1
|
Lidický O, Renešová N, Kudláčová J, Filipová M, Běláková T, Kovaříková P, Manakov D, Pankrác J, Dolníková A, Pokorná E, Klener P, Etrych T. Antibody polymer drug conjugates with increased drug to antibody ratio: CD38-targeting nanomedicines for innovative therapy of relapsed lymphomas. J Control Release 2025; 384:113876. [PMID: 40409372 DOI: 10.1016/j.jconrel.2025.113876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/30/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
The current frontline therapy for B-cell non-Hodgkin lymphomas (B-NHL), the most frequent hematologic malignancy in the Western hemisphere, is based on immunochemotherapy (ICT), i.e., a combination of genotoxic cytostatics and the anti-CD20 monoclonal antibody (mAb) rituximab. The treatment of relapsed or refractory (R/R) B-NHL remains an unmet medical need. Here, we developed and preclinically characterized a tailored hybrid mAb-polymer-drug conjugate (APDC) composed of the anti-CD38 mAb daratumumab (clinically approved for multiple myeloma therapy) and biocompatible N-(2-hydroxypropyl)methacrylamide (HPMA)-based copolymers conjugated with a cytotoxic payload monomethyl auristatine E (MMAE) via a Val-Cit-para-amino benzyl carbamate spacer cleavable by lysosomal enzymes. This innovative APDC design results in a significantly higher drug-to-antibody ratio (DAR) while maintaining a degree-of-conjugation (DOC) comparable to that of standard antibody-drug conjugates (ADCs). The enhanced anti-lymphoma efficacy of the new APDC nanotherapeutics, compared to standard ADCs, was confirmed in vivo using patient-derived lymphoma xenografts from a patient with R/R B-NHL. These APDC nanomedicines show promise as a personalized targeted chemotherapy approach.
Collapse
Affiliation(s)
- Ondřej Lidický
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, v.v.i., Heyrovského nám. 2, 162 00, Prague 6, Czech Republic
| | - Nicol Renešová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53, Prague 2, Czech Republic
| | - Júlia Kudláčová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, v.v.i., Heyrovského nám. 2, 162 00, Prague 6, Czech Republic
| | - Marcela Filipová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, v.v.i., Heyrovského nám. 2, 162 00, Prague 6, Czech Republic
| | - Tereza Běláková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, v.v.i., Heyrovského nám. 2, 162 00, Prague 6, Czech Republic
| | - Petra Kovaříková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53, Prague 2, Czech Republic; First Department of Internal Medicine-Department of Hematology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 2, 128 08, Prague 2, Czech Republic
| | - Dmitry Manakov
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53, Prague 2, Czech Republic
| | - Jan Pankrác
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00, Czech Republic
| | - Alexandra Dolníková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53, Prague 2, Czech Republic
| | - Eva Pokorná
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53, Prague 2, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53, Prague 2, Czech Republic; First Department of Internal Medicine-Department of Hematology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 2, 128 08, Prague 2, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, v.v.i., Heyrovského nám. 2, 162 00, Prague 6, Czech Republic.
| |
Collapse
|
2
|
Zhang Z, Ding C, Sun T, Wang L, Chen C. Tumor Therapy Strategies Based on Microenvironment-Specific Responsive Nanomaterials. Adv Healthc Mater 2023; 12:e2300153. [PMID: 36933000 DOI: 10.1002/adhm.202300153] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/10/2023] [Indexed: 03/19/2023]
Abstract
The tumor microenvironment (TME) is a complex and variable region characterized by hypoxia, low pH, high redox status, overexpression of enzymes, and high-adenosine triphosphate concentrations. In recent years, with the continuous in-depth study of nanomaterials, more and more TME-specific response nanomaterials are used for tumor treatment. However, the complexity of the TME causes different types of responses with various strategies and mechanisms of action. Aiming to systematically demonstrate the recent advances in research on TME-responsive nanomaterials, this work summarizes the characteristics of TME and outlines the strategies of different TME responses. Representative reaction types are illustrated and their merits and demerits are analyzed. Finally, forward-looking views on TME-response strategies for nanomaterials are presented. It is envisaged that such emerging strategies for the treatment of cancer are expected to exhibit dramatic trans-clinical capabilities, demonstrating the extensive potential for the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Zhaocong Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chengwen Ding
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Tiedong Sun
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Lei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Chunxia Chen
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
3
|
Bobde Y, Biswas S, Ghosh B. Current trends in the development of HPMA-based block copolymeric nanoparticles for their application in drug delivery. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.110018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
4
|
Randárová E, Kudláčová J, Etrych T. HPMA copolymer-antibody constructs in neoplastic treatment: an overview of therapeutics, targeted diagnostics, and drug-free systems. J Control Release 2020; 325:304-322. [DOI: 10.1016/j.jconrel.2020.06.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 12/27/2022]
|
5
|
Lidický O, Klener P, Machová D, Vočková P, Pokorná E, Helman K, Mavis C, Janoušková O, Etrych T. Overcoming resistance to rituximab in relapsed non-Hodgkin lymphomas by antibody-polymer drug conjugates actively targeted by anti-CD38 daratumumab. J Control Release 2020; 328:160-170. [PMID: 32860930 DOI: 10.1016/j.jconrel.2020.08.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 11/30/2022]
Abstract
B-cell non-Hodgkin lymphomas (B-NHL) represent the most common type of hematologic malignancies in the Western hemisphere. The therapy of all B-NHL is based on the combination of different genotoxic cytostatics and anti-CD20 monoclonal antibody (mAb) rituximab. Unfortunately, many patients relapse after the mentioned front-line treatment approaches. The therapy of patients with relapsed/refractory (R/R) B-NHL represents an unmet medical need. We designed, developed and tested novel actively targeted hybrid mAb-polymer-drug conjugate (APDC) containing anti-CD20, anti-CD38 or anti-CD19 mAbs. Biocompatible copolymers based on N-(2-hydroxypropyl)methacrylamide (HPMA) with cytostatic agent doxorubicin attached via stimuli-sensitive hydrazone bond were employed for the mAb grafting. Anti-lymphoma efficacy of the APDC nanotherapeutics was evaluated in vivo on a panel of three patient-derived lymphoma xenografts derived from two patients with R/R B-NHL and one patient with so far untreated B-NHL. In both PDX models derived from patients with R/R B-NHL, the targeting with anti-CD38 antibody daratumumab demonstrated highly improved anti-lymphoma efficacy compared to the targeting with anti-CD20 rituximab, two experimental anti-CD19 antibodies and non-targeted controls. The results represent a proof-of-concept of a new algorithm of personalized anti-tumor therapy based on highly innovative APDC biomaterials.
Collapse
Affiliation(s)
- Ondřej Lidický
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Pavel Klener
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Prague 2, Czech Republic; First Department of Internal Medicine- Hematology, University General Hospital and First Faculty of Medicine, Charles University, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Daniela Machová
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Petra Vočková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Prague 2, Czech Republic; First Department of Internal Medicine- Hematology, University General Hospital and First Faculty of Medicine, Charles University, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Eva Pokorná
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Prague 2, Czech Republic
| | - Karel Helman
- Faculty of Informatics and Statistics, University of Economics, Prague, Czech Republic
| | - Cory Mavis
- Department of Medicine Lymphoma/Myeloma, Roswell Park, Comprehensive Cancer Center, Elm & Carlton Sts, Buffalo, NY 14263, United States of America
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic.
| |
Collapse
|
6
|
Kostka L, Kotrchová L, Šubr V, Libánská A, Ferreira CA, Malátová I, Lee HJ, Barnhart TE, Engle JW, Cai W, Šírová M, Etrych T. HPMA-based star polymer biomaterials with tuneable structure and biodegradability tailored for advanced drug delivery to solid tumours. Biomaterials 2020; 235:119728. [DOI: 10.1016/j.biomaterials.2019.119728] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/27/2019] [Accepted: 12/22/2019] [Indexed: 02/03/2023]
|
7
|
May JN, Golombek SK, Baues M, Dasgupta A, Drude N, Rix A, Rommel D, von Stillfried S, Appold L, Pola R, Pechar M, van Bloois L, Storm G, Kuehne AJC, Gremse F, Theek B, Kiessling F, Lammers T. Multimodal and multiscale optical imaging of nanomedicine delivery across the blood-brain barrier upon sonopermeation. Am J Cancer Res 2020; 10:1948-1959. [PMID: 32042346 PMCID: PMC6993230 DOI: 10.7150/thno.41161] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/20/2019] [Indexed: 01/15/2023] Open
Abstract
Rationale: The blood-brain barrier (BBB) is a major obstacle for drug delivery to the brain. Sonopermeation, which relies on the combination of ultrasound and microbubbles, has emerged as a powerful tool to permeate the BBB, enabling the extravasation of drugs and drug delivery systems (DDS) to and into the central nervous system (CNS). When aiming to improve the treatment of high medical need brain disorders, it is important to systematically study nanomedicine translocation across the sonopermeated BBB. To this end, we here employed multimodal and multiscale optical imaging to investigate the impact of DDS size on brain accumulation, extravasation and penetration upon sonopermeation. Methods: Two prototypic DDS, i.e. 10 nm-sized pHPMA polymers and 100 nm-sized PEGylated liposomes, were labeled with fluorophores and intravenously injected in healthy CD-1 nude mice. Upon sonopermeation, computed tomography-fluorescence molecular tomography, fluorescence reflectance imaging, fluorescence microscopy, confocal microscopy and stimulated emission depletion nanoscopy were used to study the effect of DDS size on their translocation across the BBB. Results: Sonopermeation treatment enabled safe and efficient opening of the BBB, which was confirmed by staining extravasated endogenous IgG. No micro-hemorrhages, edema and necrosis were detected in H&E stainings. Multimodal and multiscale optical imaging showed that sonopermeation promoted the accumulation of nanocarriers in mouse brains, and that 10 nm-sized polymeric DDS accumulated more strongly and penetrated deeper into the brain than 100 nm-sized liposomes. Conclusions: BBB opening via sonopermeation enables safe and efficient delivery of nanomedicine formulations to and into the brain. When looking at accumulation and penetration (and when neglecting issues such as drug loading capacity and therapeutic efficacy) smaller-sized DDS are found to be more suitable for drug delivery across the BBB than larger-sized DDS. These findings are valuable for better understanding and further developing nanomedicine-based strategies for the treatment of CNS disorders.
