1
|
Saraei P, Ghasemi M, Talebi A, Vafaeinezhad A, Saberzadeh J. Nutritional Strategies in Oncology: A Narrative Review of Advances in Folate-Targeted Therapeutic Approaches for Cancer Treatment. Nutr Cancer 2025:1-23. [PMID: 40295145 DOI: 10.1080/01635581.2025.2497096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Folate, a water-soluble B vitamin crucial for DNA synthesis and repair, is internalized by cells through specific folate receptors (FRs), which are frequently overexpressed in various types of cancers. In this comprehensive study, we conducted a review of the literature from Google Scholar, PubMed, and Science Direct, focusing on research published between 1980 and 2024 to evaluate folate-targeted therapeutic strategies in oncology. Our study design involved a rigorous review of both preclinical and clinical research, emphasizing strategies such as folate-drug conjugates, antibody-drug conjugates, and folate-targeted nanoparticles. Key findings indicate that targeting FRs in cancers such as ovarian, breast, cervical, renal, and colorectal enhances drug delivery specificity to tumors, increases therapeutic efficacy, and decreases systemic toxicity compared to traditional chemotherapy. Several clinical trials reported improved progression-free survival and overall response rates among patients receiving folate-targeted therapies. In conclusion, our review highlights the significant potential of folate-targeted strategies in advancing precision oncology while these approaches provide substantial benefits in terms of efficacy and safety, further research is essential to refine drug design and expand clinical applications. Such initiatives will facilitate the development of more personalized cancer treatment protocols that maximize therapeutic outcomes while minimizing adverse effects.
Collapse
Affiliation(s)
- Pouya Saraei
- Student Research Committee, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Morteza Ghasemi
- Comprehensive Medical Research Center, Center for Basic Medical Sciences, Physiology Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Athar Talebi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Arefe Vafaeinezhad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Jamileh Saberzadeh
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Sahane P, Puri N, Khairnar P, Phatale V, Shukla S, Priyadarshinee A, Srivastava S. Harnessing Folate Receptors: A Comprehensive Review on the Applications of Folate-Adorned Nanocarriers for the Management of Melanoma. ACS APPLIED BIO MATERIALS 2025. [PMID: 40275606 DOI: 10.1021/acsabm.5c00077] [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: 04/26/2025]
Abstract
The advancement in exclusively tailored therapeutic delivery systems has escalated a great deal of interest in targeted delivery to augment therapeutic efficacy and to lessen adverse effects. The targeted delivery approach promisingly helps to surmount the unmet clinical needs of conventional therapies, including chemoresistance, limited penetration, and side effects. In the case of melanoma, various receptors were overexpressed on the tumor site, among which folate receptor (FR) targeting is considered to be a progressive approach for managing melanoma. FRs are the macromolecules of the glycosyl phosphatidylinositol-attached protein that possess globular assembly with a greater affinity toward specific ligands. So, the functional ligands can be utilized to design targeted nanocarriers (NCs) that can effectively bind to overexpressed FRs. Hence, folate-adorned NCs (FNCs) offer various benefits such as site-specific targeting, cargo protection, and minimizing toxicity. This review focuses on the insights and implications of FRs, targeting FRs, and mechanisms, challenges, and advantages of FNCs. Further, the applications of various FNCs, such as liposomes, polymeric NCs, albumin nanoparticles, inorganic NCs, liquid crystalline nanoparticles, and nanogels, have been elaborated for melanoma therapy. Likewise, the potential of FNCs in immunotherapy, photodynamic therapy, chemotherapy, gene therapy, photothermal therapy, and tumor imaging has been exhaustively discussed. Furthermore, translational hurdles and potential solutions are discussed in detail. The present review is expected to give thoughtful ideas to researchers, industry stakeholders, and formulation scientists for the efficacious development of FNCs.
Collapse
Affiliation(s)
- Prajakta Sahane
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Niharika Puri
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Abhipsa Priyadarshinee
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| |
Collapse
|
3
|
Zhang L, Zhou J, Obianwuna UE, Long C, Qiu K, Zhang H, Qi X, Wu S. Optimizing selenium-enriched yeast supplementation in laying hens: Enhancing egg quality, selenium concentration in eggs, antioxidant defense, and liver health. Poult Sci 2025; 104:104584. [PMID: 39615326 PMCID: PMC11648770 DOI: 10.1016/j.psj.2024.104584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025] Open
Abstract
This study evaluated the effects of selenium-enriched yeast (SY) supplementation at various levels on health and production parameters in laying hens, including egg production, egg quality, selenium (Se) concentrations in eggs, liver health, serum biochemical markers, antioxidant function, and immune responses. A total of 360 Hy-Line Brown hens (28 weeks old) were randomly assigned to four dietary groups with six replicates of 15 birds each, monitored over a 12-week feeding trial after a two-week acclimatization period. The dietary groups included a control (basal diet without selenium) and three SY-supplemented groups with Se levels of 0.3 mg/kg (SY03), 1.5 mg/kg (SY15), and 6.0 mg/kg (SY60). The results showed no significant effects of dietary SY on laying performance or feed efficiency (P > 0.05). However, the SY15 group showed significant improvements in egg quality, particularly in albumen height, Haugh Unit and yolk color (P < 0.05). Selenium concentrations in eggs, albumen