1
|
Prodi E, Neri D, De Luca R. Tumor-Homing Antibody-Cytokine Fusions for Cancer Therapy. Onco Targets Ther 2024; 17:697-715. [PMID: 39224695 PMCID: PMC11368152 DOI: 10.2147/ott.s480787] [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: 07/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Recombinant cytokine products have emerged as a promising avenue in cancer therapy due to their capacity to modulate and enhance the immune response against tumors. However, their clinical application is significantly hindered by systemic toxicities already at low doses, thus preventing escalation to therapeutically active regimens. One promising approach to overcoming these limitations is using antibody-cytokine fusion proteins (also called immunocytokines). These biopharmaceuticals leverage the targeting specificity of antibodies to deliver cytokines directly to the tumor microenvironment, thereby reducing systemic exposure and enhancing the therapeutic index. This review comprehensively examines the development and potential of antibody-cytokine fusion proteins in cancer therapy. It explores the molecular characteristics that influence the performance of these fusion proteins, and it highlights key findings from preclinical and clinical studies, illustrating the potential of immunocytokines to improve treatment outcomes in cancer patients. Recent advancements in the field, such as novel engineering strategies and combination strategies to enhance the efficacy and safety of immunocytokines, are also discussed. These innovations offer new opportunities to optimize this class of biotherapeutics, making them a more viable and effective option for cancer treatment. As the field continues to evolve, understanding the critical factors that influence the performance of immunocytokines will be essential for successfully translating these therapies into clinical practice.
Collapse
Affiliation(s)
- Eleonora Prodi
- Philochem AG, Otelfingen, 8112, Switzerland
- University of Trento, Italy, CiBIO (Department of Cellular, Computational and Integrative Biology), Povo, 38123, Trento
| | - Dario Neri
- Philogen Spa, Siena, 53100, Italy
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
2
|
Xu Y, Liu R, Li R, Zhi X, Yang P, Qian L, Sun D, Liu L, Dai Z. Manipulating Neovasculature-Targeting Capability of Biomimetic Nanodiscs for Synergistic Photoactivatable Tumor Infarction and Chemotherapy. ACS NANO 2023; 17:16192-16203. [PMID: 37555449 DOI: 10.1021/acsnano.3c05463] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Tumor infarction therapy is a promising antitumor strategy with the advantages of taking a short therapy duration, less risk of resistance, and effectiveness against a wide range of tumor types. However, its clinical application is largely hindered by tumor recurrence in the surviving rim and the potential risk of thromboembolic events due to nonspecific vasculature targeting. Herein, a neovasculature-targeting synthetic high-density lipoprotein (sHDL) nanodisc loaded with pyropheophorbide-a and camptothecin (CPN) was fabricated for photoactivatable tumor infarction and synergistic chemotherapy. By manipulating the anisotropy in ligand modification of sHDL nanodiscs, CPN modified with neovaculature-targeting peptide on the planes (PCPN) shows up to 7-fold higher cellular uptake compared with that around the edge (ECPN). PCPN can efficiently bind to endothelial cells of tumor vessels, and upon laser irradiation, massive local thrombus can be induced by the photodynamic reaction to deprive nutrition supply. Meanwhile, CPT could be released in response to the tumor reductive environment, thus killing residual tumor cells in the surviving rim to inhibit recurrence. These findings not only offer a powerful approach of synergistic cancer therapy but also suggest the potential of plane-modified sHDL nanodiscs as a versatile drug delivery nanocarrier.
Collapse
Affiliation(s)
- Yunxue Xu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Rui Li
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Xin Zhi
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Peipei Yang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Linxue Qian
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Desheng Sun
- Department of Ultrasound, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Li Liu
- Department of Ultrasound, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
3
|
Fan X, Jiang Y, Li M, Zhang Y, Tian C, Mao L, Xie H, Sun L, Yang Z, Sitti M. Scale-reconfigurable miniature ferrofluidic robots for negotiating sharply variable spaces. SCIENCE ADVANCES 2022; 8:eabq1677. [PMID: 36112686 PMCID: PMC9481141 DOI: 10.1126/sciadv.abq1677] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Magnetic miniature soft robots have shown great potential for facilitating biomedical applications by minimizing invasiveness and possible physical damage. However, researchers have mainly focused on fixed-size robots, with their active locomotion accessible only when the cross-sectional dimension of these confined spaces is comparable to that of the robot. Here, we realize the scale-reconfigurable miniature ferrofluidic robots (SMFRs) based on ferrofluid droplets and propose a series of control strategies for reconfiguring SMFR's scale and deformation to achieve trans-scale motion control by designing a multiscale magnetic miniature robot actuation (M3RA) system. The results showed that SMFRs, varying from centimeters to a few micrometers, leveraged diverse capabilities, such as locomotion in structured environments, deformation to squeeze through gaps, and even reversible scale reconfiguration for navigating sharply variable spaces. A miniature robot system with these capabilities combined is promising to be applied in future wireless medical robots inside confined regions of the human body.
Collapse
Affiliation(s)
- Xinjian Fan
- School of Mechanical and Electrical Engineering, Soochow University, No. 8, Jixue Road, Suzhou 215131, China
| | - Yihui Jiang
- School of Mechanical and Electrical Engineering, Soochow University, No. 8, Jixue Road, Suzhou 215131, China
| | - Mingtong Li
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart 70569, Germany
| | - Yunfei Zhang
- School of Mechanical and Electrical Engineering, Soochow University, No. 8, Jixue Road, Suzhou 215131, China
| | - Chenyao Tian
- State Key Laboratory of Robotics and Systems, Harbin Institute of Technology, Yikuang, Harbin 150080, China
| | - Liyang Mao
- State Key Laboratory of Robotics and Systems, Harbin Institute of Technology, Yikuang, Harbin 150080, China
| | - Hui Xie
- State Key Laboratory of Robotics and Systems, Harbin Institute of Technology, Yikuang, Harbin 150080, China
| | - Lining Sun
- School of Mechanical and Electrical Engineering, Soochow University, No. 8, Jixue Road, Suzhou 215131, China
| | - Zhan Yang
- School of Mechanical and Electrical Engineering, Soochow University, No. 8, Jixue Road, Suzhou 215131, China
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart 70569, Germany
- Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich, Switzerland
- School of Medicine and College of Engineering, Koç University, 34450 Istanbul, Turkey
| |
Collapse
|
4
|
Xie H, Li W, Liu H, Chen Y, Ma M, Wang Y, Luo Y, Song D, Hou Q, Lu W, Bai Y, Li B, Ma J, Huang C, Yang T, Liu Z, Zhao X, Ding P. Erythrocyte Membrane-Coated Invisible Acoustic-Sensitive Nanoparticle for Inducing Tumor Thrombotic Infarction by Precisely Damaging Tumor Vascular Endothelium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201933. [PMID: 35789094 DOI: 10.1002/smll.202201933] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/16/2022] [Indexed: 06/15/2023]
Abstract
Selective induction of tumor thrombus infarction is a promising antitumor strategy. Non-persistent embolism due to non-compacted thrombus and activated fibrinolytic system within the tumor large blood vessels and tumor margin recurrence are the main therapeutic bottlenecks. Herein, an erythrocyte membrane-coated invisible acoustic-sensitive nanoparticle (TXA+DOX/PFH/RBCM@cRGD) is described, which can induce tumor thrombus infarction by precisely damaging tumor vascular endothelium. It is revealed that TXA+DOX/PFH/RBCM@cRGD can effectively accumulate on the endothelial surface of tumor vessels with the help of the red blood cell membrane (RBCM) stealth coating and RGD cyclic peptide (cRGD), which can be delivered in a targeted manner as nanoparticle missiles. As a kind of phase-change material, perfluorohexane (PFH) nanodroplets possess excellent acoustic responsiveness. Acoustic-sensitive missiles can undergo an acoustic phase transition and intense cavitation with response to low-intensity focused ultrasound (LIFU), damaging the tumor vascular endothelium, rapidly initiating the coagulation cascade, and forming thromboembolism in the tumor vessels. The drugs loaded in the inner water phase are released explosively. Tranexamic acid (TXA) inhibits the fibrinolytic system, and doxorubicin (DOX) eliminates the margin survival. In summary, a stealthy and acoustically responsive multifunctional nanoparticle delivery platform is successfully developed for inducing thrombus infarction by precisely damaging tumor vascular endothelium.
Collapse
Affiliation(s)
- Huichao Xie
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wan Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongfeng Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Mengrui Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yichen Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yucen Luo
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Di Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qianqian Hou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenwen Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yu Bai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bao Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jizhuang Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chi Huang
- Ultrasound Department of Shengjing Hospital, China Medical University, Shenyang, 110016, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, 04401, USA
| | - Zhining Liu
- Ultrasound Department, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| |
Collapse
|
5
|
Bienia A, Wiecheć-Cudak O, Murzyn AA, Krzykawska-Serda M. Photodynamic Therapy and Hyperthermia in Combination Treatment-Neglected Forces in the Fight against Cancer. Pharmaceutics 2021; 13:1147. [PMID: 34452108 PMCID: PMC8399393 DOI: 10.3390/pharmaceutics13081147] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/26/2021] [Accepted: 07/16/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of death in humans. Despite the progress in cancer treatment, and an increase in the effectiveness of diagnostic methods, cancer is still highly lethal and very difficult to treat in many cases. Combination therapy, in the context of cancer treatment, seems to be a promising option that may allow minimizing treatment side effects and may have a significant impact on the cure. It may also increase the effectiveness of anti-cancer therapies. Moreover, combination treatment can significantly increase delivery of drugs to cancerous tissues. Photodynamic therapy and hyperthermia seem to be ideal examples that prove the effectiveness of combination therapy. These two kinds of therapy can kill cancer cells through different mechanisms and activate various signaling pathways. Both PDT and hyperthermia play significant roles in the perfusion of a tumor and the network of blood vessels wrapped around it. The main goal of combination therapy is to combine separate mechanisms of action that will make cancer cells more sensitive to a given therapeutic agent. Such an approach in treatment may contribute toward increasing its effectiveness, optimizing the cancer treatment process in the future.
