1
|
Wang Y, Huang R, Feng S, Mo R. Advances in nanocarriers for targeted drug delivery and controlled drug release. Chin J Nat Med 2025; 23:513-528. [PMID: 40383609 DOI: 10.1016/s1875-5364(25)60861-2] [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: 08/23/2024] [Revised: 09/13/2024] [Accepted: 12/24/2024] [Indexed: 05/20/2025]
Abstract
Nanocarrier-based drug delivery systems (nDDSs) present significant opportunities for improving disease treatment, offering advantages in drug encapsulation, solubilization, stability enhancement, and optimized pharmacokinetics and biodistribution. nDDSs, comprising lipid, polymeric, protein, and inorganic nanovehicles, can be guided by or respond to biological cues for precise disease treatment and management. Equipping nanocarriers with tissue/cell-targeted ligands enables effective navigation in complex environments, while functionalization with stimuli-responsive moieties facilitates site-specific controlled release. These strategies enhance drug delivery efficiency, augment therapeutic efficacy, and reduce side effects. This article reviews recent strategies and ongoing advancements in nDDSs for targeted drug delivery and controlled release, examining lesion-targeted nanomedicines through surface modification with small molecules, peptides, antibodies, carbohydrates, or cell membranes, and controlled-release nanocarriers responding to endogenous signals such as pH, redox conditions, enzymes, or external triggers like light, temperature, and magnetism. The article also discusses perspectives on future developments.
Collapse
Affiliation(s)
- Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Shufan Feng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
2
|
Zhang Y, Zhang Q, Li C, Zhou Z, Lei H, Liu M, Zhang D. Advances in cell membrane-based biomimetic nanodelivery systems for natural products. Drug Deliv 2024; 31:2361169. [PMID: 38828914 PMCID: PMC11149581 DOI: 10.1080/10717544.2024.2361169] [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: 06/08/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Active components of natural products, which include paclitaxel, curcumin, gambogic acid, resveratrol, triptolide and celastrol, have promising anti-inflammatory, antitumor, anti-oxidant, and other pharmacological activities. However, their clinical application is limited due to low solubility, instability, low bioavailability, rapid metabolism, short half-life, and strong off-target toxicity. To overcome these drawbacks, cell membrane-based biomimetic nanosystems have emerged that avoid clearance by the immune system, enhance targeting, and prolong drug circulation, while also improving drug solubility and bioavailability, enhancing drug efficacy, and reducing side effects. This review summarizes recent advances in the preparation and coating of cell membrane-coated biomimetic nanosystems and in their applications to disease for targeted natural products delivery. Current challenges, limitations, and prospects in this field are also discussed, providing a research basis for the development of multifunctional biomimetic nanosystems for natural products.
Collapse
Affiliation(s)
- Yifeng Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| | - Qian Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| | - Chunhong Li
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| | - Ziyun Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| | - Hui Lei
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, P. R. China
| |
Collapse
|
3
|
Nejad FS, Alizade-Harakiyan M, Haghi M, Ebrahimi R, Zangeneh MM, Farajollahi A, Fathi R, Mohammadi R, Miandoab SS, Asl MH, Divband B, Ahmadi A. Investigation of the impact of copper nanoparticles coated with ocimum bassilicum at chemoradiotherapy of colon carcinoma. Biochem Biophys Rep 2024; 39:101780. [PMID: 39044767 PMCID: PMC11263947 DOI: 10.1016/j.bbrep.2024.101780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Background Colon carcinoma poses a significant health challenge globally, particularly in developed nations where sedentary lifestyles, poor dietary choices, and genetic factors play a crucial role in its prevalence. Chemotherapy, the primary treatment method, carries severe side effects that can jeopardize patients' lives. Herbal extracts such as Ocimum Basillicum extract have shown effectiveness against cancer cells. Additionally, nanoparticles can significantly enhance drug delivery efficacy in these scenarios. Aim This article aims to investigate the impact of copper nanoparticles coated with Ocimum Bassilicum at chemoradiotherapy of Colon Carcinoma to hopefully create new treatment options with fewer side effects for patients. Methodology CuO bio-NPs were produced by the addition of 15 mL of extract dropwise to 80 mL of a 5 mM Cu (OAc)2 aqueous solution, which was then refluxed for 2 h at 100 °C. The mixture gradually became darker brown in color as a result of the heating procedure. The production of CuO NPs and the hydrogen-donating activity of antioxidant phenols within the plant are signaled by surface plasmon resonance excitation, which is the cause of this. In the cell culture, LS174t colon cancer cells were treated with OB extract, CuNPs, and OB-coated CuNPs with and without different radiation levels in order to assess cell viability, through the MTT assay, and the pro-apoptotic BAX and anti-apoptotic BCL2 expressions, through qPCR assay. Results The results demonstrate a decrease in cell viability and the expression of BCL2 and an increase in the expression of BAX especially when treated with OB-coated CuNPs and even furthermore when paired with radiation therapy. Conclusions After doing the clinical trial studies, the recent nanoparticles can be used for the treatment of Colorectal carcinoma.
Collapse
Affiliation(s)
- Farshad Seyed Nejad
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Alizade-Harakiyan
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Rokhsareh Ebrahimi
- Medicinal Chemistry Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mahdi Zangeneh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Farajollahi
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Radiation Science Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghayeh Fathi
- Polymer Research Laboratory, Department of Organic and Biochemistry, University of Tabriz, Tabriz, Iran
| | - Reza Mohammadi
- Polymer Research Laboratory, Department of Organic and Biochemistry, University of Tabriz, Tabriz, Iran
| | | | | | - Baharak Divband
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Amin Ahmadi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Pan Y, Xue Q, Yang Y, Shi T, Wang H, Song X, Luo Y, Liu W, Ren S, Cai Y, Nie Y, Song Z, Liu B, Li JP, Wei J. Glycoengineering-based anti-PD-1-iRGD peptide conjugate boosts antitumor efficacy through T cell engagement. Cell Rep Med 2024; 5:101590. [PMID: 38843844 PMCID: PMC11228665 DOI: 10.1016/j.xcrm.2024.101590] [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: 10/23/2023] [Revised: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024]
Abstract
Despite the important breakthroughs of immune checkpoint inhibitors in recent years, the objective response rates remain limited. Here, we synthesize programmed cell death protein-1 (PD-1) antibody-iRGD cyclic peptide conjugate (αPD-1-(iRGD)2) through glycoengineering methods. In addition to enhancing tissue penetration, αPD-1-(iRGD)2 simultaneously engages tumor cells and PD-1+ T cells via dual targeting, thus mediating tumor-specific T cell activation and proliferation with mild effects on non-specific T cells. In multiple syngeneic mouse models, αPD-1-(iRGD)2 effectively reduces tumor growth with satisfactory biosafety. Moreover, results of flow cytometry and single-cell RNA-seq reveal that αPD-1-(iRGD)2 remodels the tumor microenvironment and expands a population of "better effector" CD8+ tumor infiltrating T cells expressing stem- and memory-associated genes, including Tcf7, Il7r, Lef1, and Bach2. Conclusively, αPD-1-(iRGD)2 is a promising antibody conjugate therapeutic beyond antibody-drug conjugate for cancer immunotherapy.
Collapse
Affiliation(s)
- Yunfeng Pan
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qi Xue
- Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Yi Yang
- Glyco-therapy Biotechnology Co. Ltd., Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha Street, Qiantang District, Hangzhou, China
| | - Tao Shi
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hanbing Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xueru Song
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuting Luo
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenqi Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shiji Ren
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiran Cai
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yang Nie
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhentao Song
- Glyco-therapy Biotechnology Co. Ltd., Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha Street, Qiantang District, Hangzhou, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie P Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Jia Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China; Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| |
Collapse
|
5
|
Guo X, Zhang Y, Li Q, Shi F, HuangFu Y, Li J, Lao X. The influence of a modified p53 C-terminal peptide by using a tumor-targeting sequence on cellular apoptosis and tumor treatment. Apoptosis 2024; 29:865-881. [PMID: 38145442 DOI: 10.1007/s10495-023-01926-1] [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] [Accepted: 12/01/2023] [Indexed: 12/26/2023]
Abstract
The restoration of the function of p53 in tumors is a therapeutic strategy for the highly frequent mutation of the TP53 tumor suppressor gene. P460 is a wild-type peptide derived from the p53 C-terminus and has been proven to be capable of restoring the tumor suppressor function of p53. The poor accumulation of drugs in tumors is a serious hindrance to tumor treatment. For enhancing the activity of P460, the tumor-targeting sequence Arg-Gly-Asp-Arg (RGDR, C-end rule peptide) was introduced into the C-terminus of P460 to generate the new peptide P462. P462 presented better activity than P460 in inhibiting the proliferation of cancer cells and increasing the number of tumor cells undergoing apoptosis. Cell adhesion analysis and tumor imaging results revealed that P462 showed more specific and extensive binding with tumor cells and greater accumulation in tumors than the wild-type peptide. Importantly, treatment with P462 was more efficacious than that with P460 in vivo and was associated with considerably improved tumor-homing activity. This study highlights the importance of the roles of the tumor-homing sequence RGDR in the enhancement in cell attachment and tumor accumulation. The results of this work indicate that P462 could be a novel drug candidate for tumor treatment.
Collapse
Affiliation(s)
- Xiaoye Guo
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Yiming Zhang
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Qian Li
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Fangxin Shi
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Yifan HuangFu
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China.