Collapse
|
8
|
Pola R, Böhmová E, Filipová M, Pechar M, Pankrác J, Větvička D, Olejár T, Kabešová M, Poučková P, Šefc L, Zábrodský M, Janoušková O, Bouček J, Etrych T. Targeted Polymer-Based Probes for Fluorescence Guided Visualization and Potential Surgery of EGFR-Positive Head-and-Neck Tumors. Pharmaceutics 2020; 12:E31. [PMID: 31906300 PMCID: PMC7022460 DOI: 10.3390/pharmaceutics12010031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 11/16/2022] Open
Abstract
This report describes the design, synthesis and evaluation of tumor-targeted polymer probes to visualize epidermal growth factor receptor (EGFR)-positive malignant tumors for successful resection via fluorescence guided endoscopic surgery. Fluorescent polymer probes of various molecular weights enabling passive accumulation in tumors via enhanced permeability and retention were prepared and evaluated, showing an optimal molecular weight of 200,000 g/mol for passive tumor targeting. Moreover, poly(N-(2-hydroxypropyl)methacrylamide)-based copolymers labeled with fluorescent dyes were targeted with the EGFR-binding oligopeptide GE-11 (YHWYGYTPQNVI), human EGF or anti-EGFR monoclonal antibody cetuximab were all able to actively target the surface of EGFR-positive tumor cells. Nanoprobes targeted with GE-11 and cetuximab showed the best targeting profile but differed in their tumor accumulation kinetics. Cetuximab increased tumor accumulation after 15 min, whereas GE 11 needed at least 4 h. Interestingly, after 4 h, there were no significant differences in tumor targeting, indicating the potential of oligopeptide targeting for fluorescence-navigated surgery. In conclusion, fluorescent polymer probes targeted by oligopeptide GE-11 or whole antibody are excellent tools for surgical navigation during oncological surgery of head and neck squamous cell carcinoma, due to their relatively simple design, synthesis and cost, as well as optimal pharmacokinetics and accumulation in tumors.
Collapse
Affiliation(s)
- Robert Pola
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského sq. 2, 162 06 Prague 6, Czech Republic
| | - Eliška Böhmová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského sq. 2, 162 06 Prague 6, Czech Republic
| | - Marcela Filipová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského sq. 2, 162 06 Prague 6, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského sq. 2, 162 06 Prague 6, Czech Republic
| | - Jan Pankrác
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, U Nemocnice 5, 120 00 Prague 2, Czech Republic
| | - David Větvička
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Tomáš Olejár
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Martina Kabešová
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Pavla Poučková
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Luděk Šefc
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, U Nemocnice 5, 120 00 Prague 2, Czech Republic
| | - Michal Zábrodský
- Department of Otorhinolaryngology Head and Neck Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, V Úvalu 84, 150 06 Prague 5, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského sq. 2, 162 06 Prague 6, Czech Republic
| | - Jan Bouček
- Department of Otorhinolaryngology Head and Neck Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, V Úvalu 84, 150 06 Prague 5, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského sq. 2, 162 06 Prague 6, Czech Republic
| |
Collapse
|
9
|
Klener P, Etrych T, Klener P. Biological Therapy of Hematologic Malignancies: Toward a Chemotherapy- free Era. Curr Med Chem 2019; 26:1002-1018. [PMID: 28990505 DOI: 10.2174/0929867324666171006144725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
Abstract
Less than 70 years ago, the vast majority of hematologic malignancies were untreatable diseases with fatal prognoses. The development of modern chemotherapy agents, which had begun after the Second World War, was markedly accelerated by the discovery of the structure of DNA and its role in cancer biology and tumor cell division. The path travelled from the first temporary remissions observed in children with acute lymphoblastic leukemia treated with single-agent antimetabolites until the first cures achieved by multi-agent chemotherapy regimens was incredibly short. Despite great successes, however, conventional genotoxic cytostatics suffered from an inherently narrow therapeutic index and extensive toxicity, which in many instances limited their clinical utilization. In the last decade of the 20th century, increasing knowledge on the biology of certain malignancies resulted in the conception and development of first molecularly targeted agents designed to inhibit specific druggable molecules involved in the survival of cancer cells. Advances in technology and genetic engineering enabled the production of structurally complex anticancer macromolecules called biologicals, including therapeutic monoclonal antibodies, antibody-drug conjugates and antibody fragments. The development of drug delivery systems (DDSs), in which conventional drugs were attached to various types of carriers including nanoparticles, liposomes or biodegradable polymers, represented an alternative approach to the development of new anticancer agents. Despite the fact that the antitumor activity of drugs attached to DDSs was not fundamentally different, the improved pharmacokinetic profiles, decreased toxic side effects and significantly increased therapeutic indexes resulted in their enhanced antitumor efficacy compared to conventional (unbound) drugs. Approval of the first immune checkpoint inhibitor for the treatment of cancer in 2011 initiated the era of cancer immunotherapy. Checkpoint inhibitors, bispecific T-cell engagers, adoptive T-cell approaches and cancer vaccines have joined the platform so far, represented mainly by recombinant cytokines, therapeutic monoclonal antibodies and immunomodulatory agents. In specific clinical indications, conventional drugs have already been supplanted by multi-agent, chemotherapy-free regimens comprising diverse immunotherapy and/or targeted agents. The very distinct mechanisms of the anticancer activity of new immunotherapy approaches not only call for novel response criteria, but might also change fundamental treatment paradigms of certain types of hematologic malignancies in the near future.
Collapse
Affiliation(s)
- Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| | - Tomas Etrych
- Department of biomedical polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, 162 06 Prague, Czech Republic
| | - Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic
| |
Collapse
|
10
|
Theek B, Baues M, Gremse F, Pola R, Pechar M, Negwer I, Koynov K, Weber B, Barz M, Jahnen-Dechent W, Storm G, Kiessling F, Lammers T. Histidine-rich glycoprotein-induced vascular normalization improves EPR-mediated drug targeting to and into tumors. J Control Release 2018; 282:25-34. [PMID: 29730154 PMCID: PMC6130770 DOI: 10.1016/j.jconrel.2018.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 11/18/2022]
Abstract
Tumors are characterized by leaky blood vessels, and by an abnormal and heterogeneous vascular network. These pathophysiological characteristics contribute to the enhanced permeability and retention (EPR) effect, which is one of the key rationales for developing tumor-targeted drug delivery systems. Vessel abnormality and heterogeneity, however, which typically result from excessive pro-angiogenic signaling, can also hinder efficient drug delivery to and into tumors. Using histidine-rich glycoprotein (HRG) knockout and wild type mice, and HRG-overexpressing and normal t241 fibrosarcoma cells, we evaluated the effect of genetically induced and macrophage-mediated vascular normalization on the tumor accumulation and penetration of 10-20 nm-sized polymeric drug carriers based on poly(N-(2-hydroxypropyl)methacrylamide). Multimodal and multiscale optical imaging was employed to show that normalizing the tumor vasculature improves the accumulation of fluorophore-labeled polymers in tumors, and promotes their penetration out of tumor blood vessels deep into the interstitium.
