1
|
Zhang Y, Tang L, Liu H, Cheng Y. The Multiple Functions of HB-EGF in Female Reproduction and Related Cancer: Molecular Mechanisms and Targeting Strategies. Reprod Sci 2024; 31:2588-2603. [PMID: 38424408 DOI: 10.1007/s43032-024-01454-6] [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: 08/16/2023] [Accepted: 01/03/2024] [Indexed: 03/02/2024]
Abstract
Heparin-binding growth factor (HB-EGF) is a member of the epidermal growth factor (EGF) ligand family which has a crucial role in women's health. However, there is a lack of comprehensive review to summarize the significance of HB-EGF. Therefore, this work first described the expression patterns of HB-EGF in the endometrium and ovary of different species and gestational time. Then, the focus was on exploring how it promotes the successful implantation and regulates the process of decidualization and the function of ovarian granulosa cells as an intermediate molecule. Otherwise, we also focused on the clinical and prognostic significance of HB-EGF in female-related cancers (including ovarian cancer, cervical cancer, and endometrial cancer) and breast cancer. Lastly, the article also summarizes the current drugs targeting HB-EGF in the treatment of ovarian cancer and breast cancer. Overall, these studies found that the expression of HB-EGF in the endometrium is spatiotemporal and species-specific. And it mediates the dialogue between the blastocyst and endometrium, promoting synchronous development of the blastocyst and endometrium as an intermediate molecule. HB-EGF may serve as a potentially valuable prognostic clinical indicator in tumors. And the specific inhibitor of HB-EGF (CRM197) has a certain anti-tumor ability, which can exert synergistic anti-tumor effects with conventional chemotherapy drugs. However, it also suggests that more research is needed in the future to elucidate its specific mechanisms and to accommodate clinical studies with a larger sample size to clarify its clinical value.
Collapse
Affiliation(s)
- Yuwei Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Lujia Tang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
2
|
Parveen N, Abourehab MA, Shukla R, Thanikachalam PV, Jain GK, Kesharwani P. Immunoliposomes as an emerging nanocarrier for breast cancer therapy. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2022.111781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
3
|
Moudgil A, Salve R, Gajbhiye V, Chaudhari BP. Challenges and emerging strategies for next generation liposomal based drug delivery: An account of the breast cancer conundrum. Chem Phys Lipids 2023; 250:105258. [PMID: 36375540 DOI: 10.1016/j.chemphyslip.2022.105258] [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: 09/20/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
The global cancer burden is witnessing an upsurge with breast cancer surpassing other cancers worldwide. Furthermore, an escalation in the breast cancer caseload is also expected in the coming years. The conventional therapeutic regimens practiced routinely are associated with many drawbacks to which nanotechnological interventions offer a great advantage. But how eminent could liposomes and their advantages be in superseding these existing therapeutic modalities? A solution is reflected in this review that draws attention to a decade-long journey embarked upon by researchers in this wake. This text is a comprehensive discussion of liposomes, the front runners of the drug delivery systems, and their active and passive targeting approaches for breast cancer management. Active targeting has been studied over the decade by many receptors overexpressed on the breast cancer cells and passive targeting with many drug combinations. The results converge on the fact that the actively targeted formulations exhibit a superior efficacy over their non-targeted counterparts and the all liposomal formulations are efficacious over the free drugs. This undoubtedly underlines the dominion of liposomal formulations over conventional chemotherapy. These investigations have led to the development of different liposomal formulations with active and passive targeting capacities that could be explored in depth. Acknowledging and getting a deeper insight into the liposomal evolution through time also unveiled many imperfections and unchartered territories that can be explored to deliver dexterous liposomal formulations against breast cancer and more in the clinical trial pipeline.
Collapse
Affiliation(s)
- Aliesha Moudgil
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Rajesh Salve
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Bhushan P Chaudhari
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India.
| |
Collapse
|
4
|
A comprehensive review on immuno-nanomedicine for breast cancer therapy: Technical challenges and troubleshooting measures. Int Immunopharmacol 2021; 103:108433. [PMID: 34922248 DOI: 10.1016/j.intimp.2021.108433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022]
Abstract
Nanosized drug carriers have received a major attention in cancer therapeutics and theranostics. The immuno-nanomedicine is a combination of monoclonal antibody (mAb)/mAb-drug-nanoparticles. The immuno-nanomedicine offers a promising strategy to target cancer cells. However, the understating of nanotechnology, cancer biology, immunomedicine, and nanoparticle surface chemistry has provided a better clue to prepare the effective immuno-nanomedicine for cancer therapy. Moreover, the selection of nanoparticles type and its composition is essential for development of efficient drug delivery system (DDS) to target the cancer cell site. Immuno-nanomedicine works in the ligand-receptor binding mechanism through the interaction of mAb conjugated nanoparticles and specific antigen over expressed on target cancer cells. Therefore, the selection of specific receptors in the cancer cell and their ligand is important to prepare the active immuno-nanomedicines. Moreover, the factors such as drug loading, entrapment efficiency, size, shape, and ligand conjugation of a nanocarrier are considered as major factors for a better cancer cell, internalization, drug release, and cancer cell ablation. The target-based over-expression of antigen, mAb is engineered and conjugated with nanoparticles for successful targeting of the cancer cells without causing adverse effects to normal cells. Therefore, this review analyzed the fundamental factors in the immuno-nanomedicine for breast cancer and its technical challenges in the fabrication of the antibody alone/and drug conjugated nanoparticles.
Collapse
|
5
|
Rizwanullah M, Ahmad MZ, Ghoneim MM, Alshehri S, Imam SS, Md S, Alhakamy NA, Jain K, Ahmad J. Receptor-Mediated Targeted Delivery of Surface-ModifiedNanomedicine in Breast Cancer: Recent Update and Challenges. Pharmaceutics 2021; 13:2039. [PMID: 34959321 PMCID: PMC8708551 DOI: 10.3390/pharmaceutics13122039] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer therapeutic intervention continues to be ambiguous owing to the lack of strategies for targeted transport and receptor-mediated uptake of drugs by cancer cells. In addition to this, sporadic tumor microenvironment, prominent restrictions with conventional chemotherapy, and multidrug-resistant mechanisms of breast cancer cells possess a big challenge to even otherwise optimal and efficacious breast cancer treatment strategies. Surface-modified nanomedicines can expedite the cellular uptake and delivery of drug-loaded nanoparticulate constructs through binding with specific receptors overexpressed aberrantly on the tumor cell. The present review elucidates the interesting yet challenging concept of targeted delivery approaches by exploiting different types of nanoparticulate systems with multiple targeting ligands to target overexpressed receptors of breast cancer cells. The therapeutic efficacy of these novel approaches in preclinical models is also comprehensively discussed in this review. It is concluded from critical analysis of related literature that insight into the translational gap between laboratories and clinical settings would provide the possible future directions to plug the loopholes in the process of development of these receptor-targeted nanomedicines for the treatment of breast cancer.
Collapse
Affiliation(s)
- Md. Rizwanullah
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.)
