1
|
Wang S, Wang H, Drabek A, Smith WS, Liang F, Huang ZR. Unleashing the Potential: Designing Antibody-Targeted Lipid Nanoparticles for Industrial Applications with CMC Considerations and Clinical Outlook. Mol Pharm 2024; 21:4-17. [PMID: 38117251 DOI: 10.1021/acs.molpharmaceut.3c00735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Antibody-targeted lipid nanoparticles (Ab-LNPs) are rapidly gaining traction as multifaceted platforms in precision medicine, adept at delivering a diverse array of therapeutic agents, including nucleic acids and small molecules. This review provides an incisive overview of the latest developments in the field of Ab-LNP technology, with a special emphasis on pivotal design aspects such as antibody engineering, bioconjugation strategies, and advanced formulation techniques. Furthermore, it addresses critical chemistry, manufacturing, and controls (CMC) considerations and thoroughly examines the in vivo dynamics of Ab-LNPs, underscoring their promising potential for clinical application. By seamlessly blending scientific advancements with practical industrial perspectives, this review casts a spotlight on the burgeoning role of Ab-LNPs as an innovative and potent tool in the realm of targeted drug delivery.
Collapse
Affiliation(s)
- Sheryl Wang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Hong Wang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Andrew Drabek
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Wenwen Sha Smith
- FUSION BioVenture, 15 Presidential Way, Woburn, Massachusetts 01801, United States
| | - Feng Liang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Zhaohua Richard Huang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| |
Collapse
|
2
|
Moulahoum H, Ghorbanizamani F, Zihnioglu F, Timur S. Surface Biomodification of Liposomes and Polymersomes for Efficient Targeted Drug Delivery. Bioconjug Chem 2021; 32:1491-1502. [PMID: 34283580 DOI: 10.1021/acs.bioconjchem.1c00285] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chemotherapy has seen great progress in the development of performant treatment strategies. Nanovesicles such as liposomes and polymersomes demonstrated great potential in cancer therapy. However, these nanocarriers deliver their content passively, which faces a lot of constraints during blood circulation. The main challenge resides in degradation and random delivery to normal tissues. Hence, targeting drug delivery using specific molecules (such as antibodies) grafted over the surface of these nanocarriers came as the answer to overcome many problems faced before. The advantage of using antibodies is their antigen/antibody recognition, which provides a high level of specificity to reach treatment targets. This review discusses the many techniques of nanocarrier functionalization with antibodies. The aim is to recognize the various approaches by describing their advantages and deficiencies to create the most suitable drug delivery platform. Some methods are more suitable for other applications rather than drug delivery, which can explain the low success of some proposed targeted nanocarriers. In here, a critical analysis of how every method could impact the recognition and targeting capacity of some nanocarriers (liposomes and polymersomes) is discussed to make future research more impactful and advance the field of biomedicine further.
Collapse
Affiliation(s)
- Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Faezeh Ghorbanizamani
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Figen Zihnioglu
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey.,Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, 35100, Bornova, Izmir, Turkey
| |
Collapse
|
3
|
Puerto-Camacho P, Amaral AT, Lamhamedi-Cherradi SE, Menegaz BA, Castillo-Ecija H, Ordóñez JL, Domínguez S, Jordan-Perez C, Diaz-Martin J, Romero-Pérez L, Lopez-Alvarez M, Civantos-Jubera G, Robles-Frías MJ, Biscuola M, Ferrer C, Mora J, Cuglievan B, Schadler K, Seifert O, Kontermann R, Pfizenmaier K, Simón L, Fabre M, Carcaboso ÁM, Ludwig JA, de Álava E. Preclinical Efficacy of Endoglin-Targeting Antibody-Drug Conjugates for the Treatment of Ewing Sarcoma. Clin Cancer Res 2018; 25:2228-2240. [PMID: 30420447 DOI: 10.1158/1078-0432.ccr-18-0936] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/13/2018] [Accepted: 11/06/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Endoglin (ENG; CD105) is a coreceptor of the TGFβ family that is highly expressed in proliferating endothelial cells. Often coopted by cancer cells, ENG can lead to neo-angiogenesis and vasculogenic mimicry in aggressive malignancies. It exists both as a transmembrane cell surface protein, where it primarily interacts with TGFβ, and as a soluble matricellular protein (sENG) when cleaved by matrix metalloproteinase 14 (MMP14). High ENG expression has been associated with poor prognosis in Ewing sarcoma, an aggressive bone cancer that primarily occurs in adolescents and young adults. However, the therapeutic value of ENG targeting has not been fully explored in this disease. EXPERIMENTAL DESIGN We characterized the expression pattern of transmembrane ENG, sENG, and MMP14 in preclinical and clinical samples. Subsequently, the antineoplastic potential of two novel ENG-targeting monoclonal antibody-drug conjugates (ADC), OMTX503 and OMTX703, which differed only by their drug payload (nigrin-b A chain and cytolysin, respectively), was assessed in cell lines and preclinical animal models of Ewing sarcoma. RESULTS Both ADCs suppressed cell proliferation in proportion to the endogenous levels of ENG observed in vitro. Moreover, the ADCs significantly delayed tumor growth in Ewing sarcoma cell line-derived xenografts and patient-derived xenografts in a dose-dependent manner. CONCLUSIONS Taken together, these studies demonstrate potent preclinical activity of first-in-class anti-ENG ADCs as a nascent strategy to eradicate Ewing sarcoma.
Collapse
Affiliation(s)
- Pilar Puerto-Camacho
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Ana Teresa Amaral
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | | | - Brian A Menegaz
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Helena Castillo-Ecija
- Institut de Recerca Sant Joan de Déu, Pediatric Hematology and Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - José Luis Ordóñez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | | | - Carmen Jordan-Perez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Juan Diaz-Martin
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Laura Romero-Pérez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Maria Lopez-Alvarez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Gema Civantos-Jubera
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - María José Robles-Frías
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Michele Biscuola
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | | | - Jaume Mora
- Institut de Recerca Sant Joan de Déu, Pediatric Hematology and Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - Branko Cuglievan
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Keri Schadler
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | - Ángel M Carcaboso
- Institut de Recerca Sant Joan de Déu, Pediatric Hematology and Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas.
| | - Enrique de Álava
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain.
| |
Collapse
|
4
|
Gilabert-Oriol R, Ryan GM, Leung AWY, Firmino NS, Bennewith KL, Bally MB. Liposomal Formulations to Modulate the Tumour Microenvironment and Antitumour Immune Response. Int J Mol Sci 2018; 19:ijms19102922. [PMID: 30261606 PMCID: PMC6213379 DOI: 10.3390/ijms19102922] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022] Open
Abstract
Tumours are complex systems of genetically diverse malignant cells that proliferate in the presence of a heterogeneous microenvironment consisting of host derived microvasculature, stromal, and immune cells. The components of the tumour microenvironment (TME) communicate with each other and with cancer cells, to regulate cellular processes that can inhibit, as well as enhance, tumour growth. Therapeutic strategies have been developed to modulate the TME and cancer-associated immune response. However, modulating compounds are often insoluble (aqueous solubility of less than 1 mg/mL) and have suboptimal pharmacokinetics that prevent therapeutically relevant drug concentrations from reaching the appropriate sites within the tumour. Nanomedicines and, in particular, liposomal formulations of relevant drug candidates, define clinically meaningful drug delivery systems that have the potential to ensure that the right drug candidate is delivered to the right area within tumours at the right time. Following encapsulation in liposomes, drug candidates often display extended plasma half-lives, higher plasma concentrations and may accumulate directly in the tumour tissue. Liposomes can normalise the tumour blood vessel structure and enhance the immunogenicity of tumour cell death; relatively unrecognised impacts associated with using liposomal formulations. This review describes liposomal formulations that affect components of the TME. A focus is placed on formulations which are approved for use in the clinic. The concept of tumour immunogenicity, and how liposomes may enhance radiation and chemotherapy-induced immunogenic cell death (ICD), is discussed. Liposomes are currently an indispensable tool in the treatment of cancer, and their contribution to cancer therapy may gain even further importance by incorporating modulators of the TME and the cancer-associated immune response.