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, 210009, Nanjing, P.R. China.
| |
Collapse
|
6
|
Tripathy RK, Pande AH. Molecular and functional insight into anti-EGFR nanobody: Theranostic implications for malignancies. Life Sci 2024; 345:122593. [PMID: 38554946 DOI: 10.1016/j.lfs.2024.122593] [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: 01/09/2024] [Revised: 02/27/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Targeted therapy and imaging are the most popular techniques for the intervention and diagnosis of cancer. A potential therapeutic target for the treatment of cancer is the epidermal growth factor receptor (EGFR), primarily for glioblastoma, lung, and breast cancer. Over-production of ligand, transcriptional up-regulation due to autocrine/paracrine signalling, or point mutations at the genomic locus may contribute to the malfunction of EGFR in malignancies. This exploit makes use of EGFR, an established biomarker for cancer diagnostics and treatment. Despite considerable development in the last several decades in making EGFR inhibitors, they are still not free from limitations like toxicity and a short serum half-life. Nanobodies and antibodies share similar binding properties, but nanobodies have the additional advantage that they can bind to antigenic epitopes deep inside the target that conventional antibodies are unable to access. For targeted therapy, anti-EGFR nanobodies can be conjugated to various molecules such as drugs, peptides, toxins and photosensitizers. These nanobodies can be designed as novel immunoconjugates using the universal modular antibody-based platform technology (UniCAR). Furthermore, Anti-EGFR nanobodies can be expressed in neural stem cells and visualised by effective fluorescent and radioisotope labelling.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India.
| |
Collapse
|
7
|
Jin M, Liu B, Zhang Z, Mu Y, Ma L, Yao H, Wang DA. Catechin-Functionalized Cationic Lipopolymer Based Multicomponent Nanomicelles for Lung-Targeting Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2302985. [PMID: 37558506 DOI: 10.1002/adma.202302985] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/22/2023] [Indexed: 08/11/2023]
Abstract
Catechins from green tea are one of the most effective natural compounds for cancer chemoprevention and have attracted extensive research. Cancer cell-selective apoptosis-inducing properties of catechins depend on efficient intracellular delivery. However, the low bioavailability limits the application of catechins. Herein, a nano-scaled micellar composite composed of catechin-functionalized cationic lipopolymer and serum albumin is constructed. Cationic liposomes tend to accumulate in the pulmonary microvasculature due to electrostatic effects and are able to deliver the micellar system intracellularly, thus improving the bioavailability of catechins. Albumin in the system acts as a biocompatible anti-plasma absorbent, forming complexes with positively charged lipopolymer under electrostatic interactions, contributing to prolonged in vivo retention. The physicochemical properties of the nano-micellar complexes are characterized, and the antitumor properties of catechin-functionalized materials are confirmed by reactive oxygen species (ROS), caspase-3, and cell apoptosis measurements. The role of each functional module, cationic polymeric liposome, and albumin is revealed by cell penetration, in vivo animal assays, etc. This multicomponent micellar nanocomposite has the potential to become an effective vehicle for the treatment of lung diseases such as pneumonia, lung tumors, sepsis-induced lung injury, etc. This study also demonstrates that it is a great strategy to create a delivery system that is both tissue-targeted and biologically active by combining cationic liposomes with the native bioactive compound catechins.
Collapse
Affiliation(s)
- Min Jin
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518057, P. R. China
| |
Collapse
|
8
|
Abuwatfa WH, Pitt WG, Husseini GA. Scaffold-based 3D cell culture models in cancer research. J Biomed Sci 2024; 31:7. [PMID: 38221607 PMCID: PMC10789053 DOI: 10.1186/s12929-024-00994-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024] Open
Abstract
Three-dimensional (3D) cell cultures have emerged as valuable tools in cancer research, offering significant advantages over traditional two-dimensional (2D) cell culture systems. In 3D cell cultures, cancer cells are grown in an environment that more closely mimics the 3D architecture and complexity of in vivo tumors. This approach has revolutionized cancer research by providing a more accurate representation of the tumor microenvironment (TME) and enabling the study of tumor behavior and response to therapies in a more physiologically relevant context. One of the key benefits of 3D cell culture in cancer research is the ability to recapitulate the complex interactions between cancer cells and their surrounding stroma. Tumors consist not only of cancer cells but also various other cell types, including stromal cells, immune cells, and blood vessels. These models bridge traditional 2D cell cultures and animal models, offering a cost-effective, scalable, and ethical alternative for preclinical research. As the field advances, 3D cell cultures are poised to play a pivotal role in understanding cancer biology and accelerating the development of effective anticancer therapies. This review article highlights the key advantages of 3D cell cultures, progress in the most common scaffold-based culturing techniques, pertinent literature on their applications in cancer research, and the ongoing challenges.
Collapse
Affiliation(s)
- Waad H Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, P.O. Box. 26666, Sharjah, United Arab Emirates
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Ghaleb A Husseini
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, P.O. Box. 26666, Sharjah, United Arab Emirates.
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates.
| |
Collapse
|
9
|
Li S, Hoefnagel SJM, Krishnadath KK. Single domain Camelid antibody fragments for molecular imaging and therapy of cancer. Front Oncol 2023; 13:1257175. [PMID: 37746282 PMCID: PMC10514897 DOI: 10.3389/fonc.2023.1257175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Despite innovations in cancer therapeutics, cancer remains associated with high mortality and is one of biggest health challenges worldwide. Therefore, developing precise cancer imaging and effective treatments is an unmet clinical need. A relatively novel type of therapeutics are heavy chain variable domain antibody fragments (VHHs) derived from llamas. Here, we explored the suitability of VHHs for cancer imaging and therapy through reviewing the existing literature. We searched the MEDLINE, EMBASE and Cochrane databases and identified 32 papers on molecular imaging and 41 papers on therapy that were suitable for comprehensive reviewing. We found that VHHs harbor a higher specificity and affinity compared to mAbs, which contributes to high-quality imaging and less side-effects on healthy cells. The employment of VHHs in cancer imaging showed remarkably shorter times between administration and imaging. Studies showed that 18F and 99mTc are two optimal radionuclides for imaging with VHHs and that site-specific labelling is the optimal conjugation modality for VHHs with radionuclide or fluorescent molecules. We found different solutions for reducing kidney retention and immunogenicity of VHHs. VHHs as anticancer therapeutics have been tested in photodynamic therapy, targeted radionuclide therapy, immunotherapy and molecular targeted therapy. These studies showed that VHHs target unique antigen epitopes, which are distinct from the ones recognized by mAbs. This advantage means that VHHs may be more effective for targeted anticancer therapy and can be combined with mAbs. We found that high cellular internalization and specificity of VHHs contributes to the effectiveness and safety of VHHs as anticancer therapeutics. Two clinical trials have confirmed that VHHs are effective and safe for cancer imaging and therapy. Together, VHHs seem to harbor several advantages compared to mAbs and show potential for application in personalized treatment for cancer patients. VHH-based imaging and therapy are promising options for improving outcomes of cancer patients.
Collapse
Affiliation(s)
- Shulin Li
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
| | | | - Kausilia Krishnawatie Krishnadath
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
10
|
Saifi MA, Sathish G, Bazaz MR, Godugu C. Exploration of tumor penetrating peptide iRGD as a potential strategy to enhance tumor penetration of cancer nanotherapeutics. Biochim Biophys Acta Rev Cancer 2023; 1878:188895. [PMID: 37037389 DOI: 10.1016/j.bbcan.2023.188895] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023]
Abstract
Cancer therapy continues to be a huge challenge as most chemotherapeutic agents exert serious adverse effects on healthy organs. Chemotherapeutic agents lack selective targeting and even the existing target specific therapies are failing due to poor distribution into the tumor microenvironment. Nanotechnology offers multiple advantages to address the limitations encountered by conventional therapy. However, the delivery of nanotherapeutics to tumor tissue has not improved over the years partly due to the poor and inadequate distribution of nanotherapeutics into deeper tumor regions resulting in resistance and relapse. To curb the penetration concerns, iRGD was explored and found to be highly effective in improving the delivery of cancer nanomedicine. The preclinical observations are highly encouraging; however, the clinical translation is at a nascent stage. Based on this, we have made an elaborative effort to give a detailed account of various promising applications of iRGD to increase anticancer and tumor imaging potential. Importantly, we have comprehensively discussed the shortcomings and uncertainties associated with the clinical translation of iRGD-based therapeutic approaches and future directions.
Collapse
Affiliation(s)
- Mohd Aslam Saifi
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Gauri Sathish
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Mohd Rabi Bazaz
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India.
| |
Collapse
|
11
|
Saibu OA, Hammed SO, Oladipo OO, Odunitan TT, Ajayi TM, Adejuyigbe AJ, Apanisile BT, Oyeneyin OE, Oluwafemi AT, Ayoola T, Olaoba OT, Alausa AO, Omoboyowa DA. Protein-protein interaction and interference of carcinogenesis by supramolecular modifications. Bioorg Med Chem 2023; 81:117211. [PMID: 36809721 DOI: 10.1016/j.bmc.2023.117211] [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: 10/25/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023]
Abstract
Protein-protein interactions (PPIs) are essential in normal biological processes, but they can become disrupted or imbalanced in cancer. Various technological advancements have led to an increase in the number of PPI inhibitors, which target hubs in cancer cell's protein networks. However, it remains difficult to develop PPI inhibitors with desired potency and specificity. Supramolecular chemistry has only lately become recognized as a promising method to modify protein activities. In this review, we highlight recent advances in the use of supramolecular modification approaches in cancer therapy. We make special note of efforts to apply supramolecular modifications, such as molecular tweezers, to targeting the nuclear export signal (NES), which can be used to attenuate signaling processes in carcinogenesis. Finally, we discuss the strengths and weaknesses of using supramolecular approaches to targeting PPIs.
Collapse
Affiliation(s)
- Oluwatosin A Saibu
- Department of Environmental Toxicology, Universitat Duisburg-Essen, NorthRhine-Westphalia, Germany
| | - Sodiq O Hammed
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oladapo O Oladipo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Tope T Odunitan
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Temitope M Ajayi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Aderonke J Adejuyigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Boluwatife T Apanisile
- Department of Nutrition and Dietetics, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oluwatoba E Oyeneyin
- Theoretical and Computational Chemistry Unit, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Adenrele T Oluwafemi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Tolulope Ayoola
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Olamide T Olaoba
- Department of Molecular Pathogenesis and Therapeutics, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Abdullahi O Alausa
- Department of Molecular Biology and Biotechnology, ITMO University, St Petersburg, Russia
| | - Damilola A Omoboyowa
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| |
Collapse
|
12
|
Zhou S, Li C, Yuan Y, Jiang L, Chen W, Jiang X. Dendritic lipopeptide liposomes decorated with dual-targeted proteins. Biomater Sci 2022; 10:7032-7041. [PMID: 36318065 DOI: 10.1039/d2bm00952h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to their homing effects, cell and cell membrane-derived nanocarriers have been widely used to enhance drug target delivery. Inspired by the protein-anchored cell membrane architecture, we here report a tumor-targeted liposome, dtDLP, which was constructed through the electrostatic interaction between dendritic lipopeptide liposomes and a dual-targeted recombinant protein, achieving superior tumor homing, cellular endocytotic and penetration abilities. The dual-targeted recombinant protein consists of an anti-epidermal growth factor receptor single domain antibody and a peptide ligand for the integrin αvβ3. dtDLPs substantially reduced macrophage phagocytosis and increased drug internalization in both 4T1 cells and HeLa cells by providing more endocytic pathways. In addition, the dtDLPs showed great penetration ability in both multicellular spheroids and tumor tissues. Due to the improved cancer cellular uptake and tumor penetration, the dtDLPs exhibited a superior anticancer effect in both HeLa and 4T1 tumor-bearing mice. This work will be helpful for the design of cell-specific liposomes with admirable tumor targeting, endocytotic and penetration abilities.