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.)
| | - Keerti Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)—Raebareli, Lucknow 226002, India;
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| |
Collapse
|
6
|
Hirata Y, Tashima R, Mitsuhashi N, Yoneda S, Ozono M, Fukuta T, Majima E, Kogure K. A simple, fast, and orientation-controllable technology for preparing antibody-modified liposomes. Int J Pharm 2021; 607:120966. [PMID: 34352337 DOI: 10.1016/j.ijpharm.2021.120966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/06/2021] [Accepted: 07/29/2021] [Indexed: 12/22/2022]
Abstract
Modification with antibodies is a useful strategy for the delivery of nanoparticles to target cells. However, the complexity of the required chemical modifications makes them time-consuming and low efficiency, and the orientation of the antibody is challenging to control. To develop a simple, fast, effective, and orientation-controllable technology, we employed staphylococcal protein A, which can bind to the Fc region of antibodies, as a tool for conjugating antibodies to nanoparticles. Specifically, we modified the C-domain dimer of protein A to contain a lysine cluster to create a molecule, DPACK, that would electrostatically bind to anionic liposomes. Using this protein, antibody-modified liposomes can be prepared in 35 min with two steps: (1) interaction of DPACK with liposomes and (2) interaction of an antibody with DPACK-modified liposomes. Binding efficiencies of DPACK with liposomes and IgG with DPACK-modified liposomes were 75% and 72-84%, respectively. Uptake of liposomes modified with anti-epidermal growth factor receptor (EGFR) antibodies via DPACK by EGFR-expressing cancer cells was significantly higher than that of unmodified liposomes, and the liposomes accumulated in tumors and colocalized with EGFR. This simple, fast, effective and orientation-controllable technology for preparing antibody-modified liposomes will be useful for active targeting drug delivery.
Collapse
Affiliation(s)
- Yuma Hirata
- Faculty of Pharmaceutical Science, Tokushima University, Shomachi-1-78-1, Tokushima, Tokushima 770-8505, Japan
| | - Riho Tashima
- Faculty of Pharmaceutical Science, Tokushima University, Shomachi-1-78-1, Tokushima, Tokushima 770-8505, Japan
| | - Naoto Mitsuhashi
- ProteNova Co., Ltd., 1488-1 Nishimura, Higashikagawa, Kagawa 769-2604, Japan
| | - Shintaro Yoneda
- Grasuate School of Biomedical Sciences, Tokushima University, Shomachi-1-78-1, Tokushima, Tokushima 770-8505, Japan
| | - Mizune Ozono
- Grasuate School of Biomedical Sciences, Tokushima University, Shomachi-1-78-1, Tokushima, Tokushima 770-8505, Japan
| | - Tatsuya Fukuta
- Grasuate School of Biomedical Sciences, Tokushima University, Shomachi-1-78-1, Tokushima, Tokushima 770-8505, Japan; Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama-shi, Wakayama 640-8156, Japan
| | - Eiji Majima
- ProteNova Co., Ltd., 1488-1 Nishimura, Higashikagawa, Kagawa 769-2604, Japan
| | - Kentaro Kogure
- Grasuate School of Biomedical Sciences, Tokushima University, Shomachi-1-78-1, Tokushima, Tokushima 770-8505, Japan.
| |
Collapse
|
7
|
Petrilli R, Pinheiro DP, de Cássia Evangelista de Oliveira F, Galvão GF, Marques LGA, Lopez RFV, Pessoa C, Eloy JO. Immunoconjugates for Cancer Targeting: A Review of Antibody-Drug Conjugates and Antibody-Functionalized Nanoparticles. Curr Med Chem 2021; 28:2485-2520. [PMID: 32484100 DOI: 10.2174/0929867327666200525161359] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/07/2020] [Accepted: 04/16/2020] [Indexed: 11/22/2022]
Abstract
Targeted therapy has been recently highlighted due to the reduction of side effects and improvement in overall efficacy and survival from different types of cancers. Considering the approval of many monoclonal antibodies in the last twenty years, cancer treatment can be accomplished by the combination of monoclonal antibodies and small molecule chemotherapeutics. Thus, strategies to combine both drugs in a single administration system are relevant in the clinic. In this context, two strategies are possible and will be further discussed in this review: antibody-drug conjugates (ADCs) and antibody-functionalized nanoparticles. First, it is important to better understand the possible molecular targets for cancer therapy, addressing different antigens that can selectively bind to antibodies. After selecting the best target, ADCs can be prepared by attaching a cytotoxic drug to an antibody able to target a cancer cell antigen. Briefly, an ADC will be formed by a monoclonal antibody (MAb), a cytotoxic molecule (cytotoxin) and a chemical linker. Usually, surface-exposed lysine or the thiol group of cysteine residues are used as anchor sites for linker-drug molecules. Another strategy that should be considered is antibody-functionalized nanoparticles. Basically, liposomes, polymeric and inorganic nanoparticles can be attached to specific antibodies for targeted therapy. Different conjugation strategies can be used, but nanoparticles coupling between maleimide and thiolated antibodies or activation with the addition of ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC)/ N-hydroxysuccinimide (NHS) (1:5) and further addition of the antibody are some of the most used strategies. Herein, molecular targets and conjugation strategies will be presented and discussed to better understand the in vitro and in vivo applications presented. Also, the clinical development of ADCs and antibody-conjugated nanoparticles are addressed in the clinical development section. Finally, due to the innovation related to the targeted therapy, it is convenient to analyze the impact on patenting and technology. Information related to the temporal evolution of the number of patents, distribution of patent holders and also the number of patents related to cancer types are presented and discussed. Thus, our aim is to provide an overview of the recent developments in immunoconjugates for cancer targeting and highlight the most important aspects for clinical relevance and innovation.
Collapse
Affiliation(s)
- Raquel Petrilli
- University for International Integration of the Afro-Brazilian Lusophony, Institute of Health Sciences, Ceara, Brazil
| | - Daniel Pascoalino Pinheiro
- Federal University of Ceara, College of Medicine, Department of Physiology and Pharmacology, Fortaleza, Ceara, Brazil
| | | | - Gabriela Fávero Galvão
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Av. Cafe s/n, Ribeirao Preto, SP, Brazil
| | - Lana Grasiela Alves Marques
- Institute of Communication and Scientific and Technological Information in Health, Oswaldo Cruz Foundation - FIOCRUZ, Rio de Janeiro, Brazil
| | - Renata Fonseca Vianna Lopez
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Av. Cafe s/n, Ribeirao Preto, SP, Brazil
| | - Claudia Pessoa
- Federal University of Ceara, College of Medicine, Department of Physiology and Pharmacology, Fortaleza, Ceara, Brazil
| | - Josimar O Eloy
- Federal University of Ceará, College of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, Ceara, Brazil
| |
Collapse
|
8
|
Shimizu K, Agata K, Takasugi S, Goto S, Narita Y, Asai T, Magata Y, Oku N. New strategy for MS treatment with autoantigen-modified liposomes and their therapeutic effect. J Control Release 2021; 335:389-397. [PMID: 34033858 DOI: 10.1016/j.jconrel.2021.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
As current treatments for multiple sclerosis (MS) remain chemotherapeutic ones directed toward symptoms, the development of a curative treatment is urgently required. Herein, we show an autoreactive immune cell-targetable approach using autoantigen-modified liposomes for the curative treatment of MS. In these experiments, experimental autoimmune encephalomyelitis (EAE) induced by autoantigenic myelin oligodendrocyte glycoprotein (MOG) peptide was used as a model of primary progressive MS, and MOG-modified liposomes encapsulating doxorubicin (MOG-LipDOX) were used as a therapeutic drug. The results showed that the progression of encephalomyelitis symptoms was significantly suppressed by MOG-LipDOX injection, whereas the other samples failed to show any effect. Additionally, invasion of inflammatory immune cells into the spinal cord and demyelination of neurons were clearly suppressed in the MOG-LipDOX-treated mice. FACS analysis revealed that the number of both MOG-recognizable CD4+ T cells in the spleen was obviously decreased after MOG-LipDOX treatment. Furthermore, the number of effector Th17 cells in the spleen was significantly decreased and that of regulatory Treg cells was concomitantly increased. Finally, we demonstrated that myelin proteolipid protein (PLP)-modified liposomes encapsulating DOX (PLP-LipDOX) also showed the therapeutic effect on relapsing-remitting EAE. These findings indicate that autoantigen-modified liposomal drug produced a highly therapeutic effect on EAE by delivering the encapsulated drug to autoantigen-recognizable CD4+ T cells and thus suppressing autoreactive immune responses. The present study suggests that the use of these autoantigen-modified liposomes promises to be a suitable therapeutic approach for the cure of MS.
