1
|
Abou-Elnour FS, El-Habashy SE, Essawy MM, Abdallah OY. Alendronate/lactoferrin-dual decorated lipid nanocarriers for bone-homing and active targeting of ivermectin and methyl dihydrojasmonate for leukemia. BIOMATERIALS ADVANCES 2024; 162:213924. [PMID: 38875802 DOI: 10.1016/j.bioadv.2024.213924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Chronic myeloid leukemia is a hematological cancer, where disease relapse and drug resistance are caused by bone-hosted-residual leukemia cells. An innovative resolution is bone-homing and selective-active targeting of anticancer loaded-nanovectors. Herein, ivermectin (IVM) and methyl dihydrojasmonate (MDJ)-loaded nanostructured lipid carriers (IVM-NLC) were formulated then dually decorated by lactoferrin (Lf) and alendronate (Aln) to optimize (Aln/Lf/IVM-NLC) for active-targeting and bone-homing potential, respectively. Aln/Lf/IVM-NLC (1 mg) revealed nano-size (73.67 ± 0.06 nm), low-PDI (0.43 ± 0.06), sustained-release of IVM (62.75 % at 140-h) and MDJ (78.7 % at 48-h). Aln/Lf/IVM-NLC afforded substantial antileukemic-cytotoxicity on K562-cells (4.29-fold lower IC50), higher cellular uptake and nuclear fragmentation than IVM-NLC with acceptable cytocompatibility on oral-epithelial-cells (as normal cells). Aln/Lf/IVM-NLC effectively upregulated caspase-3 and BAX (4.53 and 15.9-fold higher than IVM-NLC, respectively). Bone homing studies verified higher hydroxyapatite affinity of Aln/Lf/IVM-NLC (1 mg; 22.88 ± 0.01 % at 3-h) and higher metaphyseal-binding (1.5-fold increase) than untargeted-NLC. Moreover, Aln/Lf/IVM-NLC-1 mg secured 1.35-fold higher in vivo bone localization than untargeted-NLC, with lower off-target distribution. Ex-vivo hemocompatibility and in-vivo biocompatibility of Aln/Lf/IVM-NLC (1 mg/mL) were established, with pronounced amelioration of hepatic and renal toxicity compared to higher Aln doses. The innovative Aln/Lf/IVM-NLC could serve as a promising nanovector for bone-homing, active-targeted leukemia therapy.
Collapse
Affiliation(s)
- Fatma S Abou-Elnour
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Salma E El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Marwa M Essawy
- Department of Oral Pathology, Faculty of Dentistry, Alexandria University, Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Rizzi F, Panniello A, Comparelli R, Arduino I, Fanizza E, Iacobazzi RM, Perrone MG, Striccoli M, Curri ML, Scilimati A, Denora N, Depalo N. Luminescent Alendronic Acid-Conjugated Micellar Nanostructures for Potential Application in the Bone-Targeted Delivery of Cholecalciferol. Molecules 2024; 29:2367. [PMID: 38792228 PMCID: PMC11123821 DOI: 10.3390/molecules29102367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Vitamin D, an essential micronutrient crucial for skeletal integrity and various non-skeletal physiological functions, exhibits limited bioavailability and stability in vivo. This study is focused on the development of polyethylene glycol (PEG)-grafted phospholipid micellar nanostructures co-encapsulating vitamin D3 and conjugated with alendronic acid, aimed at active bone targeting. Furthermore, these nanostructures are rendered optically traceable in the UV-visible region of the electromagnetic spectrum via the simultaneous encapsulation of vitamin D3 with carbon dots, a newly emerging class of fluorescents, biocompatible nanoparticles characterized by their resistance to photobleaching and environmental friendliness, which hold promise for future in vitro bioimaging studies. A systematic investigation is conducted to optimize experimental parameters for the preparation of micellar nanostructures with an average hydrodynamic diameter below 200 nm, ensuring colloidal stability in physiological media while preserving the optical luminescent properties of the encapsulated carbon dots. Comprehensive chemical-physical characterization of these micellar nanostructures is performed employing optical and morphological techniques. Furthermore, their binding affinity for the principal inorganic constituent of bone tissue is assessed through a binding assay with hydroxyapatite nanoparticles, indicating significant potential for active bone-targeting. These formulated nanostructures hold promise for novel therapeutic interventions to address skeletal-related complications in cancer affected patients in the future.
Collapse
Affiliation(s)
- Federica Rizzi
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| | - Annamaria Panniello
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
| | - Roberto Comparelli
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| | - Ilaria Arduino
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (I.A.); (R.M.I.)
| | - Elisabetta Fanizza
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
- Department of Chemistry, University of Bari, 70125 Bari, Italy
| | - Rosa Maria Iacobazzi
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (I.A.); (R.M.I.)
| | - Maria Grazia Perrone
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (M.G.P.); (A.S.)
| | - Marinella Striccoli
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
| | - Maria Lucia Curri
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
- Department of Chemistry, University of Bari, 70125 Bari, Italy
| | - Antonio Scilimati
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (M.G.P.); (A.S.)
| | - Nunzio Denora
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari, 70125 Bari, Italy; (I.A.); (R.M.I.)
| | - Nicoletta Depalo
- CNR-Institute for Chemical and Physical Process, 70125 Bari, Italy; (F.R.); (A.P.); (R.C.); (E.F.); (M.S.); (M.L.C.)
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), 50121 Firenze, Italy
| |
Collapse
|
3
|
Xiao B, Ackun-Farmmer MA, Adjei-Sowah E, Liu Y, Chandrasiri I, Benoit DSW. Advancing Bone-Targeted Drug Delivery: Leveraging Biological Factors and Nanoparticle Designs to Improve Therapeutic Efficacy. ACS Biomater Sci Eng 2024; 10:2224-2234. [PMID: 38537162 DOI: 10.1021/acsbiomaterials.3c01022] [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] [Indexed: 04/09/2024]
Abstract
Designing targeted drug delivery systems to effectively treat bone diseases ranging from osteoporosis to nonunion bone defects remains a significant challenge. Previously, nanoparticles (NPs) self-assembled from diblock copolymers of poly(styrene-alt-maleic anhydride)-b-poly(styrene) (PSMA-b-PS) delivering a Wnt agonist were shown to effectively target bone and improve healing via the introduction of a peptide with high affinity to tartrate-resistant acid phosphatase (TRAP), an enzyme deposited by the osteoclasts during bone remodeling. Despite these promising results, the underlying biological factors governing targeting and subsequent drug delivery system (DDS) design parameters have not been examined to enable the rational design to improve bone selectivity. Therefore, this work investigated the effect of target ligand density, the treatment window after injury, specificity of TRAP binding peptide (TBP), the extent of TRAP deposition, and underlying genetic factors (e.g., mouse strain differences) on TBP-NP targeting. Data based on in vitro binding studies and in vivo biodistribution analyses using a murine femoral fracture model suggest that TBP-NP-TRAP interactions and TBP-NP bone accumulation were ligand-density-dependent; in vitro, TRAP affinity was correlated with ligand density up to the maximum of 200,000 TBP ligands/NP, while NPs with 80,000 TBP ligands showed 2-fold increase in fracture accumulation at day 21 post injury compared with that of untargeted or scrambled controls. While fracture accumulation exhibited similar trends when injected at day 3 compared to that at day 21 postfracture, there were no significant differences observed between TBP-functionalized and control NPs, possibly due to saturation of TRAP by NPs at day 3. Leveraging a calcium-depletion diet, TRAP deposition and TBP-NP bone accumulation were positively correlated, confirming that TRAP-TBP binding leads to TBP-NP bone accumulation in vivo. Furthermore, TBP-NP exhibited similar bone accumulation in both C57BL/6 and BALB/c mouse strains versus control NPs, suggesting the broad applicability of TBP-NP regardless of the underlying genetic differences. These studies provide insight into TBP-NP design, mechanism, and therapeutic windows, which inform NP design and treatment strategies for fractures and other bone-associated diseases that leverage TRAP, such as marrow-related hematologic diseases.
Collapse
Affiliation(s)
- Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14623, United States
| | - Marian A Ackun-Farmmer
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14623, United States
| | - Emmanuela Adjei-Sowah
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14623, United States
| | - Yuxuan Liu
- Materials Science Program, University of Rochester, Rochester, New York 14623, United States
| | - Indika Chandrasiri
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14623, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14623, United States
- Department of Chemical Engineering, University of Rochester, Rochester, New York 14623, United States
- Materials Science Program, University of Rochester, Rochester, New York 14623, United States
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon 97403, United States
| |
Collapse
|
4
|
Zhai X, Peng S, Zhai C, Wang S, Xie M, Guo S, Bai J. Design of Nanodrug Delivery Systems for Tumor Bone Metastasis. Curr Pharm Des 2024; 30:1136-1148. [PMID: 38551047 DOI: 10.2174/0113816128296883240320040636] [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: 12/29/2023] [Accepted: 03/04/2024] [Indexed: 06/28/2024]
Abstract
Tumor metastasis is a complex process that is controlled at the molecular level by numerous cytokines. Primary breast and prostate tumors most commonly metastasize to bone, and the development of increasingly accurate targeted nanocarrier systems has become a research focus for more effective anti-bone metastasis therapy. This review summarizes the molecular mechanisms of bone metastasis and the principles and methods for designing bone-targeted nanocarriers and then provides an in-depth review of bone-targeted nanocarriers for the treatment of bone metastasis in the context of chemotherapy, photothermal therapy, gene therapy, and combination therapy. Furthermore, this review also discusses the treatment of metastatic and primary bone tumors, providing directions for the design of nanodelivery systems and future research.
Collapse
Affiliation(s)
- Xiaoqing Zhai
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China
| | - Shan Peng
- School of Stomatology, Weifang Medical University, Weifang 261053, China
| | - Chunyuan Zhai
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China
| | - Shuai Wang
- People's Hospital of Gaoqing County, Zibo 256399, China
| | - Meina Xie
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Shoudong Guo
- School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
5
|
Hu Q, Zhang Y, Mukerabigwi JF, Wang H, Cao Y. Polymer Conjugate as the New Promising Drug Delivery System for Combination Therapy against Cancer. Curr Top Med Chem 2024; 24:1101-1119. [PMID: 39005059 DOI: 10.2174/0115680266280603240321064308] [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: 10/09/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 07/16/2024]
Abstract
This review highlights the advantages of combination therapy using polymer conjugates as drug delivery systems for cancer treatment. In this review, the specific structures and materials of polymer conjugates, as well as the different types of combination chemotherapy strategies, are discussed. Specific targeting strategies, such as monoclonal antibody therapy and small molecule ligands, are also explored. Additionally, self-assembled polymer micelles and overcoming multidrug resistance are described as potential strategies for combination therapy. The assessment of combinational therapeutic efficacy and the challenges associated with polymer conjugates are also addressed. The future outlook aims to overcome these challenges and improve the effectiveness of drug delivery systems for combination therapy. The conclusion emphasizes the potential of polymer conjugates in combination therapy while acknowledging the need for further research and development in this field.