Collapse
Affiliation(s)
- Sensen Zhou
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Cheng Li
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Yang Yuan
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Lei Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China. .,State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| |
Collapse
|
13
|
|
14
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
15
|
Wu M, Xiao K, Liu X, Yang Y, Song G, Xiao G, Liu Q, Yuan J, Liu B. Organic Small Molecule Contrast Agent for Targeted Photoacoustic Imaging of Patient-Derived Brain Tumors. Adv Healthc Mater 2022; 11:e2201640. [PMID: 36050894 DOI: 10.1002/adhm.202201640] [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: 07/06/2022] [Revised: 08/17/2022] [Indexed: 01/28/2023]
Abstract
Traditional glioblastoma (GBM) cell lines do not maintain the heterogeneity of the original tumor, cell interactions, and therapy response, thus limiting their investigation in GBM theranostics. Herein, a kind of GBM tumor-targeting nanoparticles (NPs) TCFNP@iRGD are designed and constructed, which are generated by photoacoustic (PA) contrast agent 2-(3-cyano-4,5,5-trimethylfuran-2(5H)-ylidene) malononitrile (TCF)-OH through facile nanoprecipitation and decorated with an active targeting ligand iRGD. Their potential in GBM detection via PA imaging on glioma patient-derived cells intracranial xenograft models is evaluated for the first time. Excellent tumor-specific PA mapping performance of GBM is realized by TCFNP@iRGD, demonstrating its promising potential in the clinical diagnosis of GBM.
Collapse
Affiliation(s)
- Min Wu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.,Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, P. R. China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Xingang Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yudan Yang
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Gousheng Song
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Jian Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.,Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, P. R. China
| |
Collapse
|
16
|
Yagolovich AV, Isakova AA, Artykov AA, Vorontsova YV, Mazur DV, Antipova NV, Pavlyukov MS, Shakhparonov MI, Gileva AM, Markvicheva EA, Plotnikova EA, Pankratov AA, Kirpichnikov MP, Gasparian ME, Dolgikh DA. DR5-Selective TRAIL Variant DR5-B Functionalized with Tumor-Penetrating iRGD Peptide for Enhanced Antitumor Activity against Glioblastoma. Int J Mol Sci 2022; 23:12687. [PMID: 36293545 PMCID: PMC9604365 DOI: 10.3390/ijms232012687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
TRAIL (TNF-related apoptosis-inducing ligand) and its derivatives are potentials for anticancer therapy due to the selective induction of apoptosis in tumor cells upon binding to death receptors DR4 or DR5. Previously, we generated a DR5-selective TRAIL mutant variant DR5-B overcoming receptor-dependent resistance of tumor cells to TRAIL. In the current study, we improved the antitumor activity of DR5-B by fusion with a tumor-homing iRGD peptide, which is known to enhance the drug penetration into tumor tissues. The obtained bispecific fusion protein DR5-B-iRGD exhibited dual affinity for DR5 and integrin αvβ3 receptors. DR5-B-iRGD penetrated into U-87 tumor spheroids faster than DR5-B and demonstrated an enhanced antitumor effect in human glioblastoma cell lines T98G and U-87, as well as in primary patient-derived glioblastoma neurospheres in vitro. Additionally, DR5-B-iRGD was highly effective in a xenograft mouse model of the U-87 human glioblastoma cell line in vivo. We suggest that DR5-B-iRGD may become a promising candidate for targeted therapy for glioblastoma.
Collapse
Affiliation(s)
- Anne V. Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
- Manebio LLC, 115280 Moscow, Russia
| | - Alina A. Isakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Artem A. Artykov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Manebio LLC, 115280 Moscow, Russia
| | | | - Diana V. Mazur
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Nadezhda V. Antipova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Marat S. Pavlyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | | | - Anastasia M. Gileva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Elena A. Markvicheva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Ekaterina A. Plotnikova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, 125284 Moscow, Russia
| | - Andrey A. Pankratov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, 125284 Moscow, Russia
| | - Mikhail P. Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Marine E. Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Dmitry A. Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
17
|
Kumar A, Sharipov M, Turaev A, Azizov S, Azizov I, Makhado E, Rahdar A, Kumar D, Pandey S. Polymer-Based Hybrid Nanoarchitectures for Cancer Therapy Applications. Polymers (Basel) 2022; 14:polym14153027. [PMID: 35893988 PMCID: PMC9370428 DOI: 10.3390/polym14153027] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 12/16/2022] Open
Abstract
Globally, cancer is affecting societies and is becoming an important cause of death. Chemotherapy can be highly effective, but it is associated with certain problems, such as undesired targeting and multidrug resistance. The other advanced therapies, such as gene therapy and peptide therapy, do not prove to be effective without a proper delivery medium. Polymer-based hybrid nanoarchitectures have enormous potential in drug delivery. The polymers used in these nanohybrids (NHs) provide them with their distinct properties and also enable the controlled release of the drugs. This review features the recent use of polymers in the preparation of different nanohybrids for cancer therapy published since 2015 in some reputed journals. The polymeric nanohybrids provide an advantage in drug delivery with the controlled and targeted delivery of a payload and the irradiation of cancer by chemotherapeutical and photodynamic therapy.
Collapse
Affiliation(s)
- Arun Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India;
| | - Mirkomil Sharipov
- Department of Chemistry, Changwon National University, Changwon 51140, Korea;
| | - Abbaskhan Turaev
- Laboratory of Biological Active Macromolecular Systems, Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, Tashkent 100125, Uzbekistan;
| | - Shavkatjon Azizov
- Laboratory of Biological Active Macromolecular Systems, Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, Tashkent 100125, Uzbekistan;
- Department of Pharmaceutical Chemistry, Tashkent Pharmaceutical Institute, Tashkent 100015, Uzbekistan
- Correspondence: (S.A.); (D.K.); or (S.P.)
| | - Ismatdjan Azizov
- State Center for Expertise and Standardization of Medicines, Medical Devices, and Medical Equipment, State Unitary Enterprise, Tashkent 100002, Uzbekistan;
| | - Edwin Makhado
- Department of Chemistry, School of Physical and Mineral Sciences, University of Limpopo, Polokwane 0727, South Africa;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran;
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India;
- Correspondence: (S.A.); (D.K.); or (S.P.)
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Sciences, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea
- Correspondence: (S.A.); (D.K.); or (S.P.)
| |
Collapse
|
18
|
Tucker RP, Degen M. Revisiting the Tenascins: Exploitable as Cancer Targets? Front Oncol 2022; 12:908247. [PMID: 35785162 PMCID: PMC9248440 DOI: 10.3389/fonc.2022.908247] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
For their full manifestation, tumors require support from the surrounding tumor microenvironment (TME), which includes a specific extracellular matrix (ECM), vasculature, and a variety of non-malignant host cells. Together, these components form a tumor-permissive niche that significantly differs from physiological conditions. While the TME helps to promote tumor progression, its special composition also provides potential targets for anti-cancer therapy. Targeting tumor-specific ECM molecules and stromal cells or disrupting aberrant mesenchyme-cancer communications might normalize the TME and improve cancer treatment outcome. The tenascins are a family of large, multifunctional extracellular glycoproteins consisting of four members. Although each have been described to be expressed in the ECM surrounding cancer cells, tenascin-C and tenascin-W are currently the most promising candidates for exploitability and clinical use as they are highly expressed in various tumor stroma with relatively low abundance in healthy tissues. Here, we review what is known about expression of all four tenascin family members in tumors, followed by a more thorough discussion on tenascin-C and tenascin-W focusing on their oncogenic functions and their potential as diagnostic and/or targetable molecules for anti-cancer treatment purposes.
Collapse
Affiliation(s)
- Richard P. Tucker
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA, United States
| | - Martin Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
- *Correspondence: Martin Degen,
| |
Collapse
|
19
|
Davoodi Z, Shafiee F. Internalizing RGD, a great motif for targeted peptide and protein delivery: a review article. Drug Deliv Transl Res 2022; 12:2261-2274. [PMID: 35015253 DOI: 10.1007/s13346-022-01116-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 01/10/2023]
Abstract
Understanding that cancer is one of the most important health problems, especially in advanced societies, is not difficult. The term of targeted cancer therapy has also been well known as an ideal treatment strategy in the recent years. Peptides with ability to specifically recognize the cancer cells with suitable penetration properties have been used as the targeting motif in this regard. In the present review article, we focus on an individual RGD-derived peptide with ability to recognize the integrin receptor on the cancer cell surface like its ancestor with an additional outstanding feature to penetrate to extravascular space of tumor and ability to penetrate to cancer cells unlike the original peptide. This peptide which has been named "internalizing RGD" or "iRGD" has been the focus of researches as a new targeting motif since it was discovered. To date, many types of molecules have been associated with this peptide for their targeted delivery to cancer cells. In this review article, we have discussed a summary of penetration mechanisms of iRGD and all introduced peptides and proteins attached to this attractive cell-penetrating peptide and have expressed the results of the studies.
Collapse
Affiliation(s)
- Zeinabosadat Davoodi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran
| | - Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran.
| |
Collapse
|
20
|
Merikhian P, Darvishi B, Jalili N, Esmailinejad MR, Khatibi AS, Kalbolandi SM, Salehi M, Mosayebzadeh M, Barough MS, Majidzadeh-A K, Yadegari F, Rahbarizadeh F, Farahmand L. Recombinant nanobody against MUC1 tandem repeats inhibits growth, invasion, metastasis, and vascularization of spontaneous mouse mammary tumors. Mol Oncol 2021; 16:485-507. [PMID: 34694686 PMCID: PMC8763658 DOI: 10.1002/1878-0261.13123] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/20/2021] [Accepted: 10/19/2021] [Indexed: 11/11/2022] Open
Abstract
Alteration in glycosylation pattern of MUC1 mucin tandem repeats during carcinomas has been shown to negatively affect adhesive properties of malignant cells and enhance tumor invasiveness and metastasis. In addition, MUC1 overexpression is closely interrelated with angiogenesis, making it a great target for immunotherapy. Alongside, easier interaction of nanobodies (single-domain antibodies) with their antigens, compared to conventional antibodies, is usually associated with superior desirable results. Herein, we evaluated the preclinical efficacy of a recombinant nanobody against MUC1 tandem repeats in suppressing tumor growth, angiogenesis, invasion, and metastasis. Expressed nanobody demonstrated specificity only toward MUC1-overexpressing cancer cells and could internalize in cancer cell lines. The IC50 values (the concentration at which the nanobody exerted half of its maximal inhibitory effect) of the anti-MUC1 nanobody against MUC1-positive human cancer cell lines ranged from 1.2 to 14.3 nm. Similar concentrations could also effectively induce apoptosis in MUC1-positive cancer cells but not in normal cells or MUC1-negative human cancer cells. Immunohistochemical staining of spontaneously developed mouse breast tumors prior to in vivo studies confirmed cross-reactivity of nanobody with mouse MUC1 despite large structural dissimilarities between mouse and human MUC1 tandem repeats. In vivo, a dose of 3 µg nanobody per gram of body weight in tumor-bearing mice could attenuate tumor progression and suppress excessive circulating levels of IL-1a, IL-2, IL-10, IL-12, and IL-17A pro-inflammatory cytokines. Also, a significant decline in expression of Ki-67, MMP9, and VEGFR2 biomarkers, as well as vasculogenesis, was evident in immunohistochemically stained tumor sections of anti-MUC1 nanobody-treated mice. In conclusion, the anti-MUC1 tandem repeat nanobody of the present study could effectively overcome tumor growth, invasion, and metastasis.
