1
|
Kopeček J. Hydrophilic biomaterials: From crosslinked and self-assembled hydrogels to polymer-drug conjugates and drug-free macromolecular therapeutics. J Control Release 2024; 373:1-22. [PMID: 38734315 PMCID: PMC11384549 DOI: 10.1016/j.jconrel.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
This "Magnum Opus" accentuates my lifelong belief that the future of science is in the interdisciplinary approach to hypotheses formulation and problem solving. Inspired by the invention of hydrogels and soft contact lenses by my mentors, my six decades of research have continuously proceeded from the synthesis of biocompatible hydrogels to the development of polymer-drug conjugates, then generation of drug-free macromolecular therapeutics (DFMT) and finally to multi-antigen T cell hybridizers (MATCH). This interdisciplinary journey was inspiring; the lifetime feeling that one is a beginner in some aspects of the research is a driving force that keeps the enthusiasm high. Also, I wanted to illustrate that systematic research in one wide area can be a life-time effort without the need to jump to areas that are temporarily en-vogue. In addition to generating general scientific knowledge, hydrogels from my laboratory have been transferred to the clinic, polymer-drug conjugates to clinical trials, and drug-free macromolecular systems have an excellent potential for personalizing patient therapies. There is a limit to life but no limit to imagination. I anticipate that systematic basic research will contribute to the expansion of our knowledge and create a foundation for the design of new paradigms based on the comprehension of mechanisms of physiological processes. The emerging novel platform technologies in biomaterial-based devices and implants as well as in personalized nanomedicines will ultimately impact clinical practice.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Center for Controlled Chemical Delivery, Department of Molecular Pharmaceutics, Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
2
|
Li H, Li H, Stanton C, Ross RP, Zhao J, Chen W, Yang B. Exopolysaccharides Produced by Bifidobacterium longum subsp. longum YS108R Ameliorates DSS-Induced Ulcerative Colitis in Mice by Improving the Gut Barrier and Regulating the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7055-7073. [PMID: 38520351 DOI: 10.1021/acs.jafc.3c06421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
Ulcerative colitis (UC) is a major disease that has endangered human health. Our previous study demonstrated that Bifidobacterium longum subsp. longum YS108R, a ropy exopolysaccharide (EPS)-producing bacterium, could alleviate UC in mice, but it is unclear whether EPS is the key substance responsible for its action. In this study, we proposed to investigate the remitting effect of EPS from B. longum subsp. longum YS108R on UC in a DSS-induced UC mouse model. Water extraction and alcohol precipitation were applied to extract EPS from the supernatant of B. longum subsp. longum YS108R culture. Then the animal trial was performed, and the results indicated that YS108R EPS ameliorated colonic pathological damage and the intestinal barrier. YS108R EPS suppressed inflammation via NF-κB signaling pathway inhibition and attenuated oxidative stress via the Nrf2 signaling pathway activation. Remarkably, YS108R EPS regulated gut microbiota, as evidenced by an increase in short-chain fatty acid (SCFA)-producing bacteria and a decline in Gram-negative bacteria, resulting in an increase of propionate and butyrate and a reduction of lipopolysaccharide (LPS). Collectively, YS108R EPS manipulated the intestinal microbiota and its metabolites, which further improved the intestinal barrier and inhibited inflammation and oxidative stress, thereby alleviating UC.