Collapse
Affiliation(s)
- Roger Gilabert-Oriol
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Gemma M Ryan
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Ada W Y Leung
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6N 3P8, Canada.
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada.
| | - Natalie S Firmino
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
| | - Kevin L Bennewith
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
| | - Marcel B Bally
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6N 3P8, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
- Centre for Drug Research and Development, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
5
|
Hervé-Aubert K, Allard-Vannier E, Joubert N, Lakhrif Z, Alric C, Martin C, Viaud-Massuard MC, Dimier-Poisson I, Aubrey N, Chourpa I. Impact of Site-Specific Conjugation of ScFv to Multifunctional Nanomedicines Using Second Generation Maleimide. Bioconjug Chem 2018; 29:1553-1559. [DOI: 10.1021/acs.bioconjchem.8b00091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | | | - Nicolas Joubert
- GICC CNRS UMR 7292, Team IMT, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | - Zineb Lakhrif
- UMR Université-INRA ISP 1282, Team BioMAP, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | - Christophe Alric
- EA6295 NMNS, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | - Camille Martin
- GICC CNRS UMR 7292, Team IMT, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | | | - Isabelle Dimier-Poisson
- UMR Université-INRA ISP 1282, Team BioMAP, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | - Nicolas Aubrey
- UMR Université-INRA ISP 1282, Team BioMAP, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | - Igor Chourpa
- EA6295 NMNS, Université de Tours, 31 avenue Monge, 37200 Tours, France
| |
Collapse
|
6
|
Zhuo H, Zheng B, Liu J, Huang Y, Wang H, Zheng D, Mao N, Meng J, Zhou S, Zhong L, Zhao Y. Efficient targeted tumor imaging and secreted endostatin gene delivery by anti-CD105 immunoliposomes. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:42. [PMID: 29499713 PMCID: PMC5833054 DOI: 10.1186/s13046-018-0712-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Anti-CD105 mAb-conjugated immunoliposomes, loaded with secreted mouse endostatin gene, were developed for targeted tumor imaging and antiangiogenic gene therapy. METHODS The liposomes were investigated for size, zeta-potential, lipid content, antibody binding ability, and pcDNA loading capacity. The ability of immunoliposomes to target tumor-derived endothelial cells and perform gene transfer in vitro was measured and their basic biocompatibility was evaluated. A nude mouse/breast cancer xenograft model was used to examine the tumor internalization of fluorescent-labeled liposomes and the clinical potential of immnuoliposomes loaded with pcDNA3.1-CSF1-endostatin. RESULTS Loaded immunoliposomes were homogenously distributed with a well-defined spherical shape and bilayer, diameter of 122 ± 11 nm, and zeta potential + 1.40 mV. No significant differences were observed in body weight, liver index, oxidative stress, or liver and kidney function in mice after liposomes exposure. The addition of CD105 mAb to liposomes conferred the ability to target tumor-derived endothelial cells in vitro and in vivo. Systemic intravenous administration of fluorescent immunoliposomes in the xenograft model resulted in selective and efficient internalization in tumor vasculature. Treatment of mice with pcDNA3.1-CSF1-endostatin-loaded immunoliposomes suppressed tumor growth by 71%. CONCLUSIONS These data demonstrate the advantages of using anti-CD105 mAb-conjugated immunoliposomes to enhance tumor targeting, imaging, and gene transfer applications.
Collapse
Affiliation(s)
- Huiqin Zhuo
- Department of Gastrointestinal Surgery, Institute of Gastrointestinal Oncology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Baoshi Zheng
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Department of Cardiothoracic Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jianming Liu
- The Third Xiangya Hospital, Central South University, Changsha, 410083, China
| | - Yong Huang
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Huiling Wang
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Duo Zheng
- Department of Basic Medicine, Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Naiquan Mao
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jinyu Meng
- Biomedical Polymers Laboratory, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Sufang Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Liping Zhong
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
7
|
Seidi K, Jahanban-Esfahlan R, Zarghami N. Tumor rim cells: From resistance to vascular targeting agents to complete tumor ablation. Tumour Biol 2017; 39:1010428317691001. [DOI: 10.1177/1010428317691001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current vascular targeting strategies pursue two main goals: anti-angiogenesis agents aim to halt sprouting and the formation of new blood vessels, while vascular disrupting agents along with coaguligands seek to compromise blood circulation in the vessels. The ultimate goal of such therapies is to deprive tumor cells out of oxygen and nutrients long enough to succumb cancer cells to death. Most of vascular targeting agents presented promising therapeutic potential, but the final goal which is cure is rarely achieved. Nevertheless, in both preclinical and clinical settings, tumors tend to grow back, featuring a highly invasive, metastatic, and extremely resistant form. This review highlights the critical significance of tumor rim cells as the main factor, determining therapy success with vascular targeting agents. We present an overview of different single and combination treatments with vascular targeting agents that enable efficient targeting of tumor rim cells and long-lasting tumor cure. Understanding the nature of tumor rim cells, how they establish, how they manage to survive of vascular targeting agents, and how they contribute in tumor refractoriness, may open new avenues to the development of beneficial strategies, capable to eliminate residual rim cells, and enable tumor ablation once and forever.
Collapse
Affiliation(s)
- Khaled Seidi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Alric C, Aubrey N, Allard-Vannier É, di Tommaso A, Blondy T, Dimier-Poisson I, Chourpa I, Hervé-Aubert K. Covalent conjugation of cysteine-engineered scFv to PEGylated magnetic nanoprobes for immunotargeting of breast cancer cells. RSC Adv 2016. [DOI: 10.1039/c6ra06076e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Orientation- and site-directed covalent conjugation of cysteine-engineered scFv to PEGylated SPIONs allows antigen recognition while preserving colloidal properties of nanoprobes.