Collapse
Affiliation(s)
- Parnaz Merikhian
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Neda Jalili
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Azadeh Sharif Khatibi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shima Moradi Kalbolandi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Malihe Salehi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Marjan Mosayebzadeh
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mahdieh Shokrollahi Barough
- Cancer Immunotherapy and Regenerative Medicine, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fatemeh Yadegari
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
21
|
Ray SK, Mukherjee S. Imitating Hypoxia and Tumor Microenvironment with Immune Evasion by Employing Three Dimensional in vitro Cellular Models: Impressive Tool in Drug Discovery. Recent Pat Anticancer Drug Discov 2021; 17:80-91. [PMID: 34323197 DOI: 10.2174/1574892816666210728115605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 11/22/2022]
Abstract
The heterogeneous tumor microenvironment is exceptionally perplexing and not wholly comprehended. Different multifaceted alignments lead to the generation of oxygen destitute situations within the tumor niche that modulate numerous intrinsic tumor microenvironments. Disentangling these communications is vital for scheming practical therapeutic approaches that can successfully decrease tumor allied chemotherapy resistance by utilizing the innate capability of the immune system. Several research groups have concerned with a protruding role for oxygen metabolism along with hypoxia in the immunity of healthy tissue. Hypoxia in addition to hypoxia-inducible factors (HIFs) in the tumor microenvironment plays an important part in tumor progression and endurance. Although numerous hypoxia-focused therapies have shown promising outcomes both in vitro and in vivo these outcomes have not effectively translated into clinical preliminaries. Distinctive cell culture techniques have utilized as an in vitro model for tumor niche along with tumor microenvironment and proficient in more precisely recreating tumor genomic profiles as well as envisaging therapeutic response. To study the dynamics of tumor immune evasion, three-dimensional (3D) cell cultures are more physiologically important to the hypoxic tumor microenvironment. Recent research has revealed new information and insights into our fundamental understanding of immune systems, as well as novel results that have been established as potential therapeutic targets. There are a lot of patented 3D cell culture techniques which will be highlighted in this review. At present notable 3D cell culture procedures in the hypoxic tumor microenvironment, discourse open doors to accommodate both drug repurposing, advancement, and divulgence of new medications and will deliberate the 3D cell culture methods into standard prescription disclosure especially in the field of cancer biology which will be discussing here.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Department of Applied Sciences. Indira Gandhi Technological and Medical Sciences University, Ziro, Arunachal Pradesh-791120, India
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020, India
| |
Collapse
|
22
|
Stiltner J, McCandless K, Zahid M. Cell-Penetrating Peptides: Applications in Tumor Diagnosis and Therapeutics. Pharmaceutics 2021; 13:pharmaceutics13060890. [PMID: 34204007 PMCID: PMC8232808 DOI: 10.3390/pharmaceutics13060890] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 01/27/2023] Open
Abstract
Since their identification over twenty-five years ago, the plethora of cell-penetrating peptides (CPP) and their applications has skyrocketed. These 5 to 30 amino acid in length peptides have the unique property of breaching the cell membrane barrier while carrying cargoes larger than themselves into cells in an intact, functional form. CPPs can be conjugated to fluorophores, activatable probes, radioisotopes or contrast agents for imaging tissues, such as tumors. There is no singular mechanism for translocation of CPPs into a cell, and therefore, many CPPs are taken up by a multitude of cell types, creating the challenge of tumor-specific translocation and hindering clinical effectiveness. Varying strategies have been developed to combat this issue and enhance their diagnostic potential by derivatizing CPPs for better targeting by constructing specific cell-activated forms. These methods are currently being used to image integrin-expressing tumors, breast cancer cells, human histiocytic lymphoma and protease-secreting fibrosarcoma cells, to name a few. Additionally, identifying safe, effective therapeutics for malignant tumors has long been an active area of research. CPPs can circumvent many of the complications found in treating cancer with conventional therapeutics by targeted delivery of drugs into tumors, thereby decreasing off-target side effects, a feat not achievable by currently employed conventional chemotherapeutics. Myriad types of chemotherapeutics such as tyrosine kinase inhibitors, antitumor antibodies and nanoparticles can be functionally attached to these peptides, leading to the possibility of delivering established and novel cancer therapeutics directly to tumor tissue. While much research is needed to overcome potential issues with these peptides, they offer a significant advancement over current mechanisms to treat cancer. In this review, we present a brief overview of the research, leading to identification of CPPs with a comprehensive state-of-the-art review on the role of these novel peptides in both cancer diagnostics as well as therapeutics.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Correspondence: ; Tel.: +1-412-692-8893; Fax: 412-692-6184
| |
Collapse
|
23
|
Du S, Sha H, Ding N, Yang J, Qian H, Zhou S, Su S, Meng F, Chen H, Chen F, Zhang L, Liu B, Wei J. Lipophilic recombinant-protein insertion endows lymphocytes with enhanced targeting-infiltration ability in EGFR positive cancer. Cell Immunol 2021; 365:104376. [PMID: 33984534 DOI: 10.1016/j.cellimm.2021.104376] [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: 02/22/2021] [Revised: 04/21/2021] [Accepted: 05/02/2021] [Indexed: 12/09/2022]
Abstract
Adoptive T cell transfer is one of the most promising ways to combat solid tumors. However, the weak infiltration of T cells into tumor sites has restricted their antitumor efficacy. To overcome this obstacle, we used the lipophilic protein painting strategy to improve tumor targeting and penetrating capacity of lymphocytes for the first time. We synthesized the lipid anchor consisting of a bispecific recombinant protein iRGD-antiEGFR and DSPE-PEG derivates, then successfully inserted it into the membranes of T cells. This surface modification was non-invasive and could efficiently improve the infiltration ability of T cells into multicellular spheroids and tumor masses. The surface modified T cells also displayed superior antitumor activities in EGFR-positive tumor xenografts via systematic infusion. Moreover, the permeability and antitumor efficacy of these surface painted T cells could be remarkably enhanced when used in combination with local low-dose irradiation.
Collapse
Affiliation(s)
- Shiyao Du
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Huizi Sha
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Naiqing Ding
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Ju Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Shujuan Zhou
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Shu Su
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hong Chen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Fangjun Chen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lianru Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China.
| | - Jia Wei
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China.
| |
Collapse
|
24
|
Expression and purification of a recombinant ELRL-MAP30 with dual-targeting anti-tumor bioactivity. Protein Expr Purif 2021; 185:105893. [PMID: 33933613 DOI: 10.1016/j.pep.2021.105893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/26/2022]
Abstract
MAP30 (Momordica antiviral protein 30kD) is a single-chain Ⅰ-type ribosome inactivating protein with a variety of biological activities, including anti-tumor ability. It was reported that MAP30 would serve as a novel and relatively safe agent for prophylaxis and treatment of liver cancer. To determine whether adding two tumor targeting peptides could improve the antitumor activities of MAP30, we genetically modified MAP30 with an RGD motif and a EGFRi motif, which is a ligand with high affinity for αvβ3 integrins and with high affinity for EGFR. The recombinant protein ELRL-MAP30 (rELRL-MAP30) containing a GST-tag was expressed in E. coli. The rELRL-MAP30 was highly expressed in the soluble fraction after induction with 0.15 mM IPTG for 20 h at 16 °C. The purified rELRL-MAP30 appeared as a band on SDS-PAGE. It was identified by western blotting. Cytotoxicity of recombinant protein to HepG2, MDA-MB-231, HUVEC and MCF-7 cells was detected by MTT analysis. Half maximal inhibitory concentration (IC50) values were 54.64 μg/mL, 70.13 μg/mL, 146 μg/mL, 466.4 μg/mL, respectively. Proliferation inhibition assays indicated that rELRL-MAP30 could inhibit the growth of Human liver cancer cell HepG2 effectively. We found that rELRL-MAP30 significantly induced apoptosis in liver cancer cells, as evidenced by nuclear staining of DAPI. In addition, rELRL-MAP30 induced apoptosis in human liver cancer HepG2 cells by up-regulation of Bax as well as down-regulation of Bcl-2. Migration of cell line were markedly inhibited by rELRL-MAP30 in a dose-dependent manner compared to the recombinant MAP30 (rMAP30). In summary, the fusion protein displaying extremely potent cytotoxicity might be highly effective for tumor therapy.
Collapse
|
25
|
Abstract
Cancer is the second leading cause of death worldwide, and the search for specialised therapy options has been a challenge for decades. The emergence of active targeted therapies provides the opportunity to treat cancerous tissues without harming healthy ones due to peculiar physiological changes. Herein, peptides and peptide analogs have been gaining a lot of attention over the last decade, especially for the on-site delivery of therapeutics to target tissues in order to achieve efficient and reliable cancer treatment. Combining peptides with highly efficient drug delivery platforms could potentially eliminate off-target adverse effects encountered during active targeting of conventional chemotherapeutics. Small size, ease of production and characterisation, low immunogenicity and satisfactory binding affinity of peptides offer some advantages over other complex targeting moiety, no wonder the market of peptide-based drugs continues to expand expeditiously. It is estimated that the global peptide drug market will be worth around USD 48.04 billion by 2025, with a compound annual growth rate of 9.4%. In this review, the current state of art of peptide-based therapeutics with special interest on tumour targeting peptides has been discussed. Moreover, various active targeting strategies such as the use functionalised peptides or peptide analogs are also elaborated.