Collapse
Affiliation(s)
- Benjamin Theek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany; Department of Targeted Therapeutics, Biomaterial Science and Technology, University of Twente, Enschede, The Netherlands
| | - Maike Baues
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Felix Gremse
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Robert Pola
- Institute of Macromolecular Chemistry, Czech Academy of Science, Prague, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Czech Academy of Science, Prague, Czech Republic
| | - Inka Negwer
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Benjamin Weber
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Willi Jahnen-Dechent
- Biointerface Laboratory, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Gert Storm
- Department of Targeted Therapeutics, Biomaterial Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Aachen, Germany; Department of Targeted Therapeutics, Biomaterial Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
11
|
Hassanzadeh P, Atyabi F, Dinarvand R. Linkers: The key elements for the creation of efficient nanotherapeutics. J Control Release 2018; 270:260-267. [DOI: 10.1016/j.jconrel.2017.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/09/2017] [Accepted: 12/11/2017] [Indexed: 01/16/2023]
|
12
|
|
13
|
Mao Y, Wang X, Zheng F, Wang C, Tang Q, Tang X, Xu N, Zhang H, Zhang D, Xiong L, Liang J, Zhu J. The tumor-inhibitory effectiveness of a novel anti-Trop2 Fab conjugate in pancreatic cancer. Oncotarget 2017; 7:24810-23. [PMID: 27050150 PMCID: PMC5029744 DOI: 10.18632/oncotarget.8529] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/28/2016] [Indexed: 12/26/2022] Open
Abstract
Human trophoblastic cell surface antigen 2 (Trop2) has been reported to act oncogenically. In this study, one-step quantitative real-time polymerase chain reaction (qPCR) test and immunohistochemistry (IHC) analysis with were employed to evaluate the relationship between Trop2 expression and the clinicopathological features of patients with PC. Then a novel anti-Trop2 Fab antibody was conjugated with Doxorubicin (DOX) to form Trop2Fab-DOX, an antibody-drug conjugate. This Trop2Fab-DOX conjugate was characterized by cell ELISA and immunofluorescence assay. MTT and wound healing analyses were used to evaluate the inhibitory effect of Trop2Fab-DOX on PC cell growth in vitro, while xenograft nude mice model was established to examine the tumor-inhibitory effects of PC in vivo. High Trop2 expression was observed in PC tissues and Trop2 expression was associated with several malignant attributes of PC patients, including overall survival. Trop2Fab-DOX can bind to the Trop2-expressing PC cells and provide an improved releasing type of DOX. In addition, Trop2Fab-DOX inhibited the proliferation and suppressed the migration of PC cells in a dose-dependent manner in vitro, while inhibited the growth of PC xenografts in vivo. Trop2 is a specific marker for PC, and a novel Trop2Fab-DOX ADC has a potent antitumor activity
Collapse
Affiliation(s)
- Yuan Mao
- Department of Oncology, Jiangsu Province Geriatric Hospital, Nanjing 210024, China
| | - Xiaoying Wang
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China.,Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Feng Zheng
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Changjun Wang
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Qi Tang
- Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Xiaojun Tang
- Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Ning Xu
- Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Huiling Zhang
- Department of Gynecology and Obstetrics, Nanjing Maternal and Children Care Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Dawei Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Lin Xiong
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Jie Liang
- Department of Pathology, Wuxi Nanjing Maternal and Children Care Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China.,Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
14
|
Chytil P, Koziolová E, Etrych T, Ulbrich K. HPMA Copolymer-Drug Conjugates with Controlled Tumor-Specific Drug Release. Macromol Biosci 2017; 18. [PMID: 28805040 DOI: 10.1002/mabi.201700209] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Indexed: 11/10/2022]
Abstract
Over the past few decades, numerous polymer drug carrier systems are designed and synthesized, and their properties are evaluated. Many of these systems are based on water-soluble polymer carriers of low-molecular-weight drugs and compounds, e.g., cytostatic agents, anti-inflammatory drugs, or multidrug resistance inhibitors, all covalently bound to a carrier by a biodegradable spacer that enables controlled release of the active molecule to achieve the desired pharmacological effect. Among others, the synthetic polymer carriers based on N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers are some of the most promising carriers for this purpose. This review focuses on advances in the development of HPMA copolymer carriers and their conjugates with anticancer drugs, with triggered drug activation in tumor tissue and especially in tumor cells. Specifically, this review highlights the improvements in polymer drug carrier design with respect to the structure of a spacer to influence controlled drug release and activation, and its impact on the drug pharmacokinetics, enhanced tumor uptake, cellular trafficking, and in vivo antitumor activity.
Collapse
Affiliation(s)
- Petr Chytil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Eva Koziolová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| |
Collapse
|
15
|
A new construct of antibody-drug conjugates for treatment of B-cell non-Hodgkin's lymphomas. Eur J Pharm Sci 2017; 103:36-46. [PMID: 28249824 DOI: 10.1016/j.ejps.2017.02.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 11/20/2022]
Abstract
The aim of this study was to develop a new class of antibody-drug conjugates (ADCs) with the potential to not only enhance treatment efficacy but also improve tolerability for patients with B-cell lymphomas. Classic ADCs consist of monoclonal antibodies (mAbs) linked to drugs or toxins. They selectively deliver toxic moieties to tumor cells. As such, they greatly improve the therapeutic index compared to traditional chemotherapeutic agents. However, the therapeutic efficacy and safety of ADCs are dependent on linker stability and payload toxicity. Limited payload number on a single antibody (drug-to-antibody ratio, or DAR) has been driving investigators to use extremely toxic agents; however, even very low off-target binding of these ADCs may kill patients. Herein we report a new design of ADCs that consists of rituximab (RTX) and N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer-epirubicin conjugates. The latter was selectively attached to RTX via reduced disulfide bonds. Such design allows the introduction of a large payload of drug on the antibody without adding attachment sites and without compromising the antigen-targeting ability. The binding of the new conjugate, namely RTX-P-EPI, to Ramos cells (with high CD20 expression) was confirmed. The cytotoxicity of RTX-P-EPI against Raji and Ramos cells was also determined. Interestingly, two-fold inhibition of cell proliferation was observed when using RTX-P-EPI compared with their equivalent physical mixture of RTX and P-EPI. Treatment of male SCID mice bearing subcutaneous Ramos B-cell lymphoma tumors demonstrated that RTX-P-EPI possessed superior efficacy when compared to combination of RTX with chemotherapy EPI (RTX+EPI) and P-EPI (RTX+P-EPI), whereas single RTX and a non-specific conjugate IgG-P-EPI only showed marginal effect. The conjugate RTX-EPI in which EPI was directly attached to RTX demonstrated much less antitumor activity compared with RTX-P-EPI. The results suggest that this new design possesses synergistic potential of immunotherapy combined with established macromolecular therapy; moreover, a conventional chemo-agent could be utilized to generate highly effective ADCs and to achieve lower risk of off-target toxicity.
Collapse
|
16
|
Shen T, Xu X, Guo L, Tang H, Diao T, Gan Z, Zhang G, Yu Q. Efficient Tumor Accumulation, Penetration and Tumor Growth Inhibition Achieved by Polymer Therapeutics: The Effect of Polymer Architectures. Biomacromolecules 2016; 18:217-230. [DOI: 10.1021/acs.biomac.6b01533] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tong Shen
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Xin Xu
- Department
of Urology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Linyi Guo
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Hao Tang
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Tongxiang Diao
- Department
of Urology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Zhihua Gan
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Guan Zhang
- Department
of Urology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Qingsong Yu
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| |
Collapse
|
17
|
Sonawane SJ, Kalhapure RS, Govender T. Hydrazone linkages in pH responsive drug delivery systems. Eur J Pharm Sci 2016; 99:45-65. [PMID: 27979586 DOI: 10.1016/j.ejps.2016.12.011] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/24/2016] [Accepted: 12/09/2016] [Indexed: 01/02/2023]
Abstract
Stimuli-responsive polymeric drug delivery systems using various triggers to release the drug at the sites have become a major focus area. Among various stimuli-responsive materials, pH-responsiveness has been studied extensively. The materials used for fabricating pH-responsive drug delivery systems include a specific chemical functionality in their structure that can respond to changes in the pH of the surrounding environment. Various chemical functionalities, for example, acetal, amine, ortho ester, amine and hydrazone, have been used to design materials that are capable of releasing their payload at the acidic pH conditions of the tumor or infection sites. Hydrazone linkages are significant synthons for numerous transformations and have gained importance in pharmaceutical sciences due to their various biological and clinical applications. These linkages have been employed in various drug delivery vehicles, such as linear polymers, star shaped polymers, dendrimers, micelles, liposomes and inorganic nanoparticles, for pH-responsive drug delivery. This review paper focuses on the synthesis and characterization methods of hydrazone bond containing materials and their applications in pH-responsive drug delivery systems. It provides detailed suggestions as guidelines to materials and formulation scientists for designing biocompatible pH-responsive materials with hydrazone linkages and identifying future studies.
Collapse
Affiliation(s)
- Sandeep J Sonawane
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa
| | - Rahul S Kalhapure
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa..
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa..