Collapse
Affiliation(s)
- Huizhen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
| | - R Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
3
|
Steinhauff D, Armstrong B, Jensen MM, Ghandehari H. Development of Thermoresponsive Protein Complexes for Targeting CD20 Receptors. Macromol Biosci 2022; 22:e2200028. [PMID: 35526100 DOI: 10.1002/mabi.202200028] [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: 01/20/2022] [Revised: 04/04/2022] [Indexed: 11/07/2022]
Abstract
Therapeutics targeting cell receptors can elicits biological responses in situ. However, the ability to dictate when and where these responses occur is a current challenge. Therapeutic proteins can be combined with stimuli-responsive peptides to increase targeting and stimuli responsive behavior. To this end we have genetically engineered an elastinlike polypeptide (ELP) fusion protein for selective ELPylation. The addition of a charged foldable region provides these protein subunits with varied thermoresponsive properties from their parent ELPs. These subunits have responsive secondary structures, dependent on pH, indicating the capability to form coiled-coils with a complementary peptide tag. A Rituximab conjugate was generated herein, containing the complementary peptide. Upon mixing of the ELP and Rituximab subunits, the resulting protein complexes targeted CD20 receptors on Raji B cells, resulting in at least 2-fold increase in mean fluorescent intensities. These ELP subunits folded in vitro with complimentary the generated Rituximab conjugate. This work provides the basis for the design of a therapeutic stimuli-responsive biomacromolecule for targeting receptors. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Douglas Steinhauff
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Blair Armstrong
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Mark Martin Jensen
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Hamidreza Ghandehari
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA.,Utah Center for Nanomedicine, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 884112, USA
| |
Collapse
|
4
|
Abstract
The fields of precision imaging and drug delivery have revealed a number of tools to improve target specificity and increase efficacy in diagnosing and treating disease. Biological molecules, such as antibodies, continue to be the primary means of assuring active targeting of various payloads. However, molecular-scale recognition motifs have emerged in recent decades to achieve specificity through the design of interacting chemical motifs. In this regard, an assortment of bioorthogonal covalent conjugations offer possibilities for in situ complexation under physiological conditions. Herein, a related concept is discussed that leverages interactions from noncovalent or supramolecular motifs to facilitate in situ recognition and complex formation in the body. Classic supramolecular motifs based on host-guest complexation offer one such means of facilitating recognition. In addition, synthetic bioinspired motifs based on oligonucleotide hybridization and coiled-coil peptide bundles afford other routes to form complexes in situ. The architectures to include recognition of these various motifs for targeting enable both monovalent and multivalent presentation, seeking high affinity or engineered avidity to facilitate conjugation even under dilute conditions of the body. Accordingly, supramolecular "click chemistry" offers a complementary tool in the growing arsenal targeting improved healthcare efficacy.
Collapse
Affiliation(s)
| | | | - Matthew J. Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| |
Collapse
|
5
|
Gambles MT, Li J, Wang J, Sborov D, Yang J, Kopeček J. Crosslinking of CD38 Receptors Triggers Apoptosis of Malignant B Cells. Molecules 2021; 26:molecules26154658. [PMID: 34361811 PMCID: PMC8348492 DOI: 10.3390/molecules26154658] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/16/2023] Open
Abstract
Recently, we designed an inventive paradigm in nanomedicine—drug-free macromolecular therapeutics (DFMT). The ability of DFMT to induce apoptosis is based on biorecognition at cell surface, and crosslinking of receptors without the participation of low molecular weight drugs. The system is composed of two nanoconjugates: a bispecific engager, antibody or Fab’ fragment—morpholino oligonucleotide (MORF1) conjugate; the second nanoconjugate is a multivalent effector, human serum albumin (HSA) decorated with multiple copies of complementary MORF2. Here, we intend to demonstrate that DFMT is a platform that will be effective on other receptors than previously validated CD20. We appraised the impact of daratumumab (DARA)- and isatuximab (ISA)-based DFMT to crosslink CD38 receptors on CD38+ lymphoma (Raji, Daudi) and multiple myeloma cells (RPMI 8226, ANBL-6). The biological properties of DFMTs were determined by flow cytometry, confocal fluorescence microscopy, reactive oxygen species determination, lysosomal enlargement, homotypic cell adhesion, and the hybridization of nanoconjugates. The data revealed that the level of apoptosis induction correlated with CD38 expression, the nanoconjugates meet at the cell surface, mitochondrial signaling pathway is strongly involved, insertion of a flexible spacer in the structure of the macromolecular effector enhances apoptosis, and simultaneous crosslinking of CD38 and CD20 receptors increases apoptosis.