Collapse
Affiliation(s)
- Selin Seda Timur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - R Neslihan Gürsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
26
|
Sun S, Ding Z, Yang X, Zhao X, Zhao M, Gao L, Chen Q, Xie S, Liu A, Yin S, Xu Z, Lu X. Nanobody: A Small Antibody with Big Implications for Tumor Therapeutic Strategy. Int J Nanomedicine 2021; 16:2337-2356. [PMID: 33790553 PMCID: PMC7997558 DOI: 10.2147/ijn.s297631] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. However, the efficacy of antibody-based therapy is still confined and desperately needs further improvement. Nanobodies are the recombinant variable domains of heavy-chain-only antibodies, with many unique properties such as small size (~15kDa), excellent solubility, superior stability, ease of manufacture, quick clearance from blood, and deep tissue penetration, which gain increasing acceptance as therapeutical tools and are considered also as building blocks for chimeric antigen receptors as well as for targeted drug delivery. Thus, one of the promising novel developments that may address the deficiency of monoclonal antibody-based therapies is the utilization of nanobodies. This article provides readers the significant factors that the structural and biochemical properties of nanobodies and the research progress on nanobodies in the fields of tumor treatment, as well as their application prospect.
Collapse
Affiliation(s)
- Shuyang Sun
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Ziqiang Ding
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiaomei Yang
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xinyue Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Minlong Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Li Gao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Qu Chen
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shenxia Xie
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Aiqun Liu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shihua Yin
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhiping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xiaoling Lu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
27
|
Molecular targeted treatment and drug delivery system for gastric cancer. J Cancer Res Clin Oncol 2021; 147:973-986. [PMID: 33550445 DOI: 10.1007/s00432-021-03520-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/10/2021] [Indexed: 12/24/2022]
Abstract
Gastric cancer is still a major cancer worldwide. The early diagnosis rate of gastric cancer in most high incidence countries is low. At present, the overall treatment effect of gastric cancer is poor, and the median overall survival remains low. Most of the patients with gastric cancer are in an advanced stage when diagnosed, and drug treatment has become the main means. Thus, new targeted drugs and therapeutic strategies are the hope of improving the therapeutic effect of gastric cancer. In this review, we summarize the new methods and advances of targeted therapy for gastric cancer, including novel molecular targeted therapeutic agents and drug delivery systems, with a major focus on the development of drug delivery systems (drug carriers and targeting peptides). Elaborating these new methods and advances will contribute to the management of gastric cancer.
Collapse
|
28
|
Redmond J, McCarthy H, Buchanan P, Levingstone TJ, Dunne NJ. Advances in biofabrication techniques for collagen-based 3D in vitro culture models for breast cancer research. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111944. [PMID: 33641930 DOI: 10.1016/j.msec.2021.111944] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
Collagen is the most abundant component of the extracellular matrix (ECM), therefore it represents an ideal biomaterial for the culture of a variety of cell types. Recently, collagen-based scaffolds have shown promise as 3D culture platforms for breast cancer-based research. Two-dimensional (2D) in vitro culture models, while useful for gaining preliminary insights, are ultimately flawed as they do not adequately replicate the tumour microenvironment. As a result, they do not facilitate proper 3D cell-cell/cell-matrix interactions and often an exaggerated response to therapeutic agents occurs. The ECM plays a crucial role in the development and spread of cancer. Alterations within the ECM have a significant impact on the pathogenesis of cancer, the initiation of metastasis and ultimate progression of the disease. 3D in vitro culture models that aim to replicate the tumour microenvironment have the potential to offer a new frontier for cancer research with cell growth, morphology and genetic properties that more closely match in vivo cancers. While initial 3D in vitro culture models used in breast cancer research consisted of simple hydrogel platforms, recent advances in biofabrication techniques, including freeze-drying, electrospinning and 3D bioprinting, have enabled the fabrication of biomimetic collagen-based platforms that more closely replicate the breast cancer ECM. This review highlights the current application of collagen-based scaffolds as 3D in vitro culture models for breast cancer research, specifically for adherence-based scaffolds (i.e. matrix-assisted). Finally, the future perspectives of 3D in vitro breast cancer models and their potential to lead to an improved understanding of breast cancer diagnosis and treatment are discussed.
Collapse
Affiliation(s)
- John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland
| | - Helen McCarthy
- School of Pharmacy, Queen's University, Belfast BT9 7BL, United Kingdom; School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Paul Buchanan
- School of Nursing and Human Science, Dublin City University, Dublin 9, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
29
|
Lingasamy P, Laarmann AH, Teesalu T. Tumor Penetrating Peptide-Functionalized Tenascin-C Antibody for Glioblastoma Targeting. Curr Cancer Drug Targets 2021; 21:70-79. [PMID: 33001014 DOI: 10.2174/1568009620666201001112749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/03/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Conjugation to clinical-grade tumor penetrating iRGD peptide is a widely used strategy to improve tumor homing, extravasation, and penetration of cancer drugs and tumor imaging agents. The C domain of the extracellular matrix molecule Tenascin-C (TNC-C) is upregulated in solid tumors and represents an attractive target for clinical-grade single-chain antibody- based vehicles for tumor delivery drugs and imaging agents. OBJECTIVE To study the effect of C-terminal genetic fusion of the iRGD peptide to recombinant anti- TNC-C single-chain antibody clone G11 on systemic tumor homing and extravasation. METHODS Enzyme-linked immunosorbent assay was used to study the interaction of parental and iRGD-fused anti-TNC-C single-chain antibodies with C domain of tenascin-C and αVβ3 integrins. For systemic homing studies, fluorescein-labeled ScFV G11-iRGD and ScFV G11 antibodies were administered in U87-MG glioblastoma xenograft mice, and their biodistribution was studied by confocal imaging of tissue sections stained with markers of blood vessels and Tenascin C immunoreactivity. RESULTS In a cell-free system, iRGD fusion to ScFV G11 conferred the antibody has a robust ability to bind αVβ3 integrins. The fluorescein labeling of ScFV G11-iRGD did not affect its target binding activity. In U87-MG mice, iRGD fusion to ScFV G11 antibodies improved their homing to tumor blood vessels, extravasation, and penetration of tumor parenchyma. CONCLUSION The genetic fusion of iRGD tumor penetrating peptide to non-internalizing affinity targeting ligands may improve their tumor tropism and parenchymal penetration for more efficient delivery of imaging and therapeutic agents into solid tumor lesions.
Collapse
Affiliation(s)
- Prakash Lingasamy
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| | - Anett-Hildegard Laarmann
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| |
Collapse
|
30
|
Sharifi J, Khirehgesh MR, Safari F, Akbari B. EGFR and anti-EGFR nanobodies: review and update. J Drug Target 2020; 29:387-402. [DOI: 10.1080/1061186x.2020.1853756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jafar Sharifi
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mohammad Reza Khirehgesh
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Fatemeh Safari
- School of Paramedical Sciences, Diagnostic Laboratory Sciences and Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Akbari
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
31
|
Chen T, Liu X, Hong H, Wei H. Novel single-domain antibodies against the EGFR domain III epitope exhibit the anti-tumor effect. J Transl Med 2020; 18:376. [PMID: 33023595 PMCID: PMC7541222 DOI: 10.1186/s12967-020-02538-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/21/2020] [Indexed: 01/11/2023] Open
Abstract
Background Monoclonal antibodies (mAbs) have been used for cancer therapy. They are large and have some disadvantages limiting their use. Smaller antibody fragments are needed as their alternatives. A fully human single-domain antibody (sdAb) has a small size of only 15 kDa and consists of only the variable domain of the human antibody heavy chain (VH). It has no immunogenicity. It can easily penetrate into tumor tissues, target an epitope inaccessible to mAb and be manufactured in bacteria for a low cost. Epidermal growth factor receptor (EGFR) is over-expressed in many cancer cells and is a good target for cancer therapy. Methods The EGFR protein fragment located on the EGFR extracellular domain III was chosen to screen a human sdAb library. Five human anti-EGFR sdAbs were identified. Their specific binding to EGFR was confirmed by ELISA, Western blotting and flow cytometry. Their anti-tumor effects were tested. Results Five novel fully human anti-EGFR sdAbs were isolated. They specifically bound to EGFR, not to the seven unrelated proteins as negative controls. They also bound to the three different human cancer cell lines, but not to the two cell lines as negative controls. They inhibited cell proliferation, migration and invasion and increased apoptosis of these three cancer cell lines. Two of them were tested for their anti-tumor effect in vivo and showed the anti-tumor activity in a mouse xenograft model for human lung cancer. Immunohistochemical staining of xenograft tumors also showed that their anti-tumor effects were associated with the inhibition of cancer cell proliferation and the promotion of cancer cell apoptosis. Conclusions This study clearly demonstrated that the anti-EGFR sdAbs could inhibit cancer cell growth in vitro and tumor growth in vivo. They could be potential therapeutics for the treatment of different human cancers.
Collapse
Affiliation(s)
- Tao Chen
- Department of Cell Biology and Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xue Liu
- Department of Cell Biology and Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Haifeng Hong
- Department of Cell Biology and Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Henry Wei
- Department of Cell Biology and Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
32
|
Alzeeb G, Metges JP, Corcos L, Le Jossic-Corcos C. Three-Dimensional Culture Systems in Gastric Cancer Research. Cancers (Basel) 2020; 12:E2800. [PMID: 33003476 PMCID: PMC7601358 DOI: 10.3390/cancers12102800] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC), which includes cancer of the esophagus, the oesophagogastric junction, and the stomach fundus, is highly deadly with strong regional influence, Asia being the most affected. GC is often detected at late stages, with 30% of metastatic cases at diagnosis. Many authors have devised models to both unravel the mechanisms of GC development and to evaluate candidate therapeutics. Among these models, 2D-cell cultures are progressively replaced by 3D-cell cultures that recapitulate, much more comprehensively, tumor cellular and genetic heterogeneity, as well as responsiveness to environmental changes, such as exposure to drugs or irradiation. With respect to the specifics of GC, there are high hopes from such model systems, especially with the aim of identifying prognostic markers and novel drug targets.
Collapse
Affiliation(s)
- George Alzeeb
- Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (G.A.); (L.C.)
| | - Jean-Philippe Metges
- CHU de Brest, Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France;
| | - Laurent Corcos
- Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (G.A.); (L.C.)
- CHU de Brest, Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France;
| | | |
Collapse
|
33
|
Yang EY, Shah K. Nanobodies: Next Generation of Cancer Diagnostics and Therapeutics. Front Oncol 2020; 10:1182. [PMID: 32793488 PMCID: PMC7390931 DOI: 10.3389/fonc.2020.01182] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The development of targeted medicine has greatly expanded treatment options and spurred new research avenues in cancer therapeutics, with monoclonal antibodies (mAbs) emerging as a prevalent treatment in recent years. With mixed clinical success, mAbs still hold significant shortcomings, as they possess limited tumor penetration, high manufacturing costs, and the potential to develop therapeutic resistance. However, the recent discovery of “nanobodies,” the smallest-known functional antibody fragment, has demonstrated significant translational potential in preclinical and clinical studies. This review highlights their various applications in cancer and analyzes their trajectory toward their translation into the clinic.