Collapse
Affiliation(s)
- Christophe Alric
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Nicolas Aubrey
- Université François Rabelais de Tours
- UMR1282 INRA ‘Infectiologie et Santé Publique’
- F 37000 Tours
- France
| | - Émilie Allard-Vannier
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Anne di Tommaso
- Université François Rabelais de Tours
- UMR1282 INRA ‘Infectiologie et Santé Publique’
- F 37000 Tours
- France
| | - Thibaut Blondy
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Isabelle Dimier-Poisson
- Université François Rabelais de Tours
- UMR1282 INRA ‘Infectiologie et Santé Publique’
- F 37000 Tours
- France
| | - Igor Chourpa
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Katel Hervé-Aubert
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| |
Collapse
|
9
|
Rabenhold M, Steiniger F, Fahr A, Kontermann RE, Rüger R. Bispecific single-chain diabody-immunoliposomes targeting endoglin (CD105) and fibroblast activation protein (FAP) simultaneously. J Control Release 2015; 201:56-67. [DOI: 10.1016/j.jconrel.2015.01.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 01/09/2023]
|
10
|
Kuo YC, Hung C, Gullapalli RP, Xu S, Zhuo J, Raghavan SR, D'Souza WD. Liposomal nanoprobes that combine anti-EGFR antibodies and MRI contrast agents: synthesis and in vitro characterization. RSC Adv 2014. [DOI: 10.1039/c4ra05579a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
11
|
Toi H, Tsujie M, Haruta Y, Fujita K, Duzen J, Seon BK. Facilitation of endoglin-targeting cancer therapy by development/utilization of a novel genetically engineered mouse model expressing humanized endoglin (CD105). Int J Cancer 2014; 136:452-61. [PMID: 24866768 DOI: 10.1002/ijc.28994] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/18/2014] [Accepted: 05/15/2014] [Indexed: 01/07/2023]
Abstract
Endoglin (ENG) is a TGF-β coreceptor and essential for vascular development and angiogenesis. A chimeric antihuman ENG (hENG) monoclonal antibody (mAb) c-SN6j (also known as TRC105) shows promising safety and clinical efficacy features in multiple clinical trials of patients with various advanced solid tumors. Here we developed a novel genetically engineered mouse model to optimize the ENG-targeting clinical trials. We designed a new targeting vector that contains exons 4-8 of hENG gene to generate novel genetically engineered mice (GEMs) expressing functional human/mouse chimeric (humanized) ENG with desired epitopes. Genotyping of the generated mice confirmed that we generated the desired GEMs. Immunohistochemical analysis demonstrated that humanized ENG protein of the GEMs expresses epitopes defined by 7 of our 8 anti-hENG mAbs tested. Surprisingly the homozygous GEMs develop normally and are healthy. Established breast and colon tumors as well as metastasis and tumor microvessels in the GEMs were effectively suppressed by systemic administration of anti-hENG mAbs. Additionally, test result indicates that synergistic potentiation of antitumor efficacy can be induced by simultaneous targeting of two distinct epitopes by anti-hENG mAbs. Sorafenib and capecitabine also showed antitumor efficacy in the GEMs. The presented novel GEMs are the first GEMs that express the targetable humanized ENG. Test results indicate utility of the GEMs for the clinically relevant studies. Additionally, we generated GEMs expressing a different humanized ENG containing exons 5-6 of hENG gene, and the homozygous GEMs develop normally and are healthy.
Collapse
Affiliation(s)
- Hirofumi Toi
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY; Department of Surgery, Megumino Hospital, Eniwa, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
12
|
Targeted small interfering RNA-immunoliposomes as a promising therapeutic agent against highly pathogenic Avian Influenza A (H5N1) virus infection. Antimicrob Agents Chemother 2014; 58:2816-24. [PMID: 24614365 DOI: 10.1128/aac.02768-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
This study describes a proof-of-concept study on the use of small interfering RNA (siRNA)-immunoliposomes as a therapeutic agent against H5N1 influenza virus infection. siRNA specific for influenza virus nucleoprotein (NP) mRNA was employed as the key antiviral agent to inhibit viral replication in this study. A humanized single-chain Fv antibody (huscFv) against the hemagglutinin (HA) of H5N1 highly pathogenic avian influenza virus (HPAI) was used as the targeting molecule to HA of H5N1 virus, which is abundantly expressed on the surface of infected cells (the HA target cells). The huscFv was applied to cationic polyethylene glycol-conjugated 3β-[N-(N',N'-dimethylaminoethane) carbamoyl] cholesterol-dioleoylphosphatidyl ethanolamine (PEGylated DC-Chol-DOPE) liposomes to generate immunoliposomes for siRNA delivery. The immunoliposomes were shown to specifically bind HA-expressing Sf9 cells and demonstrated enhanced siRNA transfection efficiency. The siRNA transfection efficiency was significantly reduced after preincubation of the HA target cells with an excess amount of free huscFv. These results therefore demonstrated that the enhanced siRNA delivery by use of immunoliposomes was mediated via targeting by huscFv. Furthermore, the siRNA silencing effect was more pronounced when the immunoliposomes were administered 6 to 12 h post-H5N1 infection in MDCK cells compared with the nontargeted liposomes. This proof-of-concept study may contribute to the future design and development of an siRNA delivery system for combating viral infectious diseases in humans.
Collapse
|
13
|
Monaghan M, Greiser U, Wall JG, O’Brien T, Pandit A. Interference: an alteRNAtive therapy following acute myocardial infarction. Trends Pharmacol Sci 2012; 33:635-45. [DOI: 10.1016/j.tips.2012.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/10/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
|
14
|
Antinuclear antibodies with nucleosome-restricted specificity for targeted delivery of chemotherapeutic agents. Ther Deliv 2012; 1:257-72. [PMID: 22816132 DOI: 10.4155/tde.10.30] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Circulating antinuclear autoantibodies (ANAs) are well known to accompany various pathological conditions and can be artificially induced by immunization. Research and clinical data permit us to hypothesize a definite connection between cancer and ANAs. Based on the available data, my group's research suggested that exogenous ANAs may be used as anticancer therapeutics. Among these ANAs, nucleosome-specific ANAs may be particularly useful. Advances in cancer immunotherapy with monoclonal antibodies re-emphasized the role of humoral immunity in neoplasia control. The development of a universal antibody targeting diverse cancers is of clear importance. We showed that certain natural ANAs recognize the surface of numerous tumor cells but not normal cells via cell surface-bound nucleosomes originating from the apoptotically dying neighboring tumor cells, mediate antibody-dependent cellular cytotoxicity of tumor cells in vitro and inhibit the development of murine tumor in syngeneic mice. A single monoclonal antinuclear nucleosome-specific autoantibody, mAb 2C5, specifically recognizes multiple unrelated human tumor cell lines and accumulates at a high tumor-to-normal cell ratio in various human tumors in nude mice. Immunotherapy with mAb 2C5 resulted in significant suppression of the growth of several human tumors. In addition, mAb 2C5, when used in subtherapeutic quantities, can serve as a highly efficient specific ligand to target various drug- or diagnostic agent-loaded pharmaceutical nanocarriers, such as liposomes and polymeric micelles, to various tumors. Here, the data (accumulated predominantly in our laboratory over several years) on mAb 2C5-mediated tumor targeting of chemotherapeutic agents is reviewed.
Collapse
|
15
|
Kobayashi N, Odaka K, Uehara T, Imanaka-Yoshida K, Kato Y, Oyama H, Tadokoro H, Akizawa H, Tanada S, Hiroe M, Fukumura T, Komuro I, Arano Y, Yoshida T, Irie T. Toward in vivo imaging of heart disease using a radiolabeled single-chain Fv fragment targeting tenascin-C. Anal Chem 2011; 83:9123-30. [PMID: 22074352 DOI: 10.1021/ac202159p] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antibodies specific to a particular target molecule can be used as analytical reagents, not only for in vitro immunoassays but also for noninvasive in vivo imaging, e.g., immunoscintigraphies. In the latter case, it is important to reduce the size of antibody molecules in order to achieve suitable in vivo "diagnostic kinetics" and generate higher-resolution images. For these purposes, single-chain Fv fragments (scFvs; M(r) < 30 kDa) have greater potential than intact immunoglobulins (~150 kDa) or Fab (or Fab') fragments (~50 kDa). Our recent observation of enhanced tenascin-C (Tnc) expression at sites of cardiac repair after myocardial infarction prompted us to develop a radiolabeled scFv against Tnc for in vivo imaging of heart disease. We cloned the genes encoding the heavy and light chain variable domains of the mouse anti-Tnc monoclonal antibody 4F10, and combined them to create a single gene. The resulting scFv-4F10 gene was expressed in E. coli cells to produce soluble scFv proteins. scFv-4F10 has an affinity for Tnc (K(a) = 3.5 × 10(7) M(-1)), similar to the Fab fragment of antibody 4F10 (K(a) = 1.3 × 10(7) M(-1)) and high enough to be of practical use. A cysteine residue was then added to the C-terminus to achieve site-specific (111)In labeling via a chelating group. The resulting (111)In-labeled scFv was administered to a rat model of acute myocardial infarction. Biodistribution and quantitative autoradiographic studies indicated higher uptake of the radioactivity at the infarcted myocardium than the noninfarcted one. Single photon emission computed tomography (SPECT) provided in vivo cardiac images that coincided with the ex vivo observations. Our results will promote advances in diagnostic strategies for heart disease.
