1
|
Bashraheel SS, Al-Sulaiti H, Goda SK. Generation of Novel Tumour-Selective SEA Superantigen-Based Peptides with Improved Safety and Efficacy for Precision Cancer Immunotherapy. Int J Mol Sci 2024; 25:9423. [PMID: 39273378 PMCID: PMC11395200 DOI: 10.3390/ijms25179423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Bacterial superantigens are T-cell-stimulatory protein molecules which produce massive cytokines and cause human diseases. Due to their ability to activate up to 20% of resting T-cells, they have effectively killed T-cell-dependent tumours in vivo. However, the intrinsic toxicity of whole SAg molecules highlights the urgent need to develop more effective and safer SAg-based immunotherapy. With its unique approach, our study is a significant step towards developing safer tumour-targeted superantigen peptides (TTSP). We identified the T-cell activation function regions on the SEA superantigen and produced variants with minimal lethality, ensuring a safer approach to cancer treatment. This involved the creation of twenty 50-amino-acid-long overlapping peptides covering the full-length SEA superantigen (P1-P20). We then screened these peptides for T-cell activation, successfully isolating two peptides (P5 and P15) with significant T-cell activation. These selected peptides were used to design and synthesise tumour-targeted superantigen peptides, which were linked to a cancer-specific third loop (L3) of transforming growth factor-α (TGF-α), TGFαL3 from either a C' or N' terminal with an eight-amino-acid flexible linker in between. We also produced several P15 variants by changing single amino acids or by amino acid deletions. The novel molecules were then investigated for cytokine production and tumour-targeted killing. The findings from our previous study and the current work open up new avenues for peptide-based immunotherapy, particularly when combined with other immunotherapy techniques, thereby ensuring effective and safer cancer treatment.
Collapse
Affiliation(s)
- Sara S Bashraheel
- College of Medicine, QU Health, Qatar University (QU), Doha P.O. Box 2713, Qatar
| | - Haya Al-Sulaiti
- College of Health and Science, QU Health, Qatar University (QU), Doha P.O. Box 2713, Qatar
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Sayed K Goda
- College of Science and Technology, University of Derby, Derby DE22 1GB, UK
| |
Collapse
|
2
|
Chiyyeadu A, Asgedom G, Bruhn M, Rocha C, Schlegel TU, Neumann T, Galla M, Vollmer Barbosa P, Hoffmann M, Ehrhardt K, Ha TC, Morgan M, Schoeder CT, Pöhlmann S, Kalinke U, Schambach A. A tetravalent bispecific antibody outperforms the combination of its parental antibodies and neutralizes diverse SARS-CoV-2 variants. Clin Immunol 2024; 260:109902. [PMID: 38218210 DOI: 10.1016/j.clim.2024.109902] [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: 10/11/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
The devastating impact of COVID-19 on global health shows the need to increase our pandemic preparedness. Recombinant therapeutic antibodies were successfully used to treat and protect at-risk patients from COVID-19. However, the currently circulating Omicron subvariants of SARS-CoV-2 are largely resistant to therapeutic antibodies, and novel approaches to generate broadly neutralizing antibodies are urgently needed. Here, we describe a tetravalent bispecific antibody, A7A9 TVB, which actively neutralized many SARS-CoV-2 variants of concern, including early Omicron subvariants. Interestingly, A7A9 TVB neutralized more variants at lower concentration as compared to the combination of its parental monoclonal antibodies, A7K and A9L. A7A9 also reduced the viral load of authentic Omicron BA.1 virus in infected pseudostratified primary human nasal epithelial cells. Overall, A7A9 displayed the characteristics of a potent broadly neutralizing antibody, which may be suitable for prophylactic and therapeutic applications in the clinics, thus highlighting the usefulness of an effective antibody-designing approach.
Collapse
Affiliation(s)
- Abhishek Chiyyeadu
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Girmay Asgedom
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Matthias Bruhn
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany
| | - Cheila Rocha
- German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Tom U Schlegel
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Thomas Neumann
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Philippe Vollmer Barbosa
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Markus Hoffmann
- German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Katrin Ehrhardt
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Teng-Cheong Ha
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Clara T Schoeder
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Stefan Pöhlmann
- German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
3
|
Chuang WH, Pislyagin E, Lin LY, Menchinskaya E, Chernikov O, Kozhemyako V, Gorpenchenko T, Manzhulo I, Chaikina E, Agafonova I, Silchenko A, Avilov S, Stonik V, Tzou SC, Aminin D, Wang YM. Holothurian triterpene glycoside cucumarioside A 2-2 induces macrophages activation and polarization in cancer immunotherapy. Cancer Cell Int 2023; 23:292. [PMID: 38001420 PMCID: PMC10668486 DOI: 10.1186/s12935-023-03141-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Despite intensive developments of adoptive T cell and NK cell therapies, the efficacy against solid tumors remains elusive. Our study demonstrates that macrophage-based cell therapy could be a potent therapeutic option against solid tumors. METHODS To this end, we determine the effect of a natural triterpene glycoside, cucumarioside A2-2 (CA2-2), on the polarization of mouse macrophages into the M1 phenotype, and explore the antitumor activity of the polarized macrophage. The polarization of CA2-2-pretreated macrophages was analyzed by flow cytometry and confocal imaging. The anti-cancer activity of CA2-2 macrophages was evaluated against 4T1 breast cancer cells and EAC cells in vitro and syngeneic mouse model in vivo. RESULTS Incubation of murine macrophages with CA2-2 led to polarization into the M1 phenotype, and the CA2-2-pretreated macrophages could selectively target and kill various types of cancer in vitro. Notably, loading near-infrared (NIR) fluorochrome-labeled nanoparticles, MnMEIO-mPEG-CyTE777, into macrophages substantiated that M1 macrophages can target and penetrate tumor tissues in vivo efficiently. CONCLUSION In this study, CA2-2-polarized M1 macrophages significantly attenuated tumor growth and prolonged mice survival in the syngeneic mouse models. Therefore, ex vivo CA2-2 activation of mouse macrophages can serve as a useful model for subsequent antitumor cellular immunotherapy developments.