Collapse
Affiliation(s)
- Emily Y Yang
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Departments of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Departments of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
| |
Collapse
|
34
|
Song Y, Xu M, Li Y, Li Y, Gu W, Halimu G, Fu X, Zhang H, Zhang C. An iRGD peptide fused superantigen mutant induced tumor-targeting and T lymphocyte infiltrating in cancer immunotherapy. Int J Pharm 2020; 586:119498. [PMID: 32505575 DOI: 10.1016/j.ijpharm.2020.119498] [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: 02/05/2020] [Revised: 05/07/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022]
Abstract
Solid tumors are intrinsically resistant to immunotherapy because of the major challenges including the immunosuppression and poor penetration of drugs and lymphocytes into solid tumors due to the complicated tumor microenvironment (TME). Our previous study has created a novel superantigen mutant ST-4 to efficiently active the T lymphocytes and alleviate immune suppression. In the present study, to accumulate ST-4 into the TME, we constructed a recombinant protein, ST-4-iRGD, by fusing ST-4 to a tumor-homing peptide, iRGD. We hypothesized that ST-4-iRGD could internalize into the TME through iRGD-mediated tumor targeting and tumor tissue penetrating to activate the regional immunoreaction. The results of in vitro studies showed that ST-4-iRGD achieved improved tumor targeting and cytotoxicity in mouse B16F10 melanoma cells. The iRGD-mediated tumor tissue penetration was further confirmed by imaging and immunofluorescence studies in vivo, wherein higher distribution of ST-4-iRGD was observed in the mouse 4T1 breast tumor model. Moreover, ST-4-iRGD exhibited enhanced anti-solid tumor characteristics and induced improved lymphocyte infiltration in the B16F10 and 4T1 models. In conclusion, using iRGD to facilitate better dissemination of the therapeutic agent ST-4 throughout a solid tumor mass is feasible, and ST-4-iRGD may be a potential candidate for efficient cancer immunotherapy in the future.
Collapse
Affiliation(s)
- Yubo Song
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Mingkai Xu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China.
| | - Yongqiang Li
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Yansheng Li
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Wu Gu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Gulinare Halimu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; University of Chinese Academy of Sciences, 19 YuQuan Road, Beijing 100049, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Xuanhe Fu
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Huiwen Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| | - Chenggang Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 72 WenHua Road, Shenyang 110016, PR China; Key Laboratory of Superantigen Research, Shenyang Bureau of Science and Technology, 72 WenHua Road, Shenyang 110016, PR China
| |
Collapse
|
35
|
Lingasamy P, Tobi A, Kurm K, Kopanchuk S, Sudakov A, Salumäe M, Rätsep T, Asser T, Bjerkvig R, Teesalu T. Tumor-penetrating peptide for systemic targeting of Tenascin-C. Sci Rep 2020; 10:5809. [PMID: 32242067 PMCID: PMC7118115 DOI: 10.1038/s41598-020-62760-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/09/2020] [Indexed: 11/09/2022] Open
Abstract
Extracellular matrix in solid tumors has emerged as a specific, stable, and abundant target for affinity-guided delivery of anticancer drugs. Here we describe the homing peptide that interacts with the C-isoform of Tenascin-C (TNC-C) upregulated in malignant tissues. TNC-C binding PL3 peptide (amino acid sequence: AGRGRLVR) was identified by in vitro biopanning on recombinant TNC-C. Besides TNC-C, PL3 interacts via its C-end Rule (CendR) motif with cell-and tissue penetration receptor neuropilin-1 (NRP-1). Functionalization of iron oxide nanoworms (NWs) and metallic silver nanoparticles (AgNPs) with PL3 peptide increased tropism of systemic nanoparticles towards glioblastoma (GBM) and prostate carcinoma xenograft lesions in nude mice (eight and five-fold respectively). Treatment of glioma-bearing mice with proapoptotic PL3-guided NWs improved the survival of the mice, whereas treatment with untargeted particles had no effect. PL3-coated nanoparticles were found to accumulate in TNC-C and NRP-1-positive areas in clinical tumor samples, suggesting a translational relevance. The systemic tumor-targeting properties and binding of PL3-NPs to the clinical tumor sections, suggest that the PL3 peptide may have applications as a targeting moiety for the selective delivery of imaging and therapeutic agents to solid tumors.
Collapse
Affiliation(s)
- Prakash Lingasamy
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Allan Tobi
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kaarel Kurm
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | | | - Aleksander Sudakov
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Oxford Nanopore Technologies Ltd., Oxford, UK
| | - Markko Salumäe
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tõnu Rätsep
- Department of Neurosurgery, Tartu University Hospital, Tartu, Estonia
| | - Toomas Asser
- Department of Neurosurgery, Tartu University Hospital, Tartu, Estonia
| | - Rolf Bjerkvig
- Department of Biomedicine Translational Cancer Research, University of Bergen, Bergen, Norway
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia. .,Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA. .,Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, USA.
| |
Collapse
|
36
|
Wang J, Li N, Cao L, Gao C, Zhang Y, Shuai Q, Xie J, Luo K, Yang J, Gu Z. DOX-loaded peptide dendritic copolymer nanoparticles for combating multidrug resistance by regulating the lysosomal pathway of apoptosis in breast cancer cells. J Mater Chem B 2020; 8:1157-1170. [PMID: 31951231 DOI: 10.1039/c9tb02130b] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Multidrug resistance (MDR) is a common phenomenon in clinical oncology and is a major obstacle to cancer chemotherapy. Many nanoparticle (NP)-based drug delivery systems have been developed to overcome MDR depending on increasing intracellular drug concentrations via increased cellular uptake and rapid drug release. The objective of this work was to investigate the performance and possible mechanisms of enzyme-sensitive mPEGylated dendron-GFLG-DOX conjugate based nanoparticles for blockading the MDR phenotype of MCF-7/ADR. In vitro, mPEGylated dendron-GFLG-DOX conjugate based nanoparticles could significantly promote cellular uptake and accumulation, potent cytotoxicity and apoptosis compared to free DOX in resistant cells. mPEGylated dendron-GFLG-DOX conjugate based nanoparticles were found to translocate across the membranes of resistant cells via active endocytic pathways leading to more DOX accumulating in the nuclei of MCF-7/ADR cells. Importantly, we found that mPEGylated dendron-GFLG-DOX conjugate based nanoparticles could induce cathepsin B in the cytoplasm and enhance lysosomal-mediated cell death compared to free DOX. Furthermore, mPEGylated dendron-GFLG-DOX conjugate based nanoparticles enhanced the drug's penetration, toxicity, and growth inhibition compared to free DOX in the three-dimensional multicellular tumor spheroid model. In vivo, mPEGylated dendron-GFLG-DOX conjugate based nanoparticles significantly improved the therapeutic efficacy against MDR xenograft tumors, and showed better biocompatibility than free DOX. These results indicated that mPEGylated dendron-GFLG-DOX conjugate based nanoparticles could be used as an alternative drug delivery system for MDR tumor treatment through initiating the lysosomal apoptosis pathway.
Collapse
Affiliation(s)
- Jianxi Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Ning Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China. and School of Pharmacy, Fujian Medical University, Fuzhou 350122, P. R. China
| | - Lei Cao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Chao Gao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Yan Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Qizhi Shuai
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Jinghui Xie
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China. and College of Materials Science and Engineering, Nanjing Tech University, Nanjing 210009, P. R. China
| |
Collapse
|
37
|
Ur Rahman A, Khan S, Khan M. Transport of trans-activator of transcription (TAT) peptide in tumour tissue model: evaluation of factors affecting the transport of TAT evidenced by flow cytometry. ACTA ACUST UNITED AC 2019; 72:519-530. [PMID: 31868235 DOI: 10.1111/jphp.13221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/29/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Trans-activator of transcription (TAT), a cell penetrating peptide, has been explored to overcome resistance to penetration and transport inside the cell, therefore, suggested to be used as drug delivery vector into drug-resistant tumours. The generosity of this study was to evaluate modifiable factors (concentration, temperature, incubation time and spheroid age) on the penetration of TAT. METHODS Multicellular tumour spheroids (MCTS) used as tumour tissue models to mimic some characteristics with in-vivo tumors. Cell monolayer and 3-, 5-, 7-day-old MCTS were incubated with TAT and effects of modifiable factors were determined quantitatively through flow cytometry, based on TAT-positive cell count (%) and mean fluorescence intensity. KEY FINDINGS Enhancing TAT concentration (1, 5 and 25 µm), transport significantly increased (ANOVA, P < 0.0001) in cell monolayer and spheroids. However, rising temperature from 7 to 37°C (t, P > 0.05) and increasing incubation time; 20 min, 1 h and 3 h; (ANOVA, P > 0.05) were statistically non-significant. Moreover, TAT penetration declines as spheroids get older (ANOVA, P < 0.01). CONCLUSION While exploiting MCTS as tumour tissue model, older spheroids could be preferred to target penetration-resistant cells and mimic the in-vivo microenvironment.
Collapse
Affiliation(s)
- Aziz Ur Rahman
- Manchester Pharmacy School, The University of Manchester, Manchester, UK.,Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Shahzeb Khan
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Munasib Khan
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| |
Collapse
|
38
|
Ding J, Chen J, Gao L, Jiang Z, Zhang Y, Li M, Xiao Q, Lee SS, Chen X. Engineered nanomedicines with enhanced tumor penetration. NANO TODAY 2019; 29:100800. [DOI: 10.1016/j.nantod.2019.100800] [Citation(s) in RCA: 287] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
39
|
iRGD: A Promising Peptide for Cancer Imaging and a Potential Therapeutic Agent for Various Cancers. JOURNAL OF ONCOLOGY 2019; 2019:9367845. [PMID: 31346334 PMCID: PMC6617877 DOI: 10.1155/2019/9367845] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
Abstract
Poor penetration into the tumor parenchyma and the reduced therapeutic efficacy of anticancer drugs and other medications are the major problems in tumor treatment. A new tumor-homing and penetrating peptide, iRGD (CRGDK/RGPD/EC), can be effectively used to combine and deliver imaging agents or anticancer drugs into tumors. The different “vascular zip codes” expressed in different tissues can serve as targets for docking-based (synaptic) delivery of diagnostic and therapeutic molecules. αv-Integrins are abundantly expressed in the tumor vasculature, where they are recognized by peptides containing the RGD integrin recognition motif. The iRGD peptide follows a multistep tumor-targeting process: First, it is proteolytically cleaved to generate the CRGDK fragment by binding to the surface of cells expressing αv integrins (αvβ3 and αvβ5). Then, the fragment binds to neuropilin-1 and penetrates the tumor parenchyma more deeply. Compared with conventional RGD peptides, the affinity of iRGD for αv integrins is in the mid to low nanomolar range, and the CRGDK fragment has a stronger affinity for neuropilin-1 than that for αv integrins because of the C-terminal exposure of a conditional C-end Rule (CendR) motif (R/KXXR/K), whose receptor proved to be neuropilin-1. Consequently, these advantages facilitate the transfer of CRGDK fragments from integrins to neuropilin-1 and consequently deeper penetration into the tumor. Due to its specific binding and strong affinity, the iRGD peptide can deliver imaging agents and anticancer drugs into tumors effectively and deeply, which is useful in detecting the tumor, blocking tumor growth, and inhibiting tumor metastasis. This review aims to focus on the role of iRGD in the imaging and treatment of various cancers.