Collapse
Affiliation(s)
- M. Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; (M.T.G.); (J.L.); (J.W.)
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiahui Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; (M.T.G.); (J.L.); (J.W.)
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; (M.T.G.); (J.L.); (J.W.)
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Douglas Sborov
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; (M.T.G.); (J.L.); (J.W.)
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: (J.Y.); (J.K.)
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; (M.T.G.); (J.L.); (J.W.)
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: (J.Y.); (J.K.)
| |
Collapse
|
6
|
Rütter M, Milošević N, David A. Say no to drugs: Bioactive macromolecular therapeutics without conventional drugs. J Control Release 2020; 330:1191-1207. [PMID: 33207257 DOI: 10.1016/j.jconrel.2020.11.026] [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: 07/18/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022]
Abstract
The vast majority of nanomedicines (NM) investigated today consists of a macromolecular carrier and a drug payload (conjugated or encapsulated), with a purpose of preferential delivery of the drug to the desired site of action, either through passive accumulation, or by active targeting via ligand-receptor interaction. Several drug delivery systems (DDS) have already been approved for clinical use. However, recent reports are corroborating the notion that NM do not necessarily need to include a drug payload, but can exert biological effects through specific binding/blocking of important target proteins at the site of action. The seminal work of Kopeček et al. on N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers containing biorecognition motifs (peptides or oligonucleotides) for crosslinking cell surface non-internalizing receptors of malignant cells and inducing their apoptosis, without containing any low molecular weight drug, led to the definition of a special group of NM, termed Drug-Free Macromolecular Therapeutics (DFMT). Systems utilizing this approach are typically designed to employ pendant targeting-ligands on the same macromolecule to facilitate multivalent interactions with receptors. The lack of conventional small molecule drugs reduces toxicity and adverse effects at off-target sites. In this review, we describe different types of DFMT that possess biological activity without attached low molecular weight drugs. We classified the relevant research into several groups by their mechanisms of action, and compare the advantages and disadvantages of these different approaches. We show that identification of target sites, specificity of attached targeting ligands, binding affinity and the synthesis of carriers of defined size and ligand spacing are crucial aspects of DFMT development. We further discuss how knowledge in the field of NM accumulated in the past few decades can help in the design of a successful DFMT to speed up the translation into clinical practice.
Collapse
Affiliation(s)
- Marie Rütter
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Nenad Milošević
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel.
| |
Collapse
|
7
|
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]
|
8
|
Kopeček J, Yang J. Polymer nanomedicines. Adv Drug Deliv Rev 2020; 156:40-64. [PMID: 32735811 PMCID: PMC7736172 DOI: 10.1016/j.addr.2020.07.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
Polymer nanomedicines (macromolecular therapeutics, polymer-drug conjugates, drug-free macromolecular therapeutics) are a group of biologically active compounds that are characterized by their large molecular weight. This review focuses on bioconjugates of water-soluble macromolecules with low molecular weight drugs and selected proteins. After analyzing the design principles, different structures of polymer carriers are discussed followed by the examination of the efficacy of the conjugates in animal models and challenges for their translation into the clinic. Two innovative directions in macromolecular therapeutics that depend on receptor crosslinking are highlighted: a) Combination chemotherapy of backbone degradable polymer-drug conjugates with immune checkpoint blockade by multivalent polymer peptide antagonists; and b) Drug-free macromolecular therapeutics, a new paradigm in drug delivery.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
9
|
Zhang L, Fang Y, Li L, Yang J, Radford DC, Kopeček J. Human Serum Albumin-Based Drug-Free Macromolecular Therapeutics: Apoptosis Induction by Coiled-Coil-Mediated Cross-Linking of CD20 Antigens on Lymphoma B Cell Surface. Macromol Biosci 2018; 18:e1800224. [PMID: 30259654 PMCID: PMC6392022 DOI: 10.1002/mabi.201800224] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/10/2018] [Indexed: 01/25/2023]
Abstract
A therapeutic platform-drug-free macromolecular therapeutics (DFMT)-that induces apoptosis in B cells by cross-linking of CD20 receptors, without the need for low molecular weight cytotoxic drug, is developed. In this report, a DFMT system is synthesized and evaluated based on human serum albumin (HSA) and two complementary coiled-coil forming peptides, CCE and CCK. Fab' fragment of anti-CD20 monoclonal antibody rituximab is attached to CCE (Fab'-CCE); multiple grafts of CCK are conjugated to HSA (HSA-(CCK)7 ). The colocalization of both nanoconjugates at the surface of non-Hodgkin's lymphoma (NHL) Raji cells is demonstrated by confocal fluorescence microscopy. The colocalization leads to coiled-coil formation, CD20 cross-linking, and apoptosis induction. The apoptotic levels are evaluated by Annexin V, Caspase 3, and terminal deoxynucleotidyl transferase dUTP nick end labeling assays. Selective surface binding of DFMT to CD20+ cells is validated in experiments on a coculture of CD20+ (Raji) and CD20-(DG-75) cells. It is found that DFMT can trigger calcium influx only in Raji cells, but not in DG-75 cells. A highly specific treatment for NHL and other B cell malignancies with considerable translational potential is presented by HSA-based DFMT system.