Collapse
Affiliation(s)
- Kosuke Shimizu
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan.
| | - Kazuki Agata
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shohei Takasugi
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shungo Goto
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Yudai Narita
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Yasuhiro Magata
- Department of Molecular Imaging, Institute of Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan; Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| |
Collapse
|
9
|
Grudén S, Brunmark C, Holmqvist B, Brenndörfer ED, Johansson M, Liu J, Zhao Y, Axén N, Hassan M. Biodistribution of fluorescence-labelled EGF protein from slow release NanoZolid depots in mouse. Int J Pharm 2021; 601:120588. [PMID: 33845148 DOI: 10.1016/j.ijpharm.2021.120588] [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: 09/24/2020] [Revised: 03/05/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
AIM The study was designed to evaluate the ability of the calcium sulfate based NanoZolid® drug delivery technology to locally release the epidermal growth factor (EGF) protein while maintaining its biological activity. METHODS NanoZolid-formulated EGF protein labelled with a near infrared dye (EGF-NIR) depots or EGF-NIR dissolved in PBS were injected subcutaneously into mice bearing EGF receptor (EGFR) positive human A549 lung cancer tumors inoculated subcutaneously. The release and biodistribution of the EGF-NIR were investigated in vivo longitudinally up to 96 h post administration, utilizing whole body fluorescence imaging. In order to confirm the in vivo findings, histological analysis of tumor cryosections was performed to investigate EGF-NIR fluorescent signal and EGFR expression level by immunofluorescence labelling. RESULTS The in vivo fluorescence imaging showed a controlled release profile of the EGF-NIR loaded in the NanoZolid depots compared to free EGF-NIR. Histological analysis of the tumors further demonstrated a prevailing distribution of EGF-NIR in regions with high levels of EGFR expression. CONCLUSION Calcium sulfate based depots can be used to formulate EGF while maintaining its biological activity, e.g. receptor binding capability. This may have a good clinical potential for local delivery of biomolecules to enhance treatment efficacy and minimize systemic adverse effects.
Collapse
Affiliation(s)
- Stefan Grudén
- Experimental Cancer Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden; LIDDS AB, Virdings allé 32B, SE-754 50 Uppsala, Sweden.
| | - Charlott Brunmark
- Truly Labs AB, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden.
| | - Bo Holmqvist
- ImaGene-iT AB, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden.
| | | | | | - Jian Liu
- Truly Labs AB, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden.
| | - Ying Zhao
- Experimental Cancer Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden; Clinical Research Center (KFC) and Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | - Niklas Axén
- LIDDS AB, Virdings allé 32B, SE-754 50 Uppsala, Sweden.
| | - Moustapha Hassan
- Experimental Cancer Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden; Clinical Research Center (KFC) and Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
10
|
Juan A, Cimas FJ, Bravo I, Pandiella A, Ocaña A, Alonso-Moreno C. Antibody Conjugation of Nanoparticles as Therapeutics for Breast Cancer Treatment. Int J Mol Sci 2020; 21:E6018. [PMID: 32825618 PMCID: PMC7504566 DOI: 10.3390/ijms21176018] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer is the most common invasive tumor in women and the second leading cause of cancer-related death. Nanomedicine raises high expectations for millions of patients as it can provide better, more efficient, and affordable healthcare, and it has the potential to develop novel therapeutics for the treatment of solid tumors. In this regard, targeted therapies can be encapsulated into nanocarriers, and these nanovehicles are guided to the tumors through conjugation with antibodies-the so-called antibody-conjugated nanoparticles (ACNPs). ACNPs can preserve the chemical structure of drugs, deliver them in a controlled manner, and reduce toxicity. As certain breast cancer subtypes and indications have limited therapeutic options, this field provides hope for the future treatment of patients with difficult to treat breast cancers. In this review, we discuss the application of ACNPs for the treatment of this disease. Given the fact that ACNPs have shown clinical activity in this clinical setting, special emphasis on the role of the nanovehicles and their translation to the clinic is placed on the revision.
Collapse
Affiliation(s)
- Alberto Juan
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Francisco J. Cimas
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad Oncología Traslacional, 02071 Albacete, Spain
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer-CSIC, IBSAL- Salamanca and CIBERONC, 37007 Salamanca, Spain;
| | - Alberto Ocaña
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Experimental Therapeutics Unit, Hospital clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
11
|
Leukocyte-mimetic liposomes possessing leukocyte membrane proteins pass through inflamed endothelial cell layer by regulating intercellular junctions. Int J Pharm 2019; 563:314-323. [DOI: 10.1016/j.ijpharm.2019.04.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/14/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022]
|
12
|
Shen Y, Ruan L, Lian C, Li R, Tu Z, Liu H. Discovery of HB-EGF binding peptides and their functional characterization in ovarian cancer cell lines. Cell Death Discov 2019; 5:82. [PMID: 30937184 PMCID: PMC6433920 DOI: 10.1038/s41420-019-0163-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/03/2019] [Accepted: 02/10/2019] [Indexed: 01/29/2023] Open
Abstract
Ovarian cancer is one of the most frequent causes of cancer death among all gynecologic cancers. Though standard therapy often results in temporary clinical remission, most patients suffer from recurrence and metastasis of ovarian cancer, which highlights the need for developing new therapeutic agents targeting specific molecules. Previous studies have demonstrated that the native ligand of epidermal growth factor receptor (EGFR) and ErbB4, heparin-binding EGF-like growth factor (HB-EGF), plays a critical role in the progression of ovarian cancer and is associated with prognosis of ovarian cancer. In the current study, we tried to develop a peptide-based treatment for ovarian cancer by targeting HB-EGF. After the functions of HB-EGF in promoting migration and invasion of SKOV3 and HO-8910 cells were confirmed, phage display was used to discover peptides binding to HB-EGF. Two peptides, no. 7 and no. 29 were found mildly binding to HB-EGF. Then the effects of these peptides on HB-EGF functions were examined and both peptides no. 7 and no. 29 were found indeed inhibiting the functions of HB-EGF in promoting migration and invasion of SKOV3 and HO-8910 cells in vitro. Further mechanism investigation showed that peptides no. 7 and no. 29 inhibited HB-EGF-promoted cell migration and invasion through attenuating activation of the EGFR signaling pathway manifested by decreased p-Erk1/2 and Snail levels. More importantly, peptides no. 7 and no. 29 showed strong activities in inhibiting migration of SKOV3 cells in vivo. These results provide a proof of concept method for developing novel peptide drugs to combat ovarian cancer through interfering with HB-EGF mediated signaling pathways.
