tumors and other lesions. These peptides may be useful in targeting diagnostic and therapeutic materials into tumors and injured tissues.
Collapse
Affiliation(s)
- Jan Pilch
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Darren M. Brown
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Masanobu Komatsu
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Tero A. H. Järvinen
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Meng Yang
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111; and
| | - David Peters
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Robert M. Hoffman
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111; and
- Department of Surgery, University of California, 200 West Arbor Drive, San Diego, CA 92103-8220
| | - Erkki Ruoslahti
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| |
Collapse
|
159
|
Abstract
With 18 monoclonal antibody (mAb) products currently on the market and more than 100 in clinical trials, it is clear that engineered antibodies have come of age as biopharmaceuticals. In fact, by 2008, engineered antibodies are predicted to account for >30% of all revenues in the biotechnology market. Smaller recombinant antibody fragments (for example, classic monovalent antibody fragments (Fab, scFv)) and engineered variants (diabodies, triabodies, minibodies and single-domain antibodies) are now emerging as credible alternatives. These fragments retain the targeting specificity of whole mAbs but can be produced more economically and possess other unique and superior properties for a range of diagnostic and therapeutic applications. Antibody fragments have been forged into multivalent and multi-specific reagents, linked to therapeutic payloads (such as radionuclides, toxins, enzymes, liposomes and viruses) and engineered for enhanced therapeutic efficacy. Recently, single antibody domains have been engineered and selected as targeting reagents against hitherto immunosilent cavities in enzymes, receptors and infectious agents. Single-domain antibodies are anticipated to significantly expand the repertoire of antibody-based reagents against the vast range of novel biomarkers being discovered through proteomics. As this review aims to show, there is tremendous potential for all antibody fragments either as robust diagnostic reagents (for example in biosensors), or as nonimmunogenic in vivo biopharmaceuticals with superior biodistribution and blood clearance properties.
Collapse
Affiliation(s)
- Philipp Holliger
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK
| | | |
Collapse
|
160
|
Abstract
Monoclonal antibodies are beginning to fulfil their potential as therapeutics in different pathological conditions, with a special focus on cancer. At the same time, antiangiogenic therapy has evolved into one of the most active fields in cancer research. The logical combination of both strategies has produced a growing number of antibodies aimed to interfere with tumour angiogenesis at different steps of the angiogenic process. This development has taken advantage of recombinant technologies that have revolutionised the selection and production of monoclonal antibodies, and gene therapy approaches that achieve sustained and effective concentrations of therapeutic antibodies in vivo. Here, the current knowledge about these antibody-based antiangiogenic strategies is summarised and the authors propose a novel therapeutic approach based on the blocking of crucial binding sites present in the extracellular matrix.
Collapse
Affiliation(s)
- Laura Sanz
- Department of Immunology, Hospital Universitario Clínica Puerta de Hierro, San Martín de Porres 4, 28035 Madrid, Spain
| | | |
Collapse
|
161
|
Ebbinghaus C, Ronca R, Kaspar M, Grabulovski D, Berndt A, Kosmehl H, Zardi L, Neri D. Engineered vascular-targeting antibody-interferon-gamma fusion protein for cancer therapy. Int J Cancer 2005; 116:304-13. [PMID: 15800913 DOI: 10.1002/ijc.20952] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A number of cytokines are either approved drugs or are in advanced clinical trials, yet these biopharmaceuticals do not typically localize efficiently in solid tumors and manifest their therapeutic potential at the expense of severe side effects. The targeted delivery of cytokines to solid tumors is a promising avenue for increasing the therapeutic index of these biopharmaceuticals. We engineered a fusion protein between scFv(L19), a human antibody fragment specific to the EDB domain of fibronectin, and a cysteine-free mutant of murine interferon-gamma. The resulting fusion protein was capable of targeting new blood vessels in solid tumors, and the targeting efficiency was strikingly increased in tumor-bearing knockout mice lacking the interferon-gamma receptor. ScFv(L19)-interferon-gamma displayed a strong antitumor effect in both subcutaneous and metastatic murine F9 teratocarcinomas, but was not efficacious as single agent when used to treat C51 and CT26 tumors. The potency of this fusion protein could be substantially enhanced by combination with doxorubicin and other immunocytokines. These findings are of clinical relevance, as the EDB domain is a marker of angiogenesis, with identical sequence in mouse and man, which is abundantly expressed in a variety of aggressive solid tumors but is undetectable in most normal tissues.
Collapse
Affiliation(s)
- Christina Ebbinghaus
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
162
|
Ran S, He J, Huang X, Soares M, Scothorn D, Thorpe PE. Antitumor effects of a monoclonal antibody that binds anionic phospholipids on the surface of tumor blood vessels in mice. Clin Cancer Res 2005; 11:1551-62. [PMID: 15746060 DOI: 10.1158/1078-0432.ccr-04-1645] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We recently reported that anionic phospholipids, principally phosphatidylserine, become exposed on the external surface of viable vascular endothelial cells in tumors, possibly in response to oxidative stresses present in the tumor microenvironment. In the present study, we tested the hypothesis that a monoclonal antibody directed against anionic phospholipids might exert antitumor effects by causing vascular damage in tumors. EXPERIMENTAL DESIGN A new mouse immunoglobulin G3 monoclonal antibody, 3G4, was raised that binds anionic phospholipids in the presence of serum or beta2-glycoprotein I. The antibody was tested for its ability to localize to tumor vessels and exert antitumor effects in mice. RESULTS 3G4 recognized anionic phospholipids on the external membrane of H(2)O(2)-treated endothelial cells and in vitro. It localized specifically to tumor vascular endothelium and to necrotic tumor cells after injection into severe combined immunodeficient mice bearing orthotopic MDA-MB-435 tumors. Treatment with 3G4 retarded the growth of four different tumors in mice. It reduced the growth of established orthotopic MDA-MB-231 and MDA-MB-435 human breast tumors in mice by 75% and 65% respectively, large L540 human Hodgkin's tumors by 50%, and small syngeneic Meth A fibrosarcomas by 90%. Histologic examination revealed vascular damage, a reduction in vascular density, and a reduction in tumor plasma volume. Treatment with 3G4 induced the binding of monocytes to tumor endothelium and infiltration of macrophages into MDA-MB-435 and MDA-MB-231 tumors. No toxicity to the mice was observed. CONCLUSIONS 3G4 localizes specifically to complexes of anionic phospholipids and serum proteins on the surface of vascular endothelial cells in tumors in mice. This results in damage to tumor vasculature and suppression of tumor growth.
Collapse
Affiliation(s)
- Sophia Ran
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 2201 Inwood Road NC7.304, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
163
|
Menrad A, Menssen HD. ED-B fibronectin as a target for antibody-based cancer treatments. Expert Opin Ther Targets 2005; 9:491-500. [PMID: 15948669 DOI: 10.1517/14728222.9.3.491] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemotherapeutic agents for the treatment of solid cancers do not discriminate between malignant and normal tissue, but rather depend on the increased proliferation of tumour cells versus benign cells. To reach therapeutically active concentrations in the tumour, large doses of these rather unspecific compounds have to be given to the patient, often resulting in severe side effects. Therefore, the goal of modern cancer research is the development of highly selective compounds which are able to discriminate between tumour tissue and normal tissue. One promising approach in this direction is antibody-mediated targeted cancer therapy which may either block an important receptor-ligand interaction or deliver a therapeutically active molecule to an otherwise nonfunctional target. A prerequisite for such an approach is the tumour-selective expression of the respective target structure. This review discusses extra domain-B fibronectin as a promising target which is associated with tumour angiogenesis and tumour growth for the development of novel antibody-mediated therapies.
Collapse
Affiliation(s)
- Andreas Menrad
- Department of AntiAngiogenesis Research, Schering AG, Corporate Business Area Oncology, Germany.
| | | |
Collapse
|
164
|
Abstract
It is now accepted that the growth of solid tumours is dependent on their capacity to acquire a blood supply, and much effort has been directed towards the development of agents (known as anti-angiogenics) that disrupt this process. More recently, it has become apparent that targeted destruction of the established tumour vasculature is another avenue for exciting therapeutic opportunities. In this article, we present evidence that vascular targeting is an effective antitumour strategy in animal models, describe strategies for identifying putative tumour vascular targets and discuss future prospects for vascular targeting in the clinic.
Collapse
Affiliation(s)
- Dario Neri
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zürich, Wolfgang-Pauli-Strasse 10, CH-8093 Zürich, Switzerland.
| | | |
Collapse
|
165
|
Sanz L, Cuesta AM, Compte M, Alvarez-Vallina L. Antibody engineering: facing new challenges in cancer therapy. Acta Pharmacol Sin 2005; 26:641-8. [PMID: 15916728 DOI: 10.1111/j.1745-7254.2005.00135.x] [Citation(s) in RCA: 231] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Antibody-based therapeutics are beginning to realize the promise enclosed in their early denomination as magic bullets. Initial disappointment has turned into clinical and commercial success, and engineered antibodies currently represent over 30% of biopharmaceuticals in clinical trials. Recent structural and functional data have allowed the design of a new generation of therapeutic antibodies, with strategies ranging from complement-mediated and antibody-dependant cellular cytotoxicity enhancement to improved cytotoxic payloads using toxins, drugs, radionucleids and viral delivery. This review considers the structure of different types of recombinant antibodies, their mechanism of action and how their efficacy has been increased using a broad array of approaches. We will also focus on the additional benefits offered by the use of gene therapy methods for the in vivo production of therapeutic antibodies.
Collapse
Affiliation(s)
- Laura Sanz
- Servicio de Inmunologia, Hospital Universitario Puerta de Hierro, Madrid, Spain.
| | | | | | | |
Collapse
|
166
|
Narazaki M, Tosato G. Targeting Coagulation to the Tumor Microvasculature: Perspectives and Therapeutic Implications From Preclinical Studies. ACTA ACUST UNITED AC 2005; 97:705-7. [PMID: 15900035 DOI: 10.1093/jnci/dji152] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
167
|
Makabe K, Asano R, Ito T, Tsumoto K, Kudo T, Kumagai I. Tumor-directed lymphocyte-activating cytokines: refolding-based preparation of recombinant human interleukin-12 and an antibody variable domain-fused protein by additive-introduced stepwise dialysis. Biochem Biophys Res Commun 2005; 328:98-105. [PMID: 15670756 DOI: 10.1016/j.bbrc.2004.12.141] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Indexed: 11/27/2022]
Abstract
Integration of lymphocyte-activating cytokines (e.g., interleukin-12: IL-12) to tumor cells offers promise for cancer immunotherapy, but the preparation of such heterodimeric proteins by refolding is difficult because of subunit instability. We achieved the refolding of Escherichia coli-expressed human IL-12 by a stepwise dialysis method, preventing the formation of insoluble aggregates by adding a redox reagent and an aggregation suppressor. We also constructed a tumor-specific IL-12 protein, each subunit of which was fused with one chain of variable domain fragment (Fv) of anticarcinoembryonic antigen (CEA) antibody T84.66 (aCEA-IL12). Fusion of IL-12 with Fv greatly increased the yield of functional heterodimer. Several assays have indicated that the Fv domain and IL-12 domain of the fused protein had cognate biological activities, and it enhanced the cytotoxicity of T-LAK cells for the cancer cell line.
Collapse
Affiliation(s)
- Koki Makabe
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Aoba-yama 07, Aoba-ku, Sendai 980-8579, Japan
| | | | | | | | | | | |
Collapse
|
168
|
Rybak JN, Ettorre A, Kaissling B, Giavazzi R, Neri D, Elia G. In vivo protein biotinylation for identification of organ-specific antigens accessible from the vasculature. Nat Methods 2005; 2:291-8. [PMID: 15782212 DOI: 10.1038/nmeth745] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 02/02/2005] [Indexed: 01/07/2023]
Abstract
We describe a new methodology, based on terminal perfusion of rodents with a reactive ester derivative of biotin that enables the covalent modification of proteins readily accessible from the bloodstream. Biotinylated proteins from total organ extracts can be purified on streptavidin resin in the presence of strong detergents, digested on the resin and subjected to liquid chromatography-tandem mass spectrometry for identification. In the present study, in vivo biotinylation procedure led to the identification of hundreds of proteins in different mouse organs, including some showing a restricted pattern of expression in certain body tissues. Furthermore, biotinylation of mice with F9 subcutaneous tumors or orthotopic kidney tumors revealed both quantitative and qualitative differences in the recovery of biotinylated proteins, as compared to normal tissues. This technology is applicable to proteomic investigations of the differential expression of accessible proteins in physiological and pathological processes in animal models, and to human surgical specimens using ex vivo perfusion procedures.
Collapse
Affiliation(s)
- Jascha-N Rybak
- Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
169
|
Abstract
The advent of recombinant biotechnology and the recent sequencing of the human genome now allow for identification of scores of potential protein therapeutics along with the capacity to produce them in quantities and purities required for clinical application. Thus, clinical development of potential protein therapeutics has become as commonplace as development efforts of classical small molecule therapeutics. Whereas small molecule therapeutic lead candidates are identified through screens of large sets of possibilities, therapeutic protein candidates are defined by genetic information as a single composition (or a limited set of isoforms). Small molecule leads are identified through the combined assessment of desired selectivity, biodistribution and pharmacokinetic properties. In essence, these selection parameters emulate the actions of protein therapeutics that function as systemic hormones through their ability to target selective cells and tissues of the body via selective receptor interaction with minimal actions elsewhere. However, many, if not most, potential protein therapeutics do not normally circulate through the body to reach their target cell or tissue; rather, they are frequently synthesised at local sites, act at that site and are degraded without reaching appreciable systemic levels. Dose-limiting adverse events are associated with systemic administration of many of these proteins, restricting their clinical potential. This review examines current strategies to reduce these dose-limiting events by possibly focusing the delivery of potential protein therapeutics to discrete tissues and cells.
Collapse
Affiliation(s)
- Randall J Mrsny
- Welsh School of Pharmacy, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
170
|
Smith J, Kontermann RE, Embleton J, Kumar S. Antibody phage display technologies with special reference to angiogenesis. FASEB J 2005; 19:331-41. [PMID: 15746176 DOI: 10.1096/fj.04-2863rev] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The presence of blood vessels is a prerequisite for normal development, tissue growth, and tissue repair. However, its abnormal occurrence or absence can also potentiate disease processes. Angiogenic therapies have been used to stimulate blood vessel growth in ischemic conditions such as severe end-stage peripheral vascular disease, ischemic heart disease and stroke and for inhibition of angiogenesis in tumors. The targeting and identification of novel endothelial cell (EC) markers that can ultimately be used in angiogenic strategies is an expanding field but is limited by the availability of reagents. For instance repeated injection of mouse monoclonal antibodies (Mabs) against angiogenic EC, can result in the production of autoantibodies. Therefore, these mouse Mabs cannot be used for therapeutic purposes. Phage display technology was employed in this context to select antibodies, proteins, and peptides against known or novel EC antigens. Furthermore, technologies have been developed that enable the specific targeting of epitopes on cells including the endothelium with high-affinity/avidity antibodies. The focus for these antibody targeting strategies are markers that are unique or up-regulated on angiogenic EC including the vascular endothelial growth factor receptor (VEGFR) KDR, endoglin (CD105), and the extracellular domain B (ED-B) domain of fibronectin (FN). These markers are reviewed herein.
Collapse
Affiliation(s)
- Julia Smith
- University of Manchester, Stopford Building, Oxford Rd, Manchester, M13 9PT, UK.
| | | | | | | |
Collapse
|
171
|
Wong RJ, Chan MK, Yu Z, Ghossein RA, Ngai I, Adusumilli PS, Stiles BM, Shah JP, Singh B, Fong Y. Angiogenesis inhibition by an oncolytic herpes virus expressing interleukin 12. Clin Cancer Res 2005; 10:4509-16. [PMID: 15240543 DOI: 10.1158/1078-0432.ccr-04-0081] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Oncolytic herpes simplex viruses (HSVs) may have significant antitumor effects resulting from the direct lysis of cancer cells. HSVs may also be used to express inserted transgenes to exploit additional therapeutic strategies. The ability of an interleukin (IL)-12-expressing HSV to treat squamous cell carcinoma (SCC) by inhibition of tumor angiogenesis is investigated in this study. EXPERIMENTAL DESIGN A replication-competent, attenuated, oncolytic HSV carrying the murine IL-12 gene (NV1042), its non-cytokine-carrying analog (NV1023), or saline was used to treat established murine SCC flank tumors by intratumoral injection. The expression of secondary antiangiogenic mediators was measured. Angiogenesis inhibition was assessed by in vivo Matrigel plug assays, flank tumor subdermal vascularity, and in vitro endothelial cell tubule formation assay. RESULTS Intratumoral injections of NV1042 (2 x 10(7) plaque-forming units) into murine SCC VII flank tumors resulted in smaller tumor volumes as compared with NV1023 or saline. IL-12 and IFN-gamma expression in tumors was 440 and 2.2 pg/mg, respectively, at 24 h after NV1042 injection, but both IL-12 and IFN-gamma were undetectable (<0.2 pg/mg) after NV1023 or saline injections. Expression of two antiangiogenesis mediators, monokine induced by IFN-gamma and IFN-inducible protein 10, was elevated after NV1042 treatment. Matrigel plug assays of NV1042-transfected SCC VII tumor cells demonstrated significantly decreased hemoglobin content and microvessel density as compared with NV1023 and PBS. Excised murine flank tumors treated with NV1042 had decreased subdermal vascularity as compared with NV1023 and PBS. Both splenocytes and IL-12 expression by NV1042 were required for in vitro inhibition of endothelial tubule formation. CONCLUSIONS IL-12 expression by an oncolytic herpes virus enhances therapy of SCC through antiangiogenic mechanisms. Strategies combining HSV oncolysis with angiogenesis inhibition merit further investigation for potential clinical application.
Collapse
Affiliation(s)
- Richard J Wong
- Head and Neck Service, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Enhancement of the Antiangiogenic Activity of Interleukin-12 by Peptide Targeted Delivery of the Cytokine to αvβ3 Integrin. Mol Cancer Res 2004. [DOI: 10.1158/1541-7786.663.2.12] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
We engineered a fusion protein, mrIL-12vp [mouse recombinant interleukin (IL)-12 linked to vascular peptide], linking the vascular homing peptide CDCRGDCFC (RGD-4C), a ligand for αvβ3 integrin, to mrIL-12 to target IL-12 directly to tumor neovasculature. The fusion protein stimulated IFN-γ production in vitro and in vivo, indicating its biological activity was consistent with mrIL-12. Immunofluorescence techniques showed mrIL-12vp specifically bound to αvβ3 integrin-positive cells but not to αvβ3 integrin-negative cells. In corneal angiogenesis assays using BALB/c mice treated with either 0.5 μg/mouse/d of mrIL-12vp or mrIL-12 delivered by subcutaneous continuous infusion, mrIL-12vp inhibited corneal neovascularization by 67% compared with only a slight reduction (13%) in angiogenesis in the mrIL-12-treated animals (P = 0.008). IL-12 receptor knockout mice given mrIL-12vp showed a marked decrease in the area of corneal neovascularization compared with mice treated with mrIL-12. These results indicate that mrIL-12vp inhibits angiogenesis through IL-12-dependent and IL-12-independent mechanisms, and its augmented antiangiogenic activity may be due to suppression of endothelial cell signaling pathways by the RGD-4C portion of the fusion protein. Mice injected with NXS2 neuroblastoma cells and treated with mrIL-12vp showed significant suppression of tumor growth compared with mice treated with mrIL-12 (P = 0.03). Mice did not show signs of IL-12 toxicity when treated with mrIL-12vp, although hepatic necrosis was present in mrIL-12-treated mice. Localization of IL-12 to neovasculature significantly enhances the antiangiogenic effect, augments antitumor activity, and decreases toxicity of IL-12, offering a promising strategy for expanding development of IL-12 for treatment of cancer patients.
Collapse
|
173
|
Karmakar S, Dhar R, Das C. Inhibition of Cytotrophoblastic (JEG-3) Cell Invasion by Interleukin 12 Involves an Interferon γ-mediated Pathway. J Biol Chem 2004; 279:55297-307. [PMID: 15448160 DOI: 10.1074/jbc.m407013200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Trophoblast invasion, like tumor invasion, shares common biochemical mechanisms. However, in contrast to tumor invasion of a host tissue, trophoblastic invasion during implantation is strictly regulated, temporospatially. Factors responsible for these important regulatory processes are presently unknown; however, studies indicate that cytokines and growth factors represent in the peri-implantation uterine milieu as the possible candidates. In this study we investigated the role of interleukin (IL) 12 in regulating trophoblast invasion and the expression of trophoblast proteases (matrix metalloprotease (MMP)-2, MMP-9, and urokinase-type plasminogen activators) and their inhibitors (tissue inhibitors of metalloprotease (TIMP) 1, TIMP-2, and plasminogen activator inhibitor (PAI)-1) using an in vitro tissue culture system of human choriocarcinoma cell line JEG-3. Our major findings show an anti-invasive role of IL-12, associated with an inhibitory effect on the proteases but with an opposite up-regulating influence on the protease inhibitor, TIMP-1, whereas TIMP-2 and plasminogen activator inhibitor 1 remained unaltered. Stimulation of JEG-3 cells with IL-12 also induced interferon (IFN)-gamma production, which when neutralized using a monoclonal anti-IFN-gamma antibody, F12, abrogates its ability to down-regulate the MMPs. IL-12 also mediates an IFN-gamma-dependent up-regulation of E-cadherin, thereby implying that alteration in cell-cell adhesion besides regulating the proteases and the inhibitors possibly contributes to the observed anti-invasive role of this cytokine. TIMP-1, although stimulated by IL-12, was found to be unaltered by antibody F12, thereby implying a possibility of an IL-12-dependent-IFN-gamma independent regulation. These findings thereby suggest an important role of IL-12 in modulation of trophoblast proteases and their inhibitors besides regulating cell-cell interactions and invasion during implantation, with far reaching possibilities for understanding the mechanism(s) and regulations of invasion and metastasis.
Collapse
Affiliation(s)
- Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi-110029, India.
| | | | | |
Collapse
|
174
|
Matter CM, Schuler PK, Alessi P, Meier P, Ricci R, Zhang D, Halin C, Castellani P, Zardi L, Hofer CK, Montani M, Neri D, Lüscher TF. Molecular imaging of atherosclerotic plaques using a human antibody against the extra-domain B of fibronectin. Circ Res 2004; 95:1225-33. [PMID: 15539632 DOI: 10.1161/01.res.0000150373.15149.ff] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Current imaging modalities of human atherosclerosis, such as angiography, ultrasound, and computed tomography, visualize plaque morphology. However, methods that provide insight into plaque biology using molecular tools are still insufficient. The extra-domain B (ED-B) is inserted into the fibronectin molecule by alternative splicing during angiogenesis and tissue remodeling but is virtually undetectable in normal adult tissues. Angiogenesis and tissue repair are also hallmarks of advanced plaques. For imaging atherosclerotic plaques, the human antibody L19 (specific against ED-B) and a negative control antibody were labeled with radioiodine or infrared fluorophores and injected intravenously into atherosclerotic apolipoprotein E-null (ApoE-/-) or normal wild-type mice. Aortas isolated 4 hours, 24 hours, and 3 days after injection exhibited a selective and stable uptake of L19 when using radiographic or fluorescent imaging. L19 binding was confined to the plaques as assessed by fat staining. Comparisons between fat staining and autoradiographies 24 hours after 125I-labeled L19 revealed a significant correlation (r=0.89; P<0.0001). Minimal antibody uptake was observed in normal vessels from wild-type mice receiving the L19 antibody and in atherosclerotic vessels from ApoE-/- mice receiving the negative control antibody. Immunohistochemical studies revealed increased expression of ED-B not only in murine but also in human plaques, in which it was found predominantly around vasa vasorum and plaque matrix. In summary, we demonstrate selective targeting of atheromas in mice using the human antibody to the ED-B domain of fibronectin. Thus, our findings may set the stage for antibody-based molecular imaging of atherosclerotic plaques in the intact organism.
Collapse
Affiliation(s)
- Christian M Matter
- Cardiovascular Research, Institute of Physiology, University of Zurich and Cardiovascular Center, Zurich University Hospital, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Abstract
Vascular targeting agents (VTAs) for the treatment of cancer are designed to cause a rapid and selective shutdown of the blood vessels of tumors. Unlike antiangiogenic drugs that inhibit the formation of new vessels, VTAs occlude the pre-existing blood vessels of tumors to cause tumor cell death from ischemia and extensive hemorrhagic necrosis. Tumor selectivity is conferred by differences in the pathophysiology of tumor versus normal tissue vessels (e.g., increased proliferation and fragility, and up-regulated proteins). VTAs can kill indirectly the tumor cells that are resistant to conventional antiproliferative cancer therapies, i.e., cells in areas distant from blood vessels where drug penetration is poor, and hypoxia can lead to radiation and drug resistance. VTAs are expected to show the greatest therapeutic benefit as part of combined modality regimens. Preclinical studies have shown VTA-induced enhancement of the effects of conventional chemotherapeutic agents, radiation, hyperthermia, radioimmunotherapy, and antiangiogenic agents. There are broadly two types of VTAs, small molecules and ligand-based, which are grouped together, because they both cause acute vascular shutdown in tumors leading to massive necrosis. The small molecules include the microtubulin destabilizing drugs, combretastatin A-4 disodium phosphate, ZD6126, AVE8062, and Oxi 4503, and the flavonoid, DMXAA. Ligand-based VTAs use antibodies, peptides, or growth factors that bind selectively to tumor versus normal vessels to target tumors with agents that occlude blood vessels. The ligand-based VTAs include fusion proteins (e.g., vascular endothelial growth factor linked to the plant toxin gelonin), immunotoxins (e.g., monoclonal antibodies to endoglin conjugated to ricin A), antibodies linked to cytokines, liposomally encapsulated drugs, and gene therapy approaches. Combretastatin A-4 disodium phosphate, ZD6126, AVE8062, and DMXAA are undergoing clinical evaluation. Phase I monotherapy studies have shown that the agents are tolerated with some demonstration of single agent efficacy. Because efficacy is expected when the agents are used with conventional chemotherapeutic drugs or radiation, the results of Phase II combination studies are eagerly awaited.
Collapse
Affiliation(s)
- Philip E Thorpe
- Department of Pharmacology and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| |
Collapse
|
176
|
Brack SS, Dinkelborg LM, Neri D. Molecular targeting of angiogenesis for imaging and therapy. Eur J Nucl Med Mol Imaging 2004; 31:1327-41. [PMID: 15300415 DOI: 10.1007/s00259-004-1648-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Angiogenesis, i.e. the proliferation of new blood vessels from pre-existing ones, is an underlying process in many human diseases, including cancer, blinding ocular disorders and rheumatoid arthritis. The ability to selectively target and interfere with neovascularisation would potentially be useful in the diagnosis and treatment of angiogenesis-related diseases. This review presents the authors' views on some of the most relevant markers of angiogenesis described to date, as well as on specific ligands which have been characterised in pre-clinical animal models and/or clinical studies. Furthermore, we present an overview on technologies which are likely to have an impact on the way molecular targeting of angiogenesis is performed in the future.
Collapse
Affiliation(s)
- Simon S Brack
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | | | | |
Collapse
|
177
|
Alessi P, Ebbinghaus C, Neri D. Molecular targeting of angiogenesis. Biochim Biophys Acta Rev Cancer 2004; 1654:39-49. [PMID: 14984766 DOI: 10.1016/j.bbcan.2003.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2003] [Revised: 08/08/2003] [Accepted: 08/08/2003] [Indexed: 10/26/2022]
Abstract
The majority of pharmacological approaches for the treatment of solid tumors suffer from poor selectivity, thus limiting dose escalation (i.e., the doses of drug which are required to kill tumor cells cause unacceptable toxicities to normal tissues). The situation is made more dramatic by the fact that the majority of anticancer drugs accumulate preferentially in normal tissues rather than in neoplastic sites, due to the irregular vasculature and to the high interstitial pressure of solid tumors. One avenue towards the development of more efficacious and better tolerated anti-cancer drugs relies on the targeted delivery of therapeutic agents to the tumor environment, thus sparing normal tissues. Molecular markers which are selectively expressed in the stroma and in neo-vascular sites of aggressive solid tumors appear to be particularly suited for ligand-based tumor targeting strategies. Tumor blood vessels are accessible to agents coming from the bloodstream, and their occlusion may result in an avalanche of tumor cell death. Furthermore, endothelial cells and stromal cells are genetically more stable than tumor cells and can produce abundant markers, which are ideally suited for tumor targeting strategies. This review focuses on recent advances in the development of ligands for the selective targeting of tumor blood vessels and new blood vessels in other angiogenesis-related diseases.
Collapse
Affiliation(s)
- Patrizia Alessi
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Building 36 M14, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | | | |
Collapse
|
178
|
Abstract
PURPOSE OF REVIEW Targeting the endothelial cells that line tumor infiltrating blood vessels is a new anticancer strategy that has gained widespread support from biologists and clinicians. Here we highlight different approaches currently being used to target tumor endothelium and discuss new avenues for intervention that have been opened through the recent identification of tumor endothelial markers (TEMs). RECENT FINDINGS The ability of Avastin to prolong survival in a Phase III clinical trial of human colorectal cancer has established the validity of the anti-angiogenic approach. However, realization of the full potential of a vascular targeting strategy may require the exploitation of molecules which are highly restricted in expression to tumor endothelium. Here we explore the potential of TEMs as new targets for cancer therapy. Current knowledge of these markers and their relation to other family members in the context of tumor angiogenesis is discussed. In particular, we highlight those molecules which, by virtue of their structure, cell-surface location and expression pattern, appear to hold promise as targets for future drug development. The identification of TEM8 as the anthrax toxin receptor and the successful targeting of this receptor in preclinical tumor models make this molecule a particularly attractive candidate for future vascular targeting studies. SUMMARY Technological advances in cellular fractionation and genomics enabled the identification of several markers preferentially expressed on human tumor endothelium. Studies of these TEMs are expected to aid in our understanding of angiogenesis and could lead to the development of new imaging and diagnostic agents for cancer.
Collapse
Affiliation(s)
- Akash Nanda
- Program in Human Genetics and Molecular Biology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
179
|
|
180
|
Borsi L, Balza E, Carnemolla B, Sassi F, Castellani P, Berndt A, Kosmehl H, Biro A, Siri A, Orecchia P, Grassi J, Neri D, Zardi L. Selective targeted delivery of TNFalpha to tumor blood vessels. Blood 2003; 102:4384-92. [PMID: 12933583 DOI: 10.1182/blood-2003-04-1039] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We sought to enhance the selective toxicity of tumor necrosis factor alpha (TNFalpha) to permit its systemic use in cancer therapy. Because ligand-targeted therapeutics have proven successful in improving the selective toxicity of drugs, we prepared a fusion protein (L19mTNFalpha) composed of mouse TNFalpha and a high-affinity antibody fragment (L19 scFv) to the extradomain B (ED-B) domain of fibronectin, a marker of angiogenesis. L19mTNFalpha was expressed in mammalian cells, purified, and characterized. L19mTNFalpha was an immunoreactive and biologically active homotrimer. Radiolabeled L19mTNFalpha selectively targeted tumor neovasculature in tumor-bearing mice, where it accumulated selectively and persistently (tumor-to-blood ratio of the percentage of injected dose per gram [%ID/g] of 700, 48 hours from injection). L19mTNFalpha showed a greater anticancer therapeutic activity than both mTNFalpha and TN11mTNFalpha, a control fusion protein in which an antibody fragment, irrelevant in the tumor model used, substituted for L19. This activity was further dramatically enhanced by its combination with melphalan or the recently reported fusion protein L19-IL2. In conclusion, L19mTNFalpha allows concentrating therapeutically active doses of TNFalpha at the tumor level, thus opening new possibilities for the systemic use of TNFalpha in cancer therapy.
Collapse
Affiliation(s)
- Laura Borsi
- Laboratory of Cell Biology, Istituto Nazionale per la Ricerca sul Cancro, Largo Rosanna Benzi, 10 16132 Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Heuser C, Diehl V, Abken H, Hombach A. Anti-CD30-IL-12 antibody-cytokine fusion protein that induces IFN-gamma secretion of T cells and NK cell-mediated lysis of Hodgkin's lymphoma-derived tumor cells. Int J Cancer 2003; 106:545-552. [PMID: 12845650 DOI: 10.1002/ijc.11279] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Interleukin-12 (IL-12) is a disulfide-linked p40-p35 heterodimeric cytokine and plays a key role in linking innate cellular immunity to an adaptive Th1 response against pathogens and tumor cells and in counteracting a Th2 immune response. The pathogenesis of Hodgkin's disease (HD) is partially attributed to a Th2 dominance associated with functional anergy of T cells that accumulate in the near vicinity to the malignant Hodgkin/Reed-Sternberg (H/RS) cells. To revert Th2 polarization in the tumor lesion, we generated an anti-CD30-IL-12 antibody-cytokine fusion protein that binds to CD30 on H/RS cells and is composed of a CD30 binding domain (HRS3-scFv) linked to p40-p35 murine single chain IL-12. The HRS3-scFv-hi-IL-12 fusion protein is expressed as a 110 kD polypeptide, can be purified by affinity chromatography, and has binding specificities to both the CD30 antigen and the IL-12 receptor. After binding to CD30(+) H/RS cells, the fusion protein stimulates T cells to secrete IFN-gamma, a predominant Th1 cytokine, and induces NK cells to lyse CD30(+) cells with high efficiency. These properties make the HRS3-scFv-hi-IL-12 fusion protein suitable for the specific immunotherapy of Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Claudia Heuser
- Klinik I für Innere Medizin, Labor für Tumorgenetik, Universität zu Köln, Köln, Germany
- Zentrum für Molekulare Medizin Köln (ZMMK), Universität zu Köln, Köln, Germany
| | - Volker Diehl
- Klinik I für Innere Medizin, Labor für Tumorgenetik, Universität zu Köln, Köln, Germany
- Zentrum für Molekulare Medizin Köln (ZMMK), Universität zu Köln, Köln, Germany
| | - Hinrich Abken
- Klinik I für Innere Medizin, Labor für Tumorgenetik, Universität zu Köln, Köln, Germany
- Zentrum für Molekulare Medizin Köln (ZMMK), Universität zu Köln, Köln, Germany
| | - Andreas Hombach
- Klinik I für Innere Medizin, Labor für Tumorgenetik, Universität zu Köln, Köln, Germany
- Zentrum für Molekulare Medizin Köln (ZMMK), Universität zu Köln, Köln, Germany
| |
Collapse
|
182
|
Abstract
With the acceptance of antibodies as therapeutics, a diversity of engineered antibody forms have been created to improve their efficacy, including enhancing the effector functions of full-length antibodies, delivering toxins to kill cells or cytokines in order to stimulate the immune system, and bispecific antibodies to target multiple receptors. After years of in vitro investigation, many of these are now moving into clinical trials and are showing promise. A potential new type of effector function for antibodies, that is, the generation of reactive oxygen species that may effect inflammation or bacterial killing, has been elucidated. In addition, the field has expanded beyond a concentration on immunoglobulin G to include immunoglobulin A antibodies as potential therapeutics.
Collapse
Affiliation(s)
- Leonard Presta
- Department of Protein and Antibody Technology, DNAX Inc, 901 California Avenue, Palo Alto, CA 94304-1104, USA.
| |
Collapse
|
183
|
Birchler MT, Milisavlijevic D, Pfaltz M, Neri D, Odermatt B, Schmid S, Stoeckli SJ. Expression of the extra domain B of fibronectin, a marker of angiogenesis, in head and neck tumors. Laryngoscope 2003; 113:1231-7. [PMID: 12838025 DOI: 10.1097/00005537-200307000-00023] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES/HYPOTHESIS The extra domain B (ED-B) of fibronectin, a naturally occurring marker of tissue remodeling and angiogenesis, is expressed in the majority of aggressive solid human tumors, whereas it is not detectable in normal vessels and tissues. STUDY DESIGN In view of the diagnostic and therapeutic clinical applications of the L19 antibody, which is specific for the ED-B domain of fibronectin, a prospective immunohistochemical analysis of different head and neck tumors was performed. METHODS In all, 82 head and neck tissue biopsy specimens were immunohistochemically analyzed using the L19 antibody. They consisted of 53 different malignant tumors, 8 benign tumors, 10 nontumoral lesions, and 11 normal control tissues. RESULTS A strong positive staining with the L19 antibody could be observed in 87% of the investigated malignant tumors, in only 38% of the benign tumors, and in 20% of the nontumoral lesions (P <.0001). The extra domain B was completely absent in the normal control tissue samples. CONCLUSIONS The results show that ED-B is abundantly expressed around the neovasculature and in the stroma of the majority of malignant tumors of the head and neck but is undetectable in normal tissues. The ED-B domain of fibronectin is a good-quality tumor-stroma-associated antigen that warrants clinical trials with antibody-based pharmaceuticals, including immunoscintigraphic investigations and radioimmunoguided surgery with the radiolabeled L19 antibody.
Collapse
Affiliation(s)
- Manfred T Birchler
- Department of Otolaryngology-Head & Neck Surgery, University Hospital of Zurich, Frauenklinikstrasse 24, 8091 Zurich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
184
|
Sanz L, Alvarez-Vallina L. The extracellular matrix: a new turn-of-the-screw for anti-angiogenic strategies. Trends Mol Med 2003; 9:256-62. [PMID: 12829014 DOI: 10.1016/s1471-4914(03)00070-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Anti-angiogenic therapy is currently one of most active fields in cancer research. The initial strategies, which were aimed at inhibiting tumor vascularization, included upregulation of endogenous inhibitors and blocking of the signals delivered by angiogenic factors. However, interactions between endothelial cells and their surrounding extracellular matrix also play a crucial role in modulation of the angiogenic process. Compounds that target either the integrins implicated in these interactions or the proteases responsible for matrix remodeling have been shown to halt tumor growth in murine models and are now in clinical trials. However, little attention has been paid to integrin ligands, the extracellular matrix components that support endothelial cell survival, movement and reorganization. Here, we summarize the current knowledge about these angiogenesis inhibitors and propose a novel therapeutic approach based on the blocking of crucial binding sites present in the extracellular matrix.
Collapse
Affiliation(s)
- Laura Sanz
- Molecular Medicine Program, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
185
|
George AJT, Lee L, Pitzalis C. Isolating ligands specific for human vasculature using in vivo phage selection. Trends Biotechnol 2003; 21:199-203. [PMID: 12727380 DOI: 10.1016/s0167-7799(03)00079-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The endothelium lining blood vessels expresses molecules that are restricted in their expression to a particular tissue or organ. These molecules are attractive targets for therapy and diagnosis because they allow agents to be delivered specifically to the blood vessels supplying the desired tissue. However, it is difficult to identify these tissue-specific molecules because endothelium loses much of its tissue-specific nature when it is removed from the organ. This can be overcome by using in vivo phage selection - injecting libraries of phage bearing antibodies or peptides into an animal and isolating phage that bind to the relevant tissue. A variation on this approach, in which in vivo phage selection is performed in animals bearing human tissue xenografts, allows the isolation of peptides (and presumably other molecules) specific for human vasculature.
Collapse
Affiliation(s)
- Andrew J T George
- Department of Immunology, Division of Medicine, Imperial College London, Hammersmith Hospital, UK.
| | | | | |
Collapse
|
186
|
Scheuermann J, Volonterio A, Zerbe O, Zanda M, Neri D. Discovery and investigation of lead compounds as binders to the Extra-Domain B of the angiogenesis marker, fibronectin. Drug Dev Res 2003. [DOI: 10.1002/ddr.10161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
187
|
Yockman JW, Maheshwari A, Han SO, Kim SW. Tumor regression by repeated intratumoral delivery of water soluble lipopolymers/p2CMVmIL-12 complexes. J Control Release 2003; 87:177-86. [PMID: 12618034 DOI: 10.1016/s0168-3659(02)00362-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The recruitment of the body's own immune system is amongst the most potent defenses known against cancer. Recent attempts to harness this response have enlisted the use of the immune modulating cytokine, interleukin-12 (IL-12). The objective of this work is to investigate the organ distribution and anti-tumor response in vivo after intratumoral administration of IL-12 expression plasmid complexed with water soluble lipopolymer (WSLP). Formulations of WSLP/p2CMVmIL-12 at N/P mol ratio of 20:1 were prepared in the presence of 5% (w/v) glucose. Organ distribution data following intratumoral injection of CT-26 subcutaneous tumor-bearing BALB/c mice demonstrated enhanced retention of WSLP/p2CMVmIL-12 complexes within the tumor and limited accumulation in other organs for up to 96 h. Tumor-bearing BALB/c mice received either single or repeated intratumoral injections at 4- or 8-day intervals to examine the efficacy of single versus repeated injections on tumor regression and survival. Significant tumor growth inhibition during 4- and 8-day injection trials was observed with maximal survival in mice receiving 4-day injections of WSLP/p2CMVmIL-12 complexes. In conclusion, the water-soluble non-toxic lipopolymer complexed with p2CMVIL-12 showed enhanced transgene expression in vivo, inhibits the rate of tumor growth, and significantly increases survival.
Collapse
Affiliation(s)
- James W Yockman
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, BPRB, Room 205, Salt Lake City, UT 84112, USA
| | | | | | | |
Collapse
|
188
|
Melkko S, Halin C, Borsi L, Zardi L, Neri D. An antibody-calmodulin fusion protein reveals a functional dependence between macromolecular isoelectric point and tumor targeting performance. Int J Radiat Oncol Biol Phys 2002; 54:1485-90. [PMID: 12459375 DOI: 10.1016/s0360-3016(02)03927-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE Human monoclonal antibodies are promising agents for the development of improved anticancer therapeutics, because, unlike low-molecular-weight chemotherapeutic agents, they can selectively localize to solid tumors. In particular, the scFv(L19) antibody fragment, specific for the EDB domain of fibronectin, a marker of angiogenesis, has demonstrated an impressive tumor targeting performance in a variety of tumor-bearing animals and in patients with cancer. The purpose of this study was to develop a tumor pretargeting strategy, based on a novel anti-EDB fusion protein. METHODS AND MATERIALS We have fused the scFv(L19) to calmodulin, a small acidic protein for which specific binding peptides with a dissociation constant in the picomolar range are available. The resulting fusion protein has been expressed in mammalian cells and purified to homogeneity, before being characterized by quantitative biodistribution analysis in mice bearing the F9 murine teratocarcinoma. RESULTS Surprisingly, we have found that the fusion of scFv(L19) to calmodulin completely abrogated the tumor targeting ability of the antibody in vivo, although both scFv(L19) and calmodulin moieties within the fusion protein retained unaltered binding affinities toward their respective ligand. Furthermore, a systematic analysis of 13 derivatives of scFv(L19) recently produced in our laboratories showed that the 10 derivatives that retain the tumor targeting ability of the parental antibody have isoelectric points (pI) between 5.0 and 9.0, whereas scFv(L19)-calmodulin (pI = 4.49) and two other derivatives of scFv(L19) with pI >9.0 were unable to target tumors in vivo. CONCLUSIONS Because the EDB domain of fibronectin is a component of the modified extracellular matrix, predominantly located at the abluminal side of tumor blood vessels, our data suggest that extreme pI values of antibody-based pharmaceuticals may inhibit protein extravasation, perhaps by virtue of electrostatic interactions with endothelial cells and/or components of the extracellular matrix.
Collapse
Affiliation(s)
- Samu Melkko
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | | | | | | | | |
Collapse
|
189
|
Helguera G, Morrison SL, Penichet ML. Antibody-cytokine fusion proteins: harnessing the combined power of cytokines and antibodies for cancer therapy. Clin Immunol 2002; 105:233-46. [PMID: 12498805 DOI: 10.1006/clim.2002.5302] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- G Helguera
- Department of Microbiology, Immunology, and Molecular Genetics, Molecular Biology Institute, University of California, Los Angeles, 405 Hilgard Avenue, Los Angeles, California, 90095-1489, USA
| | | | | |
Collapse
|
190
|
Abstract
The blood vessels of individual tissues are biochemically distinct, and pathological lesions put their own signature on the vasculature. In tumors, both blood and lymphatic vessels differ from normal vessels. New methods, such as in vivo screening of phage libraries, have provided peptides and antibodies that recognize these vascular signatures and can be used in targeted delivery of therapeutic agents. Targeting a therapy to the diseased tissue enhances the efficacy of the treatment while reducing the side effects in mouse experiments. Results from drug delivery to tumor vessels have been particularly encouraging.
Collapse
Affiliation(s)
- Erkki Ruoslahti
- Cancer Research Center, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
191
|
Halin C, Niesner U, Villani ME, Zardi L, Neri D. Tumor-targeting properties of antibody-vascular endothelial growth factor fusion proteins. Int J Cancer 2002; 102:109-16. [PMID: 12385005 DOI: 10.1002/ijc.10674] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A major problem of antibody-based targeting of solid tumors is the poor penetration of antibodies into tumor tissue. Vasoactive immunoconjugates have been proposed as a means of increasing antibody uptake in tumors. In principle, VEGF (also known as vascular permeability factor) could selectively alter vascular permeability, leading to improved tumor targeting. A possible role for VEGF in the targeting of tumor neovasculature has been postulated, based on the overexpression of VEGF receptors in tumor endothelial cells. However, quantitative biodistribution studies on this topic are not available. In this report, we describe the cloning, expression, characterization and biodistribution in tumor-bearing mice of antibodies fused to either VEGF(120) or VEGF(164) The MAb fragments chosen for analysis were scFv(L19), specific for the ED-B domain of fibronectin, a marker of angiogenesis, and scFv(HyHEL-10), a negative control antibody of irrelevant specificity in mice. Neither unconjugated VEGF nor scFv(HyHEL-10)-VEGF fusion proteins showed accumulation in the tumor (tumor:blood ratios approx. 1 at 4 hr and 24 hr postinjection). By contrast, scFv(L19)-VEGF(120) but not scFv(L19)-VEGF(164) showed significant accumulation in tumors (tumor:blood ratio = 9.3 at 24 hr) but was not superior to unconjugated scFv(L19). Preinjection of unlabeled scFv(L19)-VEGF(120) prior to administration of radiolabeled fusion protein led to increased accumulation of radiolabeled scFv(L19)-VEGF(120) in the tumor but only at very high concentrations (20 microg/mouse).
Collapse
Affiliation(s)
- Cornelia Halin
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology-Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
192
|
Castellani P, Borsi L, Carnemolla B, Birò A, Dorcaratto A, Viale GL, Neri D, Zardi L. Differentiation between high- and low-grade astrocytoma using a human recombinant antibody to the extra domain-B of fibronectin. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:1695-700. [PMID: 12414516 PMCID: PMC1850782 DOI: 10.1016/s0002-9440(10)64446-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Different fibronectin (FN) isoforms are generated by the alternative splicing of the primary FN transcript. We previously demonstrated that the isoform containing the extra domain B sequence of fibronectin (B-FN), a complete type-III-homology repeat, is a marker of angiogenesis that accumulates around neovasculature only during angiogenic processes. We produced a single-chain human recombinant antibody (scFv), L19, which reacts specifically with B-FN and selectively targets tumor vasculature in vivo. We used this scFv and an antibody against a pan-endothelial marker (Factor VIII) in a double-staining procedure on specimens of low- and high-grade astrocytomas to determine the percentage of B-FN-positive vessels, (denominating the resulting value angiogenic index [AI]). Compared to vascular density and proliferative activity (evaluated using antibodies to Factor VIII and Ki67, respectively), AI correlated better with tumor grade (1.6 +/- 2.6% and 92.0 +/- 8.7% of B-FN-positive vessels in low- and high-grade astrocytomas, respectively) and was a more precise diagnostic tool than either of the two conventional methods. In fact, discriminating analysis using these three parameters showed that only AI accurately classified 100% of the cases studied, compared to 64% and 89% correctly diagnosed by vascular density and of proliferating cells, respectively.
Collapse
Affiliation(s)
- Patrizia Castellani
- Laboratory of Cell Biology, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
193
|
Borsi L, Balza E, Bestagno M, Castellani P, Carnemolla B, Biro A, Leprini A, Sepulveda J, Burrone O, Neri D, Zardi L. Selective targeting of tumoral vasculature: comparison of different formats of an antibody (L19) to the ED-B domain of fibronectin. Int J Cancer 2002; 102:75-85. [PMID: 12353237 DOI: 10.1002/ijc.10662] [Citation(s) in RCA: 270] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We recently demonstrated that a human recombinant scFv, L19, reacting with the ED-B domain of fibronectin, a marker of angiogenesis, selectively targets tumoral vasculature in vivo. Using the variable regions of L19, we constructed and expressed a human "small immunoprotein" (SIP) and a complete human IgG1 and performed biodistribution studies in tumor-bearing mice to compare the blood clearance rate, in vivo stability and performance in tumor targeting of the 3 L19 formats [dimeric scFv (scFv)(2), SIP and IgG1]. The accumulation of the different antibody formats in the tumors studied was a consequence of the clearance rate and in vivo stability of the molecules. Using the SIP, the %ID/g in tumors was 2-5 times higher than that of the (scFv)(2), reaching a maximum 4-6 hr after injection. By contrast, the accumulation of IgG1 in tumors constantly rose during the experiments. However, due to its slow clearance, the tumor-blood ratio of the %ID/g after 144 hr was only about 3 compared to a ratio of 10 for the (scFv)(2) and 70 for the SIP after the same period of time. The different in vivo behavior of these 3 completely human L19 formats could be exploited for different diagnostic and/or therapeutic purposes, depending on clinical needs and disease. Furthermore, the fact that ED-B is 100% homologous in human and mouse, which ensures that L19 reacts equally well with the human and the murine antigen, should expedite the transfer of these reagents to clinical trials.
Collapse
Affiliation(s)
- Laura Borsi
- Laboratory of Cell Biology, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Abstract
A mutant Raf-1 gene loaded onto nanoparticles, delivered to tumor vasculature with an integrin binding compound, provides effective antiangiogenic gene therapy in mice.
Collapse
|
195
|
Trepel M, Arap W, Pasqualini R. In vivo phage display and vascular heterogeneity: implications for targeted medicine. Curr Opin Chem Biol 2002; 6:399-404. [PMID: 12023122 DOI: 10.1016/s1367-5931(02)00336-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The vascular endothelium expresses differential receptors depending on the functional state and tissue localization of its cells. A method to characterize this receptor heterogeneity with phage display random peptide libraries has been developed. Using this technology, several peptide ligands have been isolated that home to tissue-specific endothelial cell receptors following intravenous administration. Such peptide ligands, or antibodies directed against specific vascular receptors, can be used to target therapeutic compounds or imaging agents to endothelial cells in vitro and in vivo. Recent advances in the field include identification of novel endothelial receptors expressed differentially in normal and pathological conditions and the isolation of peptides or antibody ligands to such receptors in in vitro assays, in animal models and in a human patient. These milestones, which extend the 'functional map' of the vasculature, should lead to clinical applications in diseases such as cancer and other conditions that exhibit distinct vascular characteristics.
Collapse
Affiliation(s)
- Martin Trepel
- University of Freiburg Medical Center, Department of Hematology and Oncology and Institute for Molecular Medicine and Cell Research, Hugstetter Strasse 55, D-79106, Freiburg, Germany
| | | | | |
Collapse
|