| |
Collapse
|
18
|
Yang J, Kopeček J. Design of smart HPMA copolymer-based nanomedicines. J Control Release 2016; 240:9-23. [DOI: 10.1016/j.jconrel.2015.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 01/13/2023]
|
19
|
Ulbrich K, Holá K, Šubr V, Bakandritsos A, Tuček J, Zbořil R. Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies. Chem Rev 2016; 116:5338-431. [DOI: 10.1021/acs.chemrev.5b00589] [Citation(s) in RCA: 1086] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Karel Ulbrich
- Institute
of Macromolecular Chemistry, The Czech Academy of Sciences, v.v.i., Heyrovsky Square 2, 162 06 Prague 6, Czech Republic
| | - Kateřina Holá
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Vladimir Šubr
- Institute
of Macromolecular Chemistry, The Czech Academy of Sciences, v.v.i., Heyrovsky Square 2, 162 06 Prague 6, Czech Republic
| | - Aristides Bakandritsos
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Jiří Tuček
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Radek Zbořil
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| |
Collapse
|
20
|
Mielańczyk A, Neugebauer D. Designing Drug Conjugates Based on Sugar Decorated V-Shape and Star Polymethacrylates: Influence of Composition and Architecture of Polymeric Carrier. Bioconjug Chem 2015; 26:2303-10. [DOI: 10.1021/acs.bioconjchem.5b00533] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Anna Mielańczyk
- Department of Physical Chemistry
and Technology of Polymers, Faculty of Chemistry, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| | - Dorota Neugebauer
- Department of Physical Chemistry
and Technology of Polymers, Faculty of Chemistry, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
| |
Collapse
|
21
|
Anti-Lymphoma Efficacy Comparison of Anti-Cd20 Monoclonal Antibody-Targeted and Non-Targeted Star-Shaped Polymer-Prodrug Conjugates. Molecules 2015; 20:19849-64. [PMID: 26556320 PMCID: PMC6331818 DOI: 10.3390/molecules201119664] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 11/17/2022] Open
Abstract
Here we describe the synthesis and biological properties of two types of star-shaped polymer-doxorubicin conjugates: non-targeted conjugate prepared as long-circulating high-molecular-weight (HMW) polymer prodrugs with a dendrimer core and a targeted conjugate with the anti-CD20 monoclonal antibody (mAb) rituximab (RTX). The copolymers were linked to the dendrimer core or to the reduced mAb via one-point attachment forming a star-shaped structure with a central antibody or dendrimer surrounded by hydrophilic polymer chains. The anticancer drug doxorubicin (DOX) was attached to the N-(2-hydroxypropyl)methacrylamide (HPMA)-based copolymer chain in star polymer systems via a pH-labile hydrazone linkage. Such polymer-DOX conjugates were fairly stable in aqueous solutions at pH 7.4, and the drug was readily released in mildly acidic environments at pH 5–5.5 by hydrolysis of the hydrazone bonds. The cytotoxicity of the polymer conjugates was tested on several CD20-positive or negative human cell lines. Similar levels of in vitro cytotoxicity were observed for all tested polymer conjugates regardless of type or structure. In vivo experiments using primary cell-based murine xenograft models of human diffuse large B-cell lymphoma confirmed the superior anti-lymphoma efficacy of the polymer-bound DOX conjugate when compared with the original drug. Targeting with RTX did not further enhance the anti-lymphoma efficacy relative to the non-targeted star polymer conjugate. Two mechanisms could play roles in these findings: changes in the binding ability to the CD-20 receptor and a significant loss of the immunological properties of RTX in the polymer conjugates.
Collapse
|
22
|
Wong PT, Choi SK. Mechanisms of Drug Release in Nanotherapeutic Delivery Systems. Chem Rev 2015; 115:3388-432. [DOI: 10.1021/cr5004634] [Citation(s) in RCA: 339] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pamela T. Wong
- Michigan
Nanotechnology Institute
for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Seok Ki Choi
- Michigan
Nanotechnology Institute
for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
23
|
Dong D, Gao W, Liu Y, Qi XR. Therapeutic potential of targeted multifunctional nanocomplex co-delivery of siRNA and low-dose doxorubicin in breast cancer. Cancer Lett 2015; 359:178-86. [DOI: 10.1016/j.canlet.2015.01.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/31/2014] [Accepted: 01/08/2015] [Indexed: 11/24/2022]
|
24
|
Moraes J, Simionca IM, Ketari H, Klok HA. Avoiding compositional drift during the RAFT copolymerization of N-(2-hydroxypropyl)methacrylamide and N-acryloxysuccinimide: towards uniform platforms for post-polymerization modification. Polym Chem 2015. [DOI: 10.1039/c5py00253b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Compositionally uniform copolymers: avoiding compositional drift during RAFT copolymerization by controlling monomer feed ratio and conversion.
Collapse
Affiliation(s)
- John Moraes
- École Polytechnique Fédérale de Lausanne (EPFL)
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques
- Laboratoire des Polymères
- Lausanne
- Switzerland
| | - Ioana-Maria Simionca
- École Polytechnique Fédérale de Lausanne (EPFL)
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques
- Laboratoire des Polymères
- Lausanne
- Switzerland
| | - Hedi Ketari
- École Polytechnique Fédérale de Lausanne (EPFL)
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques
- Laboratoire des Polymères
- Lausanne
- Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL)
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques
- Laboratoire des Polymères
- Lausanne
- Switzerland
| |
Collapse
|
25
|
Duro-Castano A, Movellan J, Vicent MJ. Smart branched polymer drug conjugates as nano-sized drug delivery systems. Biomater Sci 2015; 3:1321-34. [DOI: 10.1039/c5bm00166h] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Branched polymers own special properties derived from their intrinsic characteristics. These properties make them ideal candidates to be used as carriers for an improved generation of polymer-drug conjugates.
Collapse
Affiliation(s)
- A. Duro-Castano
- Centro de Investigación Príncipe Felipe
- Polymer Therapeutics Lab
- E-46012 Valencia
- Spain
| | - J. Movellan
- Centro de Investigación Príncipe Felipe
- Polymer Therapeutics Lab
- E-46012 Valencia
- Spain
| | - M. J. Vicent
- Centro de Investigación Príncipe Felipe
- Polymer Therapeutics Lab
- E-46012 Valencia
- Spain
| |
Collapse
|
26
|
Tappertzhofen K, Bednarczyk M, Koynov K, Bros M, Grabbe S, Zentel R. Toward Anticancer Immunotherapeutics: Well-Defined Polymer-Antibody Conjugates for Selective Dendritic Cell Targeting. Macromol Biosci 2014; 14:1444-57. [DOI: 10.1002/mabi.201400190] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/22/2014] [Indexed: 11/05/2022]
Affiliation(s)
- Kristof Tappertzhofen
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55128 Mainz Germany
| | - Monika Bednarczyk
- Department of Dermatology; University Medical Center of the Johannes Gutenberg-University; Langenbeckstrasse 1 55131 Mainz Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | - Matthias Bros
- Department of Dermatology; University Medical Center of the Johannes Gutenberg-University; Langenbeckstrasse 1 55131 Mainz Germany
| | - Stephan Grabbe
- Department of Dermatology; University Medical Center of the Johannes Gutenberg-University; Langenbeckstrasse 1 55131 Mainz Germany
- Research Center Immunology (FZI); University Medical Center of the Johannes Gutenberg-University; Langenbeckstrasse 1 55131 Mainz Germany
| | - Rudolf Zentel
- Institute of Organic Chemistry; Johannes Gutenberg-University; Duesbergweg 10-14 55128 Mainz Germany
| |
Collapse
|
27
|
Kostková H, Etrych T, Říhová B, Kostka L, Starovoytová L, Kovář M, Ulbrich K. HPMA Copolymer Conjugates of DOX and Mitomycin C for Combination Therapy: Physicochemical Characterization, Cytotoxic Effects, Combination Index Analysis, and Anti-Tumor Efficacy. Macromol Biosci 2013; 13:1648-60. [DOI: 10.1002/mabi.201300288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/25/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Hana Kostková
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic; v.v.i., Heyrovsky Sq. 2 162 06 Prague 6 Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic; v.v.i., Heyrovsky Sq. 2 162 06 Prague 6 Czech Republic
| | - Blanka Říhová
- Institute of Microbiology; Academy of Sciences of the Czech Republic; v.v.i, Vídeňská 1083 142 20 Prague 4 Czech Republic
| | - Libor Kostka
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic; v.v.i., Heyrovsky Sq. 2 162 06 Prague 6 Czech Republic
| | - Larisa Starovoytová
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic; v.v.i., Heyrovsky Sq. 2 162 06 Prague 6 Czech Republic
| | - Marek Kovář
- Institute of Microbiology; Academy of Sciences of the Czech Republic; v.v.i, Vídeňská 1083 142 20 Prague 4 Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic; v.v.i., Heyrovsky Sq. 2 162 06 Prague 6 Czech Republic
| |
Collapse
|
28
|
Chen X, Ding G, Gao Q, Sun J, Zhang Q, Du L, Qiu Z, Wang C, Zheng F, Sun B, Ni J, Feng Z, Zhu J. A human anti-c-Met Fab fragment conjugated with doxorubicin as targeted chemotherapy for hepatocellular carcinoma. PLoS One 2013; 8:e63093. [PMID: 23675455 PMCID: PMC3652865 DOI: 10.1371/journal.pone.0063093] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 03/28/2013] [Indexed: 12/15/2022] Open
Abstract