Collapse
Affiliation(s)
| | | | | | - Martyna Krzykawska-Serda
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland; (A.B.); (O.W.-C.); (A.A.M.)
| |
Collapse
|
6
|
Shimizu S, Sakai K, Chikugo T, Satou T, Shiraishi N, Mitsudomi T, Nishio K. Integrin-linked kinase pathway in heterogeneous pulmonary sarcomatoid carcinoma. Oncol Lett 2021; 21:320. [PMID: 33692852 DOI: 10.3892/ol.2021.12582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 12/14/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary sarcomatoid carcinoma (PSC) is classified as poorly differentiated, and non-small cell lung carcinomas that contained a component of sarcoma or sarcoma-like differentiation are rare. The underlying carcinogenetic mechanism governing PSC remains unclear. The current study investigated the underlying carcinogenetic mechanism of PSC based on the hypothesis that it involves the epithelial-mesenchymal transition (EMT) process. Mutation analysis of PSCs, including carcinosarcoma, pleomorphic carcinoma and epithelial carcinoma specimens, was performed using targeted deep sequencing, whole transcriptome analysis and digital spatial profiling (DSP). PSCs exhibit a distinct mutation profile, with TP53, SYNE1 and APC mutations. Therefore, clustering of the gene expression profiles allowed the PSCs to be distinguished from the epithelial carcinomas. Increased gene expression of fibronectin in PSC was an important contributor to differential profiles. Pathway analysis revealed enhanced activity of the integrin-linked kinase (ILK) signaling pathway in the PSCs. DSP analysis using 56 antibodies of marker proteins confirmed significantly higher expression of fibronectin in PSCs. Intratumor heterogeneity of fibronectin expression was observed in sarcoma components. In conclusion, epithelial-mesenchymal transition process mediated by ILK signaling may be associated with carcinogenetic mechanisms of PSC. Overexpression of fibronectin mediated by ILK signaling appears to serve a role in the EMT involved in the PSC transformation process.
Collapse
Affiliation(s)
- Shigeki Shimizu
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Takaaki Chikugo
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Takao Satou
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Naoki Shiraishi
- Department of Diagnostic Pathology, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tetsuya Mitsudomi
- Department of Thoracic Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
7
|
Ranjbar L, Maleki F, Sadeghzadeh N, Abediankenari S, Mardanshahi A, Masteri Farahani A. In vitro/in vivo assessment of the targeting ability of [ 99mTc] Tc-labeled an aptide specific to the extra domain B of fibronectin (APT EDB) for colorectal cancer. Ann Nucl Med 2020; 34:460-466. [PMID: 32390105 DOI: 10.1007/s12149-020-01472-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/26/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The APTEDB is an aptide specific to the extra domain B (EDB) of fibronectin with high affinity for EDB, which is expressed in malignant tumors including brain cancer (U87MG) and colorectal cancer (HT-29). Aim of this study was to evaluate the [99mTc] Tc-APTEDB potential as an imaging probe for colorectal cancer. METHODS Radiochemical purity was evaluated by HPLC and radio-isotope TLC scanner. Blocking study for specific binding assay and affinity calculation (Kd) on HT-29 cell lines were also carried out. Planar imaging and bio-distribution studies were performed in HT-29 tumor-bearing mice. RESULTS The APTEDB was efficiently labeled with technetium-99m in high radiochemical yield (up to 97%). Cellular binding study demonstrated specific binding of the [99mTc] Tc-APTEDB in cultured HT-29 cells. The Kd value was found to be 40.46 ± 13.39 nM. The tumor-to-muscle ratio was ~ 1.5 in ex vivo bio-distribution study at 1 h after injection. Planar imaging study showed higher activity accumulation in EDB expressing HT-29 tumor relative to muscle (used as control) (~ 1.7) at 1 h. CONCLUSIONS Although more studies are required to find out the full potential of this radio-ligand as an imaging probe, the present results nevertheless provide useful information about [99mTc] Tc-APTEDB, which might be beneficial in design and development of new [99mTc] Tc-APTEDB for efficient targeting of tumor in vivo.
Collapse
Affiliation(s)
- Leila Ranjbar
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, 48471-93698, Sari, Mazandaran, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fariba Maleki
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, 48471-93698, Sari, Mazandaran, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nourollah Sadeghzadeh
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, 48471-93698, Sari, Mazandaran, Iran.
| | - Saied Abediankenari
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Mardanshahi
- Department of Radiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Arezou Masteri Farahani
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, 48471-93698, Sari, Mazandaran, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
8
|
Lui BG, Salomon N, Wüstehube-Lausch J, Daneschdar M, Schmoldt HU, Türeci Ö, Sahin U. Targeting the tumor vasculature with engineered cystine-knot miniproteins. Nat Commun 2020; 11:295. [PMID: 31941901 PMCID: PMC6962393 DOI: 10.1038/s41467-019-13948-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 11/28/2019] [Indexed: 01/08/2023] Open
Abstract
The extra domain B splice variant (EDB) of human fibronectin selectively expressed in the tumor vasculature is an attractive target for cancer imaging and therapy. Here, we describe the generation and characterization of EDB-specific optical imaging probes. By screening combinatorial cystine-knot miniprotein libraries with phage display technology we discover exquisitely EDB-specific ligands that share a distinctive motif. Probes with a binding constant in the picomolar range are generated by chemical oligomerization of selected ligands and fluorophore conjugation. We show by fluorescence imaging that the probes stain EDB in tissue sections derived from human U-87 MG glioblastoma xenografts in mice. Moreover, we demonstrate selective accumulation and retention of intravenously administered probes in the tumor tissue of mice with U-87 MG glioblastoma xenografts by in vivo and ex vivo fluorescence imaging. These data warrants further pursuit of the selected cystine-knot miniproteins for in vivo imaging applications. Cystine-knot miniprotein are small, highly stable, disulfide-rich peptides with increasing potential as drugs and tumor imaging agents. Here the authors develop cystine-knot miniproteins targeting the vascular tumor marker EDB, and use them as probes for in vivo tumor vasculature imaging.
Collapse
|
9
|
Zhang B, Pang Z, Hu Y. Targeting hemostasis-related moieties for tumor treatment. Thromb Res 2020; 187:186-196. [PMID: 32032807 DOI: 10.1016/j.thromres.2020.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
Under normal conditions, the hemostatic system, that includes the involvement of the coagulation response and platelets, is anatomically and functionally inseparable from the vasculature. However, the hemostatic response always occurs in a wide range of tumors because of the high expression of coagulation initiator tissue factor (TF) in many tumor tissues, and due to the leakage of coagulation factors and platelets from the circulation system into the tumor interstitium through abnormal tumor vessels. Therefore, in addition to TF, these coagulation factors, platelets, the central moiety thrombin, the final product fibrin, and fibronectin, which is capable of stabilizing coagulation clots, are also abundant in tumors. These hemostasis-related moieties (HRMs), including TF, thrombin, fibrin, fibronectin, and platelets, are also closely associated with tumor progression, e.g., primary tumor growth and distal metastasis. The hemostatic response only occurs under pathological conditions, such as tumors, thrombosis, and atherosclerosis other than in normal tissues. The HRMs within tumors are also highly specific, establishing functional and therapeutic targets for tumor treatment. Therefore, strategies including active targeting to these moieties, modulation of HRMs deposited in the tumor microenvironment to improve tumor drug delivery, activation of prodrug by the coagulation complex formed during coagulation response, and direct inhibition of the tumor-promoting activity of HRMs could be designed for tumor therapy. In this review, we summarize various strategies that target HRMs for tumor treatment.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
10
|
Mashayekhi V, Hoog CO‘, Oliveira S. Vascular targeted photodynamic therapy: A review of the efforts towards molecular targeting of tumor vasculature. J PORPHYR PHTHALOCYA 2019; 23:1229-1240. [PMID: 33568892 PMCID: PMC7116708 DOI: 10.1142/s1088424619300180] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The therapeutic value of vascular targeted photodynamic therapy (VTP) for cancer has already been recognized in the clinic: TOOKAD® has been clinically approved in Europe and Israel for treatment of men with low-risk prostate cancer. When light is applied shortly after intravenous administration of the photosensitizer, the damage is primarily done to the vasculature. This results in vessel constriction, blood flow stasis, and thrombus formation. Subsequently, the tumor is killed due to oxygen and nutrient deprivation. To further increase treatment specificity and to reduce undesired side effects such as damaging to the surrounding healthy tissues, efforts have been made to selectively target the PS to the tumor vasculature, an approach named molecular targeted VTP (molVTP). Several receptors have already been explored for this approach, namely CD13, CD276, Extra domains of fibronectin (A, B), Integrin αvβ3, Neuropilin-1, Nucleolin, PDGFRβ, tissue factor, and VEGFR-2, which are overexpressed on tumor vasculature. Preclinical studies have shown promising results, further encouraging the investigation and future application of molVTP, to improve selectivity and efficacy of cancer treatment. This strategy will hopefully lead to even more selective treatments for many cancer patients.