Collapse
Affiliation(s)
- Yanting Shen
- 1School of Pharmacy, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| | - Lingling Ruan
- 1School of Pharmacy, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| | - Caixia Lian
- 2Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| | - Ruyan Li
- 2Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| | - Zhigang Tu
- 2Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| | - Hanqing Liu
- 1School of Pharmacy, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| |
Collapse
|
13
|
Tsuchida S, Matsuzaki T, Yamato M, Okuda K, Fu HY, Araki R, Sanada S, Asanuma H, Asano Y, Asakura M, Hao H, Takashima S, Kitakaze M, Sakata Y, Mekada E, Minamino T. Anti-HB-EGF Antibody-Mediated Delivery of siRNA to Atherosclerotic Lesions in Mice. Int Heart J 2018; 59:1425-1431. [DOI: 10.1536/ihj.17-644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shota Tsuchida
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Takashi Matsuzaki
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | - Masaki Yamato
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Keiji Okuda
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hai Ying Fu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Ryo Araki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shoji Sanada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Masanori Asakura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine
| | - Hiroyuki Hao
- Department of Pathology, Nihon University School of Medicine
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine
| | - Masafumi Kitakaze
- Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Eisuke Mekada
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| |
Collapse
|
14
|
Safety of novel liposomal drugs for cancer treatment: Advances and prospects. Chem Biol Interact 2018; 295:13-19. [DOI: 10.1016/j.cbi.2017.09.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 08/02/2017] [Accepted: 09/01/2017] [Indexed: 12/20/2022]
|
15
|
Okamoto A, Asai T, Hirai Y, Shimizu K, Koide H, Minamino T, Oku N. Systemic Administration of siRNA with Anti-HB-EGF Antibody-Modified Lipid Nanoparticles for the Treatment of Triple-Negative Breast Cancer. Mol Pharm 2018; 15:1495-1504. [PMID: 29502423 DOI: 10.1021/acs.molpharmaceut.7b01055] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Triple-negative breast cancer is one of the intractable cancers that are not sensitive to treatment with existing molecular-targeted drugs. Recently, there has been much interest in RNA interference-mediated treatment of triple-negative breast cancer. In the present study, we have developed lipid nanoparticles encapsulating siRNA (LNP-siRNA) decorated with an Fab' antibody against heparin-binding EGF-like growth factor (αHB-EGF LNP-siRNA). αHB-EGF LNP-siRNA targeting polo-like kinase 1 (PLK1) was prepared and evaluated for its anticancer effect using MDA-MB-231 human triple-negative breast cancer cells overexpressing HB-EGF on their cell surface. Biodistribution data of radioisotope-labeled LNP and fluorescence-labeled siRNA indicated that αHB-EGF LNP effectively delivered siRNA to tumor tissue in MDA-MB-231 carcinoma-bearing mice. Expression of PLK1 protein in the tumors was clearly suppressed after intravenous injection of αHB-EGF LNP-siPLK1. In addition, tumor growth was significantly inhibited by treatment with this formulation of siRNA and an antibody-modified carrier. These findings indicate that αHB-EGF LNP is a promising carrier for the treatment of HB-EGF-expressing cancers, including triple-negative breast cancer.
Collapse
Affiliation(s)
- Ayaka Okamoto
- Department of Medical Biochemistry, School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada, Suruga-ku , Shizuoka 422-8526 , Japan
- Japan Society for the Promotion of Science , 5-3-1 Kojimachi, Chiyoda-ku , Tokyo 102-0083 , Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada, Suruga-ku , Shizuoka 422-8526 , Japan
| | - Yusuke Hirai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada, Suruga-ku , Shizuoka 422-8526 , Japan
| | - Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada, Suruga-ku , Shizuoka 422-8526 , Japan
- Department of Molecular Imaging, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center , Hamamatsu University School of Medicine , 1-20-1 Handayama, Higashi-ku , Hamamatsu City , Shizuoka 431-3192 Japan
| | - Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada, Suruga-ku , Shizuoka 422-8526 , Japan
| | - Tetsuo Minamino
- Department of Cardiovascular Medicine, Graduate School of Medicine , Kagawa University , 1750-1 Ikenobe, Miki-cho , Kita-gun , Kagawa 761-0793 Japan
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada, Suruga-ku , Shizuoka 422-8526 , Japan
| |
Collapse
|
16
|
Yamato M, Matsuzaki T, Araki R, Tsuchida S, Okuda K, Fu HY, Sanada S, Asanuma H, Asano Y, Asakura M, Torii H, Noi K, Ogi H, Iwamoto R, Mekada E, Takashima S, Kitakaze M, Sakata Y, Minamino T. RNA Aptamer Binds Heparin-Binding Epidermal Growth Factor-Like Growth Factor with High Affinity and Specificity and Neutralizes Its Activity. INT J GERONTOL 2017. [DOI: 10.1016/j.ijge.2017.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
17
|
Kim S, Yang L, Kim S, Lee RG, Graham MJ, Berliner JA, Lusis AJ, Cai L, Temel RE, Rateri DL, Lee S. Targeting hepatic heparin-binding EGF-like growth factor (HB-EGF) induces anti-hyperlipidemia leading to reduction of angiotensin II-induced aneurysm development. PLoS One 2017; 12:e0182566. [PMID: 28792970 PMCID: PMC5549937 DOI: 10.1371/journal.pone.0182566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/20/2017] [Indexed: 01/02/2023] Open
Abstract
Objective The upregulated expression of heparin binding EGF-like growth factor (HB-EGF) in the vessel and circulation is associated with risk of cardiovascular disease. In this study, we tested the effects of HB-EGF targeting using HB-EGF-specific antisense oligonucleotide (ASO) on the development of aortic aneurysm in a mouse aneurysm model. Approach and results Low-density lipoprotein receptor (LDLR) deficient mice (male, 16 weeks of age) were injected with control and HB-EGF ASOs for 10 weeks. To induce aneurysm, the mice were fed a high fat diet (22% fat, 0.2% cholesterol; w/w) at 5 week point of ASO administration and infused with angiotensin II (AngII, 1,000ng/kg/min) for the last 4 weeks of ASO administration. We confirmed that the HB-EGF ASO administration significantly downregulated HB-EGF expression in multiple tissues including the liver. Importantly, the HB-EGF ASO administration significantly suppressed development of aortic aneurysms including thoracic and abdominal types. Interestingly, the HB-EGF ASO administration induced a remarkable anti-hyperlipidemic effect by suppressing very low density lipoprotein (VLDL) level in the blood. Mechanistically, the HB-EGF targeting suppressed hepatic VLDL secretion rate without changing heparin-releasable plasma triglyceride (TG) hydrolytic activity or fecal neutral cholesterol excretion rate. Conclusion This result suggested that the HB-EGF targeting induced protection against aneurysm development through anti-hyperlipidemic effects. Suppression of hepatic VLDL production process appears to be a key mechanism for the anti-hyperlipidemic effects by the HB-EGF targeting.