Collapse
Affiliation(s)
- Norihiro Kobayashi
- Kobe Pharmaceutical University, 4-19-1, Motoyama-Kitamachi, Higashinada-ku, Kobe 658-8558, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Seon BK, Haba A, Matsuno F, Takahashi N, Tsujie M, She X, Harada N, Uneda S, Tsujie T, Toi H, Tsai H, Haruta Y. Endoglin-targeted cancer therapy. Curr Drug Deliv 2011; 8:135-43. [PMID: 21034418 DOI: 10.2174/156720111793663570] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 07/30/2010] [Indexed: 01/30/2023]
Abstract
Vascular-targeting antiangiogenic therapy (VTAT) of cancer can be advantageous over conventional tumor cell targeted cancer therapy if an appropriate target is found. Our hypothesis is that endoglin (ENG; CD105) is an excellent target in VTAT. ENG is selectively expressed on vascular and lymphatic endothelium in tumors. This allows us to target both tumor-associated vasculature and lymphatic vessels to suppress tumor growth and metastasis. ENG is essential for angiogenesis/vascular development and a co-receptor of TGF-β. Our studies of selected anti-ENG monoclonal antibodies (mAbs) in several animal models and in vitro studies support our hypothesis. These mAbs and/or their immunoconjugates (immunotoxins and radioimmunoconjugates) induced regression of preformed tumors as well as inhibited formation of new tumors. In addition, they suppressed metastasis. Several mechanisms were involved in the suppressive activity of the naked (unconjugated) anti-ENG mAbs. These include direct growth suppression of proliferating endothelial cells, induction of apoptosis, ADCC (antibody-dependent cell-mediated cytotoxicity) and induction of T cell immunity. To facilitate clinical application, we generated a human/mouse chimeric anti-ENG mAb termed c-SN6j and performed studies of pharmacokinetics, toxicology and immunogenicity of c-SN6j in nonhuman primates. No significant toxicity was detected by several criteria and minimal immune response to the murine part of c-SN6j was detected after multiple i.v. injections. The results support our hypothesis that c-SN6j can be safely administered in cancer patients. This hypothesis is supported by the ongoing phase 1 clinical trial of c-SN6j (also known as TRC105) in patients with advanced or metastatic solid cancer in collaboration with Tracon Pharma and several oncologists (NCT00582985).
Collapse
Affiliation(s)
- Ben K Seon
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Positron emission tomography imaging of CD105 expression during tumor angiogenesis. Eur J Nucl Med Mol Imaging 2011; 38:1335-43. [PMID: 21373764 DOI: 10.1007/s00259-011-1765-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 02/16/2011] [Indexed: 01/03/2023]
Abstract
PURPOSE Overexpression of CD105 (endoglin) correlates with poor prognosis in many solid tumor types. Tumor microvessel density (MVD) assessed by CD105 staining is the current gold standard for evaluating tumor angiogenesis in the clinic. The goal of this study was to develop a positron emission tomography (PET) tracer for imaging CD105 expression. METHODS TRC105, a chimeric anti-CD105 monoclonal antibody, was conjugated to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and labeled with (64)Cu. FACS analysis and microscopy studies were performed to compare the CD105 binding affinity of TRC105 and DOTA-TRC105. PET imaging, biodistribution, blocking, and ex vivo histology studies were performed on 4T1 murine breast tumor-bearing mice to evaluate the ability of (64)Cu-DOTA-TRC105 to target tumor angiogenesis. Another chimeric antibody, cetuximab, was used as an isotype-matched control. RESULTS FACS analysis of human umbilical vein endothelial cells (HUVECs) revealed no difference in CD105 binding affinity between TRC105 and DOTA-TRC105, which was further validated by fluorescence microscopy. (64)Cu labeling was achieved with high yield and specific activity. Serial PET imaging revealed that the 4T1 tumor uptake of the tracer was 8.0 ± 0.5, 10.4 ± 2.8, and 9.7 ± 1.8%ID/g at 4, 24, and 48 h post-injection, respectively (n = 3), higher than most organs at late time points which provided excellent tumor contrast. Biodistribution data as measured by gamma counting were consistent with the PET findings. Blocking experiments, control studies with (64)Cu-DOTA-cetuximab, as well as ex vivo histology all confirmed the in vivo target specificity of (64)Cu-DOTA-TRC105. CONCLUSION This is the first successful PET imaging study of CD105 expression. Fast, prominent, persistent, and CD105-specific uptake of the tracer in the 4T1 tumor was observed. Further studies are warranted and currently underway.
Collapse
|
18
|
Rothdiener M, Beuttler J, Messerschmidt SKE, Kontermann RE. Antibody targeting of nanoparticles to tumor-specific receptors: immunoliposomes. Methods Mol Biol 2010; 624:295-308. [PMID: 20217604 DOI: 10.1007/978-1-60761-609-2_20] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Immunoliposomes generated by coupling of antibodies to the liposomal surface allow for an active tissue targeting, e.g., through binding to tumor cell-specific receptors. Instead of whole antibodies, single-chain Fv fragments (scFv), which represent the smallest part of an antibody containing the entire antigen-binding site, find increasing usage as targeting moiety. Here we provide protocols for the preparation of type II scFv immunoliposomes by the conventional coupling method as well as the post-insertion method. Furthermore protocols to analyze binding of these immunoliposomes to antigen-expressing cells as well as internalization through receptor-mediated endocytosis are included.
Collapse
Affiliation(s)
- Miriam Rothdiener
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | | | | | | |
Collapse
|
19
|
Rothdiener M, Müller D, Castro PG, Scholz A, Schwemmlein M, Fey G, Heidenreich O, Kontermann RE. Targeted delivery of SiRNA to CD33-positive tumor cells with liposomal carrier systems. J Control Release 2010; 144:251-8. [PMID: 20184933 DOI: 10.1016/j.jconrel.2010.02.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 02/11/2010] [Accepted: 02/16/2010] [Indexed: 02/09/2023]
Abstract
SiRNA molecules represent promising therapeutic molecules, e.g. for cancer therapy. However, efficient delivery into tumor cells remains a major obstacle for treatment. Here, we describe a liposomal siRNA carrier system for targeted delivery of siRNA to CD33-positive acute myeloid leukemia cells. The siRNA is directed against the t(8;21) translocation resulting in the AML1/MTG8 fusion protein. The siRNA was encapsulated in free or polyethylene imine (PEI)-complexed form into PEGylated liposomes endowed subsequently with an anti-CD33 single-chain Fv fragment (scFv) for targeted delivery. The resulting siRNA-loaded immunoliposomes (IL) and immunolipoplexes (ILP) showed specific binding and internalization by CD33-expressing myeloid leukemia cell lines (SKNO-1, Kasumi-1). Targeted delivery of AML1/MTG8 siRNA, but not of mismatch control siRNA, reduced AML1/MTG8 mRNA and protein levels and decreased leukemic clonogenicity, a hallmark of leukemic self-renewal. Although this study revealed that further modifications are necessary to increase efficacy of siRNA delivery and silencing, we were able to establish a targeted liposomal siRNA delivery system combining recombinant antibody fragments for targeted delivery with tumor cell-specific siRNA molecules as therapeutic agents.