Collapse
Affiliation(s)
- Vida Mashayekhi
- Division of Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Charlotte Op ‘t Hoog
- Division of Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Sabrina Oliveira
- Division of Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
- Pharmaceutics, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
11
|
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
|
12
|
Abstract
During cancer progression, the extracellular matrix (ECM) undergoes dramatic changes, which promote cancer cell migration and invasion. In the remodeled tumor ECM, fibronectin (FN) level is upregulated to assist tumor growth, progression, and invasion. FN serves as a central organizer of ECM molecules and mediates the crosstalk between the tumor microenvironment and cancer cells. Its upregulation is correlated with angiogenesis, cancer progression, metastasis, and drug resistance. A number of FN-targeting ligands have been developed for cancer imaging and therapy. Thus far, FN-targeting imaging agents have been tested for nuclear imaging, MRI, and fluorescence imaging, for tumor detection and localization. FN-targeting therapeutics, including nuclear medicine, chemotherapy drugs, cytokines, and photothermal moieties, were also developed in cancer therapy. Because of the prevalence of FN overexpression in cancer, FN targeting imaging agents and therapeutics have the promise of broad applications in the diagnosis, treatment, and image-guided interventions of many types of cancers. This review will summarize current understanding on the role of FN in cancer, discuss the design and development of FN-targeting agents, and highlight the applications of these FN-targeting agents in cancer imaging and therapy.
Collapse
Affiliation(s)
- Zheng Han
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
13
|
Costanza B, Umelo IA, Bellier J, Castronovo V, Turtoi A. Stromal Modulators of TGF-β in Cancer. J Clin Med 2017; 6:jcm6010007. [PMID: 28067804 PMCID: PMC5294960 DOI: 10.3390/jcm6010007] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/19/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is an intriguing cytokine exhibiting dual activities in malignant disease. It is an important mediator of cancer invasion, metastasis and angiogenesis, on the one hand, while it exhibits anti-tumor functions on the other hand. Elucidating the precise role of TGF-β in malignant development and progression requires a better understanding of the molecular mechanisms involved in its tumor suppressor to tumor promoter switch. One important aspect of TGF-β function is its interaction with proteins within the tumor microenvironment. Several stromal proteins have the natural ability to interact and modulate TGF-β function. Understanding the complex interplay between the TGF-β signaling network and these stromal proteins may provide greater insight into the development of novel therapeutic strategies that target the TGF-β axis. The present review highlights our present understanding of how stroma modulates TGF-β activity in human cancers.
Collapse
Affiliation(s)
- Brunella Costanza
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Ijeoma Adaku Umelo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Justine Bellier
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université Montpellier, Institut Régional du Cancer de Montpellier, 34298 Montpellier, France.
| |
Collapse
|
14
|
Jahanban-Esfahlan R, Seidi K, Zarghami N. Tumor vascular infarction: prospects and challenges. Int J Hematol 2017; 105:244-256. [DOI: 10.1007/s12185-016-2171-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022]
|
15
|
Chitgupi U, Qin Y, Lovell JF. Targeted Nanomaterials for Phototherapy. Nanotheranostics 2017; 1:38-58. [PMID: 29071178 PMCID: PMC5646723 DOI: 10.7150/ntno.17694] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/30/2016] [Indexed: 12/22/2022] Open
Abstract
Phototherapies involve the irradiation of target tissues with light. To further enhance selectivity and potency, numerous molecularly targeted photosensitizers and photoactive nanoparticles have been developed. Active targeting typically involves harnessing the affinity between a ligand and a cell surface receptor for improved accumulation in the targeted tissue. Targeting ligands including peptides, proteins, aptamers and small molecules have been explored for phototherapy. In this review, recent examples of targeted nanomaterials used in phototherapy are summarized.
Collapse
Affiliation(s)
| | | | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, USA
| |
Collapse
|
16
|
Fibronectin-targeted drug delivery in cancer. Adv Drug Deliv Rev 2016; 97:101-10. [PMID: 26639577 DOI: 10.1016/j.addr.2015.11.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/09/2023]
Abstract
Fibronectin is an extracellular matrix protein with pivotal physiological and pathological functions in development and adulthood. Alternative splicing of the precursor mRNA, produced from the single copy fibronectin gene, occurs at three sites coding for the EDA, EDB and IIICS domains. Fibronectin isoforms comprising the EDA or EDB domains are known as oncofetal forms due to their developmental importance and their re-expression in tumors, contrasting with restricted presence in normal adult tissues. These isoforms are also recognized as important markers of angiogenesis, a crucial physiological process in development and required by tumor cells in cancer progression. Attributed to this feature, EDA and EDB domains have been extensively used for the targeted delivery of cytokines, cytotoxic agents, chemotherapy drugs and radioisotopes to fibronectin-expressing tumors to exert therapeutic effects on primary cancers and metastatic lesions. In addition to drug delivery, the EDA and EDB domains of fibronectin have also been utilized to develop imaging strategies for tumor tissues. Furthermore, EDA and EDB based vaccines seem to be promising for the treatment and prevention of certain cancer types. In this review, we will summarize recent advances in fibronectin EDA and EDB-based therapeutic strategies developed to treat cancer.
Collapse
|
17
|
Pereira PMR, Korsak B, Sarmento B, Schneider RJ, Fernandes R, Tomé JPC. Antibodies armed with photosensitizers: from chemical synthesis to photobiological applications. Org Biomol Chem 2015; 13:2518-29. [PMID: 25612113 DOI: 10.1039/c4ob02334j] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Targeting photosensitizers to cancer cells by conjugating them with specific antibodies, able to recognize and bind to tumor-associated antigens, is today one of the most attractive strategies in photodynamic therapy (PDT). This comprehensive review updates on chemical routes available for the preparation of photo-immunoconjugates (PICs), which show dual chemical and biological functionalities: photo-properties of the photosensitizer and the immunoreactivity of the antibody. Moreover, photobiological results obtained with such photo-immunoconjugates using in vitro and in vivo cancer models are also discussed.
Collapse
Affiliation(s)
- Patricia M R Pereira
- QOPNA and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | | | | | | | | | | |
Collapse
|
18
|
Synthesis, bioanalysis and biodistribution of photosensitizer conjugates for photodynamic therapy. Bioanalysis 2014; 5:1099-114. [PMID: 23641699 DOI: 10.4155/bio.13.37] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Photodynamic therapy (PDT) was discovered in 1900 by Raab, and has since emerged as a promising tool for treating diseases characterized by unwanted cells or hyperproliferating tissue (e.g., cancer or infectious disease). PDT consists of the light excitation of a photosensitizer (PS) in the presence of O(2) to yield highly reactive oxygen species. In recent years, PDT has been improved by the synthesis of targeted bioconjugates between monoclonal antibodies and PS, and by investigating PS biodistribution and PD. Here, we provide a comprehensive review of major developments in PS-immunoconjugate-based PDT and the bioanalysis of these agents, with a specific emphasis on anticancer and antimicrobial PDT.
Collapse
|
19
|
Al-Omari S. Toward a molecular understanding of the photosensitizer-copper interaction for tumor destruction. Biophys Rev 2013; 5:305-311. [PMID: 28510111 PMCID: PMC5418419 DOI: 10.1007/s12551-013-0112-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/27/2013] [Indexed: 01/09/2023] Open
Abstract
The aim of this study was to shown that the photosensitizer in photodynamic therapy (PDT) can contribute to the dark toxicity and phototoxicity of the tumor by binding with copper. This binding process can remove the copper from the body, stopping angiogenesis as well as activating the mechanisms of cell death, such as apoptosis and necrosis. In PDT, this coupling may be considered a new route for fighting cancer in addition to those already known which involve reactive oxygen species.
Collapse
Affiliation(s)
- Saleh Al-Omari
- Department of Physics, Faculty of Science, The Hashemite University, Zarqa, 13115, Jordan.