Collapse
Affiliation(s)
- Wen-Han Chuang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS²B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Evgeny Pislyagin
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Liang-Yu Lin
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ekaterina Menchinskaya
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Oleg Chernikov
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Valery Kozhemyako
- Pacific State Medical University, Ostryakova Avenue, Building 2, Vladivostok, 690002, Russia
| | - Tatiana Gorpenchenko
- Federal Scientific Center of East Asia Terrestrial Biodiversity, Far Eastern Branch of the Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Igor Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of the Russian Academy of Science, Palchevskogo str. 17, Vladivostok, 690041, Russia
| | - Elena Chaikina
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Irina Agafonova
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Alexandra Silchenko
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Sergey Avilov
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Valentin Stonik
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Shey-Cherng Tzou
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS²B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Dmitry Aminin
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shin-Chuan 1st Road, Sanmin District, Kaohsiung City, 80708, Taiwan.
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan.
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS²B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan.
| |
Collapse
|
4
|
Du R, Zhao Z, Cui J, Li Y. Manganese-Based Nanotheranostics for Magnetic Resonance Imaging-Mediated Precise Cancer Management. Int J Nanomedicine 2023; 18:6077-6099. [PMID: 37908669 PMCID: PMC10614655 DOI: 10.2147/ijn.s426311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/20/2023] [Indexed: 11/02/2023] Open
Abstract
Manganese (Mn)-based magnetic resonance imaging (MRI) has become a competitive imaging modality for cancer diagnosis due to its advantages of non-invasiveness, high resolution and excellent biocompatibility. In recent years, a variety of Mn contrast agents based on different material systems have been synthesized, and a series of multi-purpose Mn nanocomposites have also emerged, showing satisfactory relaxation efficiency and MRI performance thus possess the transformation and application value in MRI-synergized cancer diagnosis and treatment. This tutorial review starts from the classification and properties of Mn-based nanomaterials, and then summarizes various preparation and functionalization strategies of nanosized Mn contrast agents, especially focuses on the latest progress of Mn contrast agents in MRI-synergized precise cancer theranostics. In addition, present review also discusses the current clinical transformation obstacles such as unclear molecular mechanisms, potential nanotoxicity, and scale production constraints. This paper provides evidence-based recommendations about the future prospects of multifunctional nanoplatforms, as well as technical guidance and panoramic expectations for the design of clinically meaningful cancer management programs.
Collapse
Affiliation(s)
- Ruochen Du
- Department of Laboratory Animal Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Ziwei Zhao
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Jing Cui
- College of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| | - Yanan Li
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, 030001, People’s Republic of China
| |
Collapse
|
5
|
Bashraheel SS, Goda SK. Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy. Int J Mol Sci 2023; 24:10507. [PMID: 37445686 DOI: 10.3390/ijms241310507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Bacterial superantigens (SAgs) are effective T-cell stimulatory molecules that lead to massive cytokine production. Superantigens crosslink between MHC class II molecules on the Antigen Presenting Cells (APC) and TCR on T-cells. This enables them to activate up to 20% of resting T cells, whilst conventional antigen presentation results in the activation of 0.001-0.0001% of the T cell population. These biological properties of superantigens make them attractive for use in immunotherapy. Previous studies have established the effectiveness of superantigens as therapeutic agents. This, however, was achieved with severe side effects due to the high lethality of the native toxins. Our study aims to produce superantigen-based peptides with minimum or no lethality for safer cancer treatment. In previous work, we designed and synthesized twenty overlapping SPEA-based peptides and successfully mapped regions in SPEA superantigen, causing a vasodilatory response. We screened 20 overlapping SPEA-based peptides designed and synthesized to cover the whole SPEA molecule for T-cell activation and tumor-killing ability. In addition, we designed and synthesized tumor-targeted superantigen-based peptides by fusion of TGFαL3 either from the N' or C' terminal of selected SPEA-based peptides with an eight-amino acid flexible linker in between. Our study identified parts of SPEA capable of stimulating human T-cells and producing different cytokines. We also demonstrated that the SPEA-based peptide conjugate binds specifically to cancer cells and can kill this cancer. Peptides induce T-cell activation, and tumor killing might pave the way for safer tumor-targeted superantigens (TTS). We proposed the combination of our new superantigen-based peptide conjugates with other immunotherapy techniques for effective and safer cancer treatment.
Collapse
Affiliation(s)
| | - Sayed K Goda
- College of Science and Technology, University of Derby, Derby DE22 1GB, UK
| |
Collapse
|
6
|
Subhan MA. Advances with metal oxide-based nanoparticles as MDR metastatic breast cancer therapeutics and diagnostics. RSC Adv 2022; 12:32956-32978. [PMID: 36425155 PMCID: PMC9670683 DOI: 10.1039/d2ra02005j] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
Metal oxide nanoparticles have attracted increased attention due to their emerging applications in cancer detection and therapy. This study envisioned to highlight the great potential of metal oxide NPs due to their interesting properties including high payload, response to magnetic field, affluence of surface modification to overcome biological barriers, and biocompatibility. Mammogram, ultrasound, X-ray computed tomography (CT), MRI, positron emission tomography (PET), optical or fluorescence imaging are used for breast imaging. Drug-loaded metal oxide nanoparticle delivered to the breast cancer cells leads to higher drug uptake. Thus, enhanced the cytotoxicity to target cells compared to free drug. The drug loaded metal oxide nanoparticle formulations hold great promise to enhance efficacy of breast cancer therapy including multidrug resistant (MDR) and metastatic breast cancers. Various metal oxides including magnetic metal oxides and magnetosomes are of current interests to explore cancer drug delivery and diagnostic efficacy especially for metastatic breast cancer. Metal oxide-based nanocarrier formulations are promising for their usage in drug delivery and release to breast cancer cells, cancer diagnosis and their clinical translations. Biomarker targeted therapy approaches for TNBC using metal oxide-based NPs are highly effective and promising.![]()
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| |
Collapse
|
7
|
Nguyen PV, Hervé-Aubert K, Chourpa I, Allard-Vannier E. Active targeting strategy in nanomedicines using anti-EGFR ligands - A promising approach for cancer therapy and diagnosis. Int J Pharm 2021; 609:121134. [PMID: 34571073 DOI: 10.1016/j.ijpharm.2021.121134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022]
Abstract
As active targeting using nanomedicines establishes itself as a strategy of choice in cancer therapy, several target receptors or ligands overexpressed in cancer cells have been identified and exploited. Among them, the epidermal growth factor receptor (EGFR) has emerged as one of the most promising oncomarkers for active targeting nanomedicines due to its overexpression and its active involvement in a wide range of cancer types. Henceforth, many novel EGFR-targeted nanomedicines for cancer therapy have been developed, giving encouraging results both in vitro and in vivo. This review focuses on different applications of such medicines in oncotherapy. On an important note, the contribution of EGFR-targeting ligands to final therapy efficacy along with current challenges and possible solutions or alternatives are emphasized.