Collapse
Affiliation(s)
- Qiang Hu
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yuannian Zhang
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Jean Felix Mukerabigwi
- Department of Chemistry, University of Rwanda, College of Science and Technology, Po. Box: 3900, Kigali, Rwanda
| | - Haili Wang
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yu Cao
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| |
Collapse
|
6
|
Subrahmanyam N, Yathavan B, Yu SM, Ghandehari H. Targeting Intratibial Osteosarcoma Using Water-Soluble Copolymers Conjugated to Collagen Hybridizing Peptides. Mol Pharm 2023; 20:1670-1680. [PMID: 36724294 PMCID: PMC10799843 DOI: 10.1021/acs.molpharmaceut.2c00880] [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] [Indexed: 02/03/2023]
Abstract
Osteosarcoma (OS) is the most common form of primary malignant bone cancer in adolescents. Over the years, OS prognosis has greatly improved due to adjuvant and neoadjuvant (preoperative) chemotherapeutic treatment, increasing the chances of successful surgery and reducing the need for limb amputation. However, chemotherapeutic treatment to treat OS is limited by off-target toxicities and requires improved localization at the tumor site. Collagen, the main constituent of bone tissue, is extensively degraded and remodeled in OS, leading to an increased availability of denatured (monomeric) collagen. Collagen hybridizing peptides (CHPs) comprise a class of peptides rationally designed to selectively bind to denatured collagen. In this work, we have conjugated CHPs as targeting moieties to water-soluble N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers to target OS tumors. We demonstrated increased accumulation of collagen-targeted HPMA copolymer-CHP conjugates compared to nontargeted HPMA copolymers, as well as increased retention compared to both nontargeted copolymers and CHPs, in a murine intratibial OS tumor model. Furthermore, we used microcomputed tomography analysis to evaluate the bone microarchitecture and correlated bone morphometric parameters (porosity, bone volume, and surface area) with maximum accumulation (Smax) and accumulation at 168 h postinjection (S168) of the copolymers at the tumor. Our results provide the foundation for the use of HPMA copolymer-CHP conjugates as targeted drug delivery systems in OS tumors.
Collapse
Affiliation(s)
- Nithya Subrahmanyam
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - Bhuvanesh Yathavan
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - S Michael Yu
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Hamidreza Ghandehari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
7
|
Shimina GG, Bateneva AV, Tsyplenkova ES, Gamaley SG, Esina TI, Volosnikova EA, Danilenko ED. HEMOSTIMULATING PROPERTIES OF THE CONJUGATES OF GRANULOCYTE-MACROPHAGE COLONY STIMULATING FACTOR WITH ALENDRONIC ACID. PHARMACY & PHARMACOLOGY 2022. [DOI: 10.19163/2307-9266-2022-10-5-472-482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the work is to evaluate the hemostimulating activity of recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) conjugates with alendronic acid (ALN) in the model of cytostatic myelosuppression and the dynamics of rhGM-CSF accumulation as a part of the conjugate in the bone tissue and bone marrow of mice.Materials and methods. The conjugates obtained by a solid-phase synthesis using 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide or periodate oxidation, were used. A hemostimulating activity was evaluated in a model of a cytostatic myelosuppression induced by the administration of cyclophosphamide to CBA/Calac mice. RhGM-CSF preparations were injected subcutaneously for 4-5 days at the dose of 90 µg/kg. After the injections cycle had been completed, the total leukocyte and segmented neutrophil counts were carried out in the blood samples, and the total karyocyte count was carried out in the bone marrow samples.The tissue distribution of rhGM-CSF preparations was assessed in outbred CD-1 mice after a single intravenous administration at the effective dose. The content of rhGM-CSF in blood, femoral tissue and bone marrow was determined by enzyme immunoassay.Results. RhGM-CSF conjugates with ALN have been shown to retain the ability of the original protein to increase the number of leukocytes, segmented blood neutrophils, and bone marrow karyocytes under the action of conjugates. The stimulation of the neutrophil production used to be observed at earlier times than in the case of rhGM-CSF. The increase in the total number of bone marrow cells after the introduction of all three conjugates was more pronounced compared to the original protein (by 34%). The increased hemostimulatory effect of the AEG conjugate was accompanied by a more intense accumulation of rhGM-CSF in the bone tissue and bone marrow of mice. The rhGM-CSF introduced into the conjugate was detected in the bone tissue for 24 h and it circulated in the bloodstream for a longer time compared to the original protein.Conclusion. The data obtained make it possible to conclude that further work on the development of effective hemostimulating drugs based on rhGM-CSF conjugates with ALN, is promising.
Collapse
Affiliation(s)
- G. G. Shimina
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - A. V. Bateneva
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - E. S. Tsyplenkova
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - S. G. Gamaley
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - T. I. Esina
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - E. A. Volosnikova
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| | - E. D. Danilenko
- Institute of Medical Biotechnology of State Research Center of Virology and Biotechnology “VECTOR”,
Federal Service for the Surveillance on Consumer Rights Protection and Human Well-being
| |
Collapse
|
8
|
Yadav P, Dua C, Bajaj A. Advances in Engineered Biomaterials Targeting Angiogenesis and Cell Proliferation for Cancer Therapy. CHEM REC 2022; 22:e202200152. [PMID: 36103616 DOI: 10.1002/tcr.202200152] [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: 05/31/2022] [Revised: 08/23/2022] [Indexed: 12/15/2022]
Abstract
Antiangiogenic therapy in combination with chemotherapeutic agents is an effective strategy for cancer treatment. However, this combination therapy is associated with several challenges including non-specific biodistribution leading to systemic toxicity. Biomaterial-mediated codelivery of chemotherapeutic and anti-angiogenic agents can exploit their passive and active targeting abilities, leading to improved drug accumulation at the tumor site and therapeutic outcomes. In this review, we present the progress made in the field of engineered biomaterials for codelivery of chemotherapeutic and antiangiogenic agents. We present advances in engineering of liposome/hydrogel/micelle-based biomaterials for delivery of combination of anticancer and anti-angiogenesis drugs, or combination of anticancer and siRNA targeting angiogenesis, and targeted nanoparticles. We then present our perspective on developing strategies for targeting angiogenesis and cell proliferation for cancer therapy.
Collapse
Affiliation(s)
- Poonam Yadav
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad - Gurgaon Expressway, Faridabad, 121001, India
| | - Chhavi Dua
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad - Gurgaon Expressway, Faridabad, 121001, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad - Gurgaon Expressway, Faridabad, 121001, India
| |
Collapse
|
9
|
Hu B, Zhang Y, Zhang G, Li Z, Jing Y, Yao J, Sun S. Research progress of bone-targeted drug delivery system on metastatic bone tumors. J Control Release 2022; 350:377-388. [PMID: 36007681 DOI: 10.1016/j.jconrel.2022.08.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Bone metastases are common in malignant tumors and the effect of conventional treatment is limited. How to effectively inhibit tumor bone metastasis and deliver the drug to the bone has become an urgent issue to be solved. While bone targeting drug delivery systems have obvious advantages in the treatment of bone tumors. The research on bone-targeted anti-tumor therapy has made significant progress in recent years. We introduced the related tumor pathways of bone metastases. The tumor microenvironment plays an important role in metastatic bone tumors. We introduce a drug-loading systems based on different environment-responsive nanocomposites for anti-tumor and anti-metastatic research. According to the process of bone metastases and the structure of bone tissue, we summarized the information on bone-targeting molecules. Bisphosphate has become the first choice of bone-targeted drug delivery carrier because of its affinity with hydroxyapatite in bone. Therefore, we sought to summarize the bone-targeting molecule of bisphosphate to identify the modification effect on bone-targeting. And this paper discusses the relationship between bisphosphate bone targeting molecular structure and drug delivery carriers, to provide some new ideas for the research and development of bone-targeting drug delivery carriers. Targeted therapy will make a more outstanding contribution to the treatment of tumors.
Collapse
Affiliation(s)
- Beibei Hu
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China; State Key Laboratory Breeding Base-Hebei Province, Key Laboratory of Molecular Chemistry for Drug, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Yongkang Zhang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Guogang Zhang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Zhongqiu Li
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Yongshuai Jing
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Jun Yao
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China.
| | - Shiguo Sun
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China.
| |
Collapse
|
10
|
Zhong Y, Li S. New Progress in Improving the Delivery Methods of Bisphosphonates in the Treatment of Bone Tumors. Drug Des Devel Ther 2021; 15:4939-4959. [PMID: 34916778 PMCID: PMC8672028 DOI: 10.2147/dddt.s337925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/11/2021] [Indexed: 11/23/2022] Open
Abstract
Bone tumors are tumors that occur in the bone or its accessory tissues, including primary tumors and metastatic tumors. The main mechanism of bisphosphonate is to inhibit the resorption of destructive bone, inhibit the activity of osteoclasts and reduce the concentration of blood calcium. Therefore, bisphosphonates can be used for malignant hypercalcaemia, pain caused by osteolytic bone metastasis, prevention of osteolytic bone metastasis, multiple myeloma osteopathy, improving radiosensitivity and so on. However, the traditional administration of bisphosphonates can cause a series of adverse reactions. To overcome this disadvantage, it is necessary to develop novel methods to improve the delivery of bisphosphonates. In this paper, the latest research progress of new and improved bisphosphonate drug delivery methods in the treatment of bone tumors is reviewed. At present, the main design idea is to connect bisphosphonate nanoparticles, liposomes, microspheres, microcapsules, couplings, prodrugs and bone tissue engineering to targeted anti-tumors systems, and positive progress has been made in in vitro and animal experiments. However, its safety and effectiveness in human body still need to be verified by more studies.
Collapse
Affiliation(s)
- Yu Zhong
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning Province, People's Republic of China
| | - Su Li
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning Province, People's Republic of China
| |
Collapse
|
11
|
Yang H, Yu Z, Ji S, Huo Q, Yan J, Gao Y, Niu Y, Xu M, Liu Y. Targeting bone microenvironments for treatment and early detection of cancer bone metastatic niches. J Control Release 2021; 341:443-456. [PMID: 34748870 DOI: 10.1016/j.jconrel.2021.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
Bone tissues are the main metastatic sites of many cancers, and bone metastasis is an important cause of death. When bone metastasis occurs, dynamic interactions between tumor cells and bone tissues promote changes in the tumor-bone microenvironments that are conducive to tumor growth and progression, which also promote several related diseases, including pathological fracture, bone pain, and hypercalcemia. Accordingly, it has obvious clinical benefits for improving the cure rate and reducing the occurrence of related diseases through targeting bone microenvironments for the treatment and early detection of cancer bone metastasis niches. In this review, we briefly analyzed the relationship between bone microstructures and tumor metastasis, as well as microenvironmental changes in osteoblasts, osteoclasts, immune cells, and extracellular and bone matrixes caused when metastatic tumor cells colonize bones. We also discuss novel designs in nanodrugs for inhibiting tumor proliferation and migration through targeting to tumor bone metastases and abnormal bone-microenvironment components. In addition, related researches on the early detection of bone and multi-organ metastases by nanoprobes are also introduced. And we look forward to provide some useful proposals and enlightenments on nanotechnology-based drug delivery and probes for the treatment and early detection of bone metastasis.