| |
Collapse
|
20
|
Tuncel S, Trivella A, Atilla D, Bennis K, Savoie H, Albrieux F, Delort L, Billard H, Dubois V, Ahsen V, Caldefie-Chézet F, Richard C, Boyle RW, Ducki S, Dumoulin F. Assessing the Dual Activity of a Chalcone–Phthalocyanine Conjugate: Design, Synthesis, and Antivascular and Photodynamic Properties. Mol Pharm 2013; 10:3706-16. [DOI: 10.1021/mp400207v] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sinem Tuncel
- Department
of Chemistry, Gebze Institute of Technology, P.O. Box 141, 41400 Gebze,
Kocaeli, Turkey
| | - Aurélien Trivella
- Clermont Université,
Université Blaise Pascal, Institut de Chimie de Clermont-Ferrand,
Equipe Photochimie, BP 10448, F-63000 Clermont-Ferrand, France
- CNRS, UMR 6296,
ICCF, F-63171 Aubiere, France
| | - Devrim Atilla
- Department
of Chemistry, Gebze Institute of Technology, P.O. Box 141, 41400 Gebze,
Kocaeli, Turkey
| | - Khalil Bennis
- Clermont Université,
ENSCCF, Institut de Chimie de Clermont-Ferrand, Equipe CESMA, BP 10448, F-63000 Clermont-Ferrand, France
- CNRS, UMR 6296,
ICCF, F-63171 Aubiere, France
| | - Huguette Savoie
- Department
of Chemistry, University of Hull, Kingston-upon-Hull, East Yorkshire, HU6 7RX, U.K
| | - Florian Albrieux
- Centre Commun de Spectrométrie
de Masse UMR 5246, CNRS-Université Claude Bernard Lyon 1, Université
de Lyon, Bâtiment Curien, 43, bd du 11 Novembre, 69622 Villeurbanne Cedex, France
| | - Laetitia Delort
- Clermont
Université,
Université d’Auvergne, ECREIN-UNH, BP
10448, F-63000 Clermont-Ferrand, France
- INRA, UMR 1019,
UNH, F-63009 Clermont-Ferrand, France
| | - Hermine Billard
- Clermont
Université,
Université d’Auvergne, ECREIN-UNH, BP
10448, F-63000 Clermont-Ferrand, France
- INRA, UMR 1019,
UNH, F-63009 Clermont-Ferrand, France
| | - Virginie Dubois
- Clermont
Université,
Université d’Auvergne, ECREIN-UNH, BP
10448, F-63000 Clermont-Ferrand, France
- INRA, UMR 1019,
UNH, F-63009 Clermont-Ferrand, France
| | - Vefa Ahsen
- Department
of Chemistry, Gebze Institute of Technology, P.O. Box 141, 41400 Gebze,
Kocaeli, Turkey
| | - Florence Caldefie-Chézet
- Clermont
Université,
Université d’Auvergne, ECREIN-UNH, BP
10448, F-63000 Clermont-Ferrand, France
- INRA, UMR 1019,
UNH, F-63009 Clermont-Ferrand, France
| | - Claire Richard
- Clermont Université,
Université Blaise Pascal, Institut de Chimie de Clermont-Ferrand,
Equipe Photochimie, BP 10448, F-63000 Clermont-Ferrand, France
- CNRS, UMR 6296,
ICCF, F-63171 Aubiere, France
| | - Ross W. Boyle
- Department
of Chemistry, University of Hull, Kingston-upon-Hull, East Yorkshire, HU6 7RX, U.K
| | - Sylvie Ducki
- Clermont Université,
ENSCCF, Institut de Chimie de Clermont-Ferrand, Equipe CESMA, BP 10448, F-63000 Clermont-Ferrand, France
- CNRS, UMR 6296,
ICCF, F-63171 Aubiere, France
| | - Fabienne Dumoulin
- Department
of Chemistry, Gebze Institute of Technology, P.O. Box 141, 41400 Gebze,
Kocaeli, Turkey
| |
Collapse
|
21
|
List T, Neri D. Immunocytokines: a review of molecules in clinical development for cancer therapy. Clin Pharmacol 2013; 5:29-45. [PMID: 23990735 PMCID: PMC3753206 DOI: 10.2147/cpaa.s49231] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The concept of therapeutically enhancing the immune system’s responsiveness to tumors is
long standing. Several cytokines have been investigated in clinical trials for their therapeutic
activity in cancer patients. However, substantial side effects and unfavorable pharmacokinetic
properties have been a major drawback hampering the administration of therapeutically relevant
doses. The use of recombinant antibody–cytokine fusion proteins promises to significantly
enhance the therapeutic index of cytokines by targeting them to the site of disease. This review
aims to provide a concise and complete overview of the preclinical data and clinical results
currently available for all immunocytokines having reached clinical development.
Collapse
Affiliation(s)
- Thomas List
- Department of Chemistry and Applied Biosciences, Swiss Federal institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
22
|
|
23
|
Kitagishi H, Hatada S, Itakura T, Maki Y, Maeda Y, Kano K. Cellular uptake of octaarginine-conjugated tetraarylporphyrin included by per-O-methylated β-cyclodextrin. Org Biomol Chem 2013; 11:3203-11. [PMID: 23584796 DOI: 10.1039/c3ob27248f] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This paper describes the synthesis, structural characterization and cellular uptake of a supramolecular 1 : 2 inclusion complex of meso-tetraphenylporphyrin having an octaarginine peptide chain (R8-TPP) and heptakis(2,3,6-tri-O-methyl)-β-cyclodextrin (TMe-β-CD). R8-TPP was synthesized by 2 approaches: (1) on-resin conjugation of the N-terminal of octaarginine with 5-(4-carboxyphenyl)-10,15,20-triphenylporphyrin, followed by cleavage from the resin, and (2) Michael addition reaction between 5-[4-(3-maleimidopropylamido)phenyl]-10,15,20-triphenylporphyrin and cysteine-octaarginine peptide (Cys-Arg8). The R8-TPP obtained from both the approaches formed stable inclusion complexes with TMe-β-CD by which non-substituted phenyl groups at the 10- and 20-positions were included to form trans-type 1 : 2 inclusion complexes. The complexation prevented the self-aggregation of R8-TPP, which resulted in the solubilisation of R8-TPP in aqueous media. A cellular uptake study using HeLa cells showed that R8-TPP complexed with TMe-β-CD in a serum-free medium was efficiently taken up by the cells and uniformly dispersed in the cytosol. In the serum-containing medium, the R8-TPP-TMe-β-CD complex dissociated, and the serum protein bound R8-TPP. The R8-TPP-protein complex was localized in the endosomes of the cells. The cytosol-dispersed R8-TPP showed a higher photo-induced cytotoxicity than its endosome-trapped counterpart.
Collapse
Affiliation(s)
- Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University, Tatara, Kyotanabe, Kyoto 610-0321, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Sadasivam M, Avci P, Gupta GK, Lakshmanan S, Chandran R, Huang YY, Kumar R, Hamblin MR. Self-assembled liposomal nanoparticles in photodynamic therapy. EUROPEAN JOURNAL OF NANOMEDICINE 2013; 5. [PMID: 24348377 DOI: 10.1515/ejnm-2013-0010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Photodynamic therapy (PDT) employs the combination of non-toxic photosensitizers (PS) together with harmless visible light of the appropriate wavelength to produce reactive oxygen species that kill unwanted cells. Because many PS are hydrophobic molecules prone to aggregation, numerous drug delivery vehicles have been tested to solubilize these molecules, render them biocompatible and enhance the ease of administration after intravenous injection. The recent rise in nanotechnology has markedly expanded the range of these nanoparticulate delivery vehicles beyond the well-established liposomes and micelles. Self-assembled nanoparticles are formed by judicious choice of monomer building blocks that spontaneously form a well-oriented 3-dimensional structure that incorporates the PS when subjected to the appropriate conditions. This self-assembly process is governed by a subtle interplay of forces on the molecular level. This review will cover the state of the art in the preparation and use of self-assembled liposomal nanoparticles within the context of PDT.
Collapse
Affiliation(s)
- Magesh Sadasivam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA; Department of Dermatology, Harvard Medical School, Boston, MA, USA; and Department of Dermatology, Dermatooncology and Venerology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Gaurav K Gupta
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA; and Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | | | - Rakkiyappan Chandran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ying-Ying Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA; Department of Dermatology, Harvard Medical School, Boston, MA, USA; and Pathology Department, Guangxi Medical University, Nanning, Guangxi, China
| | - Raj Kumar
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA; and Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Michael R Hamblin
- Department of Dermatology, Harvard Medical School, Boston, MA, USA; and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| |
Collapse
|
25
|
Rumie Vittar NB, Lamberti MJ, Pansa MF, Vera RE, Rodriguez ME, Cogno IS, Milla Sanabria LN, Rivarola VA. Ecological photodynamic therapy: new trend to disrupt the intricate networks within tumor ecosystem. Biochim Biophys Acta Rev Cancer 2012; 1835:86-99. [PMID: 23127970 DOI: 10.1016/j.bbcan.2012.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Revised: 10/25/2012] [Accepted: 10/26/2012] [Indexed: 12/22/2022]
Abstract
As with natural ecosystems, species within the tumor microenvironment are connected by pairwise interactions (e.g. mutualism, predation) leading to a strong interdependence of different populations on each other. In this review we have identified the ecological roles played by each non-neoplastic population (macrophages, endothelial cells, fibroblasts) and other abiotic components (oxygen, extracellular matrix) directly involved with neoplastic development. A way to alter an ecosystem is to affect other species within the environment that are supporting the growth and survival of the species of interest, here the tumor cells; thus, some features of ecological systems could be exploited for cancer therapy. We propose a well-known antitumor therapy called photodynamic therapy (PDT) as a novel modulator of ecological interactions. We refer to this as "ecological photodynamic therapy." The main goal of this new strategy is the improvement of therapeutic efficiency through the disruption of ecological networks with the aim of destroying the tumor ecosystem. It is therefore necessary to identify those interactions from which tumor cells get benefit and those by which it is impaired, and then design multitargeted combined photodynamic regimes in order to orchestrate non-neoplastic populations against their neoplastic counterpart. Thus, conceiving the tumor as an ecological system opens avenues for novel approaches on treatment strategies.