Collapse
Affiliation(s)
- Seonwook Kim
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Lihua Yang
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Seongu Kim
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Richard G. Lee
- Cardiovascular Antisense Drug Discovery Group at the Ionis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Mark J. Graham
- Cardiovascular Antisense Drug Discovery Group at the Ionis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Judith A. Berliner
- Department of Medicine-Cardiology, University of California-Los Angeles School of Medicine, Los Angeles, California, United States of America
| | - Aldons J. Lusis
- Department of Medicine-Cardiology, University of California-Los Angeles School of Medicine, Los Angeles, California, United States of America
| | - Lei Cai
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Ryan E. Temel
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Pharmacology & Nutritional Sciences at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Debra L. Rateri
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Sangderk Lee
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Pharmacology & Nutritional Sciences at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
18
|
Eloy JO, Petrilli R, Trevizan LNF, Chorilli M. Immunoliposomes: A review on functionalization strategies and targets for drug delivery. Colloids Surf B Biointerfaces 2017; 159:454-467. [PMID: 28837895 DOI: 10.1016/j.colsurfb.2017.07.085] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/26/2017] [Accepted: 07/29/2017] [Indexed: 12/31/2022]
Abstract
Nanoparticles, especially liposomes, have gained prominence in the field of drug delivery for the treatment of human diseases, particularly cancer; they provide several advantages, including controlled drug release, protection of the drug against degradation, improved pharmacokinetics, long circulation, and passive targeting to tumors and inflammatory sites due to the enhanced permeability and retention effect. The functionalization of liposomes with monoclonal antibodies or antibody fragments to generate immunoliposomes has emerged as a promising strategy for targeted delivery to and uptake by cells overexpressing the antigens to these antibodies, with a consequent reduction in side effects. In this review, we address functionalization strategies for the non-covalent and covalent attachment of monoclonal antibodies and their fragments to liposomal surfaces. The main reaction occurs between the sulfhydryl groups of thiolated antibodies and maleimide-containing liposomes. Furthermore, we explore the main targeting possibilities with these ligands for the treatment of a variety of pathologies, including HER2- and EGFR-positive cancers, inflammatory and cardiovascular diseases, infectious diseases, and autoimmune and neurodegenerative diseases, which have not previously been reviewed together. Overall, many studies have shown selective delivery of immunoliposomes to target cells, with promising in vivo results, particularly for cancer treatment. Although clinical trials have been conducted, immunoliposomes have not yet received clinical approval. However, immunoliposomes are promising formulations that are expected to become available for therapeutic use after clinical trials prove their safety and efficacy, and after scaling issues are resolved.
Collapse
Affiliation(s)
- Josimar O Eloy
- School of Pharmaceutical Sciences of Araraquara, São Paulo State University, UNESP, Department of Drugs and Medicines, Araraquara, SP, Brazil.
| | - Raquel Petrilli
- School of Pharmaceutical Sciences of Ribeirão Preto, São Paulo State University, USP, Department of Pharmaceutical Sciences, Ribeirão Preto, SP, Brazil
| | - Lucas Noboru Fatori Trevizan
- School of Pharmaceutical Sciences of Araraquara, São Paulo State University, UNESP, Department of Drugs and Medicines, Araraquara, SP, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences of Araraquara, São Paulo State University, UNESP, Department of Drugs and Medicines, Araraquara, SP, Brazil
| |
Collapse
|
19
|
Fukuta T, Asai T, Kiyokawa Y, Nakada T, Bessyo-Hirashima K, Fukaya N, Hyodo K, Takase K, Kikuchi H, Oku N. Targeted delivery of anticancer drugs to tumor vessels by use of liposomes modified with a peptide identified by phage biopanning with human endothelial progenitor cells. Int J Pharm 2017; 524:364-372. [PMID: 28359814 DOI: 10.1016/j.ijpharm.2017.03.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/15/2017] [Accepted: 03/24/2017] [Indexed: 01/29/2023]
Abstract
As tumor angiogenic vessels are critical for tumor growth and express different molecules on their surface from those on normal vessels, these vessels are expected to be an ideal target for anticancer drug delivery systems. It was previously reported that endothelial progenitor cells (EPCs) are involved in angiogenesis, tumor growth, and metastasis, and that EPCs show gene expression patterns similar to those of tumor endothelial cells. In the present study, a tumor vessel-targeting peptide, ASSHN, was identified from a phage-display peptide library by in vitro biopanning with human EPCs (hEPCs) and in vivo biopanning using angiogenesis model mice prepared by the dorsal air sac method. Phage clones displaying ASSHN peptide showed a marked affinity for hEPCs in vitro, and also for tumor vessels in vivo. PEGylated liposomes modified with the ASSHN peptide (ASSHN-Lip) were designed and prepared for the delivery of anticancer agents. Confocal images showed that ASSHN-Lip clearly bound to hEPCs in vitro and tumor vessels, and also showed extravasation from the vessels. The administration of doxorubicin-encapsulated ASSHN-Lip into Colon26 NL-17-bearing mice significantly suppressed tumor growth compared with doxorubicin-encapsulated PEGylated liposomes. These results suggest that the delivery of anticancer agents with ASSHN-Lip could be useful for targeted cancer therapy.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Japan Society for the Promotion of Science (JSPS), 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yuri Kiyokawa
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takashi Nakada
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Koji Bessyo-Hirashima
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Natsuki Fukaya
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kenji Hyodo
- Tsukuba Research Laboratories, Eisai Co. Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Kazuma Takase
- Tsukuba Research Laboratories, Eisai Co. Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Hiroshi Kikuchi
- Tsukuba Research Laboratories, Eisai Co. Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
20
|
Oku N. Innovations in Liposomal DDS Technology and Its Application for the Treatment of Various Diseases. Biol Pharm Bull 2017; 40:119-127. [PMID: 28154249 DOI: 10.1248/bpb.b16-00857] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Liposomes have been widely used as drug carriers in the field of drug delivery systems (DDS), and they are thought to be ideal nano-capsules for targeting DDS after being injected into the bloodstream. In general, DDS drugs meet the needs of aged and super-aged societies, since the administration route of drugs can be changed, the medication frequency reduced, the adverse effects of drugs suppressed, and so on. In fact, a number of liposomal drugs have been launched and used worldwide including liposomal anticancer drugs, and these drugs have appeared on the market owing to various innovations in liposomal DDS technologies. The accumulation of long-circulating liposomes in cancer tissue is driven by the enhanced permeability and retention (EPR) effect. In this review, liposome-based targeting DDS for cancer therapy is briefly discussed. Since cancer angiogenic vessels are the ideal target of drug carriers after their injection and are critical for cancer growth, damaging of these neovessels has been an approach for eradicating cancer cells. Also, the usage of liposomal DDS for the treatment of ischemic stroke is possible, since we observed that PEGylated liposomes accumulate in the site of cerebral ischemia in transient middle cerebral artery occlusion (t-MCAO) model rats. Interestingly, liposomes carrying neuroprotectants partly suppress ischemia/reperfusion injury of these model rats, suggesting that the EPR effect also works in ischemic diseases by causing an increase in the permeability of the blood vessel endothelium. The potential of liposomal DDS against life-threatening diseases might thus be attractive for supporting long-lived societies.
Collapse
Affiliation(s)
- Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
21
|
Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging. Int J Mol Sci 2017; 18:ijms18010208. [PMID: 28117680 PMCID: PMC5297838 DOI: 10.3390/ijms18010208] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Operating at multiple levels of control, mesenchymal stem cells from adipose tissue (ADSCs) communicate with organ systems to adjust immune response, provide signals for differentiation, migration, enzymatic reactions, and to equilibrate the regenerative demands of balanced tissue homeostasis. The identification of the mechanisms by which ADSCs accomplish these functions for dermatological rejuvenation and wound healing has great potential to identify novel targets for the treatment of disorders and combat aging. Herein, we review new insights into the role of adipose-derived stem cells in the maintenance of dermal and epidermal homeostasis, and recent advances in clinical applications of ADSCs related to dermatology.