Collapse
Affiliation(s)
- Hongbin Yang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, Jiangsu, China; School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Zhenyan Yu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Shuaishuai Ji
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Qiang Huo
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Juanzhu Yan
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Yue Gao
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, Jiangsu, China
| | - Yimin Niu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Neurology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Ming Xu
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, Jiangsu, China.
| | - Yang Liu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
12
|
Korake S, Shaikh A, Salve R, Gajbhiye KR, Gajbhiye V, Pawar A. Biodegradable dendritic Boltorn™ nanoconstructs: A promising avenue for cancer theranostics. Int J Pharm 2020; 594:120177. [PMID: 33333177 DOI: 10.1016/j.ijpharm.2020.120177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
The family of Boltorn™ H40 dendrimers is an imperative subclass of hyperbranched biodegradable polymers (HBPs), which has received mounting attention as a result of its inimitable chemical, physical and biodegradable properties. These properties embrace three-dimensional dendrimeric nanoarchitecture to avert tanglement between polymer branches, adequate spatial cavities for increased encapsulation of guest molecules, good solubility as well as low viscosity to improve processability, and a huge number of surface functional groups for chemical manipulations. Similarly, low toxicity, non-immunogenicity, and natural biodegradation are significant and critical advantages in therapeutic applications as compared to other dendritic polymers. All these characteristics of Boltorn™ H40 are of pronounced importance for planning and developing advanced targeted cargo delivery carriers for cancer therapy. The present review highlights the applications of Boltorn™ H40 HBPs for the transport of chemotherapeutic agents to manage various types of cancers.
Collapse
Affiliation(s)
- S Korake
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Pune 411038, India
| | - A Shaikh
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India; Savitribai Phule Pune University, Pune 411004, India
| | - R Salve
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India; Savitribai Phule Pune University, Pune 411004, India
| | - K R Gajbhiye
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Pune 411038, India
| | - V Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India; Savitribai Phule Pune University, Pune 411004, India.
| | - A Pawar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Pune 411038, India.
| |
Collapse
|
13
|
Yamashita S, Katsumi H, Shimizu E, Nakao Y, Yoshioka A, Fukui M, Kimura H, Sakane T, Yamamoto A. Dendrimer-based micelles with highly potent targeting to sites of active bone turnover for the treatment of bone metastasis. Eur J Pharm Biopharm 2020; 157:85-96. [PMID: 33039547 DOI: 10.1016/j.ejpb.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 01/29/2023]
Abstract
Bone-drug targeting therapies using nanoparticles based on targeting ligands remain challenging due to their uptake clearance at non-target sites such as the liver, kidney, and spleen. Furthermore, the distribution sites of nanoparticles in bones have not been fully investigated, thus halting the development of more effective bone metastasis treatment strategies. In this study, we developed nanoparticles self-assembled from cholesterol-terminated, polyethylene glycol-conjugated, aspartic acid (Asp)-modified polyamidoamine dendrimer (Asp-PAMAM-Micelles) with targeting to active bone turnover sites associated with bone metastasis pathogenesis. On analysis through whole-body single photon emission computed tomography/computed tomography (SPECT/CT) imaging, 111In-Asp-PAMAM-Micelles showed high specificity to active bone turnover sites (especially the joints in the lower limbs, shoulder, and pelvis) after intravenous injection in mice. The lower limb bone uptake clearance for 111In-Asp-PAMAM-Micelles encapsulating paclitaxel (PTX) was 3.5-fold higher than that for 111In-unmodified PAMAM-Micelles (PTX). 3H-PTX encapsulated Asp-PAMAM-Micelles effectively accumulated in the lower limb bones in a similar manner as the 111In-Asp-PAMAM-Micelles (PTX). In a bone metastatic tumor mouse model, the tumor growth in the lower limb bones was significantly inhibited by injection of Asp-PAMAM-Micelles (PTX) compared to unmodified PAMAM-Micelles (PTX). Our results demonstrate that Asp-PAMAM-Micelles are sophisticated drug delivery systems for highly potent targeting to active bone turnover sites.
Collapse
Affiliation(s)
- Shugo Yamashita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Erika Shimizu
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Yuto Nakao
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Ayane Yoshioka
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Minako Fukui
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Toshiyasu Sakane
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan; Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
14
|
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]
|
15
|
Gao X, Li L, Cai X, Huang Q, Xiao J, Cheng Y. Targeting nanoparticles for diagnosis and therapy of bone tumors: Opportunities and challenges. Biomaterials 2020; 265:120404. [PMID: 32987273 DOI: 10.1016/j.biomaterials.2020.120404] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022]
Abstract
A variety of targeted nanoparticles were developed for the diagnosis and therapy of orthotopic and metastatic bone tumors during the past decade. This critical review will focus on principles and methods in the design of these bone-targeted nanoparticles. Ligands including bisphosphonates, aspartic acid-rich peptides and synthetic polymers were grafted on nanoparticles such as PLGA nanoparticles, liposomes, dendrimers and inorganic nanoparticles for bone targeting. Besides, other ligands such as monoclonal antibodies, peptides and aptamers targeting biomarkers on tumor/bone cells were identified for targeted diagnosis and therapy. Examples of targeted nanoparticles for the early detection of bone metastatic tumors and the ablation of cancer via chemotherapy, photothermal therapy, gene therapy and combination therapy will be intensively reviewed. The development of multifunctional nanoparticles to break down the "vicious" cycle between tumor cell proliferation and bone resorption, and the challenges and perspectives in this area will be discussed.
Collapse
Affiliation(s)
- Xin Gao
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Lin Li
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Xiaopan Cai
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Quan Huang
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China.
| | - Jianru Xiao
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China.
| | - Yiyun Cheng
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
16
|
Barkat HA, Das SS, Barkat MA, Beg S, Hadi HA. Selective targeting of cancer signaling pathways with nanomedicines: challenges and progress. Future Oncol 2020; 16:2959-2979. [PMID: 32805124 DOI: 10.2217/fon-2020-0198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. Regardless of advances in understanding the molecular mechanics of cancer, its treatment is still lacking and the death rates for many forms of the disease remain the same as six decades ago. Although a variety of therapeutic agents and strategies have been reported, these therapies often failed to provide efficient therapy to patients as a consequence of the inability to deliver right and adequate chemotherapeutic agents to the right place. However, the situation has started to revolutionize substantially with the advent of novel 'targeted' nanocarrier-based cancer therapies. Such therapies hold great potential in cancer management as they are biocompatible, tailored to specific needs, tolerated and deliver enough drugs at the targeted site. Their use also enhances the delivery of chemotherapeutics by improving biodistribution, lowering toxicity, inhibiting degradation and increasing cellular uptake. However, in some instances, nonselective targeting is not enough and the inclusion of a ligand moiety is required to achieve tumor targeting and enhanced drug accumulation at the tumor site. This contemporary review outlines the targeting potential of nanocarriers, highlighting the essentiality of nanoparticles, tumor-associated molecular signaling pathways, and various biological and pathophysiological barriers.
Collapse
Affiliation(s)
- Harshita Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al-Batin, 39524, Saudi Arabia
| | - Sabya Sachi Das
- Department of Pharmaceutical Sciences & Technology, BIT, Mesra, Ranchi, 835215, Jharkhand, India
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al-Batin, 39524, Saudi Arabia
| | - Sarwar Beg
- Department of Pharmaceutics, Nanomedicine Research Lab, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Hazrina Ab Hadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| |
Collapse
|
17
|
Pisarevsky E, Blau R, Epshtein Y, Ben-Shushan D, Eldar-Boock A, Tiram G, Koshrovski-Michael S, Scomparin A, Pozzi S, Krivitsky A, Shenbach-Koltin G, Yeini E, Fridrich L, White R, Satchi-Fainaro R. Rational Design of Polyglutamic Acid Delivering an Optimized Combination of Drugs Targeting Mutated BRAF and MEK in Melanoma. ADVANCED THERAPEUTICS 2020; 3:2000028. [PMID: 35754977 PMCID: PMC9223483 DOI: 10.1002/adtp.202000028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Indexed: 12/17/2022]
Abstract
Targeted therapies against cancer can relieve symptoms and induce remission, however, they often present limited duration of disease control, cause side effects and often induce acquired resistance. Therefore, there is a great motivation to develop a unique delivery system, targeted to the tumor, in which we can combine several active entities, increase the therapeutic index by reducing systemic exposure, and enhance their synergistic activity. To meet these goals, we chose the biocompatible and biodegradable poly(α,L-glutamic acid) (PGA) as a nanocarrier that facilitates extravasation-dependent tumor targeting delivery. The RAS/RAF/MEK/ERK pathway when aberrantly activated in melanoma, can lead to uncontrolled cell proliferation, induced invasion, and reduced apoptosis. Here, we selected two drugs targeting this pathway; a MEK1/2 inhibitor (selumetinib; SLM) and a modified BRAF inhibitor (modified dabrafenib; mDBF), that exhibited synergism in vitro. We synthesized and characterized our nanomedicine of PGA conjugated to SLM and mDBF (PGA-SLM-mDBF). PGA-SLM-mDBF inhibited the proliferation of melanoma cells and decreased their migratory and sprouting abilities without inducing a hemolytic effect. Moreover, the polymer-2-drugs conjugate exhibited superior anti-tumor activity in comparison with the two separate polymer-drug conjugates in vitro and with free drugs in a mouse model of primary melanoma and prolonged survival at a lower dose.