Collapse
Affiliation(s)
- N Belén Rumie Vittar
- Universidad Nacional de Río Cuarto, Biología Molecular, Ruta 36 Km 601, Río Cuarto (5800), Córdoba, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
26
|
The immunocytokine L19-IL2 eradicates cancer when used in combination with CTLA-4 blockade or with L19-TNF. J Invest Dermatol 2012; 133:751-758. [PMID: 23096716 DOI: 10.1038/jid.2012.376] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Systemic high-dose IL2 promotes long-term survival in a subset of metastatic melanoma patients, but this treatment is accompanied by severe toxicities. The immunocytokine L19-IL2, in which IL2 is fused to the human L19 antibody capable of selective accumulation on tumor neovasculature, has recently shown encouraging clinical activity in patients with metastatic melanoma. In this study, we have investigated the therapeutic performance of L19-IL2, administered systemically in combination with a murine anti-CTLA-4 antibody or with a second clinical-stage immunocytokine (L19-TNF) in two syngeneic immunocompetent mouse models of cancer. We observed complete tumor eradications when L19-IL2 was used in combination with CTLA-4 blockade. Interestingly, mice cured from F9 tumors developed new lesions when rechallenged with tumor cells after therapy, whereas mice cured from CT26 tumors were resistant to tumor rechallenge. Similarly, L19-IL2 induced complete remissions when administered in a single intratumoral injection in combination with L19-TNF, whereas the two components did not lead to cures when administered as single agents. These findings provide a rationale for combination trials in melanoma, as the individual therapeutic agents have been extensively studied in clinical trials, and the antigen recognized by the L19 antibody has an identical sequence in mouse and man.
Collapse
|
27
|
Weiss A, den Bergh HV, Griffioen AW, Nowak-Sliwinska P. Angiogenesis inhibition for the improvement of photodynamic therapy: the revival of a promising idea. Biochim Biophys Acta Rev Cancer 2012; 1826:53-70. [PMID: 22465396 DOI: 10.1016/j.bbcan.2012.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Revised: 03/13/2012] [Accepted: 03/14/2012] [Indexed: 12/31/2022]
Abstract
Photodynamic therapy (PDT) is a minimally invasive form of treatment, which is clinically approved for the treatment of angiogenic disorders, including certain forms of cancer and neovascular eye diseases. Although the concept of PDT has existed for a long time now, it has never made a solid entrance into the clinical management of cancer. This is likely due to secondary tissue reactions, such as inflammation and neoangiogenesis. The recent development of clinically effective angiogenesis inhibitors has lead to the initiation of research on the combination of PDT with such angiostatic targeted therapies. Preclinical studies in this research field have shown promising results, causing a revival in the field of PDT. This review reports on the current research efforts on PDT and vascular targeted combination therapies. Different combination strategies with angiogenesis inhibition and vascular targeting approaches are discussed. In addition, the concept of increasing PDT selectivity by targeted delivery of photosensitizers is presented. Furthermore, the current insights on sequencing the therapy arms of such combinations will be discussed in light of vascular normalization induced by angiogenesis inhibition.
Collapse
Affiliation(s)
- Andrea Weiss
- Medical Photonics Group, Institute of Bioengineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | | | | | | |
Collapse
|
28
|
Tuncel S, Fournier-dit-Chabert J, Albrieux F, Ahsen V, Ducki S, Dumoulin F. Towards dual photodynamic and antiangiogenic agents: design and synthesis of a phthalocyanine-chalcone conjugate. Org Biomol Chem 2012; 10:1154-7. [DOI: 10.1039/c2ob06809e] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Shim G, Lee S, Kim YB, Kim CW, Oh YK. Enhanced tumor localization and retention of chlorin e6 in cationic nanolipoplexes potentiate the tumor ablation effects of photodynamic therapy. NANOTECHNOLOGY 2011; 22:365101. [PMID: 21841215 DOI: 10.1088/0957-4484/22/36/365101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Here we report the tumor ablation effects of the negatively charged photosensitizer chlorin e6 (Ce6) in nanocomplexes. Ce6 was complexed to cationic 1,2-dioleoyl-sn-glycero-3-ethylphosphocholine-based liposomes, forming cationic nanolipoplexes. The loading efficiency of Ce6 to cationic nanolipoplexes was greater than 90%. The degree of enhancement of cellular uptake of Ce6 by treatment in cationic nanolipoplexes increased with the concentration of Ce6, showing 18.3-fold higher uptake than free Ce6 at 15 µM. Molecular imaging revealed the preferential distribution and retention of Ce6 in SCC7 tumor tissues after intravenous administration of Ce6 in cationic nanolipoplexes. Moreover, localized illumination of mice receiving Ce6 in cationic nanolipoplexes resulted in the formation of thick scabs over tumor regions, and complete ablation of tumors after scab detachment. In contrast, continuous growth of tumors was observed in the group treated with free Ce6. Our results suggest that the cationic nanolipoplexes of Ce6 improve the therapeutic effects of photodynamic cancer therapy as compared to free Ce6.
Collapse
Affiliation(s)
- Gayong Shim
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | | | | | | |
Collapse
|
30
|
Srivatsan A, Ethirajan M, Pandey SK, Dubey S, Zheng X, Liu TH, Shibata M, Missert J, Morgan J, Pandey RK. Conjugation of cRGD peptide to chlorophyll a based photosensitizer (HPPH) alters its pharmacokinetics with enhanced tumor-imaging and photosensitizing (PDT) efficacy. Mol Pharm 2011; 8:1186-97. [PMID: 21702452 PMCID: PMC3148296 DOI: 10.1021/mp200018y] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The α(v)β(3) integrin receptor plays an important role in human metastasis and tumor-induced angiogenesis. Cyclic Arg-Gly-Asp (cRGD) peptide represents a selective α(v)β(3) integrin ligand that has been extensively used for research, therapy, and diagnosis of neoangiogenesis. For developing photosensitizers with enhanced PDT efficacy, we here report the synthesis of a series of bifunctional agents in which the 3-(1'-hexyloxyethyl)-3-devinylpyropheophorbide a (HPPH), a chlorophyll-based photosensitizer, was conjugated to cRGD and the related analogues. The cell uptake and in vitro PDT efficacy of the conjugates were studied in α(v)β(3) integrin overexpressing U87 and 4T1 cell lines whereas the in vivo PDT efficacy and fluorescence-imaging potential of the conjugates were compared with the corresponding nonconjugated photosensitizer HPPH in 4T1 tumors. Compared to HPPH, the HPPH-cRGD conjugate in which the arginine and aspartic acid moieties were available for binding to two subunits of α(v)β(3) integrin showed faster clearance, enhanced tumor imaging and enhanced PDT efficacy at 2-4 h postinjection. Molecular modeling studies also confirmed that the presence of the HPPH moiety in HPPH-cRGD conjugate does not interfere with specific recognition of cRGD by α(v)β(3) integrin. Compared to U87 and 4T1 cells the HPPH-cRGD showed significantly low photosensitizing efficacy in A431 (α(v)β(3) negative) tumor cells, suggesting possible target specificity of the conjugate.
Collapse
Affiliation(s)
- Avinash Srivatsan
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
A chemically modified antibody mediates complete eradication of tumours by selective disruption of tumour blood vessels. Br J Cancer 2011; 104:1106-15. [PMID: 21386847 PMCID: PMC3068510 DOI: 10.1038/bjc.2011.78] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background: The possibility of eradicating cancer by selective destruction of tumour blood vessels may represent an attractive therapeutic avenue, but most pharmaceutical agents investigated so far did not achieve complete cures and are not completely specific. Antibody conjugates now allow us to evaluate the impact of selective vascular shutdown on tumour viability and to study mechanisms of action. Methods: We synthesised a novel porphyrin-based photosensitiser suitable for conjugation to antibodies and assessed anticancer properties of its conjugate with L19, a clinical-stage human monoclonal antibody specific to the alternatively spliced EDB domain of fibronectin, a marker of tumour angiogenesis. Results: Here we show in two mouse model of cancer (F9 and A431) that L19 is capable of highly selective in vivo localisation around tumour blood vessels and that its conjugate with a photosensitiser allows selective disruption of tumour vasculature upon irradiation, leading to complete and long-lasting cancer eradication. Furthermore, depletion experiments revealed that natural killer cells are essential for the induction of long-lasting complete responses. Conclusions: These results reinforce the concept that vascular shutdown can induce a curative avalanche of tumour cell death. Immuno-photodynamic therapy may be particularly indicated for squamous cell carcinoma of the skin, which we show to be strongly positive for markers of angiogenesis.
Collapse
|
32
|
Bullous AJ, Alonso CMA, Boyle RW. Photosensitiser–antibody conjugates for photodynamic therapy. Photochem Photobiol Sci 2011; 10:721-50. [DOI: 10.1039/c0pp00266f] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
33
|
Constantinou A, Epenetos AA, Hreczuk-Hirst D, Jain S, Wright M, Chester KA, Deonarain MP. Site-specific polysialylation of an antitumor single-chain Fv fragment. Bioconjug Chem 2010; 20:924-31. [PMID: 19402707 DOI: 10.1021/bc8005122] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Protein pharmacokinetic modulation is becoming an important tool in the development of biotherapeutics. Proteins can be chemically or recombinantly modified to alter their half-lives and bioavailability to suit particular applications as well as improve side effect profiles. The most successful and clinically used approach to date is chemical conjugation with poly(ethylene glycol) polymers (PEGylation). Here, therapeutic protein half-life can be increased significantly while retaining biological function, reducing immunogenicity and cross-reaction. Naturally occurring alternatives to such synthetic polymers could have major advantages such as lower side effects due to biodegradability and metabolism. Polysialic acid (PSA) has been investigated as a pharmacokinetic modulatory biopolymer with many successful examples in preclinical and clinical development. Single-chain Fvs (scFvs) are a choice antibody format for human therapeutic antibody discovery. Because of their small size, they are rapidly eliminated from the circulation and often are rebuilt into larger proteins for drug development and a longer half-life. Here we show that chemical polysialylation can increase the half-life of an antiplacental alkaline (PLAP) and anticarcinoembryonic antigen (CEA) scFv (F1 and MFE-23, respectively) 3.4-4.9-fold, resulting in a 10.6-15.2-fold increase in blood exposure. Amine-directed coupling of the MFE-23 scFv reduced its immunoreactivity 20-fold which was resolved by site-specific polysialylation through an engineered C-terminal thiol residue. The site-specifically polysialylated MFE-23 scFv demonstrated up to 30-fold improved tumor uptake while displaying favorable tumor:normal tissue specificity. This suggests that engineering antibody fragments for site-specific polysialylation could be a useful approach to increase the half-life for a variety of therapeutic applications.