c-Met is over-expressed in hepatocellular carcinoma(HCC) but is absent or expressed at low levels in normal tissues. Therefore we generated a novel conjugate of a human anti-c-Met Fab fragment (MetFab) with doxorubicin (DOX) and assessed whether it had targeted antitumor activity against HCC and reduced the side-effects of DOX. The MetFab was screened from human phage library, conjugated with DOX via chemical synthesis, and the conjugation MetFab-DOX was confirmed by HPLC. The drug release patterns, the binding efficacy, and cellular distribution of MetFab-DOX were assessed. MetFab-DOX was stable at pH7.2 PBS while release doxorubicin quickly at pH4.0, the binding efficacy of MetFab-DOX was similarly as MetFab, and the cellular distribution of the MetFab-DOX is distinct from free DOX. The cytotoxicity of MetFab-DOX was analyzed by the MTT method and the nude mouse HCC model. The MetFab-DOX demonstrated cytotoxic effects on c-Met expressing-tumor cells, but not on the cells without c-Met expression. MetFab-DOX exerted anti-tumor effect and significantly reduced the side effect of free DOX in mice model. Furthermore, the localization of conjugate was confirmed by immunofluorescence staining of tumor tissue sections and optical tumor imaging, respectively, and the tissue-distribution of drug was compared between free DOX and MetFab-DOX treatment by spectrofluorometer. MetFab-DOX can localize to the tumor tissue, and the concentration of doxorubicin in the tumor was higher after MetFab-DOX administration than after DOX administration. In summary, MetFab-DOX can target c-Met expressing HCC cells effectively and have obvious antitumor activity with decreased side-effects in preclinical models of HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Survival/drug effects
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Drug Stability
- Gene Expression
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Humans
- Hydrogen-Ion Concentration
- Immunoconjugates/chemistry
- Immunoconjugates/genetics
- Immunoconjugates/pharmacology
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/genetics
- Immunoglobulin Fab Fragments/immunology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/immunology
- Liver Neoplasms, Experimental/pathology
- Male
- Mice
- Mice, Nude
- Molecular Targeted Therapy
- Peptide Library
- Proto-Oncogene Proteins c-met/antagonists & inhibitors
- Proto-Oncogene Proteins c-met/genetics
- Proto-Oncogene Proteins c-met/immunology
Collapse
Affiliation(s)
- Ximin Chen
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Guipeng Ding
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Qihe Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Jian Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Qianqian Zhang
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Lijian Du
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Zhenning Qiu
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Changjun Wang
- Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Feng Zheng
- Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Bowang Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Jian Ni
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenqing Feng
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention & Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
- * E-mail: (ZF); (JZ)
| | - Jin Zhu
- Key Laboratory of Antibody Technique of Ministry of Health, Department of Pathology, Nanjing Medical University, Nanjing, China
- Huadong Medical Institute of Biotechniques, Nanjing, China
- * E-mail: (ZF); (JZ)
| |
Collapse
|
29
|
Coyne CP, Jones T, Bear R. Anti-Neoplastic Cytotoxicity of Gemcitabine-(C 4- amide)-[anti-HER2/ neu] in Combination with Griseofulvin against Chemotherapeutic-Resistant Mammary Adenocarcinoma (SKBr-3). Med Chem 2013. [PMID: 26225219 PMCID: PMC4516389 DOI: 10.4172/2161-0444.1000141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction Gemcitabine is a pyrimidine nucleoside analog that becomes triphosphorylated and in this form it competitively inhibits cytidine incorporation into DNA strands. Diphosphorylated gemcitabine irreversibly inhibits ribonucleotide reductase thereby preventing deoxyribonucleotide synthesis. Functioning as a potent chemotherapeutic, gemcitabine decreases neoplastic cell proliferation and induces apoptosis which accounts for its effectiveness in the clinical treatment of several leukemia and carcinoma cell types. A brief plasma half-life due to rapid deamination, chemotherapeuticresistance and sequelae restricts gemcitabine utility in clinical oncology. Selective “targeted” gemcitabine delivery represents a molecular strategy for prolonging its plasma half-life and minimizing innocent tissue/organ exposure. Methods A previously described organic chemistry scheme was applied to synthesize a UV-photoactivated gemcitabine intermediate for production of gemcitabine-(C4-amide)-[anti-HER2/neu]. Immunodetection analysis (Western-blot) was applied to detect the presence of any degradative fragmentation or polymerization. Detection of retained binding-avidity for gemcitabine-(C4-amide)-[anti-HER2/neu] was determined by cell-ELISA using populations of chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3) that highly over-express the HER2/neu trophic membrane receptor. Anti-neoplastic cytotoxicity of gemcitabine-(C4-amide)-[anti-HER2/neu] and the tubulin/microtubule inhibitor, griseofulvin was established against chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3). Related investigations evaluated the potential for gemcitabine-(C4-amide)-[anti-HER2/neu] in dual combination with griseofulvin to evoke increased levels of anti-neoplastic cytotoxicity compared to gemcitabine-(C4-amide)-[anti-HER2/neu]. Results Covalent gemcitabine-(C4-amide)-[anti-HER2/neu] immunochemotherapeutic and griseofulvin exerted anti-neoplastic cytotoxicity against chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3). Covalent gemcitabine-(C4-amide)-[anti-HER2/neu] immunochemotherapeutic or gemcitabine in dual combination with griseofulvin created increased levels of anti-neoplastic cytotoxicity that were greater than was attainable with gemcitabine-(C4-amide)-[anti-HER2/neu] or gemcitabine alone. Conclusion Gemcitabine-(C4-amide)-[anti-HER2/neu] in dual combination with griseofulvin can produce enhanced levels of anti-neoplastic cytotoxicity and potentially provide a basis for treatment regimens with a wider margin-of-safety. Such benefits would be possible through the collective properties of; [i] selective “targeted” gemcitabine delivery; [ii] relatively lower toxicity of griseofulvin compared to many if not most conventional chemotherapeutics; [iii] reduced total dosage requirements faciliated by additive or synergistic anti-cancer properties; and [iv] differences in sequelae for gemcitabine-(C4-amide)-[anti-HER2/neu] compared to griseofulvin functioning as a tubulin/microtubule inhibitor.
Collapse
Affiliation(s)
- C P Coyne
- Department of Basic Sciences, College of Veterinary Medicine at Wise Center, Mississippi State University, Mississippi State, Mississippi 39762, USA
| | - Toni Jones
- Department of Basic Sciences, College of Veterinary Medicine at Wise Center, Mississippi State University, Mississippi State, Mississippi 39762, USA
| | - Ryan Bear
- Department of Basic Sciences, College of Veterinary Medicine at Wise Center, Mississippi State University, Mississippi State, Mississippi 39762, USA
| |
Collapse
|
30
|
Kunjachan S, Gremse F, Theek B, Koczera P, Pola R, Pechar M, Etrych T, Ulbrich K, Storm G, Kiessling F, Lammers T. Noninvasive optical imaging of nanomedicine biodistribution. ACS NANO 2013; 7:252-62. [PMID: 23067565 PMCID: PMC3743636 DOI: 10.1021/nn303955n] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Nanomedicines are sub-micrometer-sized carrier materials designed to improve the biodistribution of i.v. administered (chemo-) therapeutic agents. In recent years, ever more efforts in the nanomedicine field have employed optical imaging (OI) techniques to monitor biodistribution and target site accumulation. Thus far, however, the longitudinal assessment of nanomedicine biodistribution using OI has been impossible, due to limited light penetration (in the case of 2D fluorescence reflectance imaging; FRI) and to the inability to accurately allocate fluorescent signals to nonsuperficial organs (in the case of 3D fluorescence molecular tomography; FMT). Using a combination of high-resolution microcomputed tomography (μCT) and FMT, we have here set out to establish a hybrid imaging protocol for noninvasively visualizing and quantifying the accumulation of near-infrared fluorophore-labeled nanomedicines in tissues other than superficial tumors. To this end, HPMA-based polymeric drug carriers were labeled with Dy750, their biodistribution and tumor accumulation were analyzed using FMT, and the resulting data sets were fused with anatomical μCT data sets in which several different physiologically relevant organs were presegmented. The robustness of 3D organ segmentation was validated, and the results obtained using 3D CT-FMT were compared to those obtained upon standard 3D FMT and 2D FRI. Our findings convincingly demonstrate that combining anatomical μCT with molecular FMT facilitates the noninvasive assessment of nanomedicine biodistribution.
Collapse
Affiliation(s)
- Sijumon Kunjachan
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
| | - Felix Gremse
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
| | - Benjamin Theek
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
| | - Patrick Koczera
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
| | - Robert Pola
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tomas Etrych
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Gert Storm
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Fabian Kiessling
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
- Corresponding authors: ;
| | - Twan Lammers
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
- Corresponding authors: ;
| |
Collapse
|
31
|
Zhong YJ, Shao LH, Li Y. Cathepsin B-cleavable doxorubicin prodrugs for targeted cancer therapy (Review). Int J Oncol 2012; 42:373-83. [PMID: 23291656 PMCID: PMC3583876 DOI: 10.3892/ijo.2012.1754] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 12/03/2012] [Indexed: 12/02/2022] Open
Abstract
Doxorubicin (DOX) is one of the most effective cytotoxic anticancer drugs used for the treatment of hematological malignancies, as well as a broad range of solid tumors. However, the clinical applications of this drug have long been limited due to its severe dose-dependent toxicities. Therefore, DOX derivatives and analogs have been developed to address this issue. A type of DOX prodrug, cleaved by cathepsin B (Cat B), which is highly upregulated in malignant tumors and premalignant lesions, has been developed to achieve a higher DOX concentration in tumor tissue and a lower concentration in normal tissue, so as to enhance the efficacy and reduce toxicity to normal cells. In this review, we focused on Cat B-cleavable DOX prodrugs and discussed the efficacy of these prodrugs, demonstrated by preclinical and clinical developments.