Collapse
Affiliation(s)
- Libin Zhang
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yixin Fang
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD, University of Utah, Salt Lake City, UT, 84112, USA
| | - Lian Li
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD, University of Utah, Salt Lake City, UT, 84112, USA
| | | | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
10
|
Li H, Jin H, Wan W, Wu C, Wei L. Cancer nanomedicine: mechanisms, obstacles and strategies. Nanomedicine (Lond) 2018; 13:1639-1656. [PMID: 30035660 DOI: 10.2217/nnm-2018-0007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Targeting nanoparticles to cancers for improved therapeutic efficacy and decreased side effects remains a popular concept in the past decades. Although the enhanced permeability and retention effect serves as a key rationale for all the currently commercialized nanoformulations, it does not enable uniform delivery of nanoparticles to all tumorous regions in all patients with sufficient quantities. Also, the increase in overall survival is often modest. Many factors may influence the delivering process of nanoparticles, which must be taken into consideration for the promise of nanomedicine in patients to be realized. Herein, we review the mechanisms and influencing factors during the delivery of cancer therapeutics and summarize current strategies that have been developed for the fabrication of smart drug delivery systems.
Collapse
Affiliation(s)
- Huafei Li
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, PR China
- Tumor Immunology & Gene Therapy Center, Third Affiliated Hospital of the Second Military Medical University, 225 Changhai Road, Shanghai, 200438, PR China
- International Joint Cancer Institute, Translational Medicine Institute, the Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, PR China
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai, 200444, PR China
| | - Hai Jin
- Department of Thoracic Surgery/LaboratoryDiagnosis, First Affiliated Hospital of the Second Military Medical University,168 Changhai Road, Shanghai, 200438, PR China
| | - Wei Wan
- Department of Orthopedic Oncology, Spine Tumor Center, Second Affiliated Hospital of the Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, PR China
| | - Cong Wu
- Department of Thoracic Surgery/LaboratoryDiagnosis, First Affiliated Hospital of the Second Military Medical University,168 Changhai Road, Shanghai, 200438, PR China
| | - Lixin Wei
- Tumor Immunology & Gene Therapy Center, Third Affiliated Hospital of the Second Military Medical University, 225 Changhai Road, Shanghai, 200438, PR China
| |
Collapse
|
11
|
Ding L, Jiang Y, Zhang J, Klok HA, Zhong Z. pH-Sensitive Coiled-Coil Peptide-Cross-Linked Hyaluronic Acid Nanogels: Synthesis and Targeted Intracellular Protein Delivery to CD44 Positive Cancer Cells. Biomacromolecules 2018; 19:555-562. [PMID: 29284258 DOI: 10.1021/acs.biomac.7b01664] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The clinical translation of protein drugs that act intracellularly is limited by the absence of safe and efficient intracellular protein delivery vehicles. Here, pH-sensitive coiled-coil peptide-cross-linked hyaluronic acid nanogels (HA-cNGs) were designed and investigated for targeted intracellular protein delivery to CD44 overexpressing MCF-7 breast cancer cells. HA-cNGs were obtained with a small size of 176 nm from an equivalent mixture of hyaluronic acid conjugates with GY(EIAALEK)3GC (E3) and GY(KIAALKE)3GC (K3) peptides, respectively, at pH 7.4 by nanoprecipitation. Circular dichroism (CD) proved the formation of coiled-coil structures between E3 and K3 peptides at pH 7.4 while fast uncoiling at pH 5.0. HA-cNGs showed facile loading of cytochrome C (CC) and greatly accelerated CC release under mild acidic conditions (18.4%, 76.8%, and 91.4% protein release in 24 h at pH 7.4, 6.0, and 5.0, respectively). Confocal microscopy and flow cytometry displayed efficient internalization of CC-loaded HA-cNGs and effective endosomal escape of CC in MCF-7 cancer cells. Remarkably, HA-cNGs loaded with saporin, a ribosome inactivating protein, exhibited significantly enhanced apoptotic activity to MCF-7 cells with a low IC50 of 12.2 nM. These coiled-coil peptide-cross-linked hyaluronic acid nanogels have appeared as a simple and multifunctional platform for efficient intracellular protein delivery.
Collapse
Affiliation(s)
- Lingling Ding
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, People's Republic of China
| | - Yu Jiang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, People's Republic of China
| | - Jian Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, People's Republic of China
| | - Harm-Anton Klok
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, People's Republic of China.,Laboratoire des Polymères, Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL) , Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, People's Republic of China
| |
Collapse
|
12
|
Zhang L, Fang Y, Yang J, Kopeček J. Drug-free macromolecular therapeutics: Impact of structure on induction of apoptosis in Raji B cells. J Control Release 2016; 263:139-150. [PMID: 28024916 DOI: 10.1016/j.jconrel.2016.12.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 01/31/2023]
Abstract
Recently, we developed a new paradigm in macromolecular therapeutics that avoids the use of low molecular weight drugs. The activity of the "drug-free macromolecular therapeutics" is based on the biorecognition of complementary motifs at cell surface resulting in receptor crosslinking and apoptosis induction. The system is composed of two nanoconjugates: (1) a single-stranded morpholino oligonucleotide (MORF1) attached to an anti-CD20 Fab' fragment (Fab'-MORF1); (2) multiple copies of complementary oligonucleotide MORF2 grafted to a linear polymer of N-(2-hydroxypropyl)methacrylamide (HPMA) - P-(MORF2)x. The two conjugates crosslink CD20 antigens via MORF1-MORF2 hybridization at the surface of CD20+ malignant B-cells and induce apoptosis. Preclinical studies in a murine model of human non-Hodgkin's lymphoma showed cancer cells eradication and long-term survivors. The aim of this study was to determine the relationship between the detailed structure of the nanoconjugates and apoptosis induction in Raji cells to allow system optimization. The factors studied include the length of the MORF sequence, the valence of P-(MORF2)x (varying x), molecular weight of P-(MORF2)x, incorporation of a miniPEG spacer between Fab' and MORF1 and between polymer backbone and pendant MORF2, and comparison of two Fab' fragments, one from 1F5 antibody (Fab'1F5), the other from Rituximab (Fab'RTX). The results of apoptosis induction in human Burkitt's B-cell non-Hodgkin's lymphoma (NHL) Raji cells as determined using three apoptotic assays (Annexin V, Caspase 3, and TUNEL) indicated that: a) An improvement of apoptotic activity was observed for a 28 base pair MORF sequence when compared to MORFs composed of 20 and 25 base pairs. The differences depended on type of assay, concentration and exposure schedule (consecutive vs. premixed). b) The higher the valence of P-(MORF2)x the higher the levels of apoptosis. c) Higher molecular weight of P-(MORF2)x induced higher levels of apoptosis. d) A miniPEG8 spacer was effective in enhancing apoptotic levels in contrast to a miniPEG2 spacer. e) There was not a statistically significant difference when comparing Fab'1F5-MORF1 with Fab'RTX-MORF1.
Collapse
Affiliation(s)
- Libin Zhang
- Department of Pharmaceutics and Pharmaceutical Chemistry, CCCD, University of Utah, Salt Lake City, UT 84112, USA
| | - Yixin Fang
- Department of Pharmaceutics and Pharmaceutical Chemistry, CCCD, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, CCCD, University of Utah, Salt Lake City, UT 84112, USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, CCCD, University of Utah, Salt Lake City, UT 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
13
|
Chu TW, Kopeček J. Drug-Free Macromolecular Therapeutics--A New Paradigm in Polymeric Nanomedicines. Biomater Sci 2016; 3:908-22. [PMID: 26191406 DOI: 10.1039/c4bm00442f] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
This review highlights a unique research area in polymer-based nanomedicine designs. Drug-free macromolecular therapeutics induce apoptosis of malignant cells by the crosslinking of surface non-internalizing receptors. The receptor crosslinking is mediated by the biorecognition of high-fidelity natural binding motifs (such as antiparallel coiled-coil peptides or complementary oligonucleotides) that are grafted to the side chains of polymers or attached to targeting moieties against cell receptors. This approach features the absence of low-molecular-weight cytotoxic compounds. Here, we summarize the rationales, different designs, and advantages of drug-free macromolecular therapeutics. Recent developments of novel therapeutic systems for B-cell lymphomas are discussed, as well as relevant approaches for other diseases. We conclude by pointing out various potential future directions in this exciting new field.
Collapse
Affiliation(s)
- Te-Wei Chu
- Department of Pharmaceutics and Pharmaceutical Chemistry/Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry/Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA ; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
14
|
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]
|
15
|
Yang J, Shimada Y, Olsthoorn RCL, Snaar-Jagalska BE, Spaink HP, Kros A. Application of Coiled Coil Peptides in Liposomal Anticancer Drug Delivery Using a Zebrafish Xenograft Model. ACS NANO 2016; 10:7428-35. [PMID: 27504667 DOI: 10.1021/acsnano.6b01410] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The complementary coiled coil forming peptides E4 [(EIAALEK)4] and K4 [(KIAALKE)4] are known to trigger liposomal membrane fusion when tethered to lipid vesicles in the form of lipopeptides. In this study, we examined whether these coiled coil forming peptides can be used for drug delivery applications. First, we prepared E4 peptide modified liposomes containing the far-red fluorescent dye TO-PRO-3 iodide (E4-Lipo-TP3) and confirmed that E4-liposomes could deliver TP3 into HeLa cells expressing K4 peptide on the membrane (HeLa-K) under cell culture conditions in a selective manner. Next, we prepared doxorubicin-containing E4-liposomes (E4-Lipo-DOX) and confirmed that E4-liposomes could also deliver DOX into HeLa-K cells. Moreover, E4-Lipo-DOX showed enhanced cytotoxicity toward HeLa-K cells compared to free doxorubicin. To prove the suitability of E4/K4 coiled coil formation for in vivo drug delivery, we injected E4-Lipo-TP3 or E4-Lipo-DOX into zebrafish xenografts of HeLa-K. As a result, E4-liposomes delivered TP3 to the implanted HeLa-K cells, and E4-Lipo-DOX could suppress cancer proliferation in the xenograft when compared to nontargeted conditions (i.e., zebrafish xenograft with free DOX injection). These data demonstrate that coiled coil formation enables drug selectivity and efficacy in vivo. It is envisaged that these findings are a step forward toward biorthogonal targeting systems as a tool for clinical drug delivery.
Collapse
Affiliation(s)
- Jian Yang
- Leiden Institute of Chemistry-Supramolecular and Biomaterial Chemistry, Leiden University , Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Yasuhito Shimada
- Institute of Biology, Leiden University , Leiden 2311 EZ, The Netherlands
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine , Mie 514-8507, Japan
| | - René C L Olsthoorn
- Leiden Institute of Chemistry-Supramolecular and Biomaterial Chemistry, Leiden University , Einsteinweg 55, 2333CC Leiden, The Netherlands
| | | | - Herman P Spaink
- Institute of Biology, Leiden University , Leiden 2311 EZ, The Netherlands
| | - Alexander Kros
- Leiden Institute of Chemistry-Supramolecular and Biomaterial Chemistry, Leiden University , Einsteinweg 55, 2333CC Leiden, The Netherlands
| |
Collapse
|
16
|
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: 1120] [Impact Index Per Article: 124.4] [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
|
17
|
Abstract
This overview intends to demonstrate the close relationship between the design of smart biomaterials and water-soluble polymer-drug conjugates. First, the discovery and systematic studies of hydrogels based on crosslinked poly(meth)acrylic acid esters and substituted amides is described. Then, the lessons learned for the design of water-soluble polymers as drug carriers are highlighted. The current state-of-the-art in water-soluble, mainly poly[N-(2-hydroxypropyl)methacylamide (HPMA), polymer-drug conjugates is shown including the design of backbone degradable HPMA copolymer carriers. In the second part, the modern design of hybrid hydrogels focuses on the self-assembly of hybrid copolymers composed from the synthetic part (backbone) and biorecognizable grafts (coiled-coil forming peptides or morpholino oligonucleotides) is shown. The research of self-assembling hydrogels inspired the invention and design of drug-free macromolecular therapeutics - a new paradigm in drug delivery where crosslinking of non-internalizating CD20 receptors results in apoptosis in vitro and in vivo. The latter is mediated by biorecognition of complementary motifs; no low molecular weight drug is needed.
Collapse
Affiliation(s)
- Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, USA ; Department of Bioengineering, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
18
|
Kuzmov A, Minko T. Nanotechnology approaches for inhalation treatment of lung diseases. J Control Release 2015; 219:500-518. [PMID: 26297206 DOI: 10.1016/j.jconrel.2015.07.024] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/16/2015] [Accepted: 07/21/2015] [Indexed: 02/08/2023]
Abstract
Local administration of therapeutics by inhalation for treatment of lung diseases has the ability to deliver drugs, nucleic acids and peptides specifically to the site of their action and therefore enhance the efficacy of the treatment, limit the penetration of nebulized therapeutic agent(s) into the bloodstream and consequently decrease adverse systemic side effects of the treatment. Nanotechnology allows for a further enhancement of the treatment efficiency. The present review analyzes modern therapeutic approaches of inhaled nanoscale-based pharmaceutics for the detection and treatment of various lung diseases.
Collapse
Affiliation(s)
- Andriy Kuzmov
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway 08854, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick 08903, USA.
| |
Collapse
|
19
|
Yeo Y, Kim BK. Drug Carriers: Not an Innocent Delivery Man. AAPS JOURNAL 2015; 17:1096-104. [PMID: 26017163 DOI: 10.1208/s12248-015-9789-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/12/2015] [Indexed: 01/11/2023]
Abstract
Biomaterials used as drug carriers are often considered inactive and assumed to have no other roles than modifying pharmacokinetics and biodistribution of a drug. On the other hand, there are several examples in which the carrier materials show bioactivities in the body, which may have been underestimated or inadvertently ignored. This review highlights several examples where biomaterials used as drug carriers bring biological effects, known or newly discovered, and discusses their implications in development of new drug delivery systems.
Collapse
Affiliation(s)
- Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana, 47907, USA,
| | | |
Collapse
|
20
|
Zhang R, Yang J, Chu TW, Hartley JM, Kopeček J. Multimodality imaging of coiled-coil mediated self-assembly in a "drug-free" therapeutic system. Adv Healthc Mater 2015; 4:1054-65. [PMID: 25612325 DOI: 10.1002/adhm.201400679] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/15/2014] [Indexed: 01/23/2023]
Abstract
Two complementary coiled-coil peptides CCE/CCK are used to develop a "drug free" therapeutic system, which can specifically kill cancer cells without a drug. CCE is attached to the Fab' fragment of anti-CD20 1F5 antibody (Fab'-CCE), and CCK is conjugated in multiple grafts to poly[N-(2-hydroxypropyl)methacrylamide] (P-(CCK)x ). Two conjugates are consecutively administered: First, Fab'-CCE coats peptide CCE at CD20 antigen of lymphoma cell surface; second, CCE/CCK biorecognition between Fab'-CCE and P-(CCK)x leads to coiled-coil formation, CD20 crosslinking, membrane reorganization, and ultimately cell apoptosis. To prove that two conjugates can assemble at cell surface, multiple fluorescence imaging studies are performed, including 2-channel FMT, 3D confocal microscopy, and 4-color FACS. Confocal microscopy shows colocalization of two fluorescently labeled conjugates on non-Hodgkin's lymphoma (NHL) Raji cell surface, indicating "two-step" targeting specificity. The fluorescent images also reveal that these two conjugates can disrupt normal membrane lipid distribution and form lipid raft clusters, leading to cancer cell apoptosis. This "two-step" biorecognition capacity is further demonstrated in a NHL xenograft model, using fluorescent images at whole-body, tissue and cell levels. It is also found that delaying injection of P-(CCK)x can significantly enhance targeting efficacy. This high-specificity therapeutics provide a safe option to treat NHL and other B cell malignancies.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD; University of Utah; Salt Lake City UT 84112 USA
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD; University of Utah; Salt Lake City UT 84112 USA
| | - Te-Wei Chu
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD; University of Utah; Salt Lake City UT 84112 USA
| | - Jonathan M. Hartley
- Department of Bioengineering; University of Utah; Salt Lake City UT 84112 USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD; University of Utah; Salt Lake City UT 84112 USA
- Department of Bioengineering; University of Utah; Salt Lake City UT 84112 USA
| |
Collapse
|
21
|
Chu TW, Zhang R, Yang J, Chao MP, Shami PJ, Kopeček J. A Two-Step Pretargeted Nanotherapy for CD20 Crosslinking May Achieve Superior Anti-Lymphoma Efficacy to Rituximab. Theranostics 2015; 5:834-46. [PMID: 26000056 PMCID: PMC4440441 DOI: 10.7150/thno.12040] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/25/2015] [Indexed: 11/17/2022] Open
Abstract
The use of rituximab, an anti-CD20 mAb, in combination with chemotherapy is the current standard for the treatment of B-cell lymphomas. However, because of a significant number of treatment failures, there is a demand for new, improved therapeutics. Here, we designed a nanomedicine that crosslinks CD20 and directly induces apoptosis of B-cells without the need for toxins or immune effector functions. The therapeutic system comprises a pretargeting component (anti-CD20 Fab' conjugated with an oligonucleotide1) and a crosslinking component (N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer grafted with multiple complementary oligonucleotide2). Consecutive treatment with the two components resulted in CD20 clustering on the cell surface and effectively killed malignant B-cells in vivo. To enhance therapeutic efficacy, a two-step pretargeting approach was employed. We showed that the time lag between the two doses can be optimized based on pharmacokinetics and biodistribution of the Fab'-oligonucleotide1 conjugate. In a mouse model of human non-Hodgkin lymphoma (NHL), increasing the time lag from 1 h to 5 h resulted in dramatically improved tumor growth inhibition and animal survival. When the 5 h interval was used, the nanotherapy was more efficacious than rituximab and led to complete eradication of lymphoma cells with no signs of metastasis or disease recurrence. We further evaluated the nanomedicine using patient mantle cell lymphoma cells; the treatment demonstrated more potent apoptosis-inducing activity than rituximab hyper-crosslinked with secondary antibodies. In summary, our approach may constitute a novel treatment for NHL and other B-cell malignancies with significant advantages over conventional chemo-immunotherapy.
Collapse
|