Collapse
Affiliation(s)
- A Constantinou
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | | | | | | | | | | | | |
Collapse
|
34
|
Roesli C, Neri D. Methods for the identification of vascular markers in health and disease: from the bench to the clinic. J Proteomics 2010; 73:2219-29. [PMID: 20541635 DOI: 10.1016/j.jprot.2010.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 05/25/2010] [Accepted: 05/30/2010] [Indexed: 02/07/2023]
Abstract
Several diseases are characterized by changes in the molecular composition of vascular structures, thus offering the opportunity to use specific ligands (e.g., monoclonal antibodies) for imaging and therapy application. This novel pharmaceutical strategy, often referred to as "vascular targeting", promises to facilitate the discovery and development of selective biopharmaceuticals for the management of angiogenesis-related diseases. This article reviews novel biomedical applications based on vascular targeting strategies, as well as methodologies which have been used for the discovery of vascular markers of pathology.
Collapse
Affiliation(s)
- Christoph Roesli
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.
| | | |
Collapse
|
35
|
Trüssel S, Dumelin C, Frey K, Villa A, Buller F, Neri D. New strategy for the extension of the serum half-life of antibody fragments. Bioconjug Chem 2010; 20:2286-92. [PMID: 19916518 DOI: 10.1021/bc9002772] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Antibody fragments can recognize their cognate antigen with high affinity and can be produced at high yields, but generally display rapid blood clearance profiles. For pharmaceutical applications, the serum half-life of antibody fragments is often extended by chemical modification with polymers or by genetic fusion to albumin or albumin-binding polypeptides. Here, we report that the site-specific chemical modification of a C-terminal cysteine residue in scFv antibody fragments with a small organic molecule capable of high-affinity binding to serum albumin substantially extends serum half-life in rodents. The strategy was implemented using the antibody fragment F8, specific to the alternatively spliced EDA domain of fibronectin, a tumor-associated antigen. The unmodified and chemically modified scFv-F8 antibody fragments were studied by biodistribution analysis in tumor-bearing mice, exhibiting a dramatic increase in tumor uptake for the albumin-binding antibody derivative. The data presented in this paper indicate that the chemical modification of the antibody fragment with the 2-(3-maleimidopropanamido)-6-(4-(4-iodophenyl)butanamido)hexanoate albumin-binding moiety may represent a general strategy for the extension of the serum half-life of antibody fragments and for the improvement of their in vivo targeting performance.
Collapse
Affiliation(s)
- Sabrina Trüssel
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, and Philochem AG, ETH Zurich, Wolfgang-Pauli-Str. 10, CH-8093 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
36
|
Albini A, Indraccolo S, Noonan DM, Pfeffer U. Functional genomics of endothelial cells treated with anti-angiogenic or angiopreventive drugs. Clin Exp Metastasis 2010; 27:419-39. [PMID: 20383568 DOI: 10.1007/s10585-010-9312-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 02/16/2010] [Indexed: 01/28/2023]
Abstract
Angiogenesis is a highly regulated physiological process that has been studied in considerable detail given its importance in several chronic pathologies. Many endogenous factors and hormones intervene in the regulation of angiogensis and classical as well as targeted drugs have been developed for its control. Angiogenesis inhibition has come off the bench and entered into clinical application for cancer therapy, particularly for metastatic disease. While the clinical benefit is currently in terms of months, preclinical data suggest that novel drugs and drug combinations could lead to substantial improvement. The many targets of endogenous angiogenesis inhibitors reflect the complexity of the process; in contrast, current clinical therapies mainly target the vascular endothelial growth factor system. Cancer chemopreventive compounds can retard tumor insurgence and delay or prevent metastasis and many of these molecules hinder angiogenesis, a mechanism that we termed angioprevention. Angiopreventive drugs appear to prevalently act through the inhibition of the pro-inflammatory and anti-apoptotic player NFkappaB, thus contrasting inflammation dependent angiogenesis. Relatively little is known concerning the effects of these angiogenesis inhibitors on gene expression of endothelial cells, the main target of many of these molecules. Here we provide an exhaustive list of anti-angiogenic molecules, and summarize their effects, where known, on the transcriptome and functional genomics of endothelial cells. The regulation of specific genes can be crucial to preventive or therapeutic intervention. Further, novel targets might help to circumvent resistance to anti-angiogenic therapy. The studies we review are relevant not only to cancer but also to other chronic degenerative diseases involving endothelial cells, such as cardiovascular disorders, diabetes, rheumatoid arthritis and retinopaties, as well as vessel aging.
Collapse
Affiliation(s)
- Adriana Albini
- MultiMedica Castellanza (VA) and Oncology Research, IRCCS MultiMedica, 20138 Milan, Italy.
| | | | | | | |
Collapse
|
37
|
Alonso CMA, Palumbo A, Bullous AJ, Pretto F, Neri D, Boyle RW. Site-Specific and Stoichiometric Conjugation of Cationic Porphyrins to Antiangiogenic Monoclonal Antibodies. Bioconjug Chem 2010; 21:302-13. [DOI: 10.1021/bc9003537] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Cristina M. A. Alonso
- Department of Chemistry, University of Hull, Cottingham Road, HU6 7RX, Kingston-upon-Hull, United Kingdom, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, HCI G396, 8093 Zurich, Switzerland
| | - Alessandro Palumbo
- Department of Chemistry, University of Hull, Cottingham Road, HU6 7RX, Kingston-upon-Hull, United Kingdom, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, HCI G396, 8093 Zurich, Switzerland
| | - Aaron J. Bullous
- Department of Chemistry, University of Hull, Cottingham Road, HU6 7RX, Kingston-upon-Hull, United Kingdom, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, HCI G396, 8093 Zurich, Switzerland
| | - Francesca Pretto
- Department of Chemistry, University of Hull, Cottingham Road, HU6 7RX, Kingston-upon-Hull, United Kingdom, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, HCI G396, 8093 Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry, University of Hull, Cottingham Road, HU6 7RX, Kingston-upon-Hull, United Kingdom, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, HCI G396, 8093 Zurich, Switzerland
| | - Ross W. Boyle
- Department of Chemistry, University of Hull, Cottingham Road, HU6 7RX, Kingston-upon-Hull, United Kingdom, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich, Wolfgang-Pauli-Str. 10, HCI G396, 8093 Zurich, Switzerland
| |
Collapse
|
38
|
Sibani SA, McCarron PA, Woolfson AD, Donnelly RF. Photosensitiser delivery for photodynamic therapy. Part 2: systemic carrier platforms. Expert Opin Drug Deliv 2009; 5:1241-54. [PMID: 18976134 DOI: 10.1517/17425240802444673] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The treatment of solid tumours and angiogenic ocular diseases by photodynamic therapy (PDT) requires the injection of a photosensitiser (PS) to destroy target cells through a combination of visible light irradiation and molecular oxygen. There is currently great interest in the development of efficient and specific carrier delivery platforms for systemic PDT. OBJECTIVE This article aims to review recent developments in systemic carrier delivery platforms for PDT, with an emphasis on target specificity. METHODS Recent publications, spanning the last five years, concerning delivery carrier platforms for systemic PDT were reviewed, including PS conjugates, dendrimers, micelles, liposomes and nanoparticles. RESULTS/CONCLUSION PS conjugates and supramolecular delivery platforms can improve PDT selectivity by exploiting cellular and physiological specificities of the targeted tissue. Overexpression of receptors in cancer and angiogenic endothelial cells allows their targeting by affinity-based moieties for the selective uptake of PS conjugates and encapsulating delivery carriers, while the abnormal tumour neovascularisation induces a specific accumulation of heavy weighted PS carriers by enhanced permeability and retention (EPR) effect. In addition, polymeric prodrug delivery platforms triggered by the acidic nature of the tumour environment or the expression of proteases can be designed. Promising results obtained with recent systemic carrier platforms will, in due course, be translated into the clinic for highly efficient and selective PDT protocols.