Collapse
Affiliation(s)
- Yan-Jun Zhong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | | | | |
Collapse
|
32
|
Tappertzhofen K, Metz VV, Hubo M, Barz M, Postina R, Jonuleit H, Zentel R. Synthesis of Maleimide-Functionalyzed HPMA-Copolymers and in vitro Characterization of the aRAGE- and Human Immunoglobulin (huIgG)-Polymer Conjugates. Macromol Biosci 2012; 13:203-14. [DOI: 10.1002/mabi.201200344] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 10/20/2012] [Indexed: 11/05/2022]
|
33
|
Coyne CP, Jones T, Bear R. Influence of Alternative Tubulin Inhibitors on the Potency of a Epirubicin-Immunochemotherapeutic Synthesized with an Ultra Violet Light-Activated Intermediate: Influence of incorporating an internal/integral disulfide bond structure and Alternative Tubulin/Microtubule Inhibitors on the Cytotoxic Anti-Neoplastic Potency of Epirubicin-(C 3-amide)-Anti-HER2/neu Synthesized Utilizing a UV-Photoactivated Anthracycline Intermediate. CANCER AND CLINICAL ONCOLOGY 2012. [PMID: 26225190 DOI: 10.5539/cco.v1n2p49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Immunochemotherapeutics, epirubicin-(C3-amide)-SS-[anti-HER2/neu] with an internal disulfide bond, and epirubicin-(C3-amide)-[anti-HER2/neu] were synthesized utilizing succinimidyl 2-[(4,4'-azipentanamido) ethyl]-1,3'-dithioproprionate or succinimidyl 4,4-azipentanoate respectively. Western blot analysis was used to determine the presence of any immunoglobulin fragmentation or IgG-IgG polymerization. Retained HER2/neu binding characteristics of epirubicin-(C3-amide)-[anti-HER2/neu] and epirubicin-(C3-amide)-SS-[anti-HER2/neu] were validated by cell-ELISA using a mammary adenocarcinoma (SKBr-3) population that highly over-expresses trophic HER2/neu receptor complexes. Cytotoxic anti-neoplastic potency of epirubicin-(C3-amide)-[anti-HER2/neu] and epirubicin-(C3-amide)-SS-[anti-HER2/neu] between epirubicin-equivalent concentrations of 10-10 M and 10-6 M was determined by measuring the vitality/proliferation of chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3 cell type). Cytotoxic anti-neoplastic potency of benzimidazoles (albendazole, flubendazole, membendazole) and griseofulvin were assessed between 0-to-2 μg/ml and 0-to-100 μg/ml respectively while mebendazole and griseofulvin were analyzed at fixed concentrations of 0.35 μg/ml and 35 g/ml respectively in dual combination with gradient concentrations of epirubicin-(C3-amide)-[anti-HER2/neu] and epirubicin-(C3-amide)-SS-[anti-HER2/neu]. Cytotoxic anti-neoplastic potency for epirubicin-(C3-amide)-[anti-HER2/neu] and epirubicin-(C3-amide)-SS-[anti-HER2/neu] against chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3) was nearly identical at epirubicin-equivalent concentrations of 10-10 M and 10-6 M. The benzimadazoles also possessed cytotoxic anti-neoplastic activity with flubendazole and albendazole being the most and least potent respectively. Similarly, griseofulvin had cytotoxic anti-neoplastic activity and was more potent than methylselenocysteine. Both mebendazole and griseofulvin when applied in dual combination with either epirubicin-(C3-amide)-[anti-HER2/neu] or epirubicin-(C3-amide)-SS-[anti-HER2/neu] produced enhanced levels of cytotoxic anti-neoplatic potency.
Collapse
Affiliation(s)
- C P Coyne
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, USA
| | - Toni Jones
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, USA
| | - Ryan Bear
- College of Veterinary Medicine, Mississippi State University, USA
| |
Collapse
|
34
|
Coyne CP, Jones T, Bear R. Synthesis of Gemcitabine-(C 4- amide)-[anti-HER2/ neu] Utilizing a UV-Photoactivated Gemcitabine Intermediate: Cytotoxic Anti-Neoplastic Activity against Chemotherapeutic-Resistant Mammary Adenocarcinoma SKBr-3. ACTA ACUST UNITED AC 2012. [PMID: 26225216 DOI: 10.4236/jct.2012.325089] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gemcitabine is a pyrimidine nucleoside analog that becomes triphosphorylated intracellularly where it competitively inhibits cytidine incorporation into DNA strands. Another mechanism-of-action of gemcitabine (diphosphorylated form) involves irreversible inhibition of the enzyme ribonucleotide reductase thereby preventing deoxyribonucleotide synthesis. Functioning as a potent chemotherapeutic gemcitabine promote decreases in neoplastic cell proliferation and apoptosis which is frequently found to be effective for the treatment of several leukemias and a wide spectrum of carcinomas. A brief plasma half-life in part due to rapid deamination and chemotherapeutic-resistance restricts the utility of gemcit-abine in clinical oncology. Selective "targeted" delivery of gemcitabine represents a potential molecular strategy for simultaneously prolonging its plasma half-life and minimizing innocient tissues and organ systems exposure to chemotherapy. The molecular design and an organic chemistry based synthesis reaction is described that initially generates a UV-photoactivated gemcitabine intermediate. In a subsequent phase of the synthesis method the UV-photoactivated gemcitabine intermediate is covalently bonded to a monoclonal immunoglobulin yielding an end-product in the form of gemcitabine-(C4-amide)-[anti-HER2/neu]. Analysis by SDS-PAGE/chemiluminescent auto-radiography did not detect evidence of gemcitabine-(C4-amide)-[anti-HER2/neu] polymerization or degradative fragmentation while cell-ELISA demonstrated retained binding-avidity for HER2/neu trophic membrane receptor complexes highly over-expressed by chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3). Compared to chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3), the covalent immunochemotherapeutic, gemcitabine-(C4-amide)-[anti-HER2/neu] is anticipated to exert greater levels of cytotoxic anti-neoplastic potency against other neoplastic cell types like pancreatic carcinoma, small-cell lung carcinoma, neuroblastoma, glioblastoma, oral squamous cell carcinoma, cervical epitheliod carcinoma, or leukemia/lymphoid neoplastic cell types based on their reported sensitivity to gemcitabine and gemcitabine covalent conjugates.
Collapse
Affiliation(s)
- Cody P Coyne
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Oktibbeha County, USA
| | - Toni Jones
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Oktibbeha County, USA
| | - Ryan Bear
- Wise Center, Mississippi State University, Oktibbeha County, USA
| |
Collapse
|
35
|
Nanotechnology applied to overcome tumor drug resistance. J Control Release 2012; 162:45-55. [DOI: 10.1016/j.jconrel.2012.05.051] [Citation(s) in RCA: 238] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/29/2012] [Accepted: 05/31/2012] [Indexed: 01/01/2023]
|
36
|
Choi SK, Verma M, Silpe J, Moody RE, Tang K, Hanson JJ, Baker JR. A photochemical approach for controlled drug release in targeted drug delivery. Bioorg Med Chem 2012; 20:1281-90. [PMID: 22225916 PMCID: PMC3267001 DOI: 10.1016/j.bmc.2011.12.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/06/2011] [Accepted: 12/13/2011] [Indexed: 11/15/2022]
Abstract
Photochemistry provides a unique mechanism that enables the active control of drug release in cancer-targeting drug delivery. This study investigates the light-mediated release of methotrexate, an anticancer drug, using a photocleavable linker strategy based on o-nitrobenzyl protection. We evaluated two types of the o-nitrobenzyl-linked methotrexate for the drug release study and further extended the study to a fifth-generation poly(amidoamine) dendrimer carrier covalently conjugated with methotrexate via the o-nitrobenzyl linker. We performed the drug release studies by using a combination of three standard analytical methods that include UV/vis spectrometry, (1)H NMR spectroscopy, and anal. HPLC. This article reports that methotrexate is released by the photochemical mechanism in an actively controlled manner. The rate of the drug release varies in response to multiple control parameters, including linker design, light wavelength, exposure time, and the pH of the medium where the drug release occurs.
Collapse
Affiliation(s)
- Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Manisha Verma
- College of Literature, Science and The Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Justin Silpe
- College of Literature, Science and The Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan E. Moody
- College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kenny Tang
- College of Literature, Science and The Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey J. Hanson
- College of Literature, Science and The Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - James R. Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Coyne CP, Jones T, Bear R. Synthesis of a covalent epirubicin-(C(3)-amide)-anti-HER2/neu immunochemotherapeutic utilizing a UV-photoactivated anthracycline intermediate. Cancer Biother Radiopharm 2012; 27:41-55. [PMID: 22191802 PMCID: PMC4361169 DOI: 10.1089/cbr.2011.1097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The C(3)-monoamine on the carbohydrate moiety (daunosamine -NH(2)-3') of epirubicin was reacted under anhydrous conditions with succinimidyl 4,4-azipentanoate to create a covalent UV-photoactivated epirubicin-(C(3)-amide) intermediate with primary amine-reactive properties. A synthetic covalent bond between the UV-photoactivated epirubicin-(C(3)-amide) intermediate and the ɛ-amine of lysine residues within the amino acid sequence of anti-HER2/neu monoclonal immunoglobulin was subsequently created by exposure to UV light (354 nm) for 15 minutes. Size-separation by sodium dodecyl sulfate-polyacrylamide gel electrophoresis combined with immunodetection analysis and chemiluminescent autoradiographic imaging revealed a lack of IgG-IgG polymerization or degradative protein fragmentation of the covalent epirubicin-(C(3)-amide)-[anti-HER2/neu] immunochemotherapeutic. Retained binding-avidity of epirubicin-(C(3)-amide)-[anti-HER2/neu] was validated by cell-ELISA utilizing monolayer populations of chemotherapeutic-resistant mammary adenocarcinoma SKBr-3 which highly overexpress membrane-associated HER2/neu complexes. Between epirubicin-equivalent concentrations of 10(-10) to 10(-6) M the covalent epirubicin-(C(3)-amide)-[anti-HER2/neu] immunochemotherapeutic consistently evoked levels of cytotoxic anti-neoplastic potency that were highly analogous to chemotherapeutic-equivalent concentrations of epirubicin. Cytotoxic anti-neoplastic potency of epirubicin-(C(3)-amide)-[anti-HER2/neu] against chemotherapeutic-resistant mammary adenocarcinoma SKBr-3 challenged with epirubicin-(C(3)-amide)-[anti-HER2/neu] at an epirubicin-equivalent concentration of 10(-6) M was 88.5% (e.g., 11.5% residual survival). Between final epirubicin-equivalent concentrations of 10(-8) and 10(-7) M there was a marked threshold increase in the mean cytotoxic anti-neoplastic activity for epirubicin-(C(3)-amide)-[anti-HER2/neu] from 9.9% to 66.9% (90.2% to 33.1% residual survival).