Collapse
Affiliation(s)
- Miriam Rothdiener
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abu Lila AS, Ishida T, Kiwada H. Recent advances in tumor vasculature targeting using liposomal drug delivery systems. Expert Opin Drug Deliv 2010; 6:1297-309. [PMID: 19780711 DOI: 10.1517/17425240903289928] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tumor vessels possess unique physiological features that might be exploited for improved drug delivery. The targeting of liposomal anticancer drugs to tumor vasculature is increasingly recognized as an effective strategy to obtain superior therapeutic efficacy with limited host toxicity compared with conventional treatments. This review introduces recent advances in the field of liposomal targeting of tumor vasculature, along with new approaches that can be used in the design and optimization of liposomal delivery systems. In addition, cationic liposome is focused on as a promising carrier for achieving efficient vascular targeting. The clinical implications are discussed of several approaches using a single liposomal anticancer drug formulation: dual targeting, vascular targeting (targeting tumor endothelial cells) and tumor targeting (targeting tumor cells).
Collapse
Affiliation(s)
- Amr S Abu Lila
- The University of Tokushima, Institute of Health Biosciences, Department of Pharmacokinetics and Biopharmaceutics, 770-8505, Japan
| | | | | |
Collapse
|
21
|
Abstract
The paradigm of using nanoparticulate pharmaceutical carriers has been well established over the past decade, both in pharmaceutical research and in the clinical setting. Drug carriers are expected to stay in the blood for long time, accumulate in pathological sites with affected and leaky vasculature (tumors, inflammations, and infarcted areas) via the enhanced permeability and retention (EPR) effect, and facilitate targeted delivery of specific ligand-modified drugs and drug carriers into poorly accessible areas. Among various approaches to specifically target drug-loaded carrier systems to required pathological sites in the body, two seem to be most advanced--passive (EPR effect-mediated) targeting, based on the longevity of the pharmaceutical carrier in the blood and its accumulation in pathological sites with compromised vasculature, and active targeting, based on the attachment of specific ligands to the surface of pharmaceutical carriers to recognize and bind pathological cells. Here, we will consider and discuss these two targeting approaches using tumor targeting as an example.
Collapse
|
22
|
Fonsatti E, Nicolay HJM, Altomonte M, Covre A, Maio M. Targeting cancer vasculature via endoglin/CD105: a novel antibody-based diagnostic and therapeutic strategy in solid tumours. Cardiovasc Res 2009; 86:12-9. [PMID: 19812043 DOI: 10.1093/cvr/cvp332] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endoglin/CD105 is well acknowledged as being the most reliable marker of proliferation of endothelial cells, and it is overexpressed on tumour neovasculature. Our current knowledge of its structure, physiological role, and tissue distribution suggests that targeting of endoglin/CD105 is a novel and powerful diagnostic and therapeutic strategy in human malignancies, through the imaging of tumour-associated angiogenesis and the inhibition of endothelial cell functions related to tumour angiogenesis. Among biotherapeutic agents, monoclonal antibodies have shown a major impact on the clinical course of human malignancies of different histotypes. Along this line, the potential efficacy of anti-endoglin/CD105 antibodies and their derivatives for clinical purposes in cancer is supported by a large body of available pre-clinical in vitro and in vivo data. In this review, the main findings supporting the translation of antibody-based endoglin/CD105 targeting from pre-clinical studies to clinical applications in human cancer are summarized and discussed.
Collapse
Affiliation(s)
- Ester Fonsatti
- Division of Medical Oncology and Immunotherapy, Department of Oncology, Istituto Toscano Tumori, University Hospital of Siena, Strada delle Scotte 14, 53100 Siena, Italy
| | | | | | | | | |
Collapse
|
23
|
Uneda S, Toi H, Tsujie T, Tsujie M, Harada N, Tsai H, Seon BK. Anti-endoglin monoclonal antibodies are effective for suppressing metastasis and the primary tumors by targeting tumor vasculature. Int J Cancer 2009; 125:1446-53. [PMID: 19533687 DOI: 10.1002/ijc.24482] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Anti-metastatic activity of an antitumor agent is exceedingly important because metastasis is the primary cause of death for most solid cancer patients. In this report, we show that 3 anti-endoglin (ENG) monoclonal antibodies (mAbs) SN6a, SN6j and SN6k which define individually distinct epitopes of ENG of tumor vasculature are capable of suppressing tumor metastases in the multiple metastasis models. The metastasis models were generated by i.v., s.c. (into flank) or mammary gland fat pad injection of 4T1 murine mammary carcinoma cells and splenic injection of two types of colon26 murine colorectal carcinoma cells. Individual mAbs were injected i.v. via the tail vein of mice. SN6a and SN6j effectively suppressed the formation of metastatic colonies of 4T1 in the lung in all of the three 4T1 metastatic models. In addition, these mAbs were effective for suppressing the primary tumors of 4T1 in the skin and mammary fat pad. These mAbs effectively suppressed microvessel density and angiogenesis in tumors as measured by the Matrigel plug assay in mice. No significant side effects of the administered mAbs were detected. Furthermore, SN6a and SN6j extended survival of the tumor-bearing mice. SN6j, SN6k and their immunoconjugates with deglycosylated ricin A-chain were all effective for suppressing hepatic metastasis of colon26. The findings in the present study are clinically relevant in view of the ongoing clinical trial of a humanized (chimerized) form of SN6j.
Collapse
Affiliation(s)
- Shima Uneda
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Bernabeu C, Lopez-Novoa JM, Quintanilla M. The emerging role of TGF-beta superfamily coreceptors in cancer. Biochim Biophys Acta Mol Basis Dis 2009; 1792:954-73. [PMID: 19607914 DOI: 10.1016/j.bbadis.2009.07.003] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2009] [Revised: 07/02/2009] [Accepted: 07/06/2009] [Indexed: 12/23/2022]
Abstract
The transforming growth factor beta (TGF-beta) signaling pathway plays a key role in different physiological processes such as development, cellular proliferation, extracellular matrix synthesis, angiogenesis or immune responses and its deregulation may result in tumor development. The TGF-beta coreceptors endoglin and betaglycan are emerging as modulators of the TGF-beta response with important roles in cancer. Endoglin is highly expressed in the tumor-associated vascular endothelium with prognostic significance in selected neoplasias and with potential to be a prime vascular target for antiangiogenic cancer therapy. On the other hand, the expression of endoglin and betaglycan in tumor cells themselves appears to play an important role in the progression of cancer, influencing cell proliferation, motility, invasiveness and tumorigenicity. In addition, experiments in vitro and in vivo in which endoglin or betaglycan expression is modulated have provided evidence that they act as tumor suppressors. The purpose of this review was to highlight the potential of membrane and soluble forms of the endoglin and betaglycan proteins as molecular targets in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Carmelo Bernabeu
- Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain.