Collapse
|
40
|
Tabtimmai L, Suphakun P, Srisook P, Kiriwan D, Phanthong S, Kiatwuthinon P, Chaicumpa W, Choowongkomon K. Cell-penetrable nanobodies (transbodies) that inhibit the tyrosine kinase activity of EGFR leading to the impediment of human lung adenocarcinoma cell motility and survival. J Cell Biochem 2019; 120:18077-18087. [PMID: 31172597 DOI: 10.1002/jcb.29111] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/02/2019] [Accepted: 05/07/2019] [Indexed: 12/30/2022]
Abstract
Most patients suffering from non-small cell lung cancer (NSCLC) have epidermal growth factor receptor (EGFR) overexpression. Currently, EGFR tyrosine kinase inhibitors (TKIs) that act as the ATP-analogs and monoclonal antibodies (MAbs) to EGFR-ectodomain that block intracellular signaling are used for the treatment of advanced NSCLC. Unfortunately, adverse effects due to the TKI off-target and drug resistance occur in a significant number of the treated patients while some NSCLC genotypes do not respond to the therapeutic MAbs. Thus, a more effective remedy for the treatment of EGFR-overexpressed cancers is deemed necessary. In this study, VH/VH H displayed-phage clones that are bound to recombinant EGFR-TK were fished-out from a humanized-camel VH/VH H phage display library. VH/VH H of three phage-infected Escherichia coli clones (VH18, VH H35, and VH36) were linked molecularly to nonaarginine (R9) for making them cell penetrable. R9-VH18, R9-VH H35, and R9-VH36 were cytotoxic to human adenocarcinomic alveolar basal epithelial cells (A549) at the fifty percent inhibitory concentration (IC50 ) 0.181 ± 0.132, 0.00961 ± 0.00516, and 0.00996 ± 0.00752 μM, respectively, which were approximately 1000-fold more effective than small molecular TKIs. R9-VH18 and R9-VH36 also delayed cancer cell migration in a scratch-wound assay. Computerized homology modeling and intermolecular docking revealed that VH18 and VH H35 used CDR3 to interact with EGFR-TK residues close to the catalytic site, which might sterically hinder the ATP-binding of the TK; VH36 used CDR2 to bind at the asymmetric dimerization surface, which might disrupt EGFR dimerization leading to inhibition of intracellular signaling. The humanized-cell penetrable nanobodies have a high potential for developing further towards a clinical application.
Collapse
Affiliation(s)
- Lueacha Tabtimmai
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Praphasri Suphakun
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Pimonwan Srisook
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Duangnapa Kiriwan
- Genetic Engineering Interdisciplinary Program, Graduate School, Kasetsart University, Bangkok, Thailand
| | - Siratcha Phanthong
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| | - Pichamon Kiatwuthinon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
41
|
Lu M, Chen F, Cao C, Garvey CJ, Fletcher NL, Houston ZH, Lu H, Lord MS, Thurecht KJ, Stenzel MH. Importance of Polymer Length in Fructose-Based Polymeric Micelles for an Enhanced Biological Activity. Macromolecules 2019. [DOI: 10.1021/acs.macromol.8b02381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Mingxia Lu
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Fan Chen
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Cheng Cao
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
- Australia Nuclear
Science and Technology Organisation, Lucas Heights, NSW 2234, Australia
| | - Christopher J. Garvey
- Australia Nuclear
Science and Technology Organisation, Lucas Heights, NSW 2234, Australia
| | - Nicholas L. Fletcher
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Zachary H. Houston
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Hongxu Lu
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Megan S. Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Kristofer J. Thurecht
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Martina H. Stenzel
- Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
42
|
Shehzad A, Ravinayagam V, AlRumaih H, Aljafary M, Almohazey D, Almofty S, Al-Rashid NA, Al-Suhaimi EA. Application of Three-dimensional (3D) Tumor Cell Culture Systems and Mechanism of Drug Resistance. Curr Pharm Des 2019; 25:3599-3607. [PMID: 31612821 DOI: 10.2174/1381612825666191014163923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022]
Abstract
The in-vitro experimental model for the development of cancer therapeutics has always been challenging. Recently, the scientific revolution has improved cell culturing techniques by applying three dimensional (3D) culture system, which provides a similar physiologically relevant in-vivo model for studying various diseases including cancer. In particular, cancer cells exhibiting in-vivo behavior in a model of 3D cell culture is a more accurate cell culture model to test the effectiveness of anticancer drugs or characterization of cancer cells in comparison with two dimensional (2D) monolayer. This study underpins various factors that cause resistance to anticancer drugs in forms of spheroids in 3D in-vitro cell culture and also outlines key challenges and possible solutions for the future development of these systems.
Collapse
Affiliation(s)
- Adeeb Shehzad
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Vijaya Ravinayagam
- Scientific Research & Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Hamad AlRumaih
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Meneerah Aljafary
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Dana Almohazey
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sarah Almofty
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Noor A Al-Rashid
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
43
|
Peptide-based targeted therapeutics: Focus on cancer treatment. J Control Release 2018; 292:141-162. [DOI: 10.1016/j.jconrel.2018.11.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 12/14/2022]
|
44
|
Ren W, Sha H, Yan J, Wu P, Yang J, Li R, Zhang H, Yu L, Qian H, Liu B. Enhancement of radiotherapeutic efficacy for esophageal cancer by paclitaxel-loaded red blood cell membrane nanoparticles modified by the recombinant protein anti-EGFR-iRGD. J Biomater Appl 2018; 33:707-724. [PMID: 30388386 DOI: 10.1177/0885328218809019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Paclitaxel is widely used as a radiosensitizer for various tumors, including esophageal cancer, but its therapeutic effect remains to be improved. In this study, we constructed a novel nano-radiosensitizer, anti-EGFR-iRGD-conjugated (iE)-PRNPs, by conjugating the recombinant protein anti-epidermal growth factor receptor (EGFR)-internalizing arginine-glycine-aspartic (iRGD) to the surface of paclitaxel-loaded red blood cell membrane nanoparticles (PRNPs). The iE-PRNPs were confirmed to possess tumor-targeting, high penetrability, and sustained release properties that free paclitaxel does not possess. Compared with that of paclitaxel, the sensitizer enhancement ratio of iE-PRNPs was significantly increased (1.32-fold and 1.25-fold) in esophageal cancer cells with high and low expression levels of EGFR, respectively. Additionally, compared with that of unmodified PRNPs, the sensitizer enhancement ratio of iE-PRNPs in EGFR-overexpressing esophageal cancer cells was significantly increased (1.27-fold), while that of PRNPs in esophageal cancer cells with a low EGFR expression level increased slightly (1.06-fold). The improved radiosensitization effect was associated with enhanced G2/M arrest, increased reactive oxygen species, and more effective induction of DNA double-strand breaks. In summary, iE-PRNPs appear to be a novel type of radiosensitizer with the potential to overcome the bottleneck of esophageal cancer radiotherapeutic efficacy.
Collapse
Affiliation(s)
- Wei Ren
- 1 The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Huizi Sha
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Jing Yan
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Puyuan Wu
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Ju Yang
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Rutian Li
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hang Zhang
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lixia Yu
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hanqing Qian
- 2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Baorui Liu
- 1 The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,2 The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
45
|
Rodallec A, Sicard G, Giacometti S, Carré M, Pourroy B, Bouquet F, Savina A, Lacarelle B, Ciccolini J, Fanciullino R. From 3D spheroids to tumor bearing mice: efficacy and distribution studies of trastuzumab-docetaxel immunoliposome in breast cancer. Int J Nanomedicine 2018; 13:6677-6688. [PMID: 30425482 PMCID: PMC6204867 DOI: 10.2147/ijn.s179290] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose Nanoparticles are of rising interest in cancer research, but in vitro canonical cell monolayer models are not suitable to evaluate their efficacy when prototyping candidates. Here, we developed three-dimensional (3D) spheroid models to test the efficacy of trastuzumab-docetaxel immunoliposomes in breast cancer prior to further testing them in vivo. Materials and methods Immunoliposomes were synthesized using the standard thin film method and maleimide linker. Two human breast cancer cell lines varying in Her2 expression were tested: Her2+ cells derived from metastatic site: mammary breast MDA-MB-453 and triple-negative MDA-MB-231 cells. 3D spheroids were developed and tested with fluorescence detection to evaluate viability. In vivo efficacy and biodistribution studies were performed on xenograft bearing nude mice using fluorescent and bioluminescent imaging. Results In vitro, antiproliferative efficacy was dependent upon cell type, size of the spheroids, and treatment scheduling, resulting in subsequent changes between tested conditions and in vivo results. Immunoliposomes performed better than free docetaxel + free trastuzumab and ado-trastuzumab emtansine (T-DM1). On MDA-MB-453 and MDA-MB-231 cell growth was reduced by 76% and 25%, when compared to free docetaxel + free trastuzumab and by 85% and 70% when compared to T-DM1, respectively. In vivo studies showed tumor accumulation ranging from 3% up to 15% of the total administered dose in MDA-MB-453 and MDA-MB-231 bearing mice. When compared to free docetaxel + free trastuzumab, tumor growth was reduced by 89% (MDA-MB-453) and 25% (MDA-MB-231) and reduced by 66% (MDA-MB-453) and 29% (MDA-MB-231) when compared to T-DM1, an observation in line with data collected from 3D spheroids experiments. Conclusion We demonstrated the predictivity of 3D in vitro models when developing and testing nanoparticles in experimental oncology. In vitro and in vivo data showed efficient drug delivery with higher efficacy and prolonged survival with immunoliposomes when compared to current anti-Her2 breast cancer strategies.