Collapse
Affiliation(s)
- Cody P Coyne
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA.
| | | | | |
Collapse
|
38
|
Choi SK, Thomas TP, Li MH, Desai A, Kotlyar A, Baker JR. Photochemical release of methotrexate from folate receptor-targeting PAMAM dendrimer nanoconjugate. Photochem Photobiol Sci 2012; 11:653-60. [PMID: 22234658 DOI: 10.1039/c2pp05355a] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoparticle (NP)-based targeted drug delivery involves cell-specific targeting followed by a subsequent therapeutic action from the therapeutic carried by the NP system. NPs conjugated with methotrexate (MTX), a potent inhibitor of dihydrofolate reductase (DHFR) localized in cytosol, have been under investigation as a delivery system to target cancer cells to enhance the therapeutic index of methotrexate, which is otherwise non-selectively cytotoxic. Despite improved therapeutic activity from MTX-conjugated NPs in vitro and in vivo, the therapeutic action of these conjugates following cellular entry is poorly understood; in particular it is unclear whether the therapeutic activity requires release of the MTX. This study investigates whether MTX must be released from a nanoparticle in order to achieve the therapeutic activity. We report herein light-controlled release of methotrexate from a dendrimer-based conjugate and provide evidence suggesting that MTX still attached to the nanoconjugate system is fully able to inhibit the activity of its enzyme target and the growth of cancer cells.
Collapse
Affiliation(s)
- Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Synthesis and in vitro Characterization of Semitelechelic Poly[N-(2-hydroxypropyl)methacrylamide]-Trastuzumab Conjugates Targeted to Breast Cancer. Macromol Biosci 2011; 12:55-60. [DOI: 10.1002/mabi.201100152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 07/07/2011] [Indexed: 12/14/2022]
|
40
|
Yang T, Wang Y, Li Z, Dai W, Yin J, Liang L, Ying X, Zhou S, Wang J, Zhang X, Zhang Q. Targeted delivery of a combination therapy consisting of combretastatin A4 and low-dose doxorubicin against tumor neovasculature. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 8:81-92. [PMID: 21664295 DOI: 10.1016/j.nano.2011.05.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 04/15/2011] [Accepted: 05/03/2011] [Indexed: 12/12/2022]
Abstract
UNLABELLED The present study demonstrates the applicability of a novel strategy that employs targeted delivery of combined treatment against tumor neovasculature. Briefly, a ligand of integrins, cyclic arginine-glycine-aspartic acid-tyrosine-lysine pentapeptide (cRGDyK), was conjugated to the PEG end of polyethylene glycol-b-poly lactic acid (PEG-b-PLA), and doxorubicin was chemically linked to the PLA end of PEG-b-PLA. The targeted dual-drug micelle system was prepared by mixing combretastatin A4 (an antivascular agent), PEG-b-PLA, and the above two conjugates using a solution-casting method. The targeted micelles significantly enhanced cellular uptake of the drug by B16-F10 cells and human umbilical vein endothelial cells through a receptor-mediated endocytosis. The cRGDyK-modified dual-drug system achieved an optimal antitumor effect, lifespan increase, antineovasculature, antiproliferation, and apoptosis induction, revealing the advantage of active targeting and the modified combination therapy. In conclusion, the integration of targeted delivery and combination therapy against tumor neovasculature represents a promising approach for cancer treatment. FROM THE CLINICAL EDITOR A ligand of integrins was conjugated to PEG-b-PLA, and doxorubicin was chemically linked to the PLA. Efficiency was demonstrated in a cancer model. The integration of targeted delivery and combination therapy against tumor neovasculature represents a promising approach for cancer treatment.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/therapeutic use
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/chemistry
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Doxorubicin/administration & dosage
- Doxorubicin/chemistry
- Doxorubicin/therapeutic use
- Drug Carriers/chemistry
- Drug Delivery Systems
- Human Umbilical Vein Endothelial Cells
- Humans
- Lactates/chemistry
- Lactates/therapeutic use
- Melanoma, Experimental
- Mice
- Mice, Inbred C57BL
- Micelles
- Neovascularization, Pathologic/drug therapy
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/therapeutic use
- Polyethylene Glycols/chemistry
- Polyethylene Glycols/therapeutic use
- Stilbenes/administration & dosage
- Stilbenes/chemistry
- Stilbenes/therapeutic use
Collapse
Affiliation(s)
- Tingyuan Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Coyne CP, Jones T, Sygula A, Bailey J, Pinchuk L. Epirubicin-[Anti-HER2/ neu] Synthesized with an Epirubicin-(C 13- imino)-EMCS Analog: Anti-Neoplastic Activity against Chemotherapeutic-Resistant SKBr-3 Mammary Carcinoma in Combination with Organic Selenium. ACTA ACUST UNITED AC 2011; 2:22-39. [PMID: 26229727 DOI: 10.4236/jct.2011.21004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE Discover the anti-neoplastic efficacy of epirubicin-(C13-imino)-[anti-HER2/neu] against chemotherapeutic-resistant SKBr-3 mammary carcinoma and delineate the capacity of selenium to enhance it's cytotoxic anti-neoplastic potency. METHODS In molar excess, EMCH was combined with epirubicin to create a covalent epirubicin-(C13-imino)-EMCH-maleimide intermediate with sulfhydryl-reactive properties. Monoclonal immunoglobulin selective for HER2/neu was then thiolated with 2-iminothiolane at the terminal ε-amine group of lysine residues. The sulfhydryl-reactive epirubicin-(C13-imino)-EMCH intermediate was then combined with thiolated anti-HER2/neu monoclonal immunoglobulin. Western-blot analysis was utilized to characterize the molecular weight profiles while binding of epirubicin-(C13-imino)-[anti-HER2/neu] to membrane receptors was determined by cell-ELISA utilizing populations of SKBr-3 mammary carcinoma that highly over-expresses HER2/neu complexes. Anti-neoplastic potency of epirubicin-(C13-imino)-[anti-HER2/neu] between the epirubicin-equivalent concentrations of 10-12 M and 10-7 M was determined by vitality staining analysis with and without the presence of selenium (5 μM). RESULTS Epiribucin-(C13-imino)-[anti-HER2/neu] between epirubicin-equivalent concentrations of 10-8 M to 10-7 M consistently evoked higher anti-neoplastic potency than "free" non-conjugated epirubicin which corresponded with previous investigations utilizing epirubicin-(C3-amide)-[anti-HER2/neu] and epirubicin-(C3-amide)-[anti-EGFR]. Selenium at 5 mM consistently enhanced the cytotoxic anti-neoplastic potency of epirubicin-(C13-imino)-[anti-HER2/neu] at epirubicin equivalent concentrations (10-12 to 10-7 M). CONCLUSIONS Epirubicin-(C13-imino)-[anti-HER2/neu] is more potent than epirubicin against chemotherapeutic-resistant SKBr-3 mammary carcinoma and selenium enhances epirubicin-(C13-imino)-[anti-HER2/neu] potency. The methodology applied for synthesizing epirubicin-(C13-imino)-[anti-HER2/neu] is relatively time convenient and has low instrumentation requirements.