Collapse
|
22
|
Shimizu K. Development of New Liposome Targeting Strategies for Application of Disease Therapies. YAKUGAKU ZASSHI 2017; 137:43-48. [PMID: 28049894 DOI: 10.1248/yakushi.16-00232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Drug delivery systems (DDS) are based on the concept of providing the optimal amount of a drug to a specific area requiring treatment. Liposomes are lipid-based nanoparticles capable of encapsulating any drug into both their membrane and aqueous phases. They have the potential to be targeted when their surfaces are modified with functional molecules such as antibodies and peptides. Thus, liposomes have strong potential as drug carriers if designed for active targeting. Our research group has recently developed a new concept for liposome targeting called "reverse targeting DDS (RT-DDS)". RT-DDS differs from conventional active targeting in that the surface of the liposomes is modified with an antigenic molecule that is specifically recognized by antigen-specific immune cells. This review describes in detail the differences between these two DDS targeting concepts and proposes the application of RT-DDS to the treatment of allergies based on research using ovalbumin as a model allergy antigen.
Collapse
Affiliation(s)
- Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
23
|
Agarwal M, Sahoo AK, Bose B. Receptor-Mediated Enhanced Cellular Delivery of Nanoparticles Using Recombinant Receptor-Binding Domain of Diphtheria Toxin. Mol Pharm 2016; 14:23-30. [PMID: 27959571 DOI: 10.1021/acs.molpharmaceut.6b00480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Antibodies and peptides are often used to home nanoparticles (NPs) to specific cells. Here in this work, we have used recombinant receptor-binding domain of diphtheria toxin (RDT) as a homing molecule for NPs. Diphtheria toxin binds to heparin binding EGF-like growth factor (HB-EGF) through its receptor-binding domain. HB-EGF is often overexpressed as cell surface molecule in various types of cancer. We have prepared monodispersed, spherical PLGA NPs and coated these NPs with RDT. These NPs are characterized by FESEM and FT-IR spectroscopy. Using flow cytometry and fluorescence spectroscopy, we show that coating with RDT increases cellular uptake of PLGA NPs. We further show that RDT-coated nanoparticles are internalized through clathrin-dependent receptor-mediated endocytosis that can be reduced by specific inhibitor. These RDT-coated nanoparticles (RDT-NP) were further used for preferential delivery of Irinotecan, a chemotherapeutic agent, to cells overexpressing HB-EGF. We show that receptor-mediated enhanced uptake of RDT-NPs increases the potency of irinotecan in these cells.
Collapse
Affiliation(s)
- Mahesh Agarwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati , Guwahati 781039, India
| | - Amaresh Kumar Sahoo
- Centre for Nanotechnology, Indian Institute of Technology Guwahati , Guwahati 781039, India
| | - Biplab Bose
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati , Guwahati 781039, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati , Guwahati 781039, India
| |
Collapse
|
24
|
Wang S, Hüttmann G, Scholzen T, Zhang Z, Vogel A, Hasan T, Rahmanzadeh R. A light-controlled switch after dual targeting of proliferating tumor cells via the membrane receptor EGFR and the nuclear protein Ki-67. Sci Rep 2016; 6:27032. [PMID: 27246531 PMCID: PMC4887907 DOI: 10.1038/srep27032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022] Open
Abstract
Using nanotechnology for optical manipulation of molecular processes in cells with high spatial and temporal precision promises new therapeutic options. Especially tumor therapy may profit as it requires a combination of both selectivity and an effective cell killing mechanism. Here we show a dual targeting approach for selective and efficient light-controlled killing of cells which are positive for epidermal growth factor receptor (EGFR) and Ki-67. Liposomes with the covalently linked EGFR antibody Erbitux enabled selective uptake of FITC-labeled Ki-67 antibody TuBB-9 in EGFR-positive cells pre-loaded with the photoactive dye BPD. After irradiation at 690 nm, BPD disrupted the endosomal membranes and delivered the antibodies to the nucleoli of the cells. The second irradiation at 490 nm activated the FITC-labeled TuBB-9, which caused inactivation of the Ki-67 protein and subsequent cell death via apoptosis. Efficient cell killing was possible at nanomolar concentrations of TuBB-9 due to the effective transport by immune liposomes and the high efficacy of the Ki-67 light-inactivation. Delivery of the liposomal constructs and cell destruction correlated well with the EGFR expression pattern of different cell lines (HeLa, OVCAR-5, MCF-7, and human fibroblasts), demonstrating an excellent selectivity.
Collapse
Affiliation(s)
- Sijia Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Institute of Biomedical Optics, University of Lübeck, Peter-Monnik-Weg 4, 23562 Lübeck, Germany
| | - Gereon Hüttmann
- Institute of Biomedical Optics, University of Lübeck, Peter-Monnik-Weg 4, 23562 Lübeck, Germany
| | - Thomas Scholzen
- Department of Immunology and Cell Biology, Research Center Borstel, Parkallee 22, 23845 Borstel, Germany
| | - Zhenxi Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Alfred Vogel
- Institute of Biomedical Optics, University of Lübeck, Peter-Monnik-Weg 4, 23562 Lübeck, Germany
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 50 Blossom St, Boston, MA 02114, USA
| | - Ramtin Rahmanzadeh
- Institute of Biomedical Optics, University of Lübeck, Peter-Monnik-Weg 4, 23562 Lübeck, Germany
| |
Collapse
|
25
|
Lian C, Ruan L, Shang D, Wu Y, Lu Y, Lü P, Yang Y, Wei Y, Dong X, Ren D, Chen K, Liu H, Tu Z. Heparin-Binding Epidermal Growth Factor-Like Growth Factor as a Potent Target for Breast Cancer Therapy. Cancer Biother Radiopharm 2016; 31:85-90. [DOI: 10.1089/cbr.2015.1956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Caixia Lian
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Lingling Ruan
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Dongsheng Shang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yanfang Wu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yongjin Lu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Peng Lü
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yuhua Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Yajun Wei
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Xiaojing Dong
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Dewan Ren
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Zhigang Tu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
26
|
Han GY, Cui Z, Guo B, Mei XF. Self-assembled nanoparticles covalently consisting of doxorubicin and EDB fibronectin specific peptide for solid tumour treatment. RSC Adv 2016. [DOI: 10.1039/c6ra11186f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We developed a facile modality to prepare nanoparticles consisting of doxorubicin and ZD2 motif for treating solid tumours. The nanoparticles showed great preferential cellular uptake in PC3 cells, high cell suppression, and strong anti-tumour ability.
Collapse
Affiliation(s)
- G. Y. Han
- The First Affiliated Hospital of Jinzhou Medical University
- Jinzhou 121001
- China
| | - Z. Cui
- The First Affiliated Hospital of Jinzhou Medical University
- Jinzhou 121001
- China
| | - B. Guo
- The First Affiliated Hospital of Jinzhou Medical University
- Jinzhou 121001
- China
| | - X. F. Mei
- The First Affiliated Hospital of Jinzhou Medical University
- Jinzhou 121001
- China
| |
Collapse
|
27
|
Ikemoto K, Shimizu K, Ohashi K, Takeuchi Y, Shimizu M, Oku N. Bauhinia purprea agglutinin-modified liposomes for human prostate cancer treatment. Cancer Sci 2015; 107:53-9. [PMID: 26495901 PMCID: PMC4724813 DOI: 10.1111/cas.12839] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 12/31/2022] Open
Abstract
Bauhinia purprea agglutinin (BPA) is a well‐known lectin that recognizes galactosyl glycoproteins and glycolipids. In the present study, we firstly found that BPA bound to human prostate cancer specimens but not to normal prostate ones. Therefore, we sought to develop BPA‐PEG‐modified liposomes (BPA‐PEG‐LP) encapsulating anticancer drugs for the treatment of prostate cancer. We examined the tumor targetability of BPA‐PEG‐LP with human prostate cancer DU145 cells, and observed that fluorescently labeled BPA‐PEG‐LP dominantly associated with the cells via the interaction between liposome‐surface BPA and cell‐surface galactosyl molecules. We also observed that BPA‐PEG‐LP accumulated in the prostate cancer tissue after the i.v. injection to DU145 solid cancer‐bearing mice, and strongly bound to the cancer cells. In a therapeutic study, DU145 solid cancer‐bearing mice were i.v. injected thrice with BPA‐PEG‐LP encapsulating doxorubicin (BPA‐PEG‐LPDOX, 2 mg/kg/day as the DOX dosage) or PEG‐modified liposomes encapsulating DOX (PEG‐LPDOX). As a result, BPA‐PEG‐LPDOX significantly suppressed the growth of the DU145 cancer cells, whereas PEG‐LPDOX at the same dosage as DOX showed little anti‐cancer effect. The present study suggested that BPA‐PEG‐LP could be a useful drug carrier for the treatment of human prostate cancers.