Collapse
Affiliation(s)
- Stéphane A Sibani
- Queens University Belfast, Medical Biology Centre, School of Pharmacy, 97 Lisburn Road, Belfast BT9 7BL, UK
| | | | | | | |
Collapse
|
39
|
Scott N, Reynolds CB, Wright MJ, Qazi O, Fairweather N, Deonarain MP. Single-chain Fv phage display propensity exhibits strong positive correlation with overall expression levels. BMC Biotechnol 2008; 8:97. [PMID: 19113995 PMCID: PMC2630973 DOI: 10.1186/1472-6750-8-97] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 12/29/2008] [Indexed: 01/09/2023] Open
Abstract
Background Single chain Fvs (scFvs) are widely applied in research, diagnostics and therapeutic settings. Display and selection from combinatorial libraries is the main route to their discovery and many factors influence the success of this process. They exhibit low thermodynamic stability, resulting in low levels of premature cytosolic folding or aggregation which facilitates sec YEG-mediated translocation and phage in E. coli. However, there is little data analysing how this is related to and influenced by scFv protein expression. Results We characterised the relationship between overall scFv expression and display propensity for a panel of 15 anti-tetanus toxin scFvs and found a strong positive correlation (Rho = 0.88, p < 0.005) between the two parameters. Display propensity, overall expression and soluble localisation to the periplasm and extracellular fractions were clone specific characteristics which varied despite high levels of sequence homology. There was no correlation between display of scFv or its expression in non-fused (free) form with soluble scFv localisation to the periplasm or culture supernatant. This suggests that divergence in the fate of scFv-pIII and non-fused scFv after translocation to the periplasm accounts for the observed disparity. Differential degrees of periplasmic aggregation of non-fused scFv between clones may affect the partitioning of scFv in the periplasm and culture supernatant abrogating any correlation. We suggest that these factors do not apply to the scFv-pIII fusion since it remains anchored to the bacterial inner membrane as part of the innate phage packaging and budding process. Conclusion We conclude that in the absence of premature cytosolic aggregation or folding, the propensity of a scFv to be displayed on phage is directly related to its overall expression level and is thus indirectly influenced by factors such as codon bias, mRNA abundance or putative DNA motifs affecting expression. This suggests that scFvs capable of high overall expression and display levels may not produce high yields of non phage-fused soluble protein in either the periplasmic or extracellular fractions of E. coli. This should be considered when screening clones selected from combinatorial libraries for further study. The nucleotide and amino acid sequences of the anti-tetanus toxin scFvs have been deposited in the EMBL data base: accession numbers-C1: AM749134, C2: AM749135, C3: AM749136, C4: AM749137, C5: AM749138, N1: AM749139, N2: AM749140, N3: AM749141, N4: AM749142, N5: AM749143 J1; AM749144, J2: AM749145, J3: AM749146, J4: AM749147, J5: AM749148.
Collapse
Affiliation(s)
- Nathan Scott
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.
| | | | | | | | | | | |
Collapse
|
40
|
Rybak JN, Trachsel E, Scheuermann J, Neri D. Ligand-based vascular targeting of disease. ChemMedChem 2008; 2:22-40. [PMID: 17154429 DOI: 10.1002/cmdc.200600181] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This review illustrates the basic principles of ligand-based vascular targeting and presents some of the most advanced results obtained in this field, not only in terms of biopharmaceuticals, which are currently being investigated in clinical and preclinical studies, but also in terms of enabling technologies that facilitate target and ligand discovery. Whereas most of the vascular targeting research activities have so far concentrated on tumoral angiogenesis, the development of non-oncological applications has recently gained momentum and is likely to become an important area of modern pharmaceutical research.
Collapse
Affiliation(s)
- Jascha-N Rybak
- ETH Zürich, Institute of Pharmaceutical Sciences, Zürich, Switzerland
| | | | | | | |
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
|
Bhatti M, Yahioglu G, Milgrom LR, Garcia-Maya M, Chester KA, Deonarain MP. Targeted photodynamic therapy with multiply-loaded recombinant antibody fragments. Int J Cancer 2008; 122:1155-63. [PMID: 17973256 DOI: 10.1002/ijc.23206] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Current photodynamic therapy (PDT) of cancer is limited by inefficiencies involved in specifically targeting photosensitizers to tumors. Although antibodies are being explored as targeting vehicles, they present significant challenges, particularly in terms of pharmacokinetics and drug-coupling. We describe here a novel and effective system to covalently attach multiple photosensitizer molecules (both preclinical, pyropheophorbide-a and clinically approved, verteporfin photosensitizers) to single-chain Fvs. Further, we demonstrate that not only do the resulting photoimmunoconjugates retain photophysical functionality, they are more potent than either free photosensitizer, effectively killing tumor cells in vitro and in vivo. For example, treatment of human breast cancer xenografts with a photoimmunoconjugate comprising an anti-HER-2 scFv linked to 8-10 molecules of pyropheophorbide-a leads to significant tumor regression. These results give an insight into the important features that make scFvs good carriers for PDT drugs and provide proof of concept of our unique approach to targeted photodynamic therapy (tPDT). This promises to significantly improve on current photodynamic therapies for the treatment of cancer.
Collapse
Affiliation(s)
- Manpreet Bhatti
- Division of Cell and Molecular Biology, Faculty of Natural Sciences, Imperial College London, Exhibition Road, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
For the treatment of tumours and other proliferative conditions, widespread uptake of photodynamic therapy (PDT) has to some extent been hindered by its inability to target specifically photosensitisers (PSs) to localised lesions in the body. PSs may be deposited in the skin, leading to painful and disfiguring photosensitivity, sometimes for weeks after initial treatment. Targeting PSs specifically could not only avoid such side-effects, it could greatly improve PDT's therapeutic margin. This review describes photoimmunoconjugates (PICs) produced via successful combination of PSs with recombinant monoclonal antibody fragments (sc-Fvs). PICs can not only target specifically and destroy tumour cells in vitro and in vivo, but counter-intuitively, it is possible to conjugate many more PSs to an sc-Fv than to the much larger parent monoclonal antibody. The general utility of PICs is demonstrated by significant improvements to the potency and selectivity of already existing PSs. Furthermore, critical features of sc-Fvs are discussed that enable them to make effective PICs. This has implications for the future engineering of scFv carriers for PDT, in order to control the number and function of the PSs that can be coupled.
Collapse
Affiliation(s)
- Lionel R Milgrom
- Department of Life Sciences, Biochemistry Building, Imperial College London, UK.
| |
Collapse
|
44
|
Abstract
The growth of new blood vessels may be either beneficial or harmful. The angiogenic process may be measured by a variety of techniques, although it may often be the quality rather than quantity of resulting blood vessels that determines function. Endothelial cells play a key role in the initiation of angiogenesis, and vascular endothelial growth factor (VEGF) may be viewed as a prototypical direct-acting angiogenic factor. VEGF acts through multiple cell surface receptors and signaling pathways to stimulate endothelial cell proliferation, survival, and migration. By inducing other growth factor expression, VEGF stimulates a cascade of angiogenic activity. Different tissues may utilize various angiogenic pathways that are modulated by diverse host tissue responses. Furthermore, a single tissue may progress through a sequence of angiogenic pathways, for example, as acute injury progresses to chronic inflammation. The phenotype of the resulting neovasculature is critically dependent on the context in which it is formed. Biomarkers of angiogenesis are being developed as an aid to assessing human disease. Histological assessment of vascular density and angiogenic factor expression, in vivo imaging, Doppler ultrasound, and biofluid assays each may have clinical utility. Therapeutic targeting of angiogenesis will depend both on the generation of acceptable pharmacological agents and on the identification of patients who may and do gain benefit from such treatments.
Collapse
Affiliation(s)
- David A Walsh
- Academic Rheumatology, University of Nottingham Clinical Sciences Building, Nottingham City Hospital, Nottingham NG5 1PB, United Kingdom
| |
Collapse
|
45
|
Kuimova MK, Bhatti M, Deonarain M, Yahioglu G, Levitt JA, Stamati I, Suhling K, Phillips D. Fluorescence characterisation of multiply-loaded anti-HER2 single chain Fv-photosensitizer conjugates suitable for photodynamic therapy. Photochem Photobiol Sci 2007; 6:933-9. [PMID: 17721591 DOI: 10.1039/b708320c] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report the synthesis, spectroscopic properties and intracellular imaging of recombinant antibody single chain fragment (scFv) conjugates with photosensitizers used for photodynamic therapy of cancer (PDT). Two widely-studied photosensitizers have been selected: preclinical pyropheophorbide-a (PPa) and verteporfin (VP), which has been clinically approved for the treatment of acute macular degeneration (Visudyne). Pyropheophorbide-a and verteporfin have been conjugated to an anti-HER2 scFv containing on average ten photosensitizer molecules per scFv with a small contribution (<or=20%) from non-covalently bound molecules. Confocal fluorescence microscopy demonstrates good cellular uptake of PPa conjugate with the HER2-positive cell line, SKOV-3, while negligible cell uptake is demonstrated for the HER2-negative cell line, KB. For the VP conjugate, increased rate of cellular uptake and prolonged retention in SKOV-3 cells is observed compared to free photosensitizer. In clinical applications this could provide increased potency and desired selectivity towards malignant tissue, leaving surrounding healthy tissue unharmed and reducing skin photosensitivity. The present study highlights the usefulness of photosensitizer immunoconjugates with scFvs for targeted PDT.