Collapse
Affiliation(s)
- Phuoc Vinh Nguyen
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Katel Hervé-Aubert
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Igor Chourpa
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | | |
Collapse
|
8
|
Ghalamfarsa F, Khatami SH, Vakili O, Taheri-Anganeh M, Tajbakhsh A, Savardashtaki A, Fazli Y, Uonaki LR, Shabaninejad Z, Movahedpour A, Ghalamfarsa G. Bispecific antibodies in colorectal cancer therapy: recent insights and emerging concepts. Immunotherapy 2021; 13:1355-1367. [PMID: 34641708 DOI: 10.2217/imt-2021-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Colorectal cancer (CRC) is identified as a life-threatening malignancy. Despite several efforts and proceedings available for CRC therapy, it is still a health concern. Among a vast array of novel therapeutic procedures, employing bispecific antibodies (BsAbs) is currently considered to be a promising approach for cancer therapy. BsAbs, as a large family of molecules designed to realize two distinct epitopes or antigens, can be beneficial microgadgets to target the tumor-associated antigen pairs. On the other hand, applying the immune system's capabilities to attack malignant cells has been proven as a tremendous development in cancer therapeutic projects. The current study has attempted to overview some of the approved BsAbs in CRC therapy and those under clinical trials. For this purpose, reputable scientific search engines and databases, such as PubMed, ScienceDirect, Google Scholar, Scopus, etc., were explored using the keywords 'bispecific antibodies', 'colorectal cancer', 'immunotherapy' and 'tumor markers'.
Collapse
Affiliation(s)
- Farideh Ghalamfarsa
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy & Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mortaza Taheri-Anganeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yousef Fazli
- Dena Clinical Diagnostic Laboratory, Yasuj, Iran
| | - Leila Rezaei Uonaki
- Department of Biotechnology, School of Science, Shahrekord University, Shahrekord, Iran
| | - Zahra Shabaninejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghasem Ghalamfarsa
- Department of Microbiology & Immunology, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
9
|
Si Y, Pei X, Wang X, Han Q, Xu C, Zhang B. An anti-EGFR/anti- HER2 Bispecific Antibody with Enhanced Antitumor Activity Against Acquired Gefitinib-Resistant NSCLC Cells. Protein Pept Lett 2021; 28:1290-1297. [PMID: 34602035 DOI: 10.2174/0929866528666210930170624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) is a recurrent phenomenon during clinical therapy of non‑small-cell lung cancer (NSCLC). Studies have shown that HER2 is a key factor contributing to drug resistance in a variety of cancers. Furthermore, we have observed that HER2 is overexpressed in PC-9 NSCLC cells with acquired gefitinib-resistance (PC-9/GR) as compared to that in PC-9 cells. OBJECTIVE We hypothesized that blocking both EGFR and HER2 may serve as a potential strategy for treatment of NSCLC with acquired gefitinib-resistance. METHODS To target both EGFR and HER2 simultaneously, we developed a bispecific antibody HECrossMAb, which was derived from a humanized Cetuximab and Trastuzumab. The binding affinity of HECrossMAb for EGFR and HER2 was measured using enzyme-linked immunosorbent assay. The MTT assay was used to determine the effect of HECrossMAb on the proliferation of PC‑9 and PC‑9/GR cells in vitro. Finally, the effect of HECrossMAb on PI3K/AKT signaling and associated transcription factors was measured using western blot analysis. RESULTS Our results showed that HECrossMAb exerts enhanced cytotoxicity in both PC-9 and PC-9/GR cells by inhibiting the activation of PI3K/AKT signaling and expression of relevant transcription factors such as AEG-1, c-Myc, and c-Fos. CONCLUSION Our results suggest that HECrossMAb may function as a potential therapeutic agent for the treatment of NSCLC overexpressing EGFR and HER2.
Collapse
Affiliation(s)
- Yan Si
- Institutes of Physical Science and Information Technology, Anhui University, Hefei. China
| | - Xinxin Pei
- Institutes of Physical Science and Information Technology, Anhui University, Hefei. China
| | - Xiangfang Wang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei. China
| | - Qianqian Han
- Institutes of Physical Science and Information Technology, Anhui University, Hefei. China
| | - Changzhi Xu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei. China
| | - Buchang Zhang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei. China
| |
Collapse
|
10
|
Zhang L, Xu H, Cheng Z, Wei Y, Sun R, Liang Z, Hu Y, Zhao L, Lian X, Li X, Huang D. Human Cancer Cell Membrane-Cloaked Fe 3O 4 Nanocubes for Homologous Targeting Improvement. J Phys Chem B 2021; 125:7417-7426. [PMID: 34185527 DOI: 10.1021/acs.jpcb.1c04383] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Surface modification of nanoparticles with cellular protein components is a new biomimetic modification strategy, which utilizes the inherent affinity between homologous cells to introduce the same surface molecules into nanoparticles to improve the targeting performance. In this study, oleic acid (OA)-coated Fe3O4 nanocubes were prepared by a high-temperature thermal decomposition method and modified by 3, 4-dihydroxyphenylpropionic acid (DHCA); then, HeLa cell membranes were introduced onto the surface of the nanocubes through mixed coextrusion to try to endow them with the targeting function of natural cells. The results show that the prepared Fe3O4 nanocubes have high monodispersity, excellent water solubility, and biocompatibility. Moreover, the Fe3O4 nanocubes encapsulated by cellular protein show an obvious core-shell structure and the specific targeting property to HeLa cells is improved significantly, which is expected to be used in clinical targeted diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Lichuang Zhang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Hao Xu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Ziyi Cheng
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Ruize Sun
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Ziwei Liang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Yinchun Hu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Xia Li
- College of Mechanical and Vehicle Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| |
Collapse
|
11
|
Nanomedicines functionalized with anti-EGFR ligands for active targeting in cancer therapy: Biological strategy, design and quality control. Int J Pharm 2021; 605:120795. [PMID: 34119579 DOI: 10.1016/j.ijpharm.2021.120795] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.
Collapse
|
12
|
Sugita S, Yamato M, Hatabu T, Kataoka Y. Involvement of cancer-derived EMT cells in the accumulation of 18F-fluorodeoxyglucose in the hypoxic cancer microenvironment. Sci Rep 2021; 11:9668. [PMID: 33994540 PMCID: PMC8126561 DOI: 10.1038/s41598-021-88414-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
A high rate of glycolysis, one of the most common features of cancer, is used in positron emission tomography (PET) imaging to visualize tumor tissues using 18F-fluorodeoxyglucose (18F-FDG). Heterogeneous intratumoral distribution of 18F-FDG in tissues has been established in some types of cancer, and the maximum standardized uptake value (SUVmax) has been correlated with poor prognosis. However, the phenotype of cells that show high 18F-FDG accumulation in tumors remains unknown. Here, we combined quantitative micro-autoradiography with fluorescence immunohistochemistry to simultaneously visualize 18F-FDG distribution, the expression of multiple proteins, and hypoxic regions in the cancer microenvironment of a human A431 xenograft tumor in C.B-17/Icr-scid/scid mice. We found that the highest 18F-FDG accumulation was in cancer-derived cells undergoing epithelial-mesenchymal transition (EMT) in hypoxic regions, implicating these regions as a major contributor to increased glucose metabolism, as measured by 18F-FDG-PET.
Collapse
Affiliation(s)
- Sachi Sugita
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Okayama, 700-8530, Japan.,Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Masanori Yamato
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Toshimitsu Hatabu
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Okayama, 700-8530, Japan
| | - Yosky Kataoka
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan. .,Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
13
|
Wang HC, Hung HC, Huang PN, Kung YA, Tseng SN, Wang YM, Shih SR, Tsu-An Hsu J. Engineering a novel IgG-like bispecific antibody against enterovirus A71. Biochem Biophys Rep 2020; 24:100860. [PMID: 34095549 PMCID: PMC8164134 DOI: 10.1016/j.bbrep.2020.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/02/2020] [Accepted: 11/05/2020] [Indexed: 11/12/2022] Open
Abstract
Frequent outbreaks of enterovirus A71 (EVA71) occur in the Asia-Pacific area, and these are closely associated with severe neurological symptoms in young children. No effective antiviral therapy is currently available for the treatment of EVA71 infection. The development of monoclonal antibodies (mAbs) has demonstrated promise as a novel therapy for the prevention and treatment of infectious diseases. Several medical conditions have been treated using bispecific or multi-specific antibodies that recognize two or more distinct epitopes simultaneously. However, bispecific or multi-specific antibodies often encounter protein expression and product stability problems. In this study, we developed an IgG-like bispecific antibody (E18-F1) comprising two anti-EVA71 antibodies: E18 mAb and llama-derived F1 single-domain antibody. E18-F1 was demonstrated to exhibit superior binding affinity and antiviral activity compared with E18 or F1. Additionally, E18-F1 not only improved survival rate, but also reduced clinical signs in human SCARB2 receptor (hSCARB2) transgenic mice challenged with a lethal dose of EVA71. Altogether, our results reveal that E18-F1 is a simple format bispecific antibody with promising antiviral activity for EVA71.
Collapse
Affiliation(s)
- Hsiang-Ching Wang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Hui-Chen Hung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Peng-Nien Huang
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
- Division of Infectious Diseases, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-An Kung
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| | - Sung-Nien Tseng
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - John Tsu-An Hsu
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
14
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
15
|
Cheng YA, Wu TH, Wang YM, Cheng TL, Chen IJ, Lu YC, Chuang KH, Wang CK, Chen CY, Lin RA, Chen HJ, Liao TY, Liu ES, Chen FM. Humanized bispecific antibody (mPEG × HER2) rapidly confers PEGylated nanoparticles tumor specificity for multimodality imaging in breast cancer. J Nanobiotechnology 2020; 18:118. [PMID: 32854720 PMCID: PMC7457265 DOI: 10.1186/s12951-020-00680-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Developing a universal strategy to improve the specificity and sensitivity of PEGylated nanoaparticles (PEG-NPs) for assisting in the diagnosis of tumors is important in multimodality imaging. Here, we developed the anti-methoxypolyethylene glycol (mPEG) bispecific antibody (BsAb; mPEG × HER2), which has dual specificity for mPEG and human epidermal growth factor receptor 2 (HER2), with a diverse array of PEG-NPs to confer nanoparticles with HER2 specificity and stronger intensity. Result We used a one-step formulation to rapidly modify the nanoprobes with mPEG × HER2 and optimized the modified ratio of BsAbs on several PEG-NPs (Lipo-DiR, SPIO, Qdot and AuNP). The αHER2/PEG-NPs could specifically target MCF7/HER2 cells (HER2++) but not MCF7/neo1 cells (HER2+/−). The αHER2/Lipo-DiR and αHER2/SPIO could enhance the sensitivity of untargeted PEG-NPs on MCF7/HER2 (HER2++). In in vivo imaging, αHER2/Lipo-DiR and αHER2/SPIO increased the specific targeting and enhanced PEG-NPs accumulation at 175% and 187% on 24 h, respectively, in HER2-overexpressing tumors. Conclusion mPEG × HER2, therefore, provided a simple one-step formulation to confer HER2-specific targeting and enhanced sensitivity and contrast intensity on HER2 positive tumors for multimodality imaging. ![]()
Collapse
Affiliation(s)
- Yi-An Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Tung-Ho Wu
- Cardiovascular Division of Surgical Department, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd, Zuoying Dist, Kaohsiung, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, No. 1001, University Road, Hsinchu, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - I-Ju Chen
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Yun-Chi Lu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, No.172-1, Sec. 2, Keelung Rd, Taipei, Taiwan
| | - Chih-Kuang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Chiao-Yun Chen
- Department of Radiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan
| | - Rui-An Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Huei-Jen Chen
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Tzu-Yi Liao
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Fang-Ming Chen
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan. .,Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, No.68, Jhonghua 3rd Rd, Cianjin District, Kaohsiung, Taiwan. .,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan. .,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.
| |
Collapse
|
16
|
Huang C, Li H, Feng Y, Li X, Zhang Z, Jiang C, Wang J, Yang C, Fu Y, Mu M, Zhao S, Wang Z, Kuang Y, Hou H, Wang Y, Guo W, Xu J, Yang H, Zhou L, Tong A, Guo G. Combination therapy with B7H3-redirected bispecific antibody and Sorafenib elicits enhanced synergistic antitumor efficacy. Theranostics 2020; 10:10498-10512. [PMID: 32929362 PMCID: PMC7482810 DOI: 10.7150/thno.49480] [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: 06/14/2020] [Accepted: 08/05/2020] [Indexed: 02/05/2023] Open
Abstract
Rationale: Current traditional treatment options are frequently ineffective to fight against ovarian cancer due to late diagnosis and high recurrence. Therefore, there is a vital need for the development of novel therapeutic agents. B7H3, an immune checkpoint protein, is highly expressed in various cancers, representing it a promising target for cancer immunotherapy. Although targeting B7H3 by bispecific T cell-engaging antibodies (BiTE) has achieved successes in hematological malignancies during recent years, attempts to use them for the treatment of solid cancers are less favorable, in part due to the heterogeneity of tumors. Sorafenib is an unselective inhibitor of multiple kinases currently being tested in clinical trials for several tumors, including ovarian cancer which showed limited activity and inevitable side effect for ovarian cancer treatment. However, it is able to enhance antitumor immune response, which indicates sorafenib may improve the efficiency of immunotherapy. Methods: We evaluated the expression of B7H3 in ovarian cancer using online database and validated its expression of tumor tissues by immunohistochemistry staining. Then, B7H3 expression and the effects of sorafenib on ovarian cancer cell lines were determined by flow cytometry. In addition, 2D and 3D ovarian cancer models were established to test the combined therapeutic effect in vitro. Finally, the efficiency of B7H3×CD3 BiTE alone and its combination with sorafenib were evaluated both in vitro and in vivo. Results: Our data showed that B7H3 was highly expressed in ovarian cancer compared with normal samples. Treatment with sorafenib inhibited ovarian cancer cell proliferation and induced a noticeable upregulation of B7H3 expression level. Further study suggested that B7H3×CD3 BiTE was effective in mediating T cell killing to cancer cells. Combined treatment of sorafenib and B7H3×CD3 BiTE had synergistic anti-tumor effects in ovarian cancer models. Conclusions: Overall, our study indicates that combination therapy with sorafenib and B7H3×CD3 BiTE may be a new therapeutic option for the further study of preclinical treatment of OC.
Collapse
|
17
|
Zhao S, Yu X, Qian Y, Chen W, Shen J. Multifunctional magnetic iron oxide nanoparticles: an advanced platform for cancer theranostics. Theranostics 2020; 10:6278-6309. [PMID: 32483453 PMCID: PMC7255022 DOI: 10.7150/thno.42564] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Multifunctional magnetic nanoparticles and derivative nanocomposites have aroused great concern for multimode imaging and cancer synergistic therapies in recent years. Among the rest, functional magnetic iron oxide nanoparticles (Fe3O4 NPs) have shown great potential as an advanced platform because of their inherent magnetic resonance imaging (MRI), biocatalytic activity (nanozyme), magnetic hyperthermia treatment (MHT), photo-responsive therapy and drug delivery for chemotherapy and gene therapy. Magnetic Fe3O4 NPs can be synthesized through several methods and easily surface modified with biocompatible materials or active targeting moieties. The MRI capacity could be appropriately modulated to induce response between T1 and T2 modes by controlling the size distribution of Fe3O4 NPs. Besides, small-size nanoparticles are also desired due to the enhanced permeation and retention (EPR) effect, thus the imaging and therapeutic efficiency of Fe3O4 NP-based platforms can be further improved. Here, we firstly retrospect the typical synthesis and surface modification methods of magnetic Fe3O4 NPs. Then, the latest biomedical application including responsive MRI, multimodal imaging, nanozyme, MHT, photo-responsive therapy and drug delivery, the mechanism of corresponding treatments and cooperation therapeutics of multifunctional Fe3O4 NPs are also be explained. Finally, we also outline a brief discussion and perspective on the possibility of further clinical translations of these multifunctional nanomaterials. This review would provide a comprehensive reference for readers to understand the multifunctional Fe3O4 NPs in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Shengzhe Zhao
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 32500, China
- State Key Lab of Metal Matrix Composites, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xujiang Yu
- State Key Lab of Metal Matrix Composites, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuna Qian
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 32500, China
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Jianliang Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 32500, China
| |
Collapse
|
18
|
Palanisamy S, Wang YM. Superparamagnetic iron oxide nanoparticulate system: synthesis, targeting, drug delivery and therapy in cancer. Dalton Trans 2019; 48:9490-9515. [PMID: 31211303 DOI: 10.1039/c9dt00459a] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer is a global epidemic and is considered a leading cause of death. Various cancer treatments such as chemotherapy, surgery, and radiotherapy are available for the cure but those are generally associated with poor long-term survival rates. Consequently, more advanced and selective methods that have better outcomes, fewer side effects, and high efficacies are highly in demand. Among these is the use of superparamagnetic iron oxide nanoparticles (SPIONs) which act as an innovative kit for battling cancer. Low cost, magnetic properties and toxicity properties enable SPIONs to be widely utilized in biomedical applications. For example, magnetite and maghemite (Fe3O4 and γ-Fe2O3) exhibit superparamagnetic properties and are widely used in drug delivery, diagnosis, and therapy. These materials are termed SPIONs when their size is smaller than 20 nm. This review article aims to provide a brief introduction on SPIONs, focusing on their fundamental magnetism and biological applications. The quality and surface chemistry of SPIONs are crucial in biomedical applications; therefore an in-depth survey of synthetic approaches and surface modifications of SPIONs is provided along with their biological applications such as targeting, site-specific drug delivery and therapy.
Collapse
Affiliation(s)
- Sathyadevi Palanisamy
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, 75 Bo-Ai Street, Hsinchu 300, Taiwan.
| | | |
Collapse
|
19
|
Chen L, Wu Y, Wu H, Li J, Xie J, Zang F, Ma M, Gu N, Zhang Y. Magnetic targeting combined with active targeting of dual-ligand iron oxide nanoprobes to promote the penetration depth in tumors for effective magnetic resonance imaging and hyperthermia. Acta Biomater 2019; 96:491-504. [PMID: 31302299 DOI: 10.1016/j.actbio.2019.07.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 06/28/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022]
Abstract
The combination of multi-targeting magnetic nanoprobes and multi-targeting strategies has potential to facilitate magnetic resonance imaging (MRI) and magnetic induction hyperthermia of the tumor. Although the thermo-agents based on magnetic iron oxide nanoparticles (MION) have been successfully used in the form of intratumoral injection in clinical cure of glioblastoma, the tumor-targeted thermotherapy by intravenous administration remains challenging. Herein, we constructed a c(RGDyK)- and d-glucosamine-grafted bispecific molecular nanoprobe (Fe3O4@RGD@GLU) with a magnetic iron oxide core of size 22.17 nm and a biocompatible shell of DSPE-PEG2000, which can specially target the tumor vessel and cancer cells. The selection of c(RGDyK) could make the nanoprobe enter the neovascularization endotheliocyte through αvβ3-mediated endocytosis, which drastically reduced the dependence on the enhanced permeability and retention (EPR) effect in tumor. This dual-ligand nanoprobe exhibited strong magnetic properties and favorable biocompatibility. In vitro studies confirmed the anti-phagocytosis ability against macrophages and the specific targeting capability of Fe3O4@RGD@GLU. Then, the imaging effect and anti-tumor efficacy were compared using different targeting strategies with untargeted nanoprobes, dual-targeted nanoprobes, and magnetic targeting combined with dual-targeted nanoprobes. Moreover, the combination strategy of magnetic targeting and active targeting promoted the penetration depth of nanoprobes in addition to the increased accumulation in tumor tissue. Thus, the dual-targeted magnetic nanoprobe together with the combined targeting strategy could be a promising method in tumor imaging and hyperthermia through in vivo delivery of theranostic agents. STATEMENT OF SIGNIFICANCE: Magnetic induction hyperthermia based on iron oxide nanoparticles has been used in clinic for adjuvant treatment of recurrent glioblastoma. Nonetheless, this application is limited to intratumoral injection, and tumor-targeted hyperthermia by intravenous injection remains challenging. In this study, we developed a multi-targeted strategy by combining magnetic targeting with active targeting of dual-ligand magnetic nanoprobes. This combination mode acquired optimum contrast imaging effect through MRI and tumor-suppressive effect through hyperthermia under an alternating current magnetic field. The design of the nanoprobe was suitable for targeting most tumor lesions, which enabled it to be an effective theranostic agent with extensive uses. This study showed significant enhancement of the penetration depth and accumulation of nanoprobes in the tumor tissue for efficient imaging and hyperthermia.
Collapse
|
20
|
Farahavar G, Abolmaali SS, Gholijani N, Nejatollahi F. Antibody-guided nanomedicines as novel breakthrough therapeutic, diagnostic and theranostic tools. Biomater Sci 2019; 7:4000-4016. [PMID: 31355391 DOI: 10.1039/c9bm00931k] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances in nanotechnology, such as the development of various types of nanoparticles and hybrid nanomaterials, have revolutionized nanomedicine. The small size, customizable surface, enhanced solubility, and multi-functionality endow the nanoparticles with an ability to interact with complex cellular and biological functions in new ways. Furthermore, these systems can deliver drugs to specific tissues and provide a targeted therapy. For this purpose, different categories of molecules, particularly antibodies, have been used as ligands. Antibody-conjugated nanomaterials can significantly enhance the efficiency of nanomedicines, especially in the field of cancer. This review is focused on three major medical applications of antibody-conjugated nanomaterials, namely, therapeutic, diagnostic and theranostic applications. To provide comprehensive information on the topic and an overview of these hybrid nanomaterials for biomedical applications, a brief summary of nanomaterials and antibodies is given. Moreover, the review has depicted the potential applications of antibody-conjugated nanomaterials in different fields and their capabilities to empower nanomedicine, particularly in relation to the treatment and detection of malignancies.
Collapse
Affiliation(s)
- Ghazal Farahavar
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS research center, Institute of health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
21
|
Patout M, Guisier F, Brune X, Bohn P, Romieu A, Sarafan-Vasseur N, Sesboüé R, Renard PY, Thiberville L, Salaün M. Real-time molecular optical micro-imaging of EGFR mutations using a fluorescent erlotinib based tracer. BMC Pulm Med 2019; 19:3. [PMID: 30612556 PMCID: PMC6322267 DOI: 10.1186/s12890-018-0760-z] [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: 12/14/2017] [Accepted: 12/06/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND EGFR mutations are routinely explored in lung adenocarcinoma by sequencing tumoral DNA. The aim of this study was to evaluate a fluorescent-labelled erlotinib based theranostic agent for the molecular imaging of mutated EGFR tumours in vitro and ex vivo using a mice xenograft model and fibred confocal fluorescence microscopy (FCFM). METHODS The fluorescent tracer was synthesized in our laboratory by addition of fluorescein to an erlotinib molecule. Three human adenocarcinoma cell lines with mutated EGFR (HCC827, H1975 and H1650) and one with wild-type EGFR (A549) were xenografted on 35 Nude mice. MTT viability assay was performed after exposure to our tracer. In vitro imaging was performed at 1 μM tracer solution, and ex vivo imaging was performed on fresh tumours excised from mice and exposed to a 1 μM tracer solution in PBS for 1 h. Real-time molecular imaging was performed using FCFM and median fluorescence intensity (MFI) was recorded for each experiment. RESULTS MTT viability assay confirmed that addition of fluorescein to erlotinib did not suppress the cytotoxic of erlotinib on tumoral cells. In vitro FCFM imaging showed that our tracer was able to distinguish cell lines with mutated EGFR from those lines with wild-type EGFR (p < 0.001). Ex vivo FCFM imaging of xenografts with mutated EGFR had a significantly higher MFI than wild-type (p < 0.001). At a cut-off value of 354 Arbitrary Units, MFI of our tracer had a sensitivity of 100% and a specificity of 96.3% for identifying mutated EGFR tumours. CONCLUSION Real time molecular imaging using fluorescent erlotinib is able to identify ex vivo tumours with EGFR mutations.
Collapse
Affiliation(s)
- Maxime Patout
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France. .,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France. .,Service de Pneumologie, Oncologie Thoracique, Soins Intensifs Respiratoires, CHU de Rouen, 1 rue de Germont, 76031, Rouen Cedex, France.
| | - Florian Guisier
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France.,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France
| | - Xavier Brune
- Normandie University, COBRA, UMR 6014 & FR 3038; CNRS, F-76000, Rouen, France
| | - Pierre Bohn
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France
| | - Anthony Romieu
- Normandie University, COBRA, UMR 6014 & FR 3038; CNRS, F-76000, Rouen, France.,Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, University, Bourgogne Franche-Comté, 21078, Dijon, France
| | - Nasrin Sarafan-Vasseur
- Génétique du cancer et des maladies neuropsychiatriques, Normandie University, UNIROUEN, INSERM, F-76000, Rouen, France
| | - Richard Sesboüé
- Génétique du cancer et des maladies neuropsychiatriques, Normandie University, UNIROUEN, INSERM, F-76000, Rouen, France
| | - Pierre-Yves Renard
- Normandie University, COBRA, UMR 6014 & FR 3038; CNRS, F-76000, Rouen, France
| | - Luc Thiberville
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France.,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France
| | - Mathieu Salaün
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France.,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France
| |
Collapse
|
22
|
Gokduman K, Bestepe F, Li L, Yarmush ML, Usta OB. Dose-, treatment- and time-dependent toxicity of superparamagnetic iron oxide nanoparticles on primary rat hepatocytes. Nanomedicine (Lond) 2018; 13:1267-1284. [PMID: 29949471 PMCID: PMC6219434 DOI: 10.2217/nnm-2017-0387] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
AIM As a first study in literature, to investigate concentration-dependent (0-400 μg/ml) and exposure-dependent (single dosing vs cumulative dosing) effects of superparamagnetic iron oxide nanoparticles (d = 10 nm) on primary rat hepatocytes in a time-dependent manner. MATERIALS & METHODS Sandwich-cultured hepatocyte model was used to evaluate viability, hepatocyte specific functions and reactive oxygen species level. RESULTS In terms of all parameters, generally statistically more significant effects were observed in a concentration- and time-dependent manner. In terms of hepatocyte-specific functions, cumulative dosing caused significantly (p < 0.05) more deleterious effects at 48th hour. CONCLUSION A combination of various biomarkers should be employed for the evaluation of the effect of superparamagnetic iron oxide nanoparticles on liver, and each biomarker should be analyzed in a time- and exposure-dependent manner.
Collapse
Affiliation(s)
- Kurtulus Gokduman
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
| | - Furkan Bestepe
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
- School of Medicine, Ankara University, Ankara 06100, Turkey
| | - Lei Li
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
- Key Laboratory of Cryogenics, Technical Institute of Physics & Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
- Department of Biomedical Engineering, Rutgers State University, Piscataway, NJ 08854, USA
| | - O Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
| |
Collapse
|
23
|
Magnetic Nanoparticles Conjugated with Peptides Derived from Monocyte Chemoattractant Protein-1 as a Tool for Targeting Atherosclerosis. Pharmaceutics 2018; 10:pharmaceutics10020062. [PMID: 29795012 PMCID: PMC6027309 DOI: 10.3390/pharmaceutics10020062] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is a multifactorial inflammatory disease that may progress silently for long period, and it is also widely accepted as the main cause of cardiovascular diseases. To prevent atherosclerotic plaques from generating, imaging early molecular markers and quantifying the extent of disease progression are desired. During inflammation, circulating monocytes leave the bloodstream and migrate into incipient lipid accumulation in the artery wall, following conditioning by local growth factors and proinflammatory cytokines; therefore, monocyte accumulation in the arterial wall can be observed in fatty streaks, rupture-prone plaques, and experimental atherosclerosis. In this work, we synthesized monocyte-targeting iron oxide magnetic nanoparticles (MNPs), which were incorporated with the peptides derived from the chemokine receptor C-C chemokine receptor type 2 (CCR2)-binding motif of monocytes chemoattractant protein-1 (MCP-1) as a diagnostic tool for potential atherosclerosis. MCP-1-motif MNPs co-localized with monocytes in in vitro fluorescence imaging. In addition, with MNPs injection in ApoE knockout mice (ApoE KO mice), the well-characterized animal model of atherosclerosis, MNPs were found in specific organs or regions which had monocytes accumulation, especially the aorta of atherosclerosis model mice, through in vivo imaging system (IVIS) imaging and magnetic resonance imaging (MRI). We also performed Oil Red O staining and Prussian Blue staining to confirm the co-localization of MCP-1-motif MNPs and atherosclerosis. The results showed the promising potential of MCP-1-motif MNPs as a diagnostic agent of atherosclerosis.
Collapse
|
24
|
Moradi-Kalbolandi S, Hosseinzade A, Salehi M, Merikhian P, Farahmand L. Monoclonal antibody-based therapeutics, targeting the epidermal growth factor receptor family: from herceptin to Pan HER. J Pharm Pharmacol 2018; 70:841-854. [DOI: 10.1111/jphp.12911] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/25/2018] [Indexed: 12/30/2022]
Abstract
Abstract
Objectives
Monoclonal antibody-based of cancer therapy has been considered as one of the most successful therapeutic strategies for both haematologic malignancies and solid tumours in the last two decades. Epidermal growth factor receptor (EGFR) family signalling pathways play a key role in the regulation of cell proliferation, survival and differentiation. Hence, anti-EGFR family mAbs is one of the most promising approaches in cancer therapy.
Key findings
Here, recent advances in anti-EGFR mAb including approved or successfully tested in preclinical and clinical studies have been reviewed. Although we focus on monoclonal antibodies against the EGF receptor, but the mechanisms underlying the effects of EGFR-specific mAb in cancer therapy, to some extend the resistance to existing anti-EGFR therapies and some therapeutic strategies to overcome resistance such as combination of mAbs on different pathways are briefly discussed as well.
Summary
The EGFR family receptors, is considered as an attractive target for mAb development to inhibit their consecutive activities in tumour growth and resistance. However, due to resistance mechanisms, the combination therapies may become a good candidate for targeting EGFR family receptors.
Collapse
Affiliation(s)
- Shima Moradi-Kalbolandi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Aysooda Hosseinzade
- 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
| | - Parnaz Merikhian
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
25
|
Chen L, Xie J, Wu H, Zang F, Ma M, Hua Z, Gu N, Zhang Y. Improving sensitivity of magnetic resonance imaging by using a dual-targeted magnetic iron oxide nanoprobe. Colloids Surf B Biointerfaces 2018; 161:339-346. [DOI: 10.1016/j.colsurfb.2017.10.059] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/19/2017] [Accepted: 10/21/2017] [Indexed: 12/11/2022]
|
26
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
27
|
New anti-HER2 agents: from second-generation tyrosine kinases inhibitors to bifunctional antibodies. Curr Opin Oncol 2017; 29:405-410. [DOI: 10.1097/cco.0000000000000412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
28
|
|
29
|
Xiang Z, Yang X, Xu J, Lai W, Wang Z, Hu Z, Tian J, Geng L, Fang Q. Tumor detection using magnetosome nanoparticles functionalized with a newly screened EGFR/HER2 targeting peptide. Biomaterials 2016; 115:53-64. [PMID: 27888699 DOI: 10.1016/j.biomaterials.2016.11.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/19/2023]
Abstract
A novel peptide (P75) targeting EGFR and HER2 is successfully screened from a one-bead-one-compound (OBOC) library containing approximately 2 × 105 peptides built with the aid of computational simulation. In vitro and in vivo analyses show that P75 binds to human epithelial growth factor receptor (EGFR) with nanomolar affinity and to epithelial growth factor receptor-2 (HER2) with a lower affinity but comparable to other reported peptides. The peptide is used to modify the surface of magnetosome nanoparticles (NPs) for targeted magnetic resonance imaging (MRI). In vitro and in vivo fluorescence imaging results suggest peptide P75 modified magnetosomes (Mag-P75) specifically bind to MDA-MB-468 and SKBR3 cells as well as xenograft tumors with surprisingly low accumulation in other organs including liver and kidney. In vivo T2-weighted MR imaging studies of the xenograft tumors from SKBR3 and MDA-MB-468 cells show obviously negative contrast enhancement. The high affinity and specificity of P75 to EGFR and HER2 positive tumors, together with the success of peptide functionalized magnetosome NPs for targeted MRI demonstrate the potential of this peptide being used in the EGFR and HER2 positive tumors diagnosis and therapy.
Collapse
Affiliation(s)
- Zhichu Xiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Beijing 101408, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoliang Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junjie Xu
- State Key Laboratories for Agro-biotechnology and College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Wenjia Lai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Zihua Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Jiesheng Tian
- State Key Laboratories for Agro-biotechnology and College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Lingling Geng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Qiaojun Fang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| |
Collapse
|
30
|
Differential structured illumination microendoscopy for in vivo imaging of molecular contrast agents. Proc Natl Acad Sci U S A 2016; 113:10769-73. [PMID: 27621464 DOI: 10.1073/pnas.1613497113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Fiber optic microendoscopy has shown promise for visualization of molecular contrast agents used to study disease in vivo. However, fiber optic microendoscopes have limited optical sectioning capability, and image contrast is limited by out-of-focus light generated in highly scattering tissue. Optical sectioning techniques have been used in microendoscopes to remove out-of-focus light but reduce imaging speed or rely on bulky optical elements that prevent in vivo imaging. Here, we present differential structured illumination microendoscopy (DSIMe), a fiber optic system that can perform structured illumination in real time for optical sectioning without any opto-mechanical components attached to the distal tip of the fiber bundle. We demonstrate the use of DSIMe during in vivo fluorescence imaging in patients undergoing surgery for cervical adenocarcinoma in situ. Images acquired using DSIMe show greater contrast than standard microendoscopy, improving the ability to detect cellular atypia associated with neoplasia.
Collapse
|