| | | | | |
Collapse
|
25
|
Cell selection and characterization of a novel human endothelial cell specific nanobody. Mol Immunol 2009; 46:1814-23. [DOI: 10.1016/j.molimm.2009.01.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2008] [Revised: 01/20/2009] [Accepted: 01/25/2009] [Indexed: 12/11/2022]
|
26
|
Rüger R, Müller D, Fahr A, Kontermann RE. In vitro characterization of binding and stability of single-chain Fv Ni-NTA-liposomes. J Drug Target 2008; 14:576-82. [PMID: 17050123 DOI: 10.1080/10611860600864018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Recently, we presented a new method for the generation of single-chain Fv (scFv) immunoliposomes, which circumvents the necessity to introduce additional reactive groups in the protein. This method is based on immobilizing scFv fragments via their C-terminal hexahistidyl-tag on liposomes containing nickel-complexed dioleoyl-glycero-succinyl-nitrilotriacetic acid (Ni-NTA-DOGS) as an anchor lipid within the lipid bilayer. Here, we have extended this approach to various other scFv fragments and further demonstrate strong and selective binding of these liposomes to target cells in vitro. In order to evaluate suitability for in vivo applications, we investigated the influence of human plasma on stability and binding behaviour of scFv Ni-NTA-liposomes in vitro using scFv A5 directed against human endoglin (CD105) as a model antibody. We could show that the binding activity to target cells is rapidly lost in the presence of human plasma. Incorporation of polyethylene glycol (PEG) chains into the lipid bilayer did not protect against loss of binding capability. Further studies showed that loss of binding is mainly due to displacement of Ni-NTA-bound scFv fragments caused by plasma proteins. In conclusion, the system allows for a rapid and flexible generation of target cell specific immunoliposomes for in vitro applications but lacks stability for in vivo applications.
Collapse
Affiliation(s)
- Ronny Rüger
- Lehrstuhl für Pharmazeutische Technologie, Friedrich-Schiller-Universität Jena, Lessingstrasse 8, 07743 Jena, Germany
| | | | | | | |
Collapse
|
27
|
Müller D, Trunk G, Sichelstiel A, Zettlitz KA, Quintanilla M, Kontermann RE. Murine endoglin-specific single-chain Fv fragments for the analysis of vascular targeting strategies in mice. J Immunol Methods 2008; 339:90-8. [PMID: 18790696 DOI: 10.1016/j.jim.2008.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 08/20/2008] [Accepted: 08/20/2008] [Indexed: 11/30/2022]
Abstract
Endoglin has been identified as a promising cell surface antigen for vascular targeting approaches in cancer therapy, e.g. employing antibody molecules as targeting moieties. However, in vivo analysis of such strategies in mouse models requires antibodies recognizing endoglin on mouse endothelial cells. Here we describe the isolation of single-chain Fv fragments (scFvs) from phage display libraries, which bind to the extracellular region of mouse endoglin. One of these clones, scFv mE12, showed strong (K(d)=11 nM) and selective binding to purified endoglin and also to the endoglin-expressing mouse endothelioma cell line eEnd.2. This antibody recognized a linear epitope located in the N-terminal region (aa 27-361) of endoglin. Cell binding was further increased by generating a bivalent scFv-Fc fusion protein composed of scFv mE12 and the human gamma1 Fc part. Moreover, scFv mE12 was endowed with an additional cysteine residue in the linker region and applied for the generation of anti-endoglin scFv immunoliposomes capable of selectively binding to endoglin-expressing cells. Thus, anti-mouse endoglin scFv mE12 should be useful to analyze vascular targeting strategies in mice.
Collapse
Affiliation(s)
- Dafne Müller
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Stuttgart, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Vladimir Torchilin
- Northeastern University, Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, 360 Huntington Avenue, Boston, MA 02115, USA ;
| |
Collapse
|
29
|
Schiffelers RM, Storm G. Liposomal nanomedicines as anticancer therapeutics: beyond targeting tumor cells. Int J Pharm 2008; 364:258-64. [PMID: 18773947 DOI: 10.1016/j.ijpharm.2008.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 08/06/2008] [Accepted: 08/08/2008] [Indexed: 12/16/2022]
Abstract
Tumor cells have long been the primary target cell type of liposomes for anticancer therapy. At present, it appears that tumor growth and metastasis is facilitated by interactions between tumor cells and supporting cells. These supporting cells consist of adaptive and innate immune cells, endothelial cells, pericytes, fibroblasts, stromal and mesenchymal cells. Insight into the activity of these cells and communication between these cells has provided new tactics for targeting alternative cell types in tumor treatment and offered new drug classes that could be used to modulate the activity of these supporting cells. Here, we provide an overview of liposomal systems that have been designed to target supporting cells in tumor tissue and therapeutic results of these systems.
Collapse
Affiliation(s)
- Raymond M Schiffelers
- Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| | | |
Collapse
|
30
|
Dallas NA, Samuel S, Xia L, Fan F, Gray MJ, Lim SJ, Ellis LM. Endoglin (CD105): a marker of tumor vasculature and potential target for therapy. Clin Cancer Res 2008; 14:1931-7. [PMID: 18381930 DOI: 10.1158/1078-0432.ccr-07-4478] [Citation(s) in RCA: 248] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Endoglin (CD105) is an accessory protein of the transforming growth factor-beta receptor system expressed on vascular endothelial cells. Mutation of the endoglin gene is associated with hereditary hemorrhagic telangiectasias, or Osler-Weber-Rendu syndrome, and has been studied extensively in the context of this disease. The expression of endoglin is elevated on the endothelial cells of healing wounds, developing embryos, inflammatory tissues, and solid tumors. Endoglin is a marker of activated endothelium, and its vascular expression is limited to proliferating cells. Recent studies identified endoglin expression in several solid tumor types, with the level of expression correlating with various clinicopathologic factors including decreased survival and presence of metastases. Attempts to target endoglin and the cells that express this protein in tumor-bearing mice have yielded promising results.