Collapse
Affiliation(s)
- Keisuke Ikemoto
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kento Ohashi
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yoshihito Takeuchi
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
28
|
Arabi L, Badiee A, Mosaffa F, Jaafari MR. Targeting CD44 expressing cancer cells with anti-CD44 monoclonal antibody improves cellular uptake and antitumor efficacy of liposomal doxorubicin. J Control Release 2015; 220:275-286. [PMID: 26518722 DOI: 10.1016/j.jconrel.2015.10.044] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/03/2015] [Accepted: 10/24/2015] [Indexed: 12/19/2022]
Abstract
Although liposomes improve the safety and pharmacokinetic properties of free drugs, they have not sufficiently enhanced the therapeutic efficacy compared to them. To address this problem, targeted therapy of tumor cells holds great promise to further enhance therapeutic index and decreases off-target effects compared with non-targeted liposomes. In the context of antibody-mediated targeted cancer therapy, we evaluated the anti-tumor activity and therapeutic efficacy of Doxil, and that of Doxil modified with a monoclonal antibody (mAb) against CD44, which is one of the most well-known surface markers associated with Cancer Stem Cells (CSCs). Flow cytometry analyses and confocal laser scanning microscopy results showed significant enhanced cellular uptake of CD44-targeted Doxil (CD44-Doxil) in CD44-positive C-26 cells compared to Doxil. However, CD44-negative NIH-3T3 cells showed a similar uptake and in vitro cytotoxicity with both CD44-Doxil and non-targeted Doxil. In BALB/c mice bearing C-26 murine carcinoma, CD44-Doxil groups exhibited significantly higher doxorubicin concentration (than Doxil) inside the tumor cells, while their circulation time and distribution profile remained comparable. CD44-Doxil at doses of either 10 or 15 mg/kg resulted in superior tumor growth inhibition and higher inclination to tumor, indicating the potential of anti-CD44 mAb targeting in therapeutic efficacy improvement. This study provides proof-of-principle for actively tumor-targeting concept and merits further investigations.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacokinetics
- Antibiotics, Antineoplastic/pharmacology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/pharmacology
- Carcinoma/drug therapy
- Carcinoma/immunology
- Carcinoma/metabolism
- Carcinoma/pathology
- Cell Line, Tumor
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Dose-Response Relationship, Drug
- Doxorubicin/administration & dosage
- Doxorubicin/analogs & derivatives
- Doxorubicin/chemistry
- Doxorubicin/pharmacokinetics
- Doxorubicin/pharmacology
- Drug Compounding
- Female
- Hyaluronan Receptors/immunology
- Hyaluronan Receptors/metabolism
- Immunoconjugates/administration & dosage
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/pharmacology
- Mice
- Mice, Inbred BALB C
- NIH 3T3 Cells
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Polyethylene Glycols/administration & dosage
- Polyethylene Glycols/chemistry
- Polyethylene Glycols/pharmacokinetics
- Polyethylene Glycols/pharmacology
- Tissue Distribution
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Leila Arabi
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran; Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran
| | - Ali Badiee
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran; Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran.
| |
Collapse
|
29
|
Talluri SV, Kuppusamy G, Karri VVSR, Tummala S, Madhunapantula SV. Lipid-based nanocarriers for breast cancer treatment – comprehensive review. Drug Deliv 2015; 23:1291-305. [DOI: 10.3109/10717544.2015.1092183] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Siddartha Venkata Talluri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS University, Udhagamandalam, Tamil Nadu, India and
| | - Gowthamarajan Kuppusamy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS University, Udhagamandalam, Tamil Nadu, India and
| | | | - Shashank Tummala
- Department of Pharmaceutics, JSS College of Pharmacy, JSS University, Udhagamandalam, Tamil Nadu, India and
| | | |
Collapse
|
30
|
Weijer R, Broekgaarden M, Kos M, van Vught R, Rauws EA, Breukink E, van Gulik TM, Storm G, Heger M. Enhancing photodynamic therapy of refractory solid cancers: Combining second-generation photosensitizers with multi-targeted liposomal delivery. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY C-PHOTOCHEMISTRY REVIEWS 2015. [DOI: 10.1016/j.jphotochemrev.2015.05.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
31
|
Limasale YDP, Tezcaner A, Özen C, Keskin D, Banerjee S. Epidermal growth factor receptor-targeted immunoliposomes for delivery of celecoxib to cancer cells. Int J Pharm 2015; 479:364-73. [PMID: 25595386 DOI: 10.1016/j.ijpharm.2015.01.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/07/2015] [Accepted: 01/10/2015] [Indexed: 12/14/2022]
Abstract
Cyclooxygenase-2 (COX-2) is highly expressed in many different cancers. Therefore, the inhibition of the COX-2 pathway by a selective COX-2 inhibitor, celecoxib (CLX), may be an alternative strategy for cancer prevention and therapy. Liposomal drug delivery systems can be used to increase the therapeutic efficacy of CLX while minimizing its side effects. Previous studies have reported the encapsulation of CLX within the non-targeted long circulating liposomes and functional effect of these formulations against colorectal cancer cell lines. However, the selectivity and internalization of CLX-loaded liposomes can further be improved by grafting targeting ligands on their surface. Cetuximab (anti-epidermal growth factor receptor - EGFR - monoclonal antibody) is a promising targeting ligand since EGFR is highly expressed in a wide range of solid tumors. The aim of this study was to develop EGFR-targeted immunoliposomes for enhancing the delivery of CLX to cancer cells and to evaluate the functional effects of these liposomes in cancer cell lines. EGFR-targeted ILs, having an average size of 120nm, could encapsulate 40% of the CLX, while providing a sustained drug release profile. Cell association studies have also shown that the immunoliposome uptake was higher in EGFR-overexpressing cells compared to the non-targeted liposomes. In addition, the CLX-loaded-anti-EGFR immunoliposomes were significantly more toxic compared to the non-targeted ones in cancer cells with EGFR-overexpression but not in the cells with low EGFR expression, regardless of their COX-2 expression status. Thus, selective targeting of CLX with anti-EGFR immunoliposomes appears to be a promising strategy for therapy of tumors that overexpress EGFR.