Collapse
Affiliation(s)
- Anne Rodallec
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| | - Guillaume Sicard
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| | - Sarah Giacometti
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| | - Manon Carré
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| | | | | | | | - Bruno Lacarelle
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| | - Joseph Ciccolini
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| | - Raphaelle Fanciullino
- SMARTc Unit, Laboratory of Pharmacokinetics and Toxicology UFR Pharmacy, Center for Research on Cancer of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University, Marseille, France,
| |
Collapse
|
46
|
Chen H, Sha H, Zhang L, Qian H, Chen F, Ding N, Ji L, Zhu A, Xu Q, Meng F, Yu L, Zhou Y, Liu B. Lipid insertion enables targeted functionalization of paclitaxel-loaded erythrocyte membrane nanosystem by tumor-penetrating bispecific recombinant protein. Int J Nanomedicine 2018; 13:5347-5359. [PMID: 30254439 PMCID: PMC6141126 DOI: 10.2147/ijn.s165109] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background There is currently much interest in cancer cell targeting and tumor penetrating for research and therapeutic purposes. Purpose To improve targeting delivery of antitumor drugs to gastric cancer, in this study, a tumor-targeting biocompatible drug delivery system derived from erythrocyte membrane for delivering paclitaxel (PTX) was constructed. Methods Erythrocyte membrane of human red blood cells (RBCs) were used for preparing of erythrocyte membrane-derived vesicles. 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-(maleimide[polyethylene glycol]-3400) (DSPE-PEG-MAL), a phospholipid derivative, was used to insert tumor-targeting molecular into erythrocyte membrane-derived vesicles. A lipid insertion method was used to functionalize these vesicles without the need for direct chemical conjugation. Furthermore, a tumor-penetrating bispecific recombinant protein named anti-EGFR-iRGD was used for the first time in this work to enable nanosystem to target and penetrate efficiently into the tumor site. Results Paclitaxel (PTX)-loaded anti-EGFR-iRGD-modified erythrocyte membrane nano-system (anti-EGFR-iRGD-RBCm-PTX, abbreviated to PRP) were manufactured. PRP was spheroid, uniformly size, about 171.7±4.7 nm in average, could be stable in vitro for 8 days, and released PTX in a biphasic pattern. PRP showed comparable cytotoxicity toward human gastric cancer cells in vitro. In vivo studies showed that, PRP accumulated in tumor site within 2 h of administration, lasted longer than 48 h, and the tumor volume was reduced 61% by PRP treatment in Balb/c nude mice, without causing severe side effects. Conclusion PRP has potential applications in cancer treatment and as an adjunct for other anticancer strategies.
Collapse
Affiliation(s)
- Hong Chen
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People's Republic of China, .,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China, .,Department of Oncology, Yixing People's Hospital, Jiangsu, People's Republic of China,
| | - Huizi Sha
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Lianru Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Fangjun Chen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Naiqin Ding
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Liulian Ji
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Anqing Zhu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Qiuping Xu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| | - Yan Zhou
- Department of Oncology, Yixing People's Hospital, Jiangsu, People's Republic of China,
| | - Baorui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, People's Republic of China, .,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China,
| |
Collapse
|
47
|
Sokolova EA, Vodeneev VA, Deyev SM, Balalaeva IV. 3D in vitro models of tumors expressing EGFR family receptors: a potent tool for studying receptor biology and targeted drug development. Drug Discov Today 2018; 24:99-111. [PMID: 30205170 DOI: 10.1016/j.drudis.2018.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/27/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022]
Abstract
Carcinomas overexpressing EGFR family receptors are of high clinical importance, because the receptors have prognostic value and are used as molecular targets for anticancer therapy. Insufficient drug efficacy necessitates further in-depth research of the receptor biology and improvement in preclinical stages of drug evaluation. Here, we review the currently used advanced 3D in vitro models of tumors, including tumor spheroids, models in natural and synthetic matrices, tumor organoids and microfluidic-based models, as a potent tool for studying EGFR biology and targeted drug development. We are especially focused on factors that affect the biology of tumor cells, causing modification in the expression and basic phosphorylation of the receptors, crosstalk with other signaling pathways and switch between downstream cascades, resulting ultimately in the resistance to antitumor agents.
Collapse
Affiliation(s)
- Evgeniya A Sokolova
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia; Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklay St., Moscow 117997, Russia
| | - Vladimir A Vodeneev
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia
| | - Sergey M Deyev
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia; Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklay St., Moscow 117997, Russia
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia; I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya str., Moscow 119991, Russia.
| |
Collapse
|
48
|
Zhang Z, Qian H, Huang J, Sha H, Zhang H, Yu L, Liu B, Hua D, Qian X. Anti-EGFR-iRGD recombinant protein modified biomimetic nanoparticles loaded with gambogic acid to enhance targeting and antitumor ability in colorectal cancer treatment. Int J Nanomedicine 2018; 13:4961-4975. [PMID: 30214200 PMCID: PMC6124475 DOI: 10.2147/ijn.s170148] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Red blood cell membrane-coated nanoparticle (RBCm-NP) platform, which consist of natural RBCm and synthetic polymeric core, can extend circulation time in vivo with an improved biocompatibility and stability of this biomimetic nanocarrier. To achieve better bioavailability of antitumor drugs that were loaded in RBCm-NPs, the functionalization of coated RBCm with specific targeting ability is essential. Bispecific recombinant protein anti-EGFR-iRGD, containing both tumor penetrating peptide (internalizing RGD peptide) and EGFR single-domain antibody (sdAb), seems to be an optimal targeting ligand for RBCm-NPs in the treatment of multiple tumors, especially colorectal cancer with high EGFR expression. Materials and methods We modified the anti-EGFR-iRGD recombinant protein on the surface of RBCm-NPs by lipid insertion method to construct iE-RBCm-PLGA NPs and confirmed the presentation of active tumor-targeting ability in colorectal cancer models with high EGFR expression when compared with RBCm-PLGA NPs. In addition, potential anti-tumor drug gambogic acid (GA) was loaded into the NPs to endow the antitumor efficiency of iE-RBCm-GA/PLGA NPs. It was simultaneously evaluated whether GA can reach better biocompatibility benefiting from the improved antitumor efficiency of iE-RBCm-GA/PLGA NPs in colorectal cancer models. Results We successfully modified anti-EGFR-iRGD proteins on the surface of biomimetic NPs with integrated and stable "shell-core" structure. iE-RBCm-PLGA NPs showed its improved targeting ability in vitro (multicellular spheroids [MCS]) and in vivo (nude mice bearing tumors). Besides, no matter on short-term cell apoptosis at tumor site (terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling [TUNEL]) and long-term tumor inhibition, iE-RBCm-GA/PLGA NPs achieved better antitumor efficacy than free GA in spite of the similar effects of cytotoxicity and apoptosis to GA in vitro. Conclusion We expect that the bispecific biomimetic nanocarrier can extend the clinical application of many other potential antitumor drugs similar to GA and become a novel drug carrier in the colorectal cancer treatment.
Collapse
Affiliation(s)
- Zhen Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China, .,Department of Integrated Traditional Chinese Medicine and Western Medicine Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Hanqing Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Jie Huang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Huizi Sha
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Hang Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Lixia Yu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Baorui Liu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Dong Hua
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China,
| | - Xiaoping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| |
Collapse
|
49
|
Nanobodies as novel therapeutic agents in envenomation. Biochim Biophys Acta Gen Subj 2018; 1862:2955-2965. [PMID: 30309831 DOI: 10.1016/j.bbagen.2018.08.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND An effective therapy against envenoming should be a priority in view of the high number scorpion stings and snakebites. Serum therapy is still widely applied to treat the envenomation victims; however this approach suffers from several shortcomings. The employment of monoclonal antibodies might be an outcome as these molecules are at the core of a variety of applications from protein structure determination to cancer treatment. The progress of activities in the twilight zone between genetic and antibody engineering have led to the development of a unique class of antibody fragments. These molecules possess several benefits and lack many possible disadvantages over classical antibodies. Within recombinant antibody formats, nanobodies or single domain antigen binding fragments derived from heavy chain only antibodies in camelids occupy a privileged position. SCOPE OF REVIEW In this paper we will briefly review the common methods of envenomation treatment and focus on details of various in vivo research activities that investigate the performance of recombinant, monoclonal nanobodies in venom neutralization. MAJOR CONCLUSIONS Nanobodies bind to their cognate target with high specificity and affinity, they can be produced in large quantities from microbial expression systems and are very robust even when challenged with harsh environmental conditions. Upon administering, they rapidly distribute throughout the body and seem to be well tolerated in humans posing low immunogenicity. GENERAL SIGNIFICANCE Scorpion and snake envenomation is a major issue in developing countries and nanobodies as a venom-neutralizing agent can be considered as a valuable and promising candidate in envenomation therapy.
Collapse
|
50
|
Functionalized Folic Acid-Conjugated Amphiphilic Alternating Copolymer Actively Targets 3D Multicellular Tumour Spheroids and Delivers the Hydrophobic Drug to the Inner Core. NANOMATERIALS 2018; 8:nano8080588. [PMID: 30072655 PMCID: PMC6116205 DOI: 10.3390/nano8080588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/21/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022]
Abstract
Engineering of a “smart” drug delivery system to specifically target tumour cells has been at the forefront of cancer research, having been engineered for safer, more efficient and effective use of chemotherapy for the treatment of cancer. However, selective targeting and choosing the right cancer surface biomarker are critical for a targeted treatment to work. Currently, the available delivery systems use a two-dimensional monolayer of cancer cells to test the efficacy of the drug delivery system, but designing a “smart” drug delivery system to be specific for a tumour in vivo and to penetrate the inner core remains a major design challenge. These challenges can be overcome by using a study model that integrates the three-dimensional aspect of a tumour in a culture system. Here, we tested the efficacy of a functionalized folic acid-conjugated amphiphilic alternating copolymer poly(styrene-alt-maleic anhydride) (FA-DABA-SMA) via a biodegradable linker 2,4-diaminobutyric acid (DABA) to specifically target and penetrate the inner core of three-dimensional avascular human pancreatic and breast tumour spheroids in culture. The copolymer was quantitatively analyzed for its hydrophobic drug encapsulation efficiency using three different chemical drug structures with different molecular weights. Their release profiles and tumour targeting properties at various concentrations and pH environments were also characterized. Using the anticancer drug curcumin and two standard clinical chemotherapeutic hydrophobic drugs, paclitaxel and 5-fluorouracil, we tested the ability of FA-DABA-SMA nanoparticles to encapsulate the differently sized drugs and deliver them to kill monolayer pancreatic cancer cells using the WST-1 cell proliferation assay. The findings of this study revealed that the functionalized folic acid-conjugated amphiphilic alternating copolymer shows unique properties as an active “smart” tumor-targeting drug delivery system with the ability to internalize hydrophobic drugs and release the chemotherapeutics for effective killing of cancer cells. The novelty of the study is the first to demonstrate a functionalized “smart” drug delivery system encapsulated with a hydrophobic drug effectively targeting and penetrating the inner core of pancreatic and breast cancer spheroids and reducing their volumes in a dose- and time-dependent manner.
Collapse
|