Collapse
Affiliation(s)
- Nikolaos A Dallas
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77230-1402, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Tsujie M, Tsujie T, Toi H, Uneda S, Shiozaki K, Tsai H, Seon BK. Anti-tumor activity of an anti-endoglin monoclonal antibody is enhanced in immunocompetent mice. Int J Cancer 2008; 122:2266-73. [PMID: 18224682 DOI: 10.1002/ijc.23314] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In the present study, we investigated the mechanisms by which anti-endoglin (EDG; CD105) monoclonal antibodies (mAbs) suppress angiogenesis and tumor growth. Antihuman EDG mAb SN6j specifically bound to murine endothelial cells and was internalized into the cells in vitro. SN6j effectively suppressed angiogenesis in mice in the Matrigel plug assay. We found that SN6j is more effective for tumor suppression in immunocompetent mice than in SCID mice. We hypothesized that T cell immunity is important for effective antitumor efficacy of SN6j in vivo. To test this hypothesis, we investigated effects of CpG oligodeoxynucleotides (ODN) and depletion of CD4(+) T cells and/or CD8(+) T cells on antitumor efficacy of SN6j in mice. Systemic (i.v.) administration of a relatively small dose (0.6 mug/g body weight/dose) of SN6j suppressed growth of established s.c. tumors of colon-26 in BALB/c mice and improved survival of the tumor-bearing mice. Addition of CpG ODN to SN6j synergistically enhanced antitumor efficacy of SN6j. In contrast, such enhancing effects of CpG ODN were not detected in SCID mice. Antitumor efficacy of SN6j in BALB/c mice was abrogated when CD4(+) T cells and/or CD8(+) T cells were depleted; effect of CD8(+) T cell depletion was stronger. Interestingly, CD4-depletion decreased tumor growth while CD8-depletion enhanced tumor growth in the absence of SN6j. SN6j induced apoptosis in human umbilical vein endothelial cells in a dose-dependent manner which indicates an additional mechanism of antiangiogenesis by SN6j. (c) 2008 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Masanori Tsujie
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
VCAM-1 directed immunoliposomes selectively target tumor vasculature in vivo. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2008; 1778:854-63. [DOI: 10.1016/j.bbamem.2007.12.021] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 11/07/2007] [Accepted: 12/16/2007] [Indexed: 11/20/2022]
|
33
|
Messerschmidt SKE, Kolbe A, Müller D, Knoll M, Pleiss J, Kontermann RE. Novel Single-Chain Fv′ Formats for the Generation of Immunoliposomes by Site-Directed Coupling. Bioconjug Chem 2007; 19:362-9. [DOI: 10.1021/bc700349k] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sylvia K. E. Messerschmidt
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany, and Institut für Technische Biochemie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Anke Kolbe
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany, and Institut für Technische Biochemie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Dafne Müller
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany, and Institut für Technische Biochemie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Michael Knoll
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany, and Institut für Technische Biochemie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Jürgen Pleiss
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany, and Institut für Technische Biochemie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Roland E. Kontermann
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany, and Institut für Technische Biochemie, Universität Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| |
Collapse
|
34
|
Baum P, Müller D, Rüger R, Kontermann RE. Single-chain Fv immunoliposomes for the targeting of fibroblast activation protein-expressing tumor stromal cells. J Drug Target 2007; 15:399-406. [PMID: 17613658 DOI: 10.1080/10611860701453034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tumor stromal cells have gained increasing attention as possible target for cancer therapy. Fibroblast activation protein (FAP) represents a cell surface antigen selectively expressed by reactive tumor stromal fibroblasts of various cancers. Here, we describe anti-FAP immunoliposomes as carrier systems for active targeting of FAP-expressing cells. As targeting ligand we used single-chain Fv (scFv) molecules cross-reacting with human and mouse FAP. These scFv molecules were genetically modified to express an additional cysteine residue at the C-terminus allowing a defined and site-directed conjugation. Coupling to Mal-PEG(2000)-DSPE containing liposomes resulted in sterically stabilized scFv immunoliposomes showing strong and specific binding to FAP-expressing cells. These immunoliposomes were highly stable when incubated under physiological conditions (human plasma, 37 degrees C). In addition, we could show that binding to FAP-expressing cells leads to internalization of intact liposomes into the endosomal compartment. Thus, these anti-FAP scFv immunoliposomes should be suitable for target cell-specific delivery and uptake of encapsulated drugs.
Collapse
Affiliation(s)
- Patrick Baum
- Institut für Zellbiologie und Immunologie, Universität Stuttgart, Stuttgart, Germany
| | | | | | | |
Collapse
|
35
|
Hood E, Gonzalez M, Plaas A, Strom J, VanAuker M. Immuno-targeting of nonionic surfactant vesicles to inflammation. Int J Pharm 2007; 339:222-30. [PMID: 17448616 DOI: 10.1016/j.ijpharm.2006.12.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 12/21/2006] [Accepted: 12/22/2006] [Indexed: 12/24/2022]
Abstract
Niosomes composed of sorbitan monostearate (Span 60), polyoxyethylene sorbitan monostearate (Tween 61), cholesterol, and dicetyl phosphate were conjugated with a purified monoclonal antibody to CD44 (IM7) through a cyanuric chloride (CC) linkage on the polyoxyethylene group of the Tween 61 molecule. Inclusion of small amounts of Tween 61 within the surfactant component of niosomes formed using thin film hydration techniques and sonication did not hamper vesicle stability as compared to Span 60 niosomes. Conjugation was verified by UV absorbance of fluorescently tagged IM7 in non-fluorescing niosomes and fluorescent micrographs. The immuno-niosomes were incubated with synovial lining cells expressing CD44. Attachment of niosomes was evident and showed selectivity and specificity compared to controls. These findings suggest that the resulting immuno-niosomes may provide an effective method for targeted drug delivery.
Collapse
Affiliation(s)
- Elizabeth Hood
- Biomedical Engineering Program, Department of Chemical Engineering, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
36
|
Balestrieri ML, Napoli C. Novel challenges in exploring peptide ligands and corresponding tissue-specific endothelial receptors. Eur J Cancer 2007; 43:1242-50. [PMID: 17449238 DOI: 10.1016/j.ejca.2007.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 02/06/2007] [Accepted: 02/08/2007] [Indexed: 12/14/2022]
Abstract
The structural and molecular diversity of vascular endothelium may depend on the functional state and tissue localisation of its cells. Tumour vasculature expresses a number of molecular markers that distinguish it from normal vasculature. In cancer, the determinant of specific tumour vasculature heterogeneity is, in part, dictated by dysregulated expression of tumour-derived angiogenic factors. The identification of molecular 'addresses' on the surface of tumour vasculature has significantly contributed to the selection of targets, which have been used for delivering therapeutic and imaging agents in cancer. Cytotoxic drug, pro-apoptotic peptides, protease inhibitors, and gene therapy vectors have been successfully linked to peptides and delivered to tumour sites with an improved experimental therapy. Different diagnostic and therapeutic compounds can be efficiently targeted to specific receptors on vascular endothelial cells; the development of ligand-directed vector tools may promote systemic targeted gene delivery. Here, we review the very recent advances in the identification of peptide ligands and their corresponding tissue-specific endothelial receptors through the phage display technology with emphasis on ligand-directed delivery of therapeutic agents and targeted gene therapy.
Collapse
Affiliation(s)
- Maria Luisa Balestrieri
- Department of Chemical Biology and Physics; 1st School of Medicine, II University of Naples, Complesso S. Andrea delle Dame, Naples 80138, Italy
| | | |
Collapse
|
37
|
Cheng WWK, Das D, Suresh M, Allen TM. Expression and purification of two anti-CD19 single chain Fv fragments for targeting of liposomes to CD19-expressing cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:21-9. [PMID: 17046711 DOI: 10.1016/j.bbamem.2006.09.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 08/22/2006] [Accepted: 09/11/2006] [Indexed: 01/29/2023]
Abstract
Antibody-targeted liposomal anticancer drugs combine the specificity of antibodies with large payloads of entrapped drugs. We previously showed that liposomal doxorubicin (DXR) targeted via anti-CD19 monoclonal antibodies (mAb) or their Fab' fragments against the B-cell antigen CD19 led to improved therapeutic effects in murine B-cell lymphoma models relative to non-targeted liposomal DXR. We now are examining the use of anti-CD19 single chain fragments of the antibody variable region (scFv) as a targeting moiety, to test the hypothesis that scFv have advantages over full-sized mAb or Fab' fragments. We expressed two different anti-CD19 scFv constructs, HD37-C and HD37-CCH in E. coli, and purified the scFvs using two different methods. The HD37-CCH construct was selected for coupling studies due to its relative stability and activity in comparison to HD37-C. When coupled to liposomes, the HD37-CCH scFv showed increased binding in vitro to CD19-positive Raji cells, compared to non-targeted liposomes. Cytotoxicity data showed that HD37-CCH scFv-targeted liposomes loaded with DXR were more cytotoxic than non-targeted liposomal DXR. Our results suggest that anti-CD19 scFv constructs should be explored further for their potential in treating B-lymphoid leukemias and lymphomas.