Collapse
Affiliation(s)
- Marina K Kuimova
- Chemistry Department, Imperial College London, Exhibition Road, UK
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kaspar M, Trachsel E, Neri D. The antibody-mediated targeted delivery of interleukin-15 and GM-CSF to the tumor neovasculature inhibits tumor growth and metastasis. Cancer Res 2007; 67:4940-8. [PMID: 17510424 DOI: 10.1158/0008-5472.can-07-0283] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor-targeting immunocytokines represent a new class of anticancer pharmaceutical agents, which often display a superior therapeutic index compared with the corresponding unconjugated cytokines. In this article, we have studied the anticancer properties of interleukin-15 (IL-15) and granulocyte macrophage colony-stimulating factor (GM-CSF), fused to the human antibody fragment scFv(L19), specific to the EDB domain of fibronectin, a marker of angiogenesis. The immunocytokines L19-IL-15 and L19-GM-CSF were expressed in mammalian cells and purified to homogeneity, revealing no loss of cytokine activity in in vitro assays. Furthermore, the ability of the two immunocytokines to selectively localize to tumors in vivo was confirmed by biodistribution analysis with radioiodinated protein preparations. L19-IL-15 and L19-GM-CSF displayed a potent antitumor activity both in s.c. and in metastatic F9 and C51 murine models of cancer in immunocompetent mice. This therapeutic action was superior compared with IL-15-based and GM-CSF-based fusion proteins, containing antibodies of irrelevant specificity in the mouse, which were used as non-tumor-targeting controls. For both L19-IL-15 and L19-GM-CSF immunocytokines, CD8(+) T cells seemed to mostly contribute to the therapeutic action as shown by in vivo cell depletion experiments. The results presented in this article are of clinical significance, considering the fact that the sequence of EDB is identical in mouse and man and that the tumor-targeting ability of the L19 antibody has been extensively shown in clinical trials in patients with cancer.
Collapse
Affiliation(s)
- Manuela Kaspar
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | | |
Collapse
|
47
|
El-Emir E, Dearling JLJ, Huhalov A, Robson MP, Boxer G, Neri D, van Dongen GAMS, Trachsel E, Begent RHJ, Pedley RB. Characterisation and radioimmunotherapy of L19-SIP, an anti-angiogenic antibody against the extra domain B of fibronectin, in colorectal tumour models. Br J Cancer 2007; 96:1862-70. [PMID: 17519905 PMCID: PMC2359968 DOI: 10.1038/sj.bjc.6603806] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Angiogenesis is a characteristic feature of tumours and other disorders. The human monoclonal antibody L19- SIP targets the extra domain B of fibronectin, a marker of angiogenesis expressed in a range of tumours. The aim of this study was to investigate whole body distribution, tumour localisation and the potential of radioimmunotherapy with the L19-small immunoprotein (SIP) in colorectal tumours. Two colorectal tumour models with highly different morphologies, the SW1222 and LS174T xenografts, were used in this study. Localisation and retention of the L19-SIP antibody at tumour vessels was demonstrated using immunohistochemistry and Cy3-labelled L19-SIP. Whole body biodistribution studies in both tumour models were carried out with 125I-labelled L19-SIP. Finally, 131I-labelled antibody was used to investigate the potential of radioimmunotherapy in SW1222 tumours. Using immunohistochemistry, we confirmed extra domain B expression in the tumour vasculature. Immunofluorescence demonstrated localisation and retention of injected Cy3-labelled L19-SIP at the abluminal side of tumour vessels. Biodistribution studies using a 125I-labelled antibody showed selective tumour uptake in both models. Higher recorded values for localisation were found in the SW1222 tumours than in the LS174T (7.9 vs 6.6 %ID g−1), with comparable blood clearance for both models. Based on these results, a radioimmunotherapy study was performed in the SW1222 xenograft using 131I-Labelled L19-SIP (55.5 MBq), which showed selective tumour uptake, tumour growth inhibition and improved survival. Radio- and fluorescence-labelled L19-SIP showed selective localisation and retention at vessels of two colorectal xenografts. Furthermore, 131I-L19-SIP shows potential as a novel treatment of colorectal tumours, and provides the foundation to investigate combined therapies in the same tumour models.
Collapse
Affiliation(s)
- E El-Emir
- 1Cancer Research UK Targeting and Imaging Group, Department of Oncology, Royal Free and University College Medical School (UCL), Hampstead Campus, Rowland Hill Street, London NW3 2PF, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Trachsel E, Bootz F, Silacci M, Kaspar M, Kosmehl H, Neri D. Antibody-mediated delivery of IL-10 inhibits the progression of established collagen-induced arthritis. Arthritis Res Ther 2007; 9:R9. [PMID: 17261171 PMCID: PMC1860067 DOI: 10.1186/ar2115] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 01/12/2007] [Accepted: 01/29/2007] [Indexed: 11/10/2022] Open
Abstract
The antibody-mediated targeted delivery of cytokines to sites of disease is a promising avenue for cancer therapy, but it is largely unexplored for the treatment of chronic inflammatory conditions. Using both radioactive and fluorescent techniques, the human monoclonal antibodies L19 and G11 (specific to two markers of angiogenesis that are virtually undetectable in normal adult tissues) were found to selectively localize at arthritic sites in the murine collagen-induced model of rheumatoid arthritis following intravenous (i.v.) administration. The same animal model was used to study the therapeutic action of the L19 antibody fused to the cytokines IL-2, tumour necrosis factor (TNF) and IL-10. Whereas L19-IL-2 and L19-TNF treatment led to increased arthritic scores and paw swellings, the fusion protein L19-IL-10 displayed a therapeutic activity, which was superior to the activity of IL-10 fused to an antibody of irrelevant specificity in the mouse. The anti-inflammatory cytokine IL-10 has been investigated for the treatment of patients with rheumatoid arthritis, but clinical development plans have been discontinued because of a lack of efficacy. Because the antigen recognised by L19 is strongly expressed at sites of arthritis in humans and identical in both mice and humans, it suggests that the fusion protein L19-IL-10 might help overcome some of the clinical limitations of IL-10 and provide a therapeutic benefit to patients with chronic inflammatory disorders, including arthritis.
Collapse
Affiliation(s)
- Eveline Trachsel
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Wolfgang-Paulistrasse 10, CH-8093 Zurich, Switzerland
| | - Frank Bootz
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Wolfgang-Paulistrasse 10, CH-8093 Zurich, Switzerland
| | - Michela Silacci
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Wolfgang-Paulistrasse 10, CH-8093 Zurich, Switzerland
| | - Manuela Kaspar
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Wolfgang-Paulistrasse 10, CH-8093 Zurich, Switzerland
| | - Hartwig Kosmehl
- Institute of Pathology, Helios Klinikum Erfurt, Nordhaeuser Strasse 74, D-99089 Erfurt, Germany
| | - Dario Neri
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Wolfgang-Paulistrasse 10, CH-8093 Zurich, Switzerland
| |
Collapse
|
49
|
Trachsel E, Kaspar M, Bootz F, Detmar M, Neri D. A human mAb specific to oncofetal fibronectin selectively targets chronic skin inflammation in vivo. J Invest Dermatol 2006; 127:881-6. [PMID: 17185984 DOI: 10.1038/sj.jid.5700653] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The antibody-based targeted delivery of bioactive agents to sites of angiogenesis is an attractive therapeutic strategy for cancer treatment, but is largely unexplored for chronic inflammatory diseases. In this article, we show that the extra domain B (EDB) domain of fibronectin, a marker of angiogenesis, is expressed in psoriatic lesions, and that the anti-EDB human antibody L19 can selectively localize to chronically inflamed skin in vivo. The L19-based delivery of the cytokines IL10 and IL12 did not improve or worsen inflammation in a mouse model of chronic skin inflammation, which overexpressed vascular endothelial growth factor under the control of the keratin-14 promoter. By contrast, the L19-based targeted delivery of the proinflammatory cytokine IL2 or of the photosensitizer Sn(IV) chlorin e6 resulted in an increased swelling and reddening of inflamed skin. These results indicate that antibodies specific to components of the modified extracellular matrix can selectively accumulate at chronically inflamed sites in vivo. This observation now stimulates the search for bioactive molecules which can be fused to antibodies and which may confer a therapeutic benefit as a result of their preferential accumulation in psoriatic lesions and other sites of inflammation.
Collapse
Affiliation(s)
- Eveline Trachsel
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | | | | | | |
Collapse
|
50
|
Gafner V, Trachsel E, Neri D. An engineered antibody-interleukin-12 fusion protein with enhanced tumor vascular targeting properties. Int J Cancer 2006; 119:2205-12. [PMID: 16823838 DOI: 10.1002/ijc.22101] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The antibody-mediated targeted delivery of interleukin-12 (IL12) to the EDB domain of fibronectin, a marker of angiogenesis, is a promising avenue for enhancing the therapeutic index of this anti-cancer cytokine. Previous experiments, based on sequential fusion of a single-chain IL12 derivative to the anti-EDB antibody fragment scFv(L19) had yielded a therapeutic fusion protein [IL12-scFv(L19)-FLAG], which displayed an impressive therapeutic activity in murine models of cancer, in spite of a tumor uptake, which was less efficient compared to the parental unmodified scFv(L19). In this article, we describe the comparative analysis of 3 recombinant fusion proteins comprising the scFv(L19) and IL12 moieties. One of them, in which the p40 and p35 form a covalent heterodimer and in which each subunit is fused to a molecule of scFv(L19), displays an excellent tumor targeting performance in vivo, as assessed by quantitative biodistribution analysis, and a potent anti-tumor effect, superior to the one of IL12-scFv(L19)-FLAG. These results may have a clinical impact, considering the fact that the tumor targeting ability of scFv(L19) in patients with cancer has been demonstrated using scintigraphic methods, and that 2 scFv(L19)-based antibody-cytokine fusion are currently entering clinical trials.
Collapse
Affiliation(s)
- Verena Gafner
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zürich, Switzerland
| | | | | |
Collapse
|