Collapse
Affiliation(s)
- Evgeni Pisarevsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rachel Blau
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yana Epshtein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gal Shenbach-Koltin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lidar Fridrich
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Richard White
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
18
|
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
|
19
|
Rajora AK, Ravishankar D, Zhang H, Rosenholm JM. Recent Advances and Impact of Chemotherapeutic and Antiangiogenic Nanoformulations for Combination Cancer Therapy. Pharmaceutics 2020; 12:pharmaceutics12060592. [PMID: 32630584 PMCID: PMC7356724 DOI: 10.3390/pharmaceutics12060592] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Traditional chemotherapy, along with antiangiogenesis drugs (combination cancer therapy), has shown reduced tumor recurrence and improved antitumor effects, as tumor growth and metastasis are often dependent on tumor vascularization. However, the effect of combination chemotherapy, including synergism and additive and even antagonism effects, depends on drug combinations in an optimized ratio. Hence, nanoformulations are ideal, demonstrating a great potential for the combination therapy of chemo-antiangiogenesis for cancer. The rationale for designing various nanocarriers for combination therapy is derived from organic (polymer, lipid), inorganic, or hybrid materials. In particular, hybrid nanocarriers that consist of more than one material construct provide flexibility for different modes of entrapment within the same carrier—e.g., physical adsorption, encapsulation, and chemical conjugation strategies. These multifunctional nanocarriers can thus be used to co-deliver chemo- and antiangiogenesis drugs with tunable drug release at target sites. Hence, this review attempts to survey the most recent advances in nanoformulations and their impact on cancer treatment in a combined regimen—i.e., conventional cytotoxic and antiangiogenesis agents. The mechanisms and site-specific co-delivery strategies are also discussed herein, along with future prospects.
Collapse
Affiliation(s)
- Amit Kumar Rajora
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- Correspondence: (A.K.R.); (J.M.R.)
| | - Divyashree Ravishankar
- Bioscience Department, Sygnature Discovery, Bio City, Pennyfoot St, Nottingham NG1 1GR, UK;
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- Turku Bioscience Center, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- Correspondence: (A.K.R.); (J.M.R.)
| |
Collapse
|
20
|
Otaka A, Yamaguchi T, Saisho R, Hiraga T, Iwasaki Y. Bone-targeting phospholipid polymers to solubilize the lipophilic anticancer drug. J Biomed Mater Res A 2020; 108:2090-2099. [PMID: 32323471 DOI: 10.1002/jbm.a.36968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/19/2020] [Accepted: 03/28/2020] [Indexed: 12/31/2022]
Abstract
Current chemotherapy methods have limited effectiveness in eliminating bone metastasis, which leads to a poor prognosis associated with severe bone disorders. To provide regional chemotherapy for this metastatic tumor, a bone-targeting drug carrier was produced by introducing the osteotropic bisphosphonate alendronate (ALN) units into an amphiphilic phospholipid polymer, poly(2-methacryloyloxyethyl phosphorylcholine-co-n-butyl methacrylate). The polymer can form nanoparticles with a diameter of less than 30 nm; ALN units were exposed to the outer layer of the particle. A simple mixing procedure was used to encapsulate a hydrophobic anticancer drug, known as docetaxel (DTX), in the polymer nanoparticle, providing a uniform solution of a polymer-DTX complex in the aqueous phase. The complex showed anticancer activities against several breast cancer cell lines, and the complex formation did not hamper the pharmacological effect of DTX. The fluorescence observations evaluated by an in vivo imaging system and fluorescence microscopy showed that the addition of ALN to the polymer-DTX complex enhanced bone accumulation. Bone-targeting phospholipid polymers are potential solubilizing excipients used to formulate DTX and deliver the hydrophobic drug to bone tissues by blood administration.
Collapse
Affiliation(s)
| | - Tomoki Yamaguchi
- Department of Chemistry and Materials Engineering Faculty of Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| | - Ryoya Saisho
- Department of Chemistry and Materials Engineering Faculty of Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| | - Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Nagano, Japan
| | - Yasuhiko Iwasaki
- ORDIST, Kansai University, Osaka, Japan.,Department of Chemistry and Materials Engineering Faculty of Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| |
Collapse
|
21
|
Rani S, Gupta U. HPMA-based polymeric conjugates in anticancer therapeutics. Drug Discov Today 2020; 25:997-1012. [PMID: 32334073 DOI: 10.1016/j.drudis.2020.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/23/2020] [Accepted: 04/11/2020] [Indexed: 11/17/2022]
Abstract
Polymer therapeutics has gained prominence due to an attractive structural polymer chemistry and its applications in diseases therapy. In this review, we discussed the development and capabilities of N-(2-hydroxypropyl) methacrylamide (HPMA) and HPMA-drug conjugates in cancer therapy. The design, architecture, and structural properties of HPMA make it a versatile system for the synthesis of polymeric conjugations for biomedical applications. Research suggests that HPMA could be a possible alternative for polymers such polyethylene glycol (PEG) in biomedical applications. Although numerous clinical trials of HPMA-drug conjugates are ongoing, yet no product has been successfully brought to the market. Thus, further research is required to develop HPMA-drug conjugates as successful cancer therapeutics.
Collapse
Affiliation(s)
- Sarita Rani
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
22
|
Salameh JW, Zhou L, Ward SM, Santa Chalarca CF, Emrick T, Figueiredo ML. Polymer-mediated gene therapy: Recent advances and merging of delivery techniques. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1598. [PMID: 31793237 PMCID: PMC7676468 DOI: 10.1002/wnan.1598] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/02/2019] [Accepted: 09/19/2019] [Indexed: 01/01/2023]
Abstract
The ability to safely and precisely deliver genetic materials to target sites in complex biological environments is vital to the success of gene therapy. Numerous viral and nonviral vectors have been developed and evaluated for their safety and efficacy. This study will feature progress in synthetic polymers as nonviral vectors, which benefit from their chemical versatility, biocompatibility, and ability to carry both therapeutic cargo and targeting moieties. The combination of synthetic gene carrying constructs with advanced delivery techniques promises new therapeutic options for treating and curing genetic disorders. This article is characterized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Janelle W. Salameh
- The Weldon School of Biomedical Engineering and the
Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette,
Indiana
| | - Le Zhou
- Polymer Science and Engineering Department, University of
Massachusetts, Amherst, Massachusetts
| | - Sarah M. Ward
- Polymer Science and Engineering Department, University of
Massachusetts, Amherst, Massachusetts
| | | | - Todd Emrick
- Polymer Science and Engineering Department, University of
Massachusetts, Amherst, Massachusetts
| | - Marxa L. Figueiredo
- Department of Basic Medical Sciences and the
Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette,
Indiana
| |
Collapse
|
23
|
Yee EMH, Cirillo G, Brandl MB, Black DS, Vittorio O, Kumar N. Synthesis of Dextran-Phenoxodiol and Evaluation of Its Physical Stability and Biological Activity. Front Bioeng Biotechnol 2019; 7:183. [PMID: 31440502 PMCID: PMC6694440 DOI: 10.3389/fbioe.2019.00183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/15/2019] [Indexed: 12/04/2022] Open
Abstract
Phenoxodiol, an isoflavene anti-tumor agent, was conjugated on the polysaccharide dextran using immobilized laccase as biocatalyst. The success of the enzymatic conjugation was determined by UV-vis spectrophotometry and its functionalization degree was assessed by 1H NMR and was found to be 3.25 mg phenoxodiol/g of conjugate. An accelerated stability test showed that the resultant conjugate was nine times more stable than the free phenoxodiol when tested for its residual anti-oxidant activity with the Folin-Ciocalteu assay. The in vitro anti-proliferative activity of the conjugate was evaluated against neuroblastoma SKN-BE(2)C, triple-negative breast cancer MDA-MB-231, and glioblastoma U87 cancer cells. The conjugate was shown to be generally more potent than phenoxodiol against all three cell types tested. Additionally, the cytotoxicity and anti-angiogenic activity of the conjugate were also evaluated against non-malignant human lung fibroblast MRC-5 and human microvascular endothelial cells HMEC-1, respectively. The conjugate was found to be 1.5 times less toxic than phenoxodiol while mostly retaining 62% of its anti-angiogenic activity in the conjugate form. This study provides further evidence that the conjugation of natural product-derived drugs onto polysaccharide molecules such as dextran can lead to better stability and enhanced biological activity of the conjugate compared to the free drug alone.
Collapse
Affiliation(s)
- Eugene M. H. Yee
- School of Chemistry, University of New South Wales, Sydney, NSW, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Miriam B. Brandl
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, Australia
| | - David StC Black
- School of Chemistry, University of New South Wales, Sydney, NSW, Australia
| | - Orazio Vittorio
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
24
|
Zeng Y, Hoque J, Varghese S. Biomaterial-assisted local and systemic delivery of bioactive agents for bone repair. Acta Biomater 2019; 93:152-168. [PMID: 30711659 PMCID: PMC6615988 DOI: 10.1016/j.actbio.2019.01.060] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 01/05/2023]
Abstract
Although bone tissues possess an intrinsic capacity for repair, there are cases where bone healing is either impaired or insufficient, such as fracture non-union, osteoporosis, osteomyelitis, and cancers. In these cases, treatments like surgical interventions are used, either alone or in combination with bioactive agents, to promote tissue repair and manage associated clinical complications. Improving the efficacy of bioactive agents often requires carriers, with biomaterials being a pivotal player. In this review, we discuss the role of biomaterials in realizing the local and systemic delivery of biomolecules to the bone tissue. The versatility of biomaterials enables design of carriers with the desired loading efficiency, release profile, and on-demand delivery. Besides local administration, systemic administration of drugs is necessary to combat diseases like osteoporosis, warranting bone-targeting drug delivery systems. Thus, chemical moieties with the affinity towards bone extracellular matrix components like apatite minerals have been widely utilized to create bone-targeting carriers with better biodistribution, which cannot be achieved by the drugs alone. Bone-targeting carriers combined with the desired drugs or bioactive agents have been extensively investigated to enhance bone healing while minimizing off-target effects. Herein, these advancements in the field have been systematically reviewed. STATEMENT OF SIGNIFICANCE: Drug delivery is imperative when surgical interventions are not sufficient to address various bone diseases/defects. Biomaterial-assisted delivery systems have been designed to provide drugs with the desired loading efficiency, sustained release, and on-demand delivery to enhance bone healing. By surveying recent advances in the field, this review outlines the design of biomaterials as carriers for the local and systemic delivery of bioactive agents to the bone tissue. Particularly, biomaterials that bear chemical moieties with affinity to bone are attractive, as they can present the desired bioactive agents to the bone tissue efficiently and thus enhance the drug efficacy for bone repair.
Collapse
Affiliation(s)
- Yuze Zeng
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA
| | - Jiaul Hoque
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
25
|
Liu T, Romanova S, Wang S, Hyun MA, Zhang C, Cohen SM, Singh RK, Bronich TK. Alendronate-Modified Polymeric Micelles for the Treatment of Breast Cancer Bone Metastasis. Mol Pharm 2019; 16:2872-2883. [PMID: 31150251 DOI: 10.1021/acs.molpharmaceut.8b01343] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although the prognosis of patients with breast cancer continues to improve, breast cancer metastasis to bones remains high in incidence and challenging to manage. Here, we report the development of bone-homing alendronate (ALN)-anchored biodegradable polymeric micelles for the targeted treatment of metastatic cancer to bone. These micelles exhibited bone protective capacity including the recruitment, differentiation, and resorption activity of the osteoclasts. Encapsulation of docetaxel (DTX), the first-line chemotherapeutic for treatment of metastatic breast cancer, in ALN-modified micelles results in a sustained release, enhanced cytotoxicity, and improved pharmacokinetics. In the syngeneic animal model of late-stage disseminated breast cancer bone metastasis, the treatment with targeted DTX-loaded micelles attenuated the tumorigenesis and significantly improved animal lifespan compared to the conventional surfactant-based formulation (free DTX). These findings indicate potential applications of the osteotropic nanomedicines for bone metastasis treatment.