Collapse
MESH Headings
- Antibiotics, Antineoplastic/pharmacology
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/isolation & purification
- Antibody Affinity
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Binding Sites, Antibody
- Burkitt Lymphoma/immunology
- Burkitt Lymphoma/metabolism
- Burkitt Lymphoma/pathology
- Cell Line, Tumor
- Cell Survival/drug effects
- Chemistry, Pharmaceutical
- Cloning, Molecular
- Doxorubicin/pharmacology
- Drug Compounding
- Drug Delivery Systems
- Humans
- Immunoglobulin Fab Fragments/immunology
- Immunoglobulin Variable Region/immunology
- Immunoglobulin Variable Region/metabolism
- Inhibitory Concentration 50
- Liposomes
Collapse
Affiliation(s)
- W W K Cheng
- Dept. of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada T6G 2H7
| | | | | | | |
Collapse
|
38
|
Rüger R, Müller D, Fahr A, Kontermann RE. Generation of immunoliposomes using recombinant single-chain Fv fragments bound to Ni-NTA-liposomes. J Drug Target 2006; 13:399-406. [PMID: 16308208 DOI: 10.1080/10611860500353328] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Recombinant single-chain Fv antibody fragments (scFv) can be combined with liposomes to generate immunoliposomes for targeted drug delivery. Recent studies have shown that scFv molecules modified to express a C-terminal cysteine residue can be used for site-directed chemical conjugation. Here, we present a new method by immobilizing scFv fragments via their C-terminal hexahistidyl-tag on liposomes containing Ni-NTA-lipids (Ni-NTA-DOGS) in their lipid bilayer without the need to introduce additional reactive groups in the protein. Using an anti-endoglin scFv as a model antibody, we could show that scFv molecules are efficiently immobilized on the liposome surface and that these immunoliposomes bind specifically and strongly to endoglin-expressing endothelial cells. This approach allows for a rapid and flexible generation of target cell-specific immunoliposomes.
Collapse
Affiliation(s)
- Ronny Rüger
- Friedrich-Schiller-Universität Jena, Lehrstuhl für Pharmazeutische Technologie, Lessingstrasse 8, 07743 Jena, Germany
| | | | | | | |
Collapse
|
39
|
Hu H, Chen D, Liu Y, Deng Y, Yang S, Qiao M, Zhao J, Zhao X. Target ability and therapy efficacy of immunoliposomes using a humanized antihepatoma disulfide-stabilized Fv fragment on tumor cells. J Pharm Sci 2006; 95:192-9. [PMID: 16315243 DOI: 10.1002/jps.20517] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recently the use of peptides in bee venom (PBV) for cancer therapy has attracted considerable attention. However, PBV's extensive use is prohibited by its intense hemolytic activity. In this study, the sterically stabilized liposomal PBV (PBV-SL) was prepared using soybean phosphatidylcholine, cholesterol, and cholesterol-PEG-COOH. The humanized antihepatoma disulfide-stabilized Fv (hdsFv25) was reengineered, expressed, and coupled to sterically stabilized liposomes using the N-hydroxysuccinimide ester method. The hdsFv25-immunoliposomes (SIL [hdscFv25]) were immunoreactive as determined by ELISA assay. PBV-SIL [hdscFv25] can kill SMMC-7721 cells in vitro with higher efficiency than nontargeted liposomes. PBV-SIL [hdsFv25] displayed high antitumor activity and resulted in a significant reduction in tumor size compared to nontargeted liposomes and PBV. These results indicated that this strategy should be applicable to applicable in the treatment of other cancers.
Collapse
Affiliation(s)
- Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, RP China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
How, if at all, can drug delivery help to create ideal drugs? After four decades of trying, an effective site-specific drug-delivery system has not yet been developed. This review draws attention to the pharmacokinetic conditions that must be met to achieve a successful performance by site-selective drug-carrier delivery systems. In a drug-carrier approach, a drug is attached to a macromolecular carrier via a chemically labile linker. The carrier transports the drug to its site of action and releases it at the target site. For this simple approach to work, several fundamental conditions (nonspecific interactions, target site access, drug release and drug suitability) must be satisfied. The importance of these essential requirements, not always recognized in the development of drug-delivery systems, is discussed and illustrated by recent examples selected from the literature.
Collapse
Affiliation(s)
- Karel Petrak
- PJP Innovations, 707 Knox Street, Houston, Texas 77007, USA.
| |
Collapse
|
41
|
Peterson NC. Advances in monoclonal antibody technology: genetic engineering of mice, cells, and immunoglobulins. ILAR J 2005; 46:314-9. [PMID: 15953839 DOI: 10.1093/ilar.46.3.314] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The ability to produce antibodies that are directed against specific antigens has played a crucial role in advancing scientific discoveries. Recombinant technologies have extended the application of antibodies beyond the research laboratory and into the clinic for the treatment of cancer and other diseases. Creative approaches using these technologies have been used to reduce the antibody to its minimal functional size, and/or make them bifunctional (immunotoxins), bispecific, or less immunoreactive (humanized). Additionally, mice that are engineered to generate antibodies of human genomic origin have been used to produce therapeutic antibodies and are being further developed. As the research and clinical demands for antibodies continue to increase, the development of improved resources (cell lines and animals) to improve production efficiency, generate larger repertoires, and deliver greater yields of antibodies is being explored, and advances in this area are discussed further in this review.
Collapse
Affiliation(s)
- Norman C Peterson
- Department of Comparative Medicine, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
42
|
Smith J, Kontermann RE, Embleton J, Kumar S. Antibody phage display technologies with special reference to angiogenesis. FASEB J 2005; 19:331-41. [PMID: 15746176 DOI: 10.1096/fj.04-2863rev] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The presence of blood vessels is a prerequisite for normal development, tissue growth, and tissue repair. However, its abnormal occurrence or absence can also potentiate disease processes. Angiogenic therapies have been used to stimulate blood vessel growth in ischemic conditions such as severe end-stage peripheral vascular disease, ischemic heart disease and stroke and for inhibition of angiogenesis in tumors. The targeting and identification of novel endothelial cell (EC) markers that can ultimately be used in angiogenic strategies is an expanding field but is limited by the availability of reagents. For instance repeated injection of mouse monoclonal antibodies (Mabs) against angiogenic EC, can result in the production of autoantibodies. Therefore, these mouse Mabs cannot be used for therapeutic purposes. Phage display technology was employed in this context to select antibodies, proteins, and peptides against known or novel EC antigens. Furthermore, technologies have been developed that enable the specific targeting of epitopes on cells including the endothelium with high-affinity/avidity antibodies. The focus for these antibody targeting strategies are markers that are unique or up-regulated on angiogenic EC including the vascular endothelial growth factor receptor (VEGFR) KDR, endoglin (CD105), and the extracellular domain B (ED-B) domain of fibronectin (FN). These markers are reviewed herein.
Collapse
Affiliation(s)
- Julia Smith
- University of Manchester, Stopford Building, Oxford Rd, Manchester, M13 9PT, UK.
| | | | | | | |
Collapse
|