Collapse
Affiliation(s)
| | - Ayşen Tezcaner
- Department of Biotechnology, Middle East Technical University, Ankara 06800, Turkey; Department of Engineering Sciences, Middle East Technical University, Ankara 06800, Turkey; BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey
| | - Can Özen
- Department of Biotechnology, Middle East Technical University, Ankara 06800, Turkey; BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey
| | - Dilek Keskin
- Department of Biotechnology, Middle East Technical University, Ankara 06800, Turkey; Department of Engineering Sciences, Middle East Technical University, Ankara 06800, Turkey; BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey
| | - Sreeparna Banerjee
- Department of Biotechnology, Middle East Technical University, Ankara 06800, Turkey; Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey.
| |
Collapse
|
32
|
|
33
|
Liposomes as carriers of hydrophilic small molecule drugs: Strategies to enhance encapsulation and delivery. Colloids Surf B Biointerfaces 2014; 123:345-63. [DOI: 10.1016/j.colsurfb.2014.09.029] [Citation(s) in RCA: 292] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/30/2014] [Accepted: 09/14/2014] [Indexed: 12/18/2022]
|
34
|
Antibody-modified lipid nanoparticles for selective delivery of siRNA to tumors expressing membrane-anchored form of HB-EGF. Biochem Biophys Res Commun 2014; 449:460-5. [DOI: 10.1016/j.bbrc.2014.05.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 12/18/2022]
|
35
|
Alyautdin R, Khalin I, Nafeeza MI, Haron MH, Kuznetsov D. Nanoscale drug delivery systems and the blood-brain barrier. Int J Nanomedicine 2014; 9:795-811. [PMID: 24550672 PMCID: PMC3926460 DOI: 10.2147/ijn.s52236] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The protective properties of the blood–brain barrier (BBB) are conferred by the intricate architecture of its endothelium coupled with multiple specific transport systems expressed on the surface of endothelial cells (ECs) in the brain’s vasculature. When the stringent control of the BBB is disrupted, such as following EC damage, substances that are safe for peripheral tissues but toxic to neurons have easier access to the central nervous system (CNS). As a consequence, CNS disorders, including degenerative diseases, can occur independently of an individual’s age. Although the BBB is crucial in regulating the biochemical environment that is essential for maintaining neuronal integrity, it limits drug delivery to the CNS. This makes it difficult to deliver beneficial drugs across the BBB while preventing the passage of potential neurotoxins. Available options include transport of drugs across the ECs through traversing occludins and claudins in the tight junctions or by attaching drugs to one of the existing transport systems. Either way, access must specifically allow only the passage of a particular drug. In general, the BBB allows small molecules to enter the CNS; however, most drugs with the potential to treat neurological disorders other than infections have large structures. Several mechanisms, such as modifications of the built-in pumping-out system of drugs and utilization of nanocarriers and liposomes, are among the drug-delivery systems that have been tested; however, each has its limitations and constraints. This review comprehensively discusses the functional morphology of the BBB and the challenges that must be overcome by drug-delivery systems and elaborates on the potential targets, mechanisms, and formulations to improve drug delivery to the CNS.
Collapse
Affiliation(s)
- Renad Alyautdin
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh, Selangor, Malaysia
| | - Igor Khalin
- Faculty of Medicine and Defence Health, National Defence University of Malaysia (NDUM), Kuala Lumpur, Malaysia
| | - Mohd Ismail Nafeeza
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh, Selangor, Malaysia
| | | | - Dmitry Kuznetsov
- Department of Medicinal Nanobiotechnologies, N. I. Pirogoff Russian State Medical University, Moscow, Russia
| |
Collapse
|
36
|
|
37
|
Molavi O, Xiong XB, Douglas D, Kneteman N, Nagata S, Pastan I, Chu Q, Lavasanifar A, Lai R. Anti-CD30 antibody conjugated liposomal doxorubicin with significantly improved therapeutic efficacy against anaplastic large cell lymphoma. Biomaterials 2013; 34:8718-8725. [PMID: 23942212 PMCID: PMC3796131 DOI: 10.1016/j.biomaterials.2013.07.068] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 07/19/2013] [Indexed: 12/22/2022]
Abstract
The use of nano-carriers has been shown to improve the delivery and efficacy of chemotherapeutic agents in cancer patients. Recent studies suggest that decoration of the surface of nano-carriers with various targeting moieties may further improve the overall therapeutic efficacy. In this study, we compared the therapeutic efficacy of Doxil(®) (commercial doxorubicin-loaded liposomes) and that of Doxil(®) conjugated with anti-CD30 antibodies (CD30-targeted Doxil(®)) in treating anaplastic large cell lymphoma (ALCL), a type of T-cell lymphoma characterized by a high CD30 expression. Compared to Doxil(®), the CD30-targeted Doxil(®) showed a significantly higher binding affinity to ALCL cells (5.3% versus 27%, p = 0.005) and a lower inhibitory concentration at 50% (IC50) in-vitro (32.6 μg/mL versus 12.6 μg/mL, p = 0.006). In a SCID mouse xenograft model, CD30-targeted Doxil(®) inhibited tumor growth more significantly than the unconjugated formulation; specifically, tumors in mice treated with CD30-targeted Doxil(®) were significantly smaller than those in mice treated with Doxil(®) (average, 117 mm(3) versus 270 mm(3), p = 0.001) at 18 days after the tumors were inoculated. Our findings have provided the proof-of-principle of using CD30-targeted nano-carriers to treat cancers that are characterized by a high level of CD30 expression, such as ALCL.
Collapse
Affiliation(s)
- Ommoleila Molavi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Xiao-Bing Xiong
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Alberta, Canada
| | - Donna Douglas
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Norm Kneteman
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Satoshi Nagata
- Cancer Biology Research Center, Sanford Research/USD, SD, USA
| | - Ira Pastan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Quincy Chu
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Alberta, Canada
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
38
|
Sugiyama T, Asai T, Nedachi YM, Katanasaka Y, Shimizu K, Maeda N, Oku N. Enhanced active targeting via cooperative binding of ligands on liposomes to target receptors. PLoS One 2013; 8:e67550. [PMID: 23840738 PMCID: PMC3695877 DOI: 10.1371/journal.pone.0067550] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 05/21/2013] [Indexed: 11/18/2022] Open
Abstract
To achieve effective active targeting in a drug delivery system, we previously developed dual-targeting (DT) liposomes decorated with both vascular endothelial growth factor receptor-1 (VEGFR-1)-targeted APRPG and CD13-targeted GNGRG peptide ligands for tumor neovessels, and observed the enhanced suppression of tumor growth in Colon26 NL-17 tumor-bearing mice by the treatment with the DT liposomes encapsulating doxorubicin. In this present study, we examined the binding characteristics of DT liposomes having a different couple of ligands, namely, APRPG and integrin αvβ3-targeted GRGDS peptides. These DT liposomes synergistically associated to stimulated human umbilical vein endothelial cells compared with single-targeting (ST) liposomes decorated with APRPG or GRGDS. The results of a surface plasmon resonance assay showed that ST liposomes modified with APRPG or GRGDS peptide selectively bound to immobilized VEGFR-1 or integrin αvβ3, respectively. DT liposomes showed a higher affinity for a mixture of VEGFR-1 and integrin αvβ3 compared with ST liposomes, suggesting the cooperative binding of these 2 kinds of ligand on the liposomal surface. In a biodistribution assay, the DT liposomes accumulated to a significantly greater extent in the tumors of Colon26 NL-17 tumor-bearing mice compared with other liposomes. Moreover, the intratumoral distribution of the liposomes examined by confocal microscopy suggested that the DT liposomes targeted not only angiogenic endothelial cells but also tumor cells due to GRGDS-decoration. These findings suggest that "dual-targeting" augmented the affinity of the liposomes for the target cells and would thus be useful for active-targeting drug delivery for cancer treatment.
Collapse
Affiliation(s)
- Tomoki Sugiyama
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Yuki Murase Nedachi
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Yasufumi Katanasaka
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | | | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| |
Collapse
|