Collapse
Affiliation(s)
- Cody P Coyne
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, USA
| | - Toni Jones
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, USA
| | - Andrzej Sygula
- Department Organic Chemistry, Mississippi State University, Mississippi State, USA
| | - John Bailey
- College of Osteopathic Medicine, William Cary University, Hattiesburg, USA
| | - Lesya Pinchuk
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, USA
| |
Collapse
|
42
|
Coyne CP, Jones T, Pharr T. Synthesis of a covalent gemcitabine-(carbamate)-[anti-HER2/neu] immunochemotherapeutic and its cytotoxic anti-neoplastic activity against chemotherapeutic-resistant SKBr-3 mammary carcinoma. Bioorg Med Chem 2010; 19:67-76. [PMID: 21169024 DOI: 10.1016/j.bmc.2010.11.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 11/15/2010] [Accepted: 11/22/2010] [Indexed: 02/06/2023]
Abstract
UNLABELLED Gemcitabine is a potent chemotherapeutic that exerts cytotoxic activity against several leukemias and a wide spectrum of carcinomas. A brief plasma half-life in part due to rapid deamination and chemotherapeutic-resistance frequently limit the utility of gemcitabine in clinical oncology. Selective 'targeted' delivery of gemcitabine represents a potential molecular strategy for simultaneously prolonging its plasma half-life and minimizing exposure of innocent tissues and organ systems. MATERIALS AND METHODS Gemcitabine was combined in molar excess with N-[p-maleimidophenyl]-isocyanate (PMPI) so that the isocyanate moiety of PMPI which exclusively reacts with hydroxyl groups preferentially created a carbamate covalent bond at the terminal C(5)-methylhydroxy group of gemcitabine. Monoclonal immunoglobulin with binding-avidity specifically for HER2/neu was thiolated with 2-iminothiolane at the terminal ε-amine group of lysine amino acid residues. The gemcitabine-(carbamate)-PMPI intermediate with a maleimide moiety that exclusively reacts with reduced sulfhydryl groups was then combined with thiolated anti-HER2/neu monoclonal immunoglobulin. Western-blot analysis was utilized to delineate the molecular weight profile for gemcitabine-(carbamate)-[anti-HER2/neu] while cell binding characteristics were determined by cell-ELISA utilizing SKBr-3 mammary carcinoma which highly over-expresses HER2/neu receptors. Cytotoxic anti-neoplastic potency of gemcitabine-(carbamate)-[anti-HER2/neu] between the gemcitabine-equivalent concentrations of 10(-12) and 10(-6)M was determined utilizing vitality staining analysis of chemotherapeutic-resistant SKBr-3 mammary carcinoma. RESULTS Gemcitabine-(carbamate)-[anti-HER2/neu] was synthesized at a molar incorporation index of 1:1.1 (110%) and had a molecular weight of 150kDa that was indistinguishable from reference control immunoglobulin fractions. Cell-ELISA detected progressive increases in SKBr-3 mammary carcinoma associated immunoglobulin with corresponding increases in covalent gemcitabine immunochemotherapeutic concentrations. The in vitro cytotoxic anti-neoplastic potency of gemcitabine-(carbamate)-[anti-HER2/neu] was approximately 20% and 32% at 10(-7) and 10(-6)M (gemcitabine-equivalent concentrations) after a 182-h incubation period. DISCUSSION The investigations describes for the first time a methodology for synthesizing a gemcitabine anti-HER2/neu immunochemotherapeutic by creating a covalent bond structure between the C(5)-methylhydroxy group of gemcitabine and thiolated lysine amino acid residues of monoclonal antibody or other biologically active protein fractions. Gemcitabine-(carbamate)-[anti-HER2/neu] possessed binding-avidity at HER2/neu receptors highly over-expressed by chemotherapeutic-resistant SKBr-3 mammary carcinoma. Alternatively, gemcitabine can be covalently linked at its C(5)-methylhydroxy group to monoclonal immunoglobulin fractions that possess binding-avidity for other receptors and membrane complexes uniquely highly over-expressed by a variety of neoplastic cell types. Compared to chemotherapeutic-resistant SKBr-3 mammary carcinoma, gemcitabine-(carbamate)-[anti-HER2/neu] immunochemotherapeutic is anticipated to exert higher levels of cytotoxic anti-neoplastic potency against other neoplastic cell types like pancreatic carcinoma, small-cell lung carcinoma, neuroblastoma, glioblastoma, oral squamous cell carcinoma, cervical epithelioid carcinoma, or leukemia/lymphoid neoplastic cell types based on their reportedly greater sensitivity to gemcitabine and gemcitabine covalent conjugates.
Collapse
Affiliation(s)
- C P Coyne
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, United States.
| | | | | |
Collapse
|
43
|
Chytil P, Etrych T, Kříž J, Šubr V, Ulbrich K. N-(2-Hydroxypropyl)methacrylamide-based polymer conjugates with pH-controlled activation of doxorubicin for cell-specific or passive tumour targeting. Synthesis by RAFT polymerisation and physicochemical characterisation. Eur J Pharm Sci 2010; 41:473-82. [DOI: 10.1016/j.ejps.2010.08.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 08/02/2010] [Indexed: 11/30/2022]
|
44
|
Etrych T, Chytil P, Kovář L, Říhová B, Ulbrich K. Micellar and Antibody-Targeted Polymer Therapeutics. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/masy.200900066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Etrych T, Kovář L, Šubr V, Braunová A, Pechar M, Chytil P, Říhova B, Ulbrich K. High-molecular-weight Polymers Containing Biodegradable Disulfide Bonds: Synthesis and In Vitro Verification of Intracellular Degradation. J BIOACT COMPAT POL 2010. [DOI: 10.1177/0883911509353485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The synthesis, physico-chemical behavior, and in vitro intracellular degradation of new biodegradable graft, diblock or multiblock polymer carriers that were designed to deliver bioactive compounds by passive tumor targeting were investigated. The graft polymer carriers consisted of the N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer backbone grafted with a semitelechelic HPMA copolymer. The diblock polymer carriers were prepared by condensation of two semitelechelic HPMA copolymers. The multiblock polymer drug carrier was prepared by oxidative polycondensation of PEG-bis-cysteine. In all three carrier systems, the single polymers were linked via biodegradable disulfide bonds forming the graft, diblock or multiblock polymers. These polymers are potential polymer carriers for solid tumor-specific drug delivery with subsequent intracellular degradation to short polymer fragments that can be excreted by glomerular filtration. Prolonged blood circulation, accumulation in solid tumors, and drug release from these carriers have been reported. Here, degradation of the polymers in model buffer solutions mimicking intracellular environment as well as after incubation with EL4 T-cell lymphoma cancer cells were investigated. In both cases, degradation resulted in polymer fragments of molecular weight below the renal threshold.
Collapse
Affiliation(s)
- Tomáš Etrych
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic,
| | - Lubomír Kovář
- Institute of Microbiology, Academy of Sciences of the Czech Republic Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Vladimír Šubr
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Alena Braunová
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Petr Chytil
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Blanka Říhova
- Institute of Microbiology, Academy of Sciences of the Czech Republic Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| |
Collapse
|
46
|
Vetvicka D, Hruby M, Hovorka O, Etrych T, Vetrik M, Kovar L, Kovar M, Ulbrich K, Rihova B. Biological evaluation of polymeric micelles with covalently bound doxorubicin. Bioconjug Chem 2010; 20:2090-7. [PMID: 19835372 DOI: 10.1021/bc900212k] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The main limitation of contemporary anticancer chemotherapy remains to be the insufficient specificity of the drugs for tumor tissue, which decreases the maximum tolerated dose due to severe side effects. Micellar drug delivery systems based on amphiphilic block copolymers with a very narrow size distribution (10 to 100 nm in diameter) is a novel innovative approach. Here, we report biological and pharmacological properties of polymeric micellar conjugate containing doxorubicin (DOX) covalently bound via hydrolytically cleavable hydrazone bonds to the micelle core. The system had a very low systemic toxicity (almost 20 times lower than free DOX) and long circulation in the bloodstream (with half of the dose after 24 h). Significant accumulation of tested micelles within the tumor was confirmed by fluorescent whole body imaging. Our new micellar system showed promising therapeutic activity against established murine EL-4 T-cell lymphoma; it was found that it is able to completely cure about 75% of tumor-bearing mice (with doses of either 1 x 150 mg DOX kg(-1) or 2 x 75 mg DOX kg(-1), administered i.v.). Moreover, treatment with micelles in cured mice induced tumor-specific resistance. Up to 80% of these mice survived rechallenge with original but not with distinct tumor cells.
Collapse
Affiliation(s)
- David Vetvicka
- Institute of Microbiology AS CR, v.v.i., Videnska 1083, 14220 Prague 4, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Etrych T, Strohalm J, Kovář L, Kabešová M, Říhová B, Ulbrich K. HPMA copolymer conjugates with reduced anti-CD20 antibody for cell-specific drug targeting. I. Synthesis and in vitro evaluation of binding efficacy and cytostatic activity. J Control Release 2009; 140:18-26. [DOI: 10.1016/j.jconrel.2009.07.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 07/13/2009] [Accepted: 07/15/2009] [Indexed: 11/27/2022]
|
48
|
Injac R, Strukelj B. Recent advances in protection against doxorubicin-induced toxicity. Technol Cancer Res Treat 2009; 7:497-516. [PMID: 19044329 DOI: 10.1177/153303460800700611] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Anthracycline antibiotics are among the most effective and commonly used anticancer drugs. Unfortunately, their clinical use is restricted by dose-dependent toxicity. Doxorubicin is an anthracycline antibiotic and cytotoxic (antineoplastic) agent. It is commonly used against ovarian, breast, lung, uterine and cervical cancers, Hodgkin's disease, soft tissue and primary bone sarcomas, as well against in several other cancer types. It has been shown that free radicals are involved in doxorubicin-induced toxicity. Doxorubicin causes the generation of free radicals and the induction of oxidative stress, associated with cellular injury. This review illustrates recent applications of different natural products, drugs, drug delivery systems, and approaches for protection against doxorubicin-induced toxicity (2006-present).
Collapse
Affiliation(s)
- R Injac
- University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia.
| | | |
Collapse
|
49
|
Etrych T, Mrkvan T, Chytil P, Koňák Č, Říhová B, Ulbrich K. N-(2-hydroxypropyl)methacrylamide-based polymer conjugates with pH-controlled activation of doxorubicin. I. New synthesis, physicochemical characterization and preliminary biological evaluation. J Appl Polym Sci 2008. [DOI: 10.1002/app.28466] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|