Collapse
|
26
|
Maso K, Grigoletto A, Vicent MJ, Pasut G. Molecular platforms for targeted drug delivery. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:1-50. [DOI: 10.1016/bs.ircmb.2019.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Adjei IM, Temples MN, Brown SB, Sharma B. Targeted Nanomedicine to Treat Bone Metastasis. Pharmaceutics 2018; 10:E205. [PMID: 30366428 PMCID: PMC6320768 DOI: 10.3390/pharmaceutics10040205] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Bone metastases are common complications of solid tumors, particularly those of the prostate, breast, and lungs. Bone metastases can lead to painful and devastating skeletal-related events (SREs), such as pathological fractures and nerve compressions. Despite advances in treatment for cancers in general, options for bone metastases remain inadequate and generally palliative. Anticancer drugs (chemotherapy and radiopharmaceuticals) do not achieve therapeutic concentrations in the bone and are associated with dose-limiting side effects to healthy tissues. Nanomedicines, with their tunable characteristics, have the potential to improve drug targeting to bone metastases while decreasing side effects for their effective treatment. In this review, we present the current state of the art for nanomedicines to treat bone metastases. We also discuss new treatment modalities enhanced by nanomedicine and their effects on SREs and disease progression.
Collapse
Affiliation(s)
- Isaac M Adjei
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Madison N Temples
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Shannon B Brown
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| |
Collapse
|
28
|
Deshantri AK, Varela Moreira A, Ecker V, Mandhane SN, Schiffelers RM, Buchner M, Fens MHAM. Nanomedicines for the treatment of hematological malignancies. J Control Release 2018; 287:194-215. [PMID: 30165140 DOI: 10.1016/j.jconrel.2018.08.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022]
Abstract
Hematological malignancies (HM) are a collection of malignant transformations originating from cells in the primary or secondary lymphoid organs. Leukemia, lymphoma, and multiple myeloma comprise the three major types of HM. Current treatment consists of bone marrow transplantation, radiotherapy, immunotherapy and chemotherapy. Although, many chemotherapeutic drugs are clinically available for the treatment of HM, the use of these agents is limited due to dose-related toxicity and lack of specificity to tumor tissue. Moreover, the poor pharmacokinetic profile of most of the chemotherapeutics requires high dosage and frequent administration to maintain therapeutic levels at the target site, both increasing adverse effects. This underlines an urgent need for a suitable drug delivery system to improve efficacy, safety, and pharmacokinetic properties of conventional therapeutics. Nanomedicines have proven to enhance these properties for anticancer therapeutics. The most extensively studied nanomedicine systems are lipid-based nanoparticles and polymeric nanoparticles. Typically, nanomedicines are small sub-micron sized particles in the size range of 20-200 nm. The biocompatible and biodegradable nature of nanomedicines makes them attractive vehicles to improve drug delivery. Their small size allows them to extravasate and accumulate at malignant sites passively by means of the enhanced permeability and retention (EPR) effect, resulting from rapid angiogenesis and inflammation. Moreover, the specificity to the target tissue can be further enhanced by surface modification of nanoparticles. This review describes currently available therapies as well as limitations and potential advantages of nanomedicine formulations for treatment of various types of HM. Additionally, recent investigational and approved nanomedicine formulations and their limited applications in HM are discussed.
Collapse
Affiliation(s)
- Anil K Deshantri
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd, India
| | - Aida Varela Moreira
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Veronika Ecker
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sanjay N Mandhane
- Biological Research Pharmacology Department, Sun Pharma Advanced Research Company Ltd, India
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maike Buchner
- Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Marcel H A M Fens
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
29
|
Iwasaki Y, Yokota A, Otaka A, Inoue N, Yamaguchi A, Yoshitomi T, Yoshimoto K, Neo M. Bone-targeting poly(ethylene sodium phosphate). Biomater Sci 2018; 6:91-95. [PMID: 29184942 DOI: 10.1039/c7bm00930e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Poly(ethylene sodium phosphate) (PEP·Na) showed excellent cytocompatibility and in vivo bone affinity. Moreover, PEP·Na did not interact with thrombin, which is a coagulation-related protein. Because immobilization of therapeutic agents and imaging probes on PEP·Na is easily performed, PEP·Na is a promising polymer for bone-targeted therapies.
Collapse
Affiliation(s)
- Yasuhiko Iwasaki
- Department of Chemistry and Materials Engineering, Kansai University, 3-3-35, Yamate-cho, Suita-shi, Osaka 564-8680, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Wang Z, Li Y, Zhou F, Piao Z, Hao J. β-elemene enhances anticancer bone neoplasms efficacy of paclitaxel through regulation of GPR124 in bone neoplasms cells. Oncol Lett 2018; 16:2143-2150. [PMID: 30008912 PMCID: PMC6036473 DOI: 10.3892/ol.2018.8909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/24/2018] [Indexed: 12/19/2022] Open
Abstract
The purpose of the present study was to investigate the anticancer effects of β-elemene and paclitaxel for bone neoplasms. MTT assay, reverse transcription-quantitative polymerase chain reaction, western blotting, flow cytometry and immunostaining were used to analyze the combined effects of β-elemene and paclitaxel both in vitro and in vivo. The results demonstrated that combined treatment of β-elemene and paclitaxel (β-elemene-paclitaxel) significantly inhibited growth and aggressiveness of U-2OS cells compared with either β-elemene or paclitaxel treatment alone. It was demonstrated that β-elemene promoted paclitaxel-induced apoptosis of U-2OS cells. Anti-apoptosis B-cell lymphoma (Bcl)-2 and Bcl-w genes were downregulated and pro-apoptotic Bcl-2-associated agonist of cell death and caspase-3 genes were upregulated in U-2OS cells following treatment with β-elemene-paclitaxel. Treatment of β-elemene-paclitaxel arrested the cell cycle and decreased cyclin-dependent kinase, cyclin-B1, P21 and P27 expression levels and decreased resistant genes alterations of ATP binding cassette subfamily B member 1, LDL receptor related protein and TS in U-2OS cells. Results demonstrated that β-elemene-paclitaxel decreased G-protein coupled receptor 124 (GPR124), vascular endothelial growth factor receptor, matrix metallopeptidase (MMP)-3, MMP-9 expression levels and increased endostatin, TIMP metallopeptidase inhibitor (TIMP)-1, TIMP-2 expression in U-2OS cells. In vivo assay demonstrated that β-elemene-paclitaxel treatment inhibited tumor growth of BALB/c-nu/nu nude mice and prolonged survival rate of tumor-bearing mice. Immunostaining demonstrated that β-elemene-paclitaxel treatment increased apoptotic bodies, GPR124 and increased endostatin, TIMP-1 and TIMP-2 expression in tumor tissues. In conclusion, these results suggest that the combined treatment of β-elemene-paclitaxel is more effective at inhibiting bone neoplasm growth than β-elemene or paclitaxel single treatment GPR124.
Collapse
Affiliation(s)
- Zongze Wang
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| | - Ying Li
- Renal Department of Internal Medicine, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Fengxin Zhou
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| | - Zhe Piao
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| | - Jian Hao
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| |
Collapse
|
31
|
Farrell KB, Karpeisky A, Thamm DH, Zinnen S. Bisphosphonate conjugation for bone specific drug targeting. Bone Rep 2018; 9:47-60. [PMID: 29992180 PMCID: PMC6037665 DOI: 10.1016/j.bonr.2018.06.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/26/2022] Open
Abstract
Bones provide essential functions and are sites of unique biochemistry and specialized cells, but can also be sites of disease. The treatment of bone disorders and neoplasia has presented difficulties in the past, and improved delivery of drugs to bone remains an important goal for achieving effective treatments. Drug targeting strategies have improved drug localization to bone by taking advantage of the high mineral concentration unique to the bone hydroxyapatite matrix, as well as tissue-specific cell types. The bisphosphonate molecule class binds specifically to hydroxyapatite and inhibits osteoclast resorption of bone, providing direct treatment for degenerative bone disorders, and as emerging evidence suggests, cancer. These bone-binding molecules also provide the opportunity to deliver other drugs specifically to bone by bisphosphonate conjugation. Bisphosphonate bone-targeted therapies have been successful in treatment of osteoporosis, primary and metastatic neoplasms of the bone, and other bone disorders, as well as refining bone imaging. In this review, we focus upon the use of bisphosphonate conjugates with antineoplastic agents, and overview bisphosphonate based imaging agents, nanoparticles, and other drugs. We also discuss linker design potential and the current state of bisphosphonate conjugate research progress. Ongoing investigations continue to expand the possibilities for bone-targeted therapeutics and for extending their reach into clinical practice.
Collapse
Affiliation(s)
- Kristen B Farrell
- MBC Pharma Inc., 12635 East Montview Blvd., Aurora, CO 80045-0100, United States of America
| | - Alexander Karpeisky
- MBC Pharma Inc., 12635 East Montview Blvd., Aurora, CO 80045-0100, United States of America
| | - Douglas H Thamm
- Flint Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523-1620, United States of America
| | - Shawn Zinnen
- MBC Pharma Inc., 12635 East Montview Blvd., Aurora, CO 80045-0100, United States of America
| |
Collapse
|
32
|
Targeted drug delivery for tumor therapy inside the bone marrow. Biomaterials 2018; 155:191-202. [DOI: 10.1016/j.biomaterials.2017.11.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/26/2017] [Accepted: 11/21/2017] [Indexed: 12/18/2022]
|
33
|
Goldman E, Zinger A, da Silva D, Yaari Z, Kajal A, Vardi-Oknin D, Goldfeder M, Schroeder JE, Shainsky-Roitman J, Hershkovitz D, Schroeder A. Nanoparticles target early-stage breast cancer metastasis in vivo. NANOTECHNOLOGY 2017; 28:43LT01. [PMID: 28872058 DOI: 10.1088/1361-6528/aa8a3d] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Despite advances in cancer therapy, treating cancer after it has metastasized remains an unmet clinical challenge. In this study we demonstrate that 100 nm liposomes target triple-negative murine breast-cancer metastases post intravenous administration. Metastatic breast cancer was induced in BALB/c mice either experimentally, by a tail vein injection of 4T1 cells, or spontaneously, after implanting a primary tumor xenograft. To track their biodistribution in vivo the liposomes were labeled with multi-modal diagnostic agents, including indocyanine green and rhodamine for whole-animal fluorescent imaging, gadolinium for magnetic resonance imaging (MRI), and europium for a quantitative biodistribution analysis. The accumulation of liposomes in the metastases peaked at 24 h post the intravenous administration, similar to the time they peaked in the primary tumor. The efficiency of liposomal targeting to the metastatic tissue exceeded that of a non-liposomal agent by 4.5-fold. Liposomes were detected at very early stages in the metastatic progression, including metastatic lesions smaller than 2 mm in diameter. Surprisingly, while nanoparticles target breast cancer metastasis, they may also be found in elevated levels in the pre-metastatic niche, several days before metastases are visualized by MRI or histologically in the tissue. This study highlights the promise of diagnostic and therapeutic nanoparticles for treating metastatic cancer, possibly even for preventing the onset of the metastatic dissemination by targeting the pre-metastatic niche.
