1
|
Singerer I, Tempel L, Gruen K, Heiß J, Gutte C, Matasci M, Schrepper A, Bauer R, Berndt A, Jung C, Schulze PC, Neri D, Franz M. Extra domain A-containing fibronectin in pulmonary hypertension and treatment effects of a function-blocking antibody. Cardiovasc Res 2024; 120:1485-1497. [PMID: 39023231 DOI: 10.1093/cvr/cvae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/07/2024] [Accepted: 06/01/2024] [Indexed: 07/20/2024] Open
Abstract
AIMS Pulmonary vascular and right ventricular (RV) remodelling processes are important for development and progression of pulmonary hypertension (PH). The current study analysed the functional role of the extra domain A-containing fibronectin (ED-A+ Fn) for the development of PH by comparing ED-A+ Fn knockout (KO) and wild-type (WT) mice as well as the effects of an antibody-based therapeutic approach in a model of monocrotaline (MCT)-induced PH, which will be validated in a model of Sugen 5416/hypoxia-induced PH. METHODS AND RESULTS PH was induced using MCT (PH mice). Sixty-nine mice were divided into the following groups: sham-treated controls (WT: n = 7; KO: n = 7), PH mice without specific treatment (WT: n = 12; KO: n = 10), PH mice treated with a dual endothelin receptor antagonist (macitentan; WT: n = 6; KO: n = 11), WT PH mice treated with the F8 antibody, specifically recognizing ED-A+ Fn, (n = 8), and WT PH mice treated with an antibody of irrelevant antigen specificity (KSF, n = 8). Compared to controls, WT_PH mice showed a significant elevation of the RV systolic pressure (P = 0.04) and RV functional impairment including increased basal RV (P = 0.016) diameter or tricuspid annular plane systolic excursion (P = 0.008). In contrast, KO PH did not show such effects compared to controls (P = n.s.). In WT_PH mice treated with F8, haemodynamic and echocardiographic parameters were significantly improved compared to untreated WT_PH mice or those treated with the KSF antibody (P < 0.05). On the microscopic level, KO_PH mice showed significantly less tissue damage compared to the WT_PH mice (P = 0.008). Furthermore, lung tissue damage could significantly be reduced after F8 treatment (P = 0.04). Additionally, these findings could be verified in the Sugen 5416/hypoxia mouse model, in which F8 significantly improved echocardiographic, haemodynamic, and histologic parameters. CONCLUSION ED-A+ Fn is of crucial importance for PH pathogenesis representing a promising therapeutic target in PH. We here show a novel therapeutic approach using antibody-mediated functional blockade of ED-A+ Fn capable of attenuating and partially reversing PH-associated tissue remodelling.
Collapse
MESH Headings
- Animals
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/immunology
- Fibronectins/metabolism
- Fibronectins/genetics
- Mice, Knockout
- Disease Models, Animal
- Monocrotaline
- Ventricular Function, Right/drug effects
- Ventricular Remodeling/drug effects
- Mice, Inbred C57BL
- Pyrimidines/pharmacology
- Pulmonary Artery/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Male
- Endothelin Receptor Antagonists/pharmacology
- Vascular Remodeling/drug effects
- Antibodies, Blocking/pharmacology
- Antibodies, Monoclonal/pharmacology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/pathology
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Isabell Singerer
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Cardiovascular Center Rotenburg, Klinikum Hersfeld-Rotenburg, Heinz-Meise-Str. 100, 36199 Rotenburg an der Fulda, Germany
| | - Laura Tempel
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Katja Gruen
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Judith Heiß
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Clara Gutte
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | | | - Andrea Schrepper
- Department of Cardiothoracic Surgery, University Hospital Jena, Jena, Germany
| | - Reinhard Bauer
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, University Hospital Jena, Jena, Germany
| | - Alexander Berndt
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | | | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Cardiovascular Center Rotenburg, Klinikum Hersfeld-Rotenburg, Heinz-Meise-Str. 100, 36199 Rotenburg an der Fulda, Germany
| |
Collapse
|
2
|
Wu G, Liu Z, Mu C, Song D, Wang J, Meng X, Li Z, Qing H, Dong Y, Xie HY, Pang DW. Enhanced Proliferation of Visualizable Mesenchymal Stem Cell-Platelet Hybrid Cell for Versatile Intracerebral Hemorrhage Treatment. ACS NANO 2023; 17:7352-7365. [PMID: 37037487 DOI: 10.1021/acsnano.2c11329] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The intrinsic features and functions of platelets and mesenchymal stem cells (MSCs) indicate their great potential in the treatment of intracerebral hemorrhage (ICH). However, neither of them can completely overcome ICH because of the stealth process and the complex pathology of ICH. Here, we fabricate hybrid cells for versatile and highly efficient ICH therapy by fusing MSCs with platelets and loading with lysophosphatidic acid-modified PbS quantum dots (LPA-QDs). The obtained LPA-QDs@FCs (FCs = fusion cells) not only inherit the capabilities of both platelets and MSCs but also exhibit clearly enhanced proliferation activated by LPA. After systemic administration, many proliferating LPA-QDs@FCs rapidly accumulate in ICH areas for responding to the vascular damage and inflammation and then efficiently prevent both the primary and secondary injuries of ICH but with no obvious side effects. Moreover, the treatment process can be tracked by near-infrared II fluorescence imaging with highly spatiotemporal resolution, providing a promising solution for ICH therapy.
Collapse
Affiliation(s)
- Guanghao Wu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhenya Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
| | - Changwen Mu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Da Song
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaxin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, P. R. China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, P. R. China
| | - Ziyuan Li
- Department of Biomedical Engineering, Peking University, Beijing 100871, P. R. China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yuping Dong
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
3
|
Oliveira MC, Correia JDG. Clinical application of radioiodinated antibodies: where are we? Clin Transl Imaging 2022. [DOI: 10.1007/s40336-021-00477-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
4
|
Serum Liberation of Fetal Fibronectin Variants in Patients with Pulmonary Hypertension: ED-A + Fn as Promising Novel Biomarker of Pulmonary Vascular and Right Ventricular Myocardial Remodeling. J Clin Med 2021; 10:jcm10122559. [PMID: 34207881 PMCID: PMC8229629 DOI: 10.3390/jcm10122559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
Background and Aims: Pulmonary Hypertension (PH) represents an aetiologically and clinically heterogeneous disorder accompanied by a severely impaired prognosis. Key steps of PH pathogenesis are vascular and right ventricular myocardial remodelling entailing the re-occurrence of fetal variants of the cell adhesion modulating protein fibronectin (Fn) being virtually absent in healthy adult tissues. These variants are liberated into circulation and are therefore qualified as excellent novel serum biomarkers. Moreover, these molecules might serve as promising therapeutic targets. The current study was aimed at quantifying the serum levels of two functionally important fetal Fn variants (ED-A+ and ED-B+ Fn) in patients suffering from PH due to different aetiologies compared to healthy controls. Methods: Serum levels of ED-A+ and ED-B+ Fn were quantified using novel ELISA protocols established and validated in our group in 80 PH patients and 40 controls. Results were analysed with respect to clinical, laboratory, echocardiographic and functional parameters. Results: Serum levels of ED-A+ Fn (p = 0.001) but not ED-B+ Fn (p = 0.722) were significantly increased in PH patients compared to healthy controls. Thus, the following analyses were performed only for ED-A+ Fn. When dividing PH patients into different aetiological groups according to current ESC guidelines, the increase in ED-A+ Fn in PH patients compared to controls remained significant for group 1 (p = 0.032), 2 (p = 0.007) and 3 (p = 0.001) but not for group 4 (p = 0.156). Correlation analysis revealed a significant relation between ED-A+ Fn and brain natriuretic peptide (BNP) (r = 0.310; p = 0.002), six minutes’ walk test (r = −0.275; p = 0.02) and systolic pulmonary artery pressure (PAPsys) (r = 0.364; p < 0.001). By logistic regression analysis (backward elimination WALD) including a variety of potentially relevant patients’ characteristics, only chronic kidney disease (CKD) (OR: 8.866; CI: 1.779–44.187; p = 0.008), C reactive protein (CRP) (OR: 1.194; CI: 1.011–1.410; p = 0.037) and ED-A+ Fn (OR: 1.045; CI: 1.011–1.080; p = 0.009) could be identified as independent predictors of the presence of PH. Conclusions: Against the background of our results, ED-A+ Fn could serve as a promising novel biomarker of PH with potential value for initial diagnosis and aetiological differentiation. Moreover, it might contribute to more precise risk stratification of PH patients. Beyond that, the future role of ED-A+ Fn as a therapeutic target has to be evaluated in further studies.
Collapse
|
5
|
Ferrari M, Onuoha SC, Fossati-Jimack L, Nerviani A, Alves PL, Pagani S, Deantonio C, Colombo F, Santoro C, Sblattero D, Pitzalis C. Novel Bispecific Antibody for Synovial-Specific Target Delivery of Anti-TNF Therapy in Rheumatoid Arthritis. Front Immunol 2021; 12:640070. [PMID: 33679801 PMCID: PMC7933454 DOI: 10.3389/fimmu.2021.640070] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/26/2021] [Indexed: 12/29/2022] Open
Abstract
Biologic drugs, especially anti-TNF, are considered as the gold standard therapy in rheumatoid arthritis. However, non-uniform efficacy, incidence of infections, and high costs are major concerns. Novel tissue-specific agents may overcome the current limitations of systemic administration, providing improved potency, and safety. We developed a bispecific antibody (BsAb), combining human arthritic joint targeting, via the synovial-specific single-chain variable fragment (scFv)-A7 antibody, and TNFα neutralization, via the scFv-anti-TNFα of adalimumab, with the binding/blocking capacity comparable to adalimumab -immunoglobulin G (IgG). Tissue-targeting capacity of the BsAb was confirmed on the human arthritic synovium in vitro and in a synovium xenograft Severe combined immune deficient (SCID) mouse model. Peak graft accumulation occurred at 48 h after injection with sustained levels over adalimumab-IgG for 7 days and increased therapeutic effect, efficiently decreasing tissue cellularity, and markers of inflammation with higher potency compared to the standard treatment. This study provides the first description of a BsAb capable of drug delivery, specifically to the disease tissue, and a strong evidence of improved therapeutic effect on the human arthritic synovium, with applications to other existing biologics.
Collapse
Affiliation(s)
- Mathieu Ferrari
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Shimobi C Onuoha
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Liliane Fossati-Jimack
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alessandra Nerviani
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Pedro L Alves
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Sara Pagani
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Cecilia Deantonio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases, University of Eastern Piedmont, Novara, Italy
| | - Federico Colombo
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Claudio Santoro
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases, University of Eastern Piedmont, Novara, Italy
| | | | - Costantino Pitzalis
- Department of Experimental Medicine and Rheumatology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
6
|
Gouyou B, Ongaro T, Cazzamalli S, De Luca R, Kerschenmeyer A, Valet P, Villa A, Neri D, Matasci M. Antibody-based delivery of interleukin-9 to neovascular structures: Therapeutic evaluation in cancer and arthritis. Exp Biol Med (Maywood) 2021; 246:940-951. [PMID: 33475433 DOI: 10.1177/1535370220981578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interleukin-9 is a cytokine with multiple functions, including the ability to activate group 2 innate lymphoid cells, which has been postulated to be therapeutically active in mouse models of arthritis. Similarly, interleukin-9 has been suggested to play an important role in tumor immunity. Here, we describe the cloning, expression, and characterization of three fusion proteins based on murine interleukin-9 and the F8 antibody, specific to the alternatively spliced EDA domain of fibronectin. EDA is strongly expressed in cancer and in various arthritic conditions, while being undetectable in the majority of healthy organs. Interleukin-9-based fusion proteins with an irrelevant antibody specific to hen egg lysozyme served as negative control in our study. The fusion proteins were characterized by quantitative biodistribution analysis in tumor-bearing mice using radioiodinated protein preparations. The highest tumor uptake and best tumor:organ ratios were observed for a format, in which the interleukin-9 moiety was flanked by two units of the F8 antibody in single-chain Fv format. Biological activity of interleukin-9 was retained when the payload was fused to antibodies. However, the targeted delivery of interleukin-9 to the disease site resulted in a modest anti-tumor activity in three different murine models of cancer (K1735M2, CT26, and F9), while no therapeutic benefit was observed in a collagen induced model of arthritis. Collectively, these results confirm the possibility to deliver interleukin-9 to the site of disease but cast doubts about the alleged therapeutic activity of this cytokine in cancer and arthritis, which has been postulated in previous publications.
Collapse
Affiliation(s)
| | - Tiziano Ongaro
- Philochem AG, Libernstrasse 3, Otelfingen 8112, Switzerland
| | | | | | | | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Cedex 4, Toulouse 31432, France
| | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich 8093, Switzerland
| | - Mattia Matasci
- Philochem AG, Libernstrasse 3, Otelfingen 8112, Switzerland
| |
Collapse
|
7
|
Franz M, Grün K, Betge S, Rohm I, Ndongson-Dongmo B, Bauer R, Schulze PC, Lichtenauer M, Petersen I, Neri D, Berndt A, Jung C. Lung tissue remodelling in MCT-induced pulmonary hypertension: a proposal for a novel scoring system and changes in extracellular matrix and fibrosis associated gene expression. Oncotarget 2018; 7:81241-81254. [PMID: 27835899 PMCID: PMC5348389 DOI: 10.18632/oncotarget.13220] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/27/2016] [Indexed: 11/25/2022] Open
Abstract
Pulmonary hypertension (PH) is associated with vasoconstriction and remodelling. We studied lung tissue remodelling in a rat model of PH with special focus on histology and extracellular matrix (ECM) remodelling. After induction of PH by monocrotaline, lung tissue was analysed histologically, by gene expression analysis and immunofluorescence labelling of ED-A domain containing fibronectin (ED-A+ Fn), B domain containing tenascin-C (B+ Tn-C) as well as alpha-smooth muscle actin (α-SMA). Serum concentrations of ED-A+ Fn were determined by ELISA. Systolic right ventricular pressure (RVPsys) values were significantly elevated in PH (n = 18; 75 ± 26.4 mmHg) compared to controls (n = 10; 29 ± 19.3 mmHg; p = 0.015). The histological sum-score was significantly increased in PH (8.0 ± 2.2) compared to controls (2.5 ± 1.6; p < 0.001). Gene expression analysis revealed relevant induction of several key genes of extracellular matrix remodelling. Increased protein deposition of ED-A+ Fn but not of B+ Tn-C and α-SMA in lung tissue was found in PH (2.88 ± 3.19 area%) compared to controls (1.32 ± 0.16 area%; p = 0.030). Serum levels of ED-A+ Fn were significantly higher in PH (p = 0.007) positively correlating with RVPsys (r = 0.618, p = 0.019). We here present a novel histological scoring system to assess lung tissue remodelling in PH. Gene expression analysis revealed induction of candidate genes involved in collagen matrix turnover, fibrosis and vascular remodelling. The stable increased tissue deposition of ED-A+ Fn in PH as well as its dynamics in serum suggests a role as a promising novel biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Marcus Franz
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Katja Grün
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Stefan Betge
- Department of Angiology, Cardiovascular Center Bad Bevensen, Bad Bevensen, Germany
| | - Ilonka Rohm
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Bernadin Ndongson-Dongmo
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | | | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Iver Petersen
- Institute of Pathology, Jena University Hospital, Jena, Germany
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | - Christian Jung
- Department of Internal Medicine, Division of Cardiology, Pulmonology and Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
8
|
Rohm I, Grün K, Müller LM, Kretzschmar D, Fritzenwanger M, Yilmaz A, Lauten A, Jung C, Schulze PC, Berndt A, Franz M. Increased Serum Levels of Fetal Tenascin-C Variants in Patients with Pulmonary Hypertension: Novel Biomarkers Reflecting Vascular Remodeling and Right Ventricular Dysfunction? Int J Mol Sci 2017; 18:ijms18112371. [PMID: 29117120 PMCID: PMC5713340 DOI: 10.3390/ijms18112371] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 10/31/2017] [Accepted: 11/04/2017] [Indexed: 11/16/2022] Open
Abstract
Pulmonary vascular remodeling is a pathophysiological feature that common to all classes of pulmonary hypertension (PH) and right ventricular dysfunction, which is the major prognosis-limiting factor. Vascular, as well as cardiac tissue remodeling are associated with a re-expression of fetal variants of cellular adhesion proteins, including tenascin-C (Tn-C). We analyzed circulating levels of the fetal Tn-C splicing variants B⁺ and C⁺ Tn-C in serum of PH patients to evaluate their potential as novel biomarkers reflecting vascular remodeling and right ventricular dysfunction. Serum concentrations of B⁺ and C⁺ Tn-C were determined in 80 PH patients and were compared to 40 healthy controls by enzyme-linked immunosorbent assay. Clinical, laboratory, echocardiographic, and functional data were correlated with Tn-C levels. Serum concentrations of both Tn-C variants were significantly elevated in patients with PH (p < 0.05). Significant correlations could be observed between Tn-C and echocardiographic parameters, including systolic pulmonary artery pressure (B⁺ Tn-C: r = 0.31, p < 0.001, C⁺ Tn-C: r = 0.26, p = 0.006) and right atrial area (B⁺ Tn-C: r = 0.46, p < 0.001, C⁺ Tn-C: r = 0.49, p < 0.001), and laboratory values like BNP (B⁺ Tn-C: r = 0.45, p < 0.001, C⁺ Tn-C: r = 0.42, p < 0.001). An inverse correlation was observed between Tn-C variants and 6-minute walk distance as a functional parameter (B⁺ Tn-C: r = -0.54, p < 0.001, C⁺ Tn-C: r = -0.43, p < 0.001). In a multivariate analysis, B⁺ Tn-C, but not C⁺ Tn-C, was found to be an independent predictor of pulmonary hypertension. Both fetal Tn-C variants may represent novel biomarkers that are capable of estimating both pulmonary vascular remodeling and right ventricular load. The potential beneficial impact of Tn-C variants for risk stratification in patients with PH needs further investigation.
Collapse
Affiliation(s)
- Ilonka Rohm
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| | - Katja Grün
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| | - Linda Marleen Müller
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| | - Michael Fritzenwanger
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| | - Atilla Yilmaz
- Department of Internal Medicine II, Division of Cardiology, Elisabeth Klinikum Schmalkalden, 98574 Schmalkalden, Germany.
| | - Alexander Lauten
- Department of Cardiology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Christian Jung
- Department of Internal Medicine, Division of Cardiology, University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany.
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| | - Alexander Berndt
- Institute of Pathology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany.
| | - Marcus Franz
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany.
| |
Collapse
|
9
|
Lv L, Jiang Y, Liu X, Wang B, Lv W, Zhao Y, Shi H, Hu Q, Xin H, Xu Q, Gu Z. Enhanced Antiglioblastoma Efficacy of Neovasculature and Glioma Cells Dual Targeted Nanoparticles. Mol Pharm 2016; 13:3506-3517. [DOI: 10.1021/acs.molpharmaceut.6b00523] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Lingyan Lv
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Department
of Pharmacy, Zhangjiagang Hospital of Traditional Chinese Medicine, Affiliated Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Yan Jiang
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xin Liu
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Baoyan Wang
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wei Lv
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yue Zhao
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Huihui Shi
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Quanyin Hu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Division
of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery,
Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Hongliang Xin
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qunwei Xu
- Department
of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhen Gu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Division
of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery,
Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
10
|
Detection of Soluble ED-A(+) Fibronectin and Evaluation as Novel Serum Biomarker for Cardiac Tissue Remodeling. DISEASE MARKERS 2016; 2016:3695454. [PMID: 27635109 PMCID: PMC5007333 DOI: 10.1155/2016/3695454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/10/2016] [Indexed: 01/22/2023]
Abstract
Background and Aims. Fibronectin containing the extra domain A (ED-A+ Fn) was proven to serve as a valuable biomarker for cardiac remodeling. The study was aimed at establishing an ELISA to determine ED-A+ Fn in serum of heart failure patients. Methods. ED-A+ Fn was quantified in serum samples from 114 heart failure patients due to ischemic (ICM, n = 44) and dilated (DCM, n = 39) cardiomyopathy as well as hypertensive heart disease (HHD, n = 31) compared to healthy controls (n = 12). Results. In comparison to healthy volunteers, heart failure patients showed significantly increased levels of ED-A+ Fn (p < 0.001). In particular in ICM patients there were significant associations between ED-A+ Fn serum levels and clinical parameters, for example, increased levels with rising NYHA class (p = 0.013), a negative correlation with left ventricular ejection fraction (p = 0.026, r: −0.353), a positive correlation with left atrial diameter (p = 0.008, r: 0.431), and a strong positive correlation with systolic pulmonary artery pressure (p = 0.002, r: 0.485). In multivariate analysis, ED-A+ Fn was identified as an independent predictor of an ischemic heart failure etiology. Conclusions. The current study could clearly show that ED-A+ Fn is a promising biomarker in cardiovascular diseases, especially in heart failure patients due to an ICM. We presented a valid ELISA method, which could be applied for further studies investigating the value of ED-A+ Fn.
Collapse
|
11
|
Acker G, Palumbo A, Neri D, Vajkoczy P, Czabanka M. F8-SIP mediated targeted photodynamic therapy leads to microvascular dysfunction and reduced glioma growth. J Neurooncol 2016; 129:33-8. [PMID: 27188647 DOI: 10.1007/s11060-016-2143-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
The extra domain A (ED A) of fibronectin has been identified as a tumor vessel specific neovascular marker in glioma. Antibody based vascular targeting against ED A of fibronectin allows precise accumulation of photosensitizer in glioma microvasculature and thereby promises to overcome drawbacks of current photodynamic therapy (PDT) for glioma treatment. Our aim was to characterize microcirculatory consequences of F8-small immunoprotein (SIP) mediated PDT by intravital microscopy (IVM) and to analyze the effects on glioma growth. For IVM SF126 glioma cells were implanted into dorsal skinfold-chamber of nude mice. PDT was performed after intravenous injection of photosensitizer (PS)-coupled F8-SIP or PBS (n = 4). IVM was performed before and after PDT for 4 days. Analysis included total and functional (TVD, FVD) vessel densities, perfusion index (PI), microvascular permeability and blood flow rate (Q). To assess tumor growth SF126 glioma cells were implanted subcutaneously. PDT was performed as a single and repetitive treatment after PS-F8-SIP injection (n = 5). Subcutaneous tumors were treated after uncoupled F8-SIP injection as control group (n = 5). PDT induced microvascular stasis and thrombosis with reduced FVD (24 h: 115.98 ± 0.7 vs. 200.8 ± 61.9 cm/cm(2)) and PI (39 ± 11 vs. 70 ± 10 %), whereas TVD was not altered (298 ± 39.2 vs. 278.2 ± 51 cm/cm(2)). Microvascular dysfunction recovered 4 days after treatment. Microvascular dysfunction led to a temporary reduction of glioma growth in the first 48 h after treatment with complete recovery 5 days after treatment. Repetitive PDT resulted in sustained reduction of tumor growth. F8-SIP mediated PDT leads to microvascular dysfunction and reduced glioma growth in a preclinical glioma model with recovery of microcirculation 4 days after treatment. Repetitive application of PDT overcomes microvascular recovery and leads to prolonged antiglioma effects.
Collapse
Affiliation(s)
- G Acker
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - A Palumbo
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH, Zurich, Switzerland
| | - D Neri
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH, Zurich, Switzerland
| | - P Vajkoczy
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - M Czabanka
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
12
|
Franz M, Jung C, Lauten A, Figulla HR, Berndt A. Tenascin-C in cardiovascular remodeling: potential impact for diagnosis, prognosis estimation and targeted therapy. Cell Adh Migr 2015; 9:90-5. [PMID: 25562641 DOI: 10.1080/19336918.2014.1000075] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Fetal variants of tenascin-C are not expressed in healthy adult myocardium. But, there is a relevant re-occurrence during pathologic cardiac tissue and vascular remodeling. Thus, these molecules, in particular B and C domain containing tenascin-C, might qualify as promising novel biomarkers for diagnosis and prognosis estimation. Since a stable extracellular deposition of fetal tenascin-C variants is present in diseased cardiac tissue, the molecules are excellent target structures for antibody-based delivery of diagnostic (e.g., radionuclides) or therapeutic (bioactive payloads) agents directly to the site of disease. Against the background that fetal tenascin-C variants are functionally involved in cardiovascular tissue remodeling, therapeutic functional blocking strategies could be experimentally tested in the future.
Collapse
Affiliation(s)
- Marcus Franz
- a Department of Internal Medicine I; Jena University Hospital ; Jena , Germany
| | | | | | | | | |
Collapse
|
13
|
Feng X, Gao X, Kang T, Jiang D, Yao J, Jing Y, Song Q, Jiang X, Liang J, Chen J. Mammary-Derived Growth Inhibitor Targeting Peptide-Modified PEG–PLA Nanoparticles for Enhanced Targeted Glioblastoma Therapy. Bioconjug Chem 2015. [DOI: 10.1021/acs.bioconjchem.5b00379] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xingye Feng
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Xiaoling Gao
- Department
of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China
| | - Ting Kang
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Di Jiang
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Jianhui Yao
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Yixian Jing
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Qingxiang Song
- Department
of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China
| | - Xinguo Jiang
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Jianying Liang
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Jun Chen
- Key
Laboratory of Smart Drug Delivery, Ministry of Education, School of
Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, PR China
| |
Collapse
|
14
|
Gu G, Hu Q, Feng X, Gao X, Menglin J, Kang T, Jiang D, Song Q, Chen H, Chen J. PEG-PLA nanoparticles modified with APTEDB peptide for enhanced anti-angiogenic and anti-glioma therapy. Biomaterials 2014; 35:8215-26. [PMID: 24974009 DOI: 10.1016/j.biomaterials.2014.06.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022]
Abstract
Tumor neovasculature and tumor cells dual-targeting chemotherapy can not only destroy the tumor neovasculature, cut off the supply of nutrition and starve the tumor cells, but also directly kill tumor cells, holding great potential in overcoming the drawbacks of anti-angiogenic therapy only and improving the anti-glioma efficacy. In the present study, by taking advantage of the specific expression of fibronectin extra domain B (EDB) on both glioma neovasculature endothelial cells and glioma cells, we constructed EDB-targeted peptide APTEDB-modified PEG-PLA nanoparticles (APT-NP) for paclitaxel (PTX) loading to enable tumor neovasculature and tumor cells dual-targeting chemotherapy. PTX-loaded APT-NP showed satisfactory encapsulated efficiency, loading capacity and size distribution. In human umbilical vein endothelial cells, APT-NP exhibited significantly elevated cellular accumulation via energy-dependent, caveolae and lipid raft-involved endocytosis, and improved PTX-induced apoptosis therein. Both in vitro tube formation assay and in vivo matrigel angiogenesis analysis confirmed that APT-NP significantly improved the antiangiogenic ability of PTX. In U87MG cells, APT-NP showed elevated cellular internalization and also enhanced the cytotoxicity of the loaded PTX. Following intravenous administration, as shown by both in vivo live animal imaging and tissue distribution analysis, APT-NP achieved a much higher and specific accumulation within the glioma. As a result, APT-NP-PTX exhibited improved anti-glioma efficacy over unmodified nanoparticles and Taxol(®) in both subcutaneous and intracranial U87MG xenograft models. These findings collectively indicated that APTEDB-modified nanoparticles might serve as a promising nanocarrier for tumor cells and neovasculature dual-targeting chemotherapy and hold great potential in improving the efficacy anti-glioma therapy.
Collapse
Affiliation(s)
- Guangzhi Gu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China; Shanghai Institute for Food and Drug Control (SIFDC), 479 Futexi First Road, Shanghai 200131, PR China
| | - Quanyin Hu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China
| | - Xingye Feng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China
| | - Xiaoling Gao
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, PR China
| | - Jiang Menglin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China
| | - Ting Kang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China
| | - Di Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China
| | - Qingxiang Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, PR China
| | - Hongzhuan Chen
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, PR China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, PR China.
| |
Collapse
|
15
|
Hemmerle T, Neri D. The dose-dependent tumor targeting of antibody-IFNγ fusion proteins reveals an unexpected receptor-trapping mechanism in vivo. Cancer Immunol Res 2014; 2:559-67. [PMID: 24795141 DOI: 10.1158/2326-6066.cir-13-0182] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cytokines often display substantial toxicities at low concentrations, preventing their escalation for therapeutic treatment of cancer. Fusion proteins comprising cytokines and recombinant antibodies may improve the anticancer activity of proinflammatory cytokines. Murine IFNγ was appended in the diabody format at the C-terminus of the F8 antibody, generating the F8-IFNγ fusion protein. The F8 antibody is specific for the extra-domain A (EDA) of fibronectin, a tumor-associated antigen that is expressed in the vasculature and stroma of almost all tumor types. Tumor-targeting properties were measured in vivo using a radioiodinated preparation of the fusion protein. Therapy experiments were performed in three syngeneic murine models of cancer [F9 teratocarcinoma, WEHI-164 fibrosarcoma, and Lewis lung carcinoma (LLC)]. F8-IFNγ retained the biologic activity of both the antibody and the cytokine moiety in vitro, but, unlike the parental F8 antibody, it did not preferentially localize to the tumors in vivo. However, when unlabeled F8-IFNγ was administered before radioiodinated F8-IFNγ, a selective accumulation at the tumor site was observed. F8-IFNγ showed dose-dependent anticancer activity with a clear superiority over untargeted recombinant IFNγ. The anticancer activity was potentiated by combining with F8-IL4 without additional toxicities, whereas combination of F8-IFNγ with F8-TNF was lethal in all mice. Unlike other antibody-cytokine fusions, the use of IFNγ as payload for anticancer therapy is associated with a receptor-trapping mechanism, which can be overcome by the administration of a sufficiently large amount of the fusion protein without any detectable toxicity at the doses used.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/pharmacology
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Biological Availability
- Biomarkers, Tumor/metabolism
- Cricetinae
- Dose-Response Relationship, Immunologic
- Female
- Fibronectins/metabolism
- Interferon-gamma/immunology
- Male
- Mice, Inbred C57BL
- Neoplasms/metabolism
- Neoplasms/therapy
- Receptors, Interferon/immunology
- Recombinant Fusion Proteins/pharmacokinetics
- Recombinant Fusion Proteins/pharmacology
- Tumor Cells, Cultured
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Teresa Hemmerle
- Authors' Affiliation: Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Dario Neri
- Authors' Affiliation: Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| |
Collapse
|
16
|
Mullen L, Adams G, Layward L, Vessillier S, Annenkov A, Mittal G, Rigby A, Sclanders M, Baker D, Gould D, Chernajovsky Y. Latent cytokines for targeted therapy of inflammatory disorders. Expert Opin Drug Deliv 2013; 11:101-10. [PMID: 24294995 DOI: 10.1517/17425247.2014.863872] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The use of cytokines as therapeutic agents is important, given their potent biological effects. However, this very potency, coupled with the pleiotropic nature and short half-life of these molecules, has limited their therapeutic use. Strategies to increase the half-life and to decrease toxicity are necessary to allow effective treatment with these molecules. AREAS COVERED A number of strategies are used to overcome the natural limitations of cytokines, including PEGylation, encapsulation in liposomes, fusion to targeting peptides or antibodies and latent cytokines. Latent cytokines are engineered using the latency-associated peptide of transforming growth factor-β to produce therapeutic cytokines/peptides that are released only at the site of disease by cleavage with disease-induced matrix metalloproteinases. The principles underlying the latent cytokine technology are described and are compared to other methods of cytokine delivery. The potential of this technology for developing novel therapeutic strategies for the treatment of diseases with an inflammatory-mediated component is discussed. EXPERT OPINION Methods of therapeutic cytokine delivery are addressed. The latent cytokine technology holds significant advantages over other methods of drug delivery by providing simultaneously increased half-life and localised drug delivery without systemic effects. Cytokines that failed clinical trials should be reassessed using this delivery system.
Collapse
Affiliation(s)
- Lisa Mullen
- Queen Mary University of London, William Harvey Research Institute, Bone and Joint Research Unit, Barts and The London Medical School , Charterhouse Square , London EC1M 6BQ, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Qin S, Fite BZ, Gagnon MKJ, Seo JW, Curry FR, Thorsen F, Ferrara KW. A physiological perspective on the use of imaging to assess the in vivo delivery of therapeutics. Ann Biomed Eng 2013; 42:280-98. [PMID: 24018607 DOI: 10.1007/s10439-013-0895-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/14/2013] [Indexed: 12/25/2022]
Abstract
Our goal is to provide a physiological perspective on the use of imaging to optimize and monitor the accumulation of nanotherapeutics within target tissues, with an emphasis on evaluating the pharmacokinetics of organic particles. Positron emission tomography (PET), magnetic resonance imaging (MRI) and ultrasound technologies, as well as methods to label nanotherapeutic constructs, have created tremendous opportunities for preclinical optimization of therapeutics and for personalized treatments in challenging disease states. Within the methodology summarized here, the accumulation of the construct is estimated directly from the image intensity. Particle extravasation is then estimated based on classical physiological measures. Specifically, the transport of nanotherapeutics is described using the concept of apparent permeability, which is defined as the net flux of solute across a blood vessel wall per unit surface area of the blood vessel and per unit solute concentration difference across the blood vessel wall. The apparent permeability to small molecule MRI constructs is accurately shown to be far larger than that estimated for proteins such as albumin or nanoconstructs such as liposomes. Further, the quantitative measurements of vascular permeability are shown to facilitate detection of the transition from a pre-malignant to a malignant cancer and to quantify the delivery enhancement resulting from interventions such as ultrasound. While PET-based estimates facilitate quantitative comparisons of many constructs, high field MRI proves useful in the visualization of model drugs within small lesions and in the evaluation of the release and intracellular trafficking of nanoparticles and cargo.
Collapse
Affiliation(s)
- Shengping Qin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA,
| | | | | | | | | | | | | |
Collapse
|
18
|
Sapra P, Shor B. Monoclonal antibody-based therapies in cancer: advances and challenges. Pharmacol Ther 2013; 138:452-69. [PMID: 23507041 DOI: 10.1016/j.pharmthera.2013.03.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/19/2013] [Indexed: 12/25/2022]
Abstract
Conventional anticancer therapeutics often suffer from lack of specificity, resulting in toxicities to normal healthy tissues and poor therapeutic index. Antibody-mediated delivery of anticancer drugs or toxins to tumor cells through tumor selective or overexpressed antigens is progressively being recognized as an effective strategy for increasing the therapeutic index of anticancer drugs. In this review we focus on three therapeutic modalities in the field of antibody-mediated targeting, including antibody-drug conjugates (ADCs), immunotoxins (ITs) and immunoliposomes (ILs). Design considerations for development of each of the above therapeutic modalities are discussed. Furthermore, an overview of ADCs, ITs or ILs approved for use in clinical oncology and those currently in clinical development is provided. Challenges encountered by the field of antibody-based targeting are discussed and concepts around development of the next generation of antibody therapeutics are presented.
Collapse
Affiliation(s)
- Puja Sapra
- Bioconjugates Discovery and Development, Oncology Research Unit, Pfizer Worldwide Research and Development, 401 North Middletown Road, Pearl River, NY, 10965, USA.
| | | |
Collapse
|
19
|
Fibronectin extra domain B-specific aptide conjugated nanoparticles for targeted cancer imaging. J Control Release 2012; 163:111-8. [DOI: 10.1016/j.jconrel.2012.08.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 07/26/2012] [Accepted: 08/26/2012] [Indexed: 11/22/2022]
|
20
|
Franz M, Neri D, Berndt A. Chronic cardiac allograft rejection: critical role of ED-A(+) fibronectin and implications for targeted therapy strategies. J Pathol 2012; 226:557-61. [PMID: 22102444 DOI: 10.1002/path.3968] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic cardiac allograft rejection is characterized by cardiac allograft vasculopathy (CAV) and cardiac interstitial fibrosis (CIF) causing severe long-term complications after heart transplantation and determining allograft function and patients' prognosis. Until now, there have been no sufficient preventive or therapeutic strategies. CAV and CIF are accompanied by changes in the extracellular matrix, including re-expression of the fetal fibronectin splice variant known as ED-A(+) fibronectin. This molecule has been shown to be crucial for the development of myofibroblasts (MyoFbs) as the main cell type in CIF and for the activation of vascular smooth muscle cells (VSMCs) as the main cell type in CAV. Relevant re-expression and protein deposition of ED-A(+) fibronectin has been demonstrated in animal models of chronic rejection, with spatial association to CAV and CIF, and a quantitative correlation to the rejection grade. The paper by Booth et al published in this issue of The Journal of Pathology could prove for the first time the functional importance of ED-A(+) fibronectin for the development of CIF as a main component of chronic cardiac rejection. Thus, promising conclusions for the development of new diagnostic, preventive, and therapeutic strategies for chronic cardiac rejection focusing on ED-A(+) fibronectin can be suggested.
Collapse
Affiliation(s)
- Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Erlanger Allee 101, 07740 Jena, Germany.
| | | | | |
Collapse
|
21
|
Kamperidis P, Kamalati T, Ferrari M, Jones M, Garrood T, Smith MD, Diez-Posada S, Hughes C, Finucane C, Mather S, Nissim A, George AJT, Pitzalis C. Development of a novel recombinant biotherapeutic with applications in targeted therapy of human arthritis. ACTA ACUST UNITED AC 2012; 63:3758-67. [PMID: 21953304 DOI: 10.1002/art.30650] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To isolate recombinant antibodies with specificity for human arthritic synovium and to develop targeting reagents with joint-specific delivery capacity for therapeutic and/or diagnostic applications. METHODS In vivo single-chain Fv (scFv) antibody phage display screening using a human synovial xenograft model was used to isolate antibodies specific to the microvasculature of human arthritic synovium. Single-chain Fv antibody tissue-specific reactivity was assessed by immunostaining of synovial tissues from normal controls and from patients with rheumatoid arthritis and osteoarthritis, normal human tissue arrays, and tissues from other patients with inflammatory diseases displaying neovasculogenesis. In vivo scFv antibody tissue-specific targeting capacity was examined in the human synovial xenograft model using both (125)I-labeled and biotinylated antibody. RESULTS We isolated a novel recombinant human antibody, scFv A7, with specificity for the microvasculature of human arthritic synovium. We showed that in vivo, this antibody could efficiently target human synovial microvasculature in SCID mice transplanted with human arthritic synovial xenografts. Our results demonstrated that scFv A7 antibody had no reactivity with the microvasculature or with other cellular components found in a comprehensive range of normal human tissues including normal human synovium. Further, we showed that the reactivity of the scFv A7 antibody was not a common feature of neovasculogenesis associated with chronic inflammatory conditions. CONCLUSION Here we report for the first time the identification of an scFv antibody, A7, that specifically recognizes an epitope expressed in the microvasculature of human arthritic synovium and that has the potential to be developed as a joint-specific pharmaceutical.
Collapse
Affiliation(s)
- Panagiotis Kamperidis
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sapra P, Hooper AT, O'Donnell CJ, Gerber HP. Investigational antibody drug conjugates for solid tumors. Expert Opin Investig Drugs 2011; 20:1131-49. [DOI: 10.1517/13543784.2011.582866] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
23
|
Baldinger A, Brehm BR, Richter P, Bossert T, Gruen K, Hekmat K, Kosmehl H, Neri D, Figulla HR, Berndt A, Franz M. Comparative analysis of oncofetal fibronectin and tenascin-C expression in right atrial auricular and left ventricular human cardiac tissue from patients with coronary artery disease and aortic valve stenosis. Histochem Cell Biol 2011; 135:427-41. [PMID: 21479812 DOI: 10.1007/s00418-011-0809-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2011] [Indexed: 12/19/2022]
Abstract
Aortic valve stenosis (AVS) and coronary artery disease (CAD) are accompanied by changes in the cardiac extra cellular matrix (cECM) including the re-expression of oncofetal fibronectin (Fn) and tenascin-C (Tn-C) variants. Human antibodies against these variants are usable for targeted therapy. Aim of the study was the comparative analysis of cECM remodelling in tissue samples from right atrial auricle (RAA) and left ventricular septum (LVS). RAA and LVS specimens from 30 patients (17 × AVS; 13 × AVS+CAD) were analysed with respect to histological changes and ECM remodelling using PCR based ECM gene expression profiling. Re-expression of ED-A(+) Fn and A1(+) Tn-C was investigated on the mRNA and on the protein level. For immunofluorescence, human recombinant small immunoprotein (SIP) format antibodies were used. There was a positive correlation of the grade of histological changes in RAA and corresponding LVS samples (r = 0.695). ECM gene expression levels were higher in LVS compared to RAA. For 24 genes, a corresponding relevant (>2.5-fold) up- or down-regulation in RAA and LVS occurred. Using SIP antibodies, a positive correlation of protein deposition levels in RAA and corresponding LVS (r = 0.818) could be shown for ED-A(+) Fn. Cardiac tissue remodelling is likely a process involving the entire heart reflected by intra-individually comparable histology and cECM changes in RAA and LVS samples. ED-A(+) Fn might be an excellent target for an antibody-mediated delivery of diagnostic or therapeutic agents. The RAA is a valuable and representative tool to evaluate cardiac remodelling and to plan individualized therapy.
Collapse
Affiliation(s)
- Anja Baldinger
- Department of Internal Medicine I, University Hospital of Jena, Erlanger Allee 101, 07740, Jena, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Turtoi A, De Pauw E, Castronovo V. Innovative proteomics for the discovery of systemically accessible cancer biomarkers suitable for imaging and targeted therapies. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:12-8. [PMID: 21224037 DOI: 10.1016/j.ajpath.2010.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 08/10/2010] [Accepted: 08/30/2010] [Indexed: 12/30/2022]
Abstract
The discovery of biomarkers that are readily accessible through the circulating blood and are selectively overexpressed in pathological tissues has become a major research objective, particularly in the field of oncology. Indisputably, this group of molecules has a high potential to serve as an innovative tool for effective imaging and targeted cancer therapy approaches. In this attractive therapeutic concept, specific cancer proteins are reached by intravenously administered ligands that are coupled to cytotoxic drugs. Such compounds are able to induce cancer destruction while sparing normal tissues. Owing to the performance of mass spectrometry technology, current high-throughput proteomic analysis allows for the identification of a high number of proteins that are differentially expressed in the cancerous tissues. However, such approaches provide no information regarding the effective accessibility of the >biomarkers and, therefore, the possibility for these discovered proteins to be targeted. To bypass this major limitation, which clearly slows the discovery of such biomarkers, innovative methodological strategies have been developed to enrich the clinical specimens before the mass spectrometry analysis. The focus is laid on the group of proteins that are necessarily located either at the exterior face of the plasma membrane or in the extracellular matrix. The present review addresses the current technologies meant for the discovery and analysis of accessible antigens from clinically relevant samples.
Collapse
Affiliation(s)
- Andrei Turtoi
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | | | | |
Collapse
|
25
|
Zhang JM, Shi XL, Zhu J, Wu YF. Inhibitory effects of a chimeric anti-VEGFR-2 Fab antibody on tumor growth and angiogenesis in an orthotopic xenograft mouse model of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2010; 18:3621-3626. [DOI: 10.11569/wcjd.v18.i34.3621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the inhibitory effects of a mouse chimeric anti-VEGFR-2 Fab antibody on tumor growth and angiogenesis in an orthotopic xenograft mouse model of hepatocellular carcinoma.
METHODS: The antigen-binding activity of cFab antibody was assessed by ELISA. An orthotopic xenograft mouse model of hepatocellular carcinoma was created by direct injection of H22 cells into the liver parenchyma of ICR mice. Twenty-four ICR mice bearing orthotopic HCC were divided into two groups: control group (treated with normal saline) and cFab group (treated with cFab antibody). After the treatment, tumor weight and mouse survival time were compared between the two groups. Histological changes and microvessel density (MVD) in tumors were also evaluated.
RESULTS: The chimeric anti-VEGFR-2 Fab antibody exhibited a strong binding activity to VEGFR-2. Tumor size in the cFab group was significantly decreased compared with the control group (313.9 mm3 ± 41.9 mm3vs 635.4 mm3 ± 70.1 mm3, P < 0.05); and the mice of the cFab group had a significantly longer survival time (20.0 d vs 13.0 d, Long-rank test χ2 = 4.611, P = 0.032). The MVD in the cFab group was also significantly lower than that in the control group (34.48 ± 1.39 vs 9.56 ± 1.26,P < 0.05).
CONCLUSION: The chimeric anti-VEGFR-2 Fab antibody can inhibit tumor growth in an orthotopic xenograft mouse model of hepatocellular carcinoma possibly by inhibiting tumor angiogenesis.
Collapse
|
26
|
Antiangiogenic Activity of a Neutralizing Human Single-Chain Antibody Fragment against Fibroblast Growth Factor Receptor 1. Mol Cancer Ther 2010; 9:3244-53. [DOI: 10.1158/1535-7163.mct-10-0417] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Strassberger V, Fugmann T, Neri D, Roesli C. Chemical proteomic and bioinformatic strategies for the identification and quantification of vascular antigens in cancer. J Proteomics 2010; 73:1954-73. [DOI: 10.1016/j.jprot.2010.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 05/27/2010] [Accepted: 05/27/2010] [Indexed: 10/19/2022]
|
28
|
Franz M, Brehm BR, Richter P, Gruen K, Neri D, Kosmehl H, Hekmat K, Renner A, Gummert J, Figulla HR, Berndt A. Changes in extra cellular matrix remodelling and re-expression of fibronectin and tenascin-C splicing variants in human myocardial tissue of the right atrial auricle: implications for a targeted therapy of cardiovascular diseases using human SIP format antibodies. J Mol Histol 2010; 41:39-50. [PMID: 20232238 DOI: 10.1007/s10735-010-9260-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 02/26/2010] [Indexed: 11/29/2022]
Abstract
Cardiovascular diseases are accompanied by changes in the extracellular matrix (ECM) including the re-expression of fibronectin and tenascin-C splicing variants. Using human recombinant small immunoprotein (SIP) format antibodies, a molecular targeting of these proteins is of therapeutic interest. Tissue samples of the right atrial auricle from patients with coronary artery disease and valvular heart disease were analysed by PCR based ECM gene expression profiling. Moreover, the re-expression of fibronectin and tenascin-C splicing variants was investigated by immunofluoerescence labelling. We demonstrated changes in ECM gene expression depending on histological damage or underlying cardiac disease. An increased expression of fibronectin and tenascin-C mRNA in association to histological damage and in valvular heart disease compared to coronary artery disease could be shown. There was a distinct re-expression of ED-A containing fibronectin and A1 domain containing tenascin-C detectable with human recombinant SIP format antibodies in diseased myocardium. ED-A containing fibronectin showed a clear vessel positivity. For A1 domain containing tenascin-C, there was a particular positivity in areas of interstitial and perivascular fibrosis. Right atrial myocardial tissue is a valuable model to investigate cardiac ECM remodelling. Human recombinant SIP format antibodies usable for an antibody-mediated targeted delivery of drugs might offer completely new therapeutic options in cardiac diseases.
Collapse
Affiliation(s)
- Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Erlanger Allee 101, 07740, Jena, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sgier D, Zuberbuehler K, Pfaffen S, Neri D. Isolation and characterization of an inhibitory human monoclonal antibody specific to the urokinase-type plasminogen activator, uPA. Protein Eng Des Sel 2010; 23:261-9. [DOI: 10.1093/protein/gzp089] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
30
|
Abstract
The inhibition of angiogenesis represents a major step toward a more selective and better-tolerated therapy of cancer. An alternative way to take advantage of a tumor's absolute dependence on a functional neovasculature is illustrated by the strategy of "antibody-based vascular tumor targeting." This technology aims at the selective delivery of bioactive molecules to the tumor site by their conjugation to a carrier antibody reactive with a tumor-associated vascular antigen. A number of high-affinity monoclonal antibodies are nowadays available which have demonstrated a remarkable ability to selectively localize to the tumor vasculature. Indeed, some of them have already progressed from preclinical animal experiments to clinical studies in patients with cancer, acting as vehicles for the site-specific pharmacodelivery of proinflammatory cytokines or radionuclides.In this chapter, we present a selection of well-characterized markers of angiogenesis which have proven to be suitable targets for antibody-based vascular targeting approaches. Furthermore, different transcriptomic and proteomic methodologies for the discovery of novel vascular tumor markers are described. In the last two sections, we focus on the discussion of antibody-based vascular tumor targeting strategies for imaging and therapy applications in oncology.
Collapse
Affiliation(s)
- Christoph Schliemann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zürich, Zürich, Switzerland
| | | |
Collapse
|
31
|
Borgia B, Roesli C, Fugmann T, Schliemann C, Cesca M, Neri D, Giavazzi R. A proteomic approach for the identification of vascular markers of liver metastasis. Cancer Res 2009; 70:309-18. [PMID: 19996283 DOI: 10.1158/0008-5472.can-09-2939] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular proteins expressed at liver metastasis sites could serve as prognostic markers or as targets for pharmacodelivery applications. We employed a proteomic approach to define such proteins in three syngeneic mouse models of liver metastasis. Vascular structures were biotinylated in vivo by a terminal perfusion technique, followed by mass spectrometric analysis of accessible biotinylated proteins. In this manner, we identified 12 proteins for which expression was selectively associated with liver metastasis, confirming this association by tissue immunofluorescence or in vivo localization with radiolabeled antibodies. In summary, our findings identify vascular proteins that may have prognostic or drug-targeting use in addressing liver metastases, a common issue in many advanced cancers.
Collapse
Affiliation(s)
- Beatrice Borgia
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
32
|
Pfaffen S, Hemmerle T, Weber M, Neri D. Isolation and characterization of human monoclonal antibodies specific to MMP-1A, MMP-2 and MMP-3. Exp Cell Res 2009; 316:836-47. [PMID: 19913533 DOI: 10.1016/j.yexcr.2009.11.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 11/04/2009] [Accepted: 11/09/2009] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs), a group of more than 20 zinc-containing endopeptidases, are up-regulated in many diseases, but the use of MMP inhibitors for therapeutic purposes has often been disappointing, possibly for the limited specificity of the drugs used in clinical trials. In principle, individual MMPs could be specifically drugged by monoclonal antibodies, either by inhibition of their catalytic activity or by antibody-based pharmacodelivery strategies. In this article we describe the isolation and affinity maturation of recombinant antibodies (SP1, SP2, SP3) specific to the murine catalytic domains of MMP-1A, MMP-2 and MMP-3. These novel reagents allowed a systematic comparative immunofluorescence analysis of the expression patterns of their cognate antigens in a variety of healthy, cancerous and arthritic murine tissues. While all three MMPs were strongly expressed in tumor and arthritis specimens, MMP-1A was completely undetectable in the normal tissues tested, while MMP-2 and MMP-3 exhibited a weak expression in certain normal tissues (e.g., liver). The new antibodies may serve as building blocks for the development of antibody-based therapy strategies in mouse models of pathology.
Collapse
Affiliation(s)
- Stefanie Pfaffen
- ETH Zürich HCI G396, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Wolfgang-Pauli-Strasse 10, CH-8093 Zürich, Switzerland
| | | | | | | |
Collapse
|
33
|
Pedretti M, Rancic Z, Soltermann A, Herzog BA, Schliemann C, Lachat M, Neri D, Kaufmann PA. Comparative immunohistochemical staining of atherosclerotic plaques using F16, F8 and L19: Three clinical-grade fully human antibodies. Atherosclerosis 2009; 208:382-9. [PMID: 19699478 DOI: 10.1016/j.atherosclerosis.2009.07.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/29/2009] [Accepted: 07/23/2009] [Indexed: 01/04/2023]
Abstract
OBJECTIVE F16, F8 and L19 are three fully human monoclonal antibodies, specific to splice isoforms of tenascin-C and fibronectin, which stain sites of active tissue remodeling and which are currently in Phase I and II clinical trials as radio-immunoconjugates and immunocytokines in patients with cancer and arthritis. The characterization of atherosclerosis using these antibodies may open novel pharmacodelivery options for the imaging and treatment of cardiovascular conditions. It may also allow a better assessment of the corresponding immunoconjugates in polymorbid patients with atherosclerotic plaques. METHODS We performed a comparative immunohistochemical analysis with the F16, F8 and L19 antibodies in 28 freshly frozen human carotid plaques and in 11 normal arteries. Furthermore, we assessed the localization of the antibodies in relation to the infiltrating macrophages, vasa vasorum and Ki67-positive proliferating cells of the plaque. RESULTS The F16 antibody, specific to the extra-domain A1 of tenascin-C, stained plaques with a selective and intense pattern, while F8 and L19, specific to the EDA and EDB domains of fibronectin, respectively, exhibited a less selective and intense staining. In immunofluorescence, F16 was found to bind regions rich in macrophages, vasa vasorum and proliferating cells, while showing no detectable vs. weak staining of normal arteries and of quiescent plaque structures. CONCLUSION The human monoclonal antibody F16 stains areas of active tissue remodeling in atherosclerotic plaques and may thus deserve to be investigated as a suitable building block for the development of radiopharmaceuticals for plaque imaging or for the antibody-based targeted delivery of therapeutic agents to atherosclerotic lesions.
Collapse
Affiliation(s)
- Marta Pedretti
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Ahlskog JKJ, Schliemann C, Mårlind J, Qureshi U, Ammar A, Pedley RB, Neri D. Human monoclonal antibodies targeting carbonic anhydrase IX for the molecular imaging of hypoxic regions in solid tumours. Br J Cancer 2009; 101:645-57. [PMID: 19623173 PMCID: PMC2736829 DOI: 10.1038/sj.bjc.6605200] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background: Hypoxia, which is commonly observed in areas of primary tumours and of metastases, influences response to treatment. However, its characterisation has so far mainly been restricted to the ex vivo analysis of tumour sections using monoclonal antibodies specific to carbonic anhydrase IX (CA IX) or by pimonidazole staining, after the intravenous administration of this 2-nitroimidazole compound in experimental animal models. Methods: In this study, we describe the generation of high-affinity human monoclonal antibodies (A3 and CC7) specific to human CA IX, using phage technology. Results: These antibodies were able to stain CA IX ex vivo and to target the cognate antigen in vivo. In one of the two animal models of colorectal cancer studied (LS174T), CA IX imaging closely matched pimonidazole staining, with a preferential staining of tumour areas characterised by little vascularity and low perfusion. In contrast, in a second animal model (SW1222), distinct staining patterns were observed for pimonidazole and CA IX targeting. We observed a complementary pattern of tumour regions targeted in vivo by the clinical-stage vascular-targeting antibody L19 and the anti-CA IX antibody A3, indicating that a homogenous pattern of in vivo tumour targeting could be achieved by a combination of the two antibodies. Conclusion: The new human anti-CA IX antibodies are expected to be non-immunogenic in patients with cancer and may serve as broadly applicable reagents for the non-invasive imaging of hypoxia and for pharmacodelivery applications.
Collapse
Affiliation(s)
- J K J Ahlskog
- Department of Chemistry and Applied Biosciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, Zurich CH-8093, Switzerland
| | | | | | | | | | | | | |
Collapse
|
35
|
Roesli C, Borgia B, Schliemann C, Gunthert M, Wunderli-Allenspach H, Giavazzi R, Neri D. Comparative analysis of the membrane proteome of closely related metastatic and nonmetastatic tumor cells. Cancer Res 2009; 69:5406-14. [PMID: 19491280 DOI: 10.1158/0008-5472.can-08-0999] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The identification of proteins that are preferentially expressed on the membrane of metastatic tumor cells is of fundamental importance in cancer research. Here, we report the systematic comparison of the membrane proteome of two closely related murine teratocarcinoma cell lines (F9B9 and F9DR), of which only one (F9DR) is capable of forming liver metastases in vivo. The proteomic methodology used in this study featured the surface protein biotinylation on tumor cells followed by protein purification on streptavidin resin and relative quantification of corresponding tryptic peptides by mass spectrometric procedures. The study allowed the identification of 998 proteins and the determination of their relative abundance. Proteins previously known to be associated with metastatic spread were found to be either up-regulated (e.g., synaptojanin-2) or down-regulated (e.g., Ceacam1) in F9DR cells. A dramatic increase in abundance at the cell membrane was observed for a broad variety of proteins (e.g., high-mobility group protein B1), which were mainly thought to reside in intracellular compartments, a finding that was confirmed using confocal laser scanning microscopy and immunochemical analysis of cell cultures. Furthermore, we showed by microautoradiographic analysis that certain target proteins can readily be reached by intravenously administered radiolabeled antibodies. Finally, we showed that the most promising antigens for antibody-based pharmacodelivery approaches are strongly and selectively expressed on the surface of tumor cells in three different syngeneic mouse models of liver metastases. Taken together, our results indicate that the expression of intracellular proteins on the membrane of metastatic cells is a feature much more common than previously expected.
Collapse
Affiliation(s)
- Christoph Roesli
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
36
|
Tijink BM, Perk LR, Budde M, Stigter-van Walsum M, Visser GWM, Kloet RW, Dinkelborg LM, Leemans CR, Neri D, van Dongen GAMS. (124)I-L19-SIP for immuno-PET imaging of tumour vasculature and guidance of (131)I-L19-SIP radioimmunotherapy. Eur J Nucl Med Mol Imaging 2009; 36:1235-44. [PMID: 19259661 PMCID: PMC2709218 DOI: 10.1007/s00259-009-1096-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 02/09/2009] [Indexed: 11/21/2022]
Abstract
Purpose The human monoclonal antibody (MAb) fragment L19-SIP is directed against extra domain B (ED-B) of fibronectin, a marker of tumour angiogenesis. A clinical radioimmunotherapy (RIT) trial with 131I-L19-SIP was recently started. In the present study, after GMP production of 124I and efficient production of 124I-L19-SIP, we aimed to demonstrate the suitability of 124I-L19-SIP immuno-PET for imaging of angiogenesis at early-stage tumour development and as a scouting procedure prior to clinical 131I-L19-SIP RIT. Methods 124I was produced in a GMP compliant way via 124Te(p,n)124I reaction and using a TERIMO™ module for radioiodine separation. L19-SIP was radioiodinated by using a modified version of the IODO-GEN method. The biodistribution of coinjected 124I- and 131I-L19-SIP was compared in FaDu xenograft-bearing nude mice, while 124I PET images were obtained from mice with tumours of <50 to ∼700 mm3. Results 124I was produced highly pure with an average yield of 15.4 ± 0.5 MBq/μAh, while separation yield was ∼90% efficient with <0.5% loss of TeO2. Overall labelling efficiency, radiochemical purity and immunoreactive fraction were for 124I-L19-SIP: ∼80 , 99.9 and >90%, respectively. Tumour uptake was 7.3 ± 2.1, 10.8 ± 1.5, 7.8 ± 1.4, 5.3 ± 0.6 and 3.1 ± 0.4%ID/g at 3, 6, 24, 48 and 72 h p.i., resulting in increased tumour to blood ratios ranging from 6.0 at 24 h to 45.9 at 72 h p.i.. Fully concordant labelling and biodistribution results were obtained with 124I- and 131I-L19-SIP. Immuno-PET with 124I-L19-SIP using a high-resolution research tomograph PET scanner revealed clear delineation of the tumours as small as 50 mm3 and no adverse uptake in other organs. Conclusions 124I-MAb conjugates for clinical immuno-PET can be efficiently produced. Immuno-PET with 124I-L19-SIP appeared qualified for sensitive imaging of tumour neovasculature and for predicting 131I-L19-SIP biodistribution.
Collapse
Affiliation(s)
- Bernard M Tijink
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center, De Boelelaan 1117, P.O. Box 7057, 1007 MB, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Foillard S, Jin ZH, Garanger E, Boturyn D, Favrot MC, Coll JL, Dumy P. Synthesis and biological characterisation of targeted pro-apoptotic peptide. Chembiochem 2009; 9:2326-32. [PMID: 18712748 DOI: 10.1002/cbic.200800327] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We report herein the synthesis and in vitro assay of new, multimeric RGD-peptide conjugates for cell-targeted drug delivery. We generated a peptide scaffold comprising two functional domains, one a tumour blood vessel "homing" motif and the other a programmed cell-death-inducing peptide sequence. RGD peptides were selected to direct the molecular conjugate to alpha(V)beta(3) integrin-containing tumour cells. The pro-apoptotic (Lys-Leu-Ala-Lys-Leu-Ala-Lys)(2) peptide was found to be nontoxic outside cells, but toxic when internalized into targeted cells as it disrupted the mitochondrial membrane. The synthesis of these targeted pro-apoptotic conjugates was carried out by assembling three different units (that is, scaffold, RGD units and pro-apoptotic peptide) through chemoselective ligations. We show that one compound displays significant biological effect in alpha(V)beta(3) integrin-containing tumour cells.
Collapse
Affiliation(s)
- Stéphanie Foillard
- Département de Chimie Moléculaire, UMR CNRS-UJF 5250, ICMG FR 2607, 301, rue de la chimie, BP 53, FR 38041 Grenoble Cedex 9, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Mårlind J, Kaspar M, Trachsel E, Sommavilla R, Hindle S, Bacci C, Giovannoni L, Neri D. Antibody-mediated delivery of interleukin-2 to the stroma of breast cancer strongly enhances the potency of chemotherapy. Clin Cancer Res 2008; 14:6515-24. [PMID: 18927291 DOI: 10.1158/1078-0432.ccr-07-5041] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE There is an interest in the discovery of biopharmaceuticals, which are well tolerated and which potentiate the action of anthracyclines and taxanes in breast cancer therapy. EXPERIMENTAL DESIGN We have produced a recombinant fusion protein, composed of the human antibody fragment scFv(F16) fused to human interleukin-2 (F16-IL2), and tested its therapeutic performance in the MDA-MB-231 xenograft model of human breast cancer. The F16 antibody is specific to the alternatively spliced A1 domain of tenascin-C, which is virtually undetectable in normal tissues but is strongly expressed in the neovasculature and stroma of breast cancer. RESULTS When used as monotherapy, F16-IL2 displayed a strikingly superior therapeutic benefit compared with unconjugated recombinant IL-2. The administration of doxorubicin either before (8 days, 24 h, or 2 h) or simultaneously with the injection of F16-IL2 did not decrease the accumulation of immunocytokine in the tumor as measured by quantitative biodistribution analysis. Therapy experiments, featuring five once per week coadministrations of 20 mug F16-IL2 and doxorubicin, showed a statistically significant reduction of tumor growth rate and prolongation of survival at a 4 mg/kg doxorubicin dose but not at a 1 mg/kg dose. By contrast, combination of F16-IL2 with paclitaxel (5 and 1 mg/kg) exhibited a significant therapeutic benefit compared with paclitaxel alone at both dose levels. F16-IL2, alone or in combination with doxorubicin, was well tolerated in cynomolgus monkeys at doses equivalent to the ones now used in clinical studies. CONCLUSIONS F16-IL2 may represent a new useful biopharmaceutical for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jessica Mårlind
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Contino-Pépin C, Parat A, Périno S, Lenoir C, Vidal M, Galons H, Karlik S, Pucci B. Preliminary biological evaluations of new thalidomide analogues for multiple sclerosis application. Bioorg Med Chem Lett 2008; 19:878-81. [PMID: 19103485 DOI: 10.1016/j.bmcl.2008.11.118] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 11/26/2008] [Accepted: 11/27/2008] [Indexed: 01/08/2023]
Abstract
The present work deals with the synthesis of a new series of thalidomide derivatives for therapeutic applications. These compounds were evaluated in vitro on a human endothelial cell line EA.hy926 for their antiproliferative potential and in vivo on an experimental animal multiple sclerosis model called EAE as angiogenesis inhibitors. The preliminary results obtained on EAE assays seem to validate that anti-angiogenesis compounds could be promising tools for the treatment of MS.
Collapse
Affiliation(s)
- Christiane Contino-Pépin
- Université d'Avignon, Laboratoire de Chimie Bioorganique et des Systèmes Moléculaires Vectoriels, 33 rue Louis Pasteur, 84000 Avignon, France.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Ferrara KW. Driving delivery vehicles with ultrasound. Adv Drug Deliv Rev 2008; 60:1097-102. [PMID: 18479775 DOI: 10.1016/j.addr.2008.03.002] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Accepted: 03/04/2008] [Indexed: 12/22/2022]
Abstract
Therapeutic applications of ultrasound have been considered for over 40 years, with the mild hyperthermia and associated increases in perfusion produced by ultrasound harnessed in many of the earliest treatments. More recently, new mechanisms for ultrasound-based or ultrasound-enhanced therapies have been described, and there is now great momentum and enthusiasm for the clinical translation of these techniques. This dedicated issue of Advanced Drug Delivery Reviews, entitled "Ultrasound for Drug and Gene Delivery," addresses the mechanisms by which ultrasound can enhance local drug and gene delivery and the applications that have been demonstrated at this time. In this commentary, the identified mechanisms, delivery vehicles, applications and current bottlenecks for translation of these techniques are summarized.
Collapse
|
41
|
Villa A, Trachsel E, Kaspar M, Schliemann C, Sommavilla R, Rybak JN, Rösli C, Borsi L, Neri D. A high-affinity human monoclonal antibody specific to the alternatively spliced EDA domain of fibronectin efficiently targets tumor neo-vasculature in vivo. Int J Cancer 2008; 122:2405-13. [PMID: 18271006 DOI: 10.1002/ijc.23408] [Citation(s) in RCA: 182] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The alternatively spliced extra-domain B of fibronectin is one of the best characterized markers of tumor angiogenesis. Similarly, the extra-domain A (EDA), which can also be inserted in the fibronectin transcript by a mechanism of alternative splicing, has been shown to preferentially accumulate around new blood vessels in certain tumors, but this antigen has not been investigated so far as a target for antibody-based biomolecular intervention. We here describe the generation of 3 human monoclonal antibodies (named F8, B7 and D5), which recognize the same epitope of EDA, but which differ in terms of their dissociation constant to the human antigen (K(D) = 3.1, 16 and 17 nM, measured for monomeric preparations of the F8, B7 and D5 antibodies, respectively, in recombinant scFv format). When the 3 antibody fragments were cloned and expressed with a 5 amino acid linker, the 3 resulting homodimeric antibody preparations displayed comparable tumor: organ ratios in quantitative biodistribution studies, performed in immunocompetent 129SvEv mice, bearing subcutaneous syngeneic F9 murine tumors. The percent injected dose per gram (%ID/g) values in tumors 24 hr after intravenous injection were 9.3, 10.2 and 13 for F8, B7 and D5, respectively. The F8 antibody may serve as useful building block for the development of antibody-based targeted anti-cancer therapeutics. Preclinical and clinical investigations are facilitated by the fact that F8 recognizes the human and mouse antigen with comparable affinity, and by the observation that EDA over-expression is detectable not only in solid tumors, but also in hematological malignancies.
Collapse
Affiliation(s)
- Alessandra Villa
- Philochem AG, c/o ETH Zürich, Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rybak JN, Roesli C, Kaspar M, Villa A, Neri D. The extra-domain A of fibronectin is a vascular marker of solid tumors and metastases. Cancer Res 2007; 67:10948-57. [PMID: 18006840 DOI: 10.1158/0008-5472.can-07-1436] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One of the most promising new avenues for the development of more selective and efficacious cancer therapies relies on the antibody-mediated targeted delivery of bioactive agents (e.g., cytokines) to the tumor environment. The identification of quantitative differences in the expression of accessible vascular proteins in metastatic lesions and host organs facilitate the development of antibody-based strategies, which should be highly efficient and selective, considering the fact that an over-exuberant neovasculature is a characteristic feature of aggressive cancers, and that tumor blood vessels are readily accessible for i.v. administered therapeutic agents. Metastasis is the main cause of death in cancer. The availability of metastasis-specific antigens accessible from the bloodstream will allow a selective delivery of therapeutic agents to metastatic lesions using antibodies as vehicles. Using a combination of vascular biotinylation of 129Sv mice bearing F9 liver metastases and mass spectrometry, we have identified 435 accessible proteins in metastasis and host organ specimens, of which 117 were exclusively detected in metastases. In particular, we found that the alternatively spliced extra-domain A (EDA) of fibronectin is strongly expressed in the neovasculature of liver metastases, while being undetectable in most normal organs. A human antibody to EDA was used to show EDA expression in the neovasculature of metastases and primary tumors of human cancer patients and to target mouse liver metastases and subcutaneous tumors in vivo. Human antibody fragments specific to the EDA domain of fibronectin promise to serve as general vehicles for the efficient and selective delivery of imaging agents or therapeutic molecules to metastatic sites.
Collapse
Affiliation(s)
- Jascha-N Rybak
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | | | | | | |
Collapse
|