Collapse
Affiliation(s)
- Evgeniya Goldman
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel. The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vogus DR, Krishnan V, Mitragotri S. A review on engineering polymer drug conjugates to improve combination chemotherapy. Curr Opin Colloid Interface Sci 2017. [DOI: 10.1016/j.cocis.2017.08.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Wu VM, Mickens J, Uskoković V. Bisphosphonate-Functionalized Hydroxyapatite Nanoparticles for the Delivery of the Bromodomain Inhibitor JQ1 in the Treatment of Osteosarcoma. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25887-25904. [PMID: 28731328 PMCID: PMC5794714 DOI: 10.1021/acsami.7b08108] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Osteosarcoma (OS) is one of the most common neoplasia among children, and its survival statistics have been stagnating since the combinatorial anticancer therapy triad was first introduced. Here, we report on the assessment of the effect of hydroxyapatite (HAp) nanoparticles loaded with medronate, the simplest bisphosphonate, as a bone-targeting agent and JQ1, a small-molecule bromodomain inhibitor, as a chemotherapeutic in different 2D and 3D K7M2 OS in vitro models. Both additives decreased the crystallinity of HAp, but the effect was more intense for medronate because of its higher affinity for HAp. As the result of PO43--NH+ binding, JQ1 shielded the surface phosphates of HAp and pushed its surface charge to more positive values, exhibiting the opposite effect from calcium-blocking medronate. In contrast to the faster and more exponential release of JQ1 from monetite, its release from HAp nanoparticles followed a zero-order kinetics, but 98% of the payload was released after 48 h. The apoptotic effect of HAp nanoparticles loaded with JQ1, with medronate and with both JQ1 and medronate, was selective in 2D culture: pronounced against the OS cells and nonexistent against the healthy fibroblasts. While OS cell invasion was significantly inhibited by all of the JQ1-containing HAp formulations, that is, with and without medronate, all of the combinations of the targeting compound, medronate, and the chemotherapeutic, JQ1, delivered using HAp, but not HAp alone, inhibited OS cell migration from the tumor spheroids. JQ1 delivered using HAp had an effect on tumor migration, invasion, and apoptosis even at extremely low, subnanomolar concentrations, at which no effect of JQ1 per se was observed, meaning that this form of delivery could help achieve a multifold increase of this drug's efficacy. More than 80% of OS cells internalized JQ1-loaded HAp nanoparticles after 24 h of coincubation, suggesting that this augmentation of the activity of JQ1 may be due to the intracellular delivery and sustained release of the drug enabled by HAp. In addition to the reduction of the OS cell viability, the reduction of the migration and invasion radii was observed in OS tumor spheroids challenged with even JQ1-free medronate-functionalized HAp nanoparticles, demonstrating a definite anticancer activity of medronate alone when combined with HAp. The effect of medronate-functionalized JQ1-loaded HAp nanoparticles was most noticeable against OS cells differentiated into an osteoblastic lineage, in which case they surpassed in effect pure JQ1 and medronate-free compositions. The activity of JQ1 was mediated through increased Ezrin expression and decreased RUNX2 expression and was MYC and FOSL1 independent, but these patterns of gene expression changed in cells challenged with the nanoparticulate form of delivery, having been accompanied by the upregulation of RUNX2 and downregulation of Ezrin in OS cells treated with medronate-functionalized JQ1-loaded HAp nanoparticles.
Collapse
Affiliation(s)
- Victoria M. Wu
- Advanced Materials and Nanobiotechnology Laboratory, Department of Biomedical and Pharmaceutical Sciences, Center for Targeted Drug Delivery, Chapman University School of Pharmacy, 9401 Jeronimo Road, Irvine, California 92618-1908, United States
- Advanced Materials and Nanobiotechnology Laboratory, Department of Bioengineering, University of Illinois, Chicago, Illinois 60607-7052, United States
| | - Jarrett Mickens
- Advanced Materials and Nanobiotechnology Laboratory, Department of Bioengineering, University of Illinois, Chicago, Illinois 60607-7052, United States
| | - Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, Department of Biomedical and Pharmaceutical Sciences, Center for Targeted Drug Delivery, Chapman University School of Pharmacy, 9401 Jeronimo Road, Irvine, California 92618-1908, United States
- Advanced Materials and Nanobiotechnology Laboratory, Department of Bioengineering, University of Illinois, Chicago, Illinois 60607-7052, United States
- Corresponding Author:
| |
Collapse
|
36
|
Eldar-Boock A, Blau R, Ryppa C, Baabur-Cohen H, Many A, Vicent MJ, Kratz F, Sanchis J, Satchi-Fainaro R. Integrin-targeted nano-sized polymeric systems for paclitaxel conjugation: a comparative study. J Drug Target 2017; 25:829-844. [PMID: 28737432 DOI: 10.1080/1061186x.2017.1358727] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The generation of rationally designed polymer therapeutics via the conjugation of low molecular weight anti-cancer drugs to water-soluble polymeric nanocarriers aims to improve the therapeutic index. Here, we focus on applying polymer therapeutics to target two cell compartments simultaneously - tumour cells and angiogenic endothelial cells. Comparing different polymeric backbones carrying the same therapeutic agent and targeting moiety may shed light on any correlation between the choice of polymer and the anti-cancer activity of the conjugate. Here, we compared three paclitaxel (PTX)-bound conjugates with poly-l-glutamic acid (PGA, 4.9 mol%), 2-hydroxypropylmethacrylamide (HPMA, 1.2 mol%) copolymer, or polyethyleneglycol (PEG, 1:1 conjugate). PGA and HPMA copolymers are multivalent polymers that allow the conjugation of multiple compounds within the same polymer backbone, while PEG is a bivalent commercially available Food and Drug Administration (FDA)-approved polymer. We further conjugated PGA-PTX and PEG-PTX with the integrin αvβ3-targeting moiety RGD (5.5 mol% and 1:1 conjugate, respectively). We based our selection on the overexpression of integrin αvβ3 on angiogenic endothelial cells and several types of cancer cells. Our findings suggest that the polymer structure has major effect on the conjugate's activity on different tumour compartments. A multivalent PGA-PTX-E-[c(RGDfK)2] conjugate displayed a stronger inhibitory effect on the endothelial compartment, showing a 50% inhibition of the migration of human umbilical vein endothelial cell cells, while a PTX-PEG-E-[c(RGDfK)2] conjugate possessed enhanced anti-cancer activity on MDA-MB-231 tumour cells (IC50 = 20 nM versus IC50 300 nM for the PGA conjugate).
Collapse
Affiliation(s)
- Anat Eldar-Boock
- a Department of Physiology and Pharmacology, Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Rachel Blau
- a Department of Physiology and Pharmacology, Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | | | - Hemda Baabur-Cohen
- a Department of Physiology and Pharmacology, Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Ariel Many
- c Sourasky Medical Center , Lis Maternity Hospital , Tel Aviv , Israel
| | - María Jesús Vicent
- d Polymer Therapeutics Lab , Centro de Investigación Príncipe Felipe , Valencia , Spain
| | | | - Joaquin Sanchis
- d Polymer Therapeutics Lab , Centro de Investigación Príncipe Felipe , Valencia , Spain
| | - Ronit Satchi-Fainaro
- a Department of Physiology and Pharmacology, Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
37
|
Li Y, Hu H, Zhou Q, Ao Y, Xiao C, Wan J, Wan Y, Xu H, Li Z, Yang X. α-Amylase- and Redox-Responsive Nanoparticles for Tumor-Targeted Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:19215-19230. [PMID: 28513132 DOI: 10.1021/acsami.7b04066] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Paclitaxel (PTX) is an effective antineoplastic agent and shows potent antitumor activity against a wide spectrum of cancers. Yet, the wide clinical use of PTX is limited by its poor aqueous solubility and the side effects associated with its current therapeutic formulation. To tackle these obstacles, we report, for the first time, α-amylase- and redox-responsive nanoparticles based on hydroxyethyl starch (HES) for the tumor-targeted delivery of PTX. PTX is conjugated onto HES by a redox-sensitive disulfide bond to form HES-SS-PTX, which was confirmed by results from NMR, high-performance liquid chromatography-mass spectrometry, and Fourier transform infrared spectrometry. The HES-SS-PTX conjugates assemble into stable and monodispersed nanoparticles (NPs), as characterized with Dynamic light scattering, transmission electron microscopy, and atomic force microscopy. In blood, α-amylase will degrade the HES shell and thus decrease the size of the HES-SS-PTX NPs, facilitating NP extravasation and penetration into the tumor. A pharmacokinetic study demonstrated that the HES-SS-PTX NPs have a longer half-life than that of the commercial PTX formulation (Taxol). As a consequence, HES-SS-PTX NPs accumulate more in the tumor compared with the extent of Taxol, as shown in an in vivo imaging study. Under reductive conditions, the HES-SS-PTX NPs could disassemble quickly as evidenced by their triggered collapse, burst drug release, and enhanced cytotoxicity against 4T1 tumor cells in the presence of a reducing agent. Collectively, the HES-SS-PTX NPs show improved in vivo antitumor efficacy (63.6 vs 52.4%) and reduced toxicity in 4T1 tumor-bearing mice compared with those of Taxol. These results highlight the advantages of HES-based α-amylase- and redox-responsive NPs, showing their great clinical translation potential for cancer chemotherapy.
Collapse
Affiliation(s)
- Yihui Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Hang Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Qing Zhou
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Yanxiao Ao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Chen Xiao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Jiangling Wan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Ying Wan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Huibi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
- Wuhan Institute of Biotechnology , High Tech Road 666, East Lake High Tech Zone, Wuhan, 430040, P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, P. R. China
| |
Collapse
|
38
|
Hodgins NO, Wang JTW, Al-Jamal KT. Nano-technology based carriers for nitrogen-containing bisphosphonates delivery as sensitisers of γδ T cells for anticancer immunotherapy. Adv Drug Deliv Rev 2017; 114:143-160. [PMID: 28694026 DOI: 10.1016/j.addr.2017.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022]
Abstract
Nitrogen containing bisphosphonates (N-BPs) including zoledronate (ZOL) and alendronate (ALD) inhibit farnesyl diphosphate synthase, and have been shown to have a cytotoxic affect against cancer cells as a monotherapy and to also sensitise tumour cells to destruction by γδ T cells. γδ T cells are a subset of human T lymphocytes and have a diverse range of roles in the immune system including the recognition and destruction of cancer cells. This property of γδ T cells can be harnessed for use in cancer immunotherapy through in vivo expansion or the adoptive transfer of ex vivo activated γδ T cells. The use of N-BPs with γδ T cells has been shown to have a synergistic effect in in vitro, animal and clinical studies. N-BPs have limited in vivo activity due to rapid clearance from the circulation. By encapsulating N-BPs in liposomes (L) it is possible to increase the levels of N-BPs at non-osseous tumour sites. L-ZOL and L-ALD have been shown to have different toxicological profiles than free ZOL or ALD. Both L-ALD and L-ZOL led to increased spleen weight, leucocytosis, neutrophilia and lymphocytopenia in mice after intravenous injection. L-ALD was shown to be better tolerated than L-ZOL in murine studies. Biodistribution studies have been performed in order to better understand the interaction of N-BPs and γδ T cells in vivo. Additionally, in vivo therapy studies have shown that mice treated with both L-ALD and γδ T cells had a significant reduction in tumour growth compared to mice treated with L-ALD or γδ T cells alone. The use of ligand-targeted liposomes may further increase the efficacy of this combinatory immunotherapy. Liposomes targeting the αvβ6 integrin receptor using the peptide A20FMDV2 had a greater ability than untargeted liposomes in sensitising cancer cells to destruction by γδ T cells in αvβ6 positive cancer cell lines.
Collapse
|
39
|
Hu XX, He PP, Qi GB, Gao YJ, Lin YX, Yang C, Yang PP, Hao H, Wang L, Wang H. Transformable Nanomaterials as an Artificial Extracellular Matrix for Inhibiting Tumor Invasion and Metastasis. ACS NANO 2017; 11:4086-4096. [PMID: 28334523 DOI: 10.1021/acsnano.7b00781] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Tumor metastasis is one of the big challenges in cancer treatment and is often associated with high patient mortality. Until now, there is an agreement that tumor invasion and metastasis are related to degradation of extracellular matrix (ECM) by enzymes. Inspired by the formation of natural ECM and the in situ self-assembly strategy developed in our group, herein, we in situ constructed an artificial extracellular matrix (AECM) based on transformable Laminin (LN)-mimic peptide 1 (BP-KLVFFK-GGDGR-YIGSR) for inhibition of tumor invasion and metastasis. The peptide 1 was composed of three modules including (i) the hydrophobic bis-pyrene (BP) unit for forming and tracing nanoparticles; (ii) the KLVFF peptide motif that was inclined to form and stabilize fibrous structures through intermolecular hydrogen bonds; and (iii) the Y-type RGD-YIGSR motif, derived from LN conserved sequence, served as ligands to bind cancer cell surfaces. The peptide 1 formed nanoparticles (1-NPs) by the rapid precipitation method, owing to strong hydrophobic interactions of BP. Upon intravenous injection, 1-NPs effectively accumulated in the tumor site due to the enhanced permeability and retention (EPR) effect and/or targeting capability of RGD-YIGSR. The accumulated 1-NPs simultaneously transformed into nanofibers (1-NFs) around the solid tumor and further entwined to form AECM upon binding to receptors on the tumor cell surfaces. The AECM stably existed in the primary tumor site over 72 h, which consequently resulted in efficiently inhibiting the lung metastasis in breast and melanoma tumor models. The inhibition rates in two tumor models were 82.3% and 50.0%, respectively. This in vivo self-assembly strategy could be widely utilized to design effective drug-free biomaterials for inhibiting the tumor invasion and metastasis.
Collapse
Affiliation(s)
- Xiao-Xue Hu
- National Engineering Research Center of Industrial Crystallization Technology, State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, P. R. China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Ping-Ping He
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Guo-Bin Qi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yu-Juan Gao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yao-Xin Lin
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Chao Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Pei-Pei Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Hongxun Hao
- National Engineering Research Center of Industrial Crystallization Technology, State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, P. R. China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| |
Collapse
|
40
|
Karacivi M, Sumer Bolu B, Sanyal R. Targeting to the Bone: Alendronate-Directed Combretastatin A-4 Bearing Antiangiogenic Polymer-Drug Conjugates. Mol Pharm 2017; 14:1373-1383. [PMID: 28358515 DOI: 10.1021/acs.molpharmaceut.6b01173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Selective targeting of tumor site with chemotherapeutic agents appears to be one of the most effective methods to address many of the problems encountered with conventional chemotherapy. In this work, poly(oligoethylene glycol)methacrylate (POEGMA) based bone-targeting polymers bearing an antiangiogenic drug combretastatin A4 (CA4) were synthesized using free radical polymerization. Targeted and nontargeting copolymers were evaluated for their bone targeting efficiency, cytotoxicities against endothelial cells, namely, HUVECs and U2-OS and Saos-2 cancerous cell lines, as well as their antiangiogenic activity against endothelial cell tube formation by HUVECs. It is observed that the drug conjugated polymers conjugated with the bisphosphonate groups containing drug alendronate (ALN) have remarkably high affinity for bone mineral when compared to the polymer-drug conjugates devoid of the bisphosphonate groups. Both targeted and nontargeted polymer-drug conjugates show a sustained drug release in rat plasma with an overall release of 80-93% over 5 days. In vitro studies revealed high levels of cytotoxicity of the polymer-drug conjugates against HUVECs and U2-OS, and moderate cytotoxicity toward Saos-2. Importantly, the CA4 conjugated copolymers displayed excellent level of antiangiogenic activity as deduced from in vitro endothelial cell tube formation assay using HUVECs. Overall, a novel bone-targeting antiangiogenic polymer-drug conjugate that can be further elaborated to carry additional anticancer drugs is disclosed.
Collapse
Affiliation(s)
- Merve Karacivi
- Department of Chemistry and ‡Center for Life Science and Technologies, Bogazici University , Istanbul 34342, Turkey
| | - Burcu Sumer Bolu
- Department of Chemistry and ‡Center for Life Science and Technologies, Bogazici University , Istanbul 34342, Turkey
| | - Rana Sanyal
- Department of Chemistry and ‡Center for Life Science and Technologies, Bogazici University , Istanbul 34342, Turkey
| |
Collapse
|
41
|
Nadar RA, Margiotta N, Iafisco M, van den Beucken JJJP, Boerman OC, Leeuwenburgh SCG. Bisphosphonate-Functionalized Imaging Agents, Anti-Tumor Agents and Nanocarriers for Treatment of Bone Cancer. Adv Healthc Mater 2017; 6. [PMID: 28207199 DOI: 10.1002/adhm.201601119] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Indexed: 12/14/2022]
Abstract
Bone metastases result from the invasion of primary tumors to bone. Current treatment modalities include local treatments such as surgery and radiotherapy, while systemic treatments include chemotherapy and (palliative) treatment of skeletal metastases. Nevertheless, once bone metastases have been established they remain incurable leading to morbidity and mortality. Bisphosphonates are a well-established class of drugs, which are increasingly applied in the treatment of bone cancers owing to their effective inhibition of tumor cells and suppression of bone metastases. The increased understanding of the mechanism of action of bisphosphonates on bone and tumor cells has prompted the development of novel bisphosphonate-functionalized imaging and therapeutic agents. This review provides an update on the preclinical efficacy of bisphosphonate-functionalized fluorophore, anti-tumor agents and nanocarriers for the treatment of bone metastases. After an overview of the general characteristics of bisphosphonates and their mechanisms of action, an outline is provided on the various conjugation strategies that have become available to functionalize imaging agents, anti-tumor agents and nanocarriers with bisphosphonates. Finally, the efficacy of these bisphosphonate-modified agents and carriers in preclinical studies is evaluated by reviewing their potential to target tumors and inhibit tumor growth in clinically relevant animal models for the treatment of bone cancer.
Collapse
Affiliation(s)
- Robin A. Nadar
- Department of Biomaterials; Radboud University Medical Center; Philips van Leydenlaan 25 6525 EX Nijmegen The Netherlands
| | - Nicola Margiotta
- Dipartimento di Chimica; Università degli Studi di Bari Aldo Moro; Via E. Orabona 4 70125 Bari Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics (ISTEC); National Research Council (CNR); Via Granarolo 64 48018 Faenza Italy
| | | | - Otto C. Boerman
- Department of Nuclear Medicine; Radboud University Medical Center; Geert Grooteplein Zuid 10 6525 AG Nijmegen The Netherlands
| | - Sander C. G. Leeuwenburgh
- Department of Biomaterials; Radboud University Medical Center; Philips van Leydenlaan 25 6525 EX Nijmegen The Netherlands
| |
Collapse
|
42
|
Lin TH, Pajarinen J, Lu L, Nabeshima A, Cordova LA, Yao Z, Goodman SB. NF-κB as a Therapeutic Target in Inflammatory-Associated Bone Diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:117-154. [PMID: 28215222 DOI: 10.1016/bs.apcsb.2016.11.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammation is a defensive mechanism for pathogen clearance and maintaining tissue homeostasis. In the skeletal system, inflammation is closely associated with many bone disorders including fractures, nonunions, periprosthetic osteolysis (bone loss around orthopedic implants), and osteoporosis. Acute inflammation is a critical step for proper bone-healing and bone-remodeling processes. On the other hand, chronic inflammation with excessive proinflammatory cytokines disrupts the balance of skeletal homeostasis involving osteoblastic (bone formation) and osteoclastic (bone resorption) activities. NF-κB is a transcriptional factor that regulates the inflammatory response and bone-remodeling processes in both bone-forming and bone-resorption cells. In vitro and in vivo evidences suggest that NF-κB is an important potential therapeutic target for inflammation-associated bone disorders by modulating inflammation and bone-remodeling process simultaneously. The challenges of NF-κB-targeting therapy in bone disorders include: (1) the complexity of canonical and noncanonical NF-κB pathways; (2) the fundamental roles of NF-κB-mediated signaling for bone regeneration at earlier phases of tissue damage and acute inflammation; and (3) the potential toxic effects on nontargeted cells such as lymphocytes. Recent developments of novel inhibitors with differential approaches to modulate NF-κB activity, and the controlled release (local) or bone-targeting drug delivery (systemic) strategies, have largely increased the translational application of NF-κB therapy in bone disorders. Taken together, temporal modulation of NF-κB pathways with the combination of recent advanced bone-targeting drug delivery techniques is a highly translational strategy to reestablish homeostasis in the skeletal system.
Collapse
Affiliation(s)
- T-H Lin
- Stanford University, Stanford, CA, United States
| | - J Pajarinen
- Stanford University, Stanford, CA, United States
| | - L Lu
- Stanford University, Stanford, CA, United States
| | - A Nabeshima
- Stanford University, Stanford, CA, United States
| | - L A Cordova
- Stanford University, Stanford, CA, United States; Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Z Yao
- Stanford University, Stanford, CA, United States
| | - S B Goodman
- Stanford University, Stanford, CA, United States.
| |
Collapse
|
43
|
Rudnick-Glick S, Corem-Salkmon E, Grinberg I, Margel S. Targeted drug delivery of near IR fluorescent doxorubicin-conjugated poly(ethylene glycol) bisphosphonate nanoparticles for diagnosis and therapy of primary and metastatic bone cancer in a mouse model. J Nanobiotechnology 2016; 14:80. [PMID: 27919267 PMCID: PMC5139040 DOI: 10.1186/s12951-016-0233-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/26/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Most primary and metastatic bone tumors demonstrate increased osteoclast activity and bone resorption. Current treatment is based on a combination of surgery, radiotherapy and chemotherapy. Severe side effects are associated with chemotherapy due to use of high dosage and nonspecific uptake. Bisphosphonates have a strong affinity to Ca2+ ions and are widely used in the treatment of bone disorders. RESULTS We have engineered a unique biodegradable bisphosphonate nanoparticle (NPs) bearing two functional surface groups: (1) primary amine groups for covalent attachment of a dye/drug (e.g. NIR dye Cy 7 or doxorubicin); (2) bisphosphonate groups for targeting and chelation to bone hydroxyapatite. In addition, these engineered NPs contain high polyethyleneglycol (PEG) concentration in order to increase their blood half life time. In vitro experiments on Saos-2 human osteosarcoma cell line, demonstrated that at a tenth of the concentration, doxorubicin-conjugated bisphosphonate NPs achieved a similar uptake to free doxorubicin. In vivo targeting experiments using the NIR fluorescence bisphosphonate NPs on both Soas-2 human osteosarcoma xenograft mouse model and orthotopic bone metastases mCherry-labeled 4T1 breast cancer mouse model confirmed specific targeting. In addition, therapeutic in vivo experiments using doxorubicin-conjugated bisphosphonate NPs demonstrated a 40% greater inhibition of tumor growth in Saos-2 human osteosarcoma xenograft mouse model when compared to free doxorubicin. CONCLUSIONS In this research we have shown the potential use of doxorubicin-conjugated BP NPs for the targeting and treatment of primary and metastatic bone tumors. The targeted delivery of doxorubicin to the tumor significantly increased the efficacy of the anti-cancer drug, thus enabling the effective use of a lower concentration of doxorubicin. Furthermore, the targeting ability of the BP NPs in an orthotopic xenograft mouse model reinforced our findings that these BP NPs have the potential to be used for the treatment of primary and metastatic bone cancer.
Collapse
Affiliation(s)
- S. Rudnick-Glick
- Department of Chemistry, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, 52900 Ramat Gan, Israel
| | - E. Corem-Salkmon
- Department of Chemistry, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, 52900 Ramat Gan, Israel
| | - I. Grinberg
- Department of Chemistry, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, 52900 Ramat Gan, Israel
| | - S. Margel
- Department of Chemistry, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, 52900 Ramat Gan, Israel
| |
Collapse
|
44
|
Abstract
Background:
Bisphosphonates are drugs commonly used for the medication and prevention of diseases caused by decreased mineral density. Despite such important medicinal use, they display a variety of physiologic activities, which make them promising anti-cancer, anti-protozoal, antibacterial and antiviral agents.
Objective:
To review physiological activity of bisphosphonates with special emphasis on their ongoing and potential applications in medicine and agriculture.
Method:
Critical review of recent literature data.
Results:
Comprehensive review of activities revealed by bisphosphonates.
Conclusion:
although bisphosphonates are mostly recognized by their profound effects on bone physiology their medicinal potential has not been fully evaluated yet. Literature data considering enzyme inhibition suggest possibilities of far more wide application of these compounds. These applications are, however, limited by their low bioavailability and therefore intensive search for new chemical entities overcoming this shortage are carried out.
Collapse
|
45
|
Meng Z, Lv Q, Lu J, Yao H, Lv X, Jiang F, Lu A, Zhang G. Prodrug Strategies for Paclitaxel. Int J Mol Sci 2016; 17:E796. [PMID: 27223283 PMCID: PMC4881612 DOI: 10.3390/ijms17050796] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/04/2016] [Accepted: 05/11/2016] [Indexed: 01/08/2023] Open
Abstract
Paclitaxel is an anti-tumor agent with remarkable anti-tumor activity and wide clinical uses. However, it is also faced with various challenges especially for its poor water solubility and low selectivity for the target. To overcome these disadvantages of paclitaxel, approaches using small molecule modifications and macromolecule modifications have been developed by many research groups from all over the world. In this review, we discuss the different strategies especially prodrug strategies that are currently used to make paclitaxel more effective.
Collapse
Affiliation(s)
- Ziyuan Meng
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Quanxia Lv
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Jun Lu
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Houzong Yao
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Xiaoqing Lv
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Feng Jiang
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
- The State Key Laboratory Base of Novel Functional Materials and Preparation Science, Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Aiping Lu
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Ge Zhang
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| |
Collapse
|
46
|
Cole LE, Vargo-Gogola T, Roeder RK. Targeted delivery to bone and mineral deposits using bisphosphonate ligands. Adv Drug Deliv Rev 2016; 99:12-27. [PMID: 26482186 DOI: 10.1016/j.addr.2015.10.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/01/2015] [Accepted: 10/09/2015] [Indexed: 01/07/2023]
Abstract
The high concentration of mineral present in bone and pathological calcifications is unique compared with all other tissues and thus provides opportunity for targeted delivery of pharmaceutical drugs, including radiosensitizers and imaging probes. Targeted delivery enables accumulation of a high local dose of a therapeutic or imaging contrast agent to diseased bone or pathological calcifications. Bisphosphonates (BPs) are the most widely utilized bone-targeting ligand due to exhibiting high binding affinity to hydroxyapatite mineral. BPs can be conjugated to an agent that would otherwise have little or no affinity for the sites of interest. This article summarizes the current state of knowledge and practice for the use of BPs as ligands for targeted delivery to bone and mineral deposits. The clinical history of BPs is briefly summarized to emphasize the success of these molecules as therapeutics for metabolic bone diseases. Mechanisms of binding and the relative binding affinity of various BPs to bone mineral are introduced, including common methods for measuring binding affinity in vitro and in vivo. Current research is highlighted for the use of BP ligands for targeted delivery of BP conjugates in various applications, including (1) therapeutic drug delivery for metabolic bone diseases, bone cancer, other bone diseases, and engineered drug delivery platforms; (2) imaging probes for scintigraphy, fluorescence, positron emission tomography, magnetic resonance imaging, and computed tomography; and (3) radiotherapy. Last, and perhaps most importantly, key structure-function relationships are considered for the design of drugs with BP ligands, including the tether length between the BP and drug, the size of the drug, the number of BP ligands per drug, cleavable tethers between the BP and drug, and conjugation schemes.
Collapse
Affiliation(s)
- Lisa E Cole
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Tracy Vargo-Gogola
- Department of Biochemistry and Molecular Biology, Indiana University Simon Cancer Center, Indiana University School of Medicine-South Bend, South Bend, IN 46617, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ryan K Roeder
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
47
|
Targeted Delivery Systems for Molecular Therapy in Skeletal Disorders. Int J Mol Sci 2016; 17:428. [PMID: 27011176 PMCID: PMC4813278 DOI: 10.3390/ijms17030428] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
Abstract
Abnormalities in the integral components of bone, including bone matrix, bone mineral and bone cells, give rise to complex disturbances of skeletal development, growth and homeostasis. Non-specific drug delivery using high-dose systemic administration may decrease therapeutic efficacy of drugs and increase the risk of toxic effects in non-skeletal tissues, which remain clinical challenges in the treatment of skeletal disorders. Thus, targeted delivery systems are urgently needed to achieve higher drug delivery efficiency, improve therapeutic efficacy in the targeted cells/tissues, and minimize toxicities in non-targeted cells/tissues. In this review, we summarize recent progress in the application of different targeting moieties and nanoparticles for targeted drug delivery in skeletal disorders, and also discuss the advantages, challenges and perspectives in their clinical translation.
Collapse
|
48
|
Doppalapudi S, Jain A, Domb AJ, Khan W. Biodegradable polymers for targeted delivery of anti-cancer drugs. Expert Opin Drug Deliv 2016; 13:891-909. [DOI: 10.1517/17425247.2016.1156671] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Sindhu Doppalapudi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Anjali Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Abraham J. Domb
- School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, and Jerusalem College of Engineering (JCE), Jerusalem, Israel
| | - Wahid Khan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
49
|
Palao-Suay R, Gómez-Mascaraque L, Aguilar M, Vázquez-Lasa B, Román JS. Self-assembling polymer systems for advanced treatment of cancer and inflammation. Prog Polym Sci 2016. [DOI: 10.1016/j.progpolymsci.2015.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
50
|
Lv M, Li X, Huang Y, Wang N, Zhu X, Sun J. Inhibition of fibrous dysplasia via blocking Gsα with suramin sodium loaded with an alendronate-conjugated polymeric drug delivery system. Biomater Sci 2016; 4:1113-22. [DOI: 10.1039/c6bm00091f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A bone-targeted polymeric drug delivery system was constructed to inhibit fibrous dysplasia efficiently via blocking Gsα with suramin sodium.
Collapse
Affiliation(s)
- Mingming Lv
- Department of Oral Maxillofacial-Head Neck Oncology
- Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology
- Shanghai 200011
| | - Xiao Li
- Department of Oral and Maxillofacial Surgery
- The First Affiliated Hospital of Harbin Medical University
- Harbin 150001
- People's Republic of China
| | - Yu Huang
- School of Chemistry and Chemical Engineering
- Shanghai Key Lab of Electrical Insulation and Thermal Aging
- Shanghai Jiao Tong University
- Shanghai 200240
- People's Republic of China
| | - Nan Wang
- School of Chemistry and Chemical Engineering
- Shanghai Key Lab of Electrical Insulation and Thermal Aging
- Shanghai Jiao Tong University
- Shanghai 200240
- People's Republic of China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering
- Shanghai Key Lab of Electrical Insulation and Thermal Aging
- Shanghai Jiao Tong University
- Shanghai 200240
- People's Republic of China
| | - Jian Sun
- Department of Oral Maxillofacial-Head Neck Oncology
- Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology
- Shanghai 200011
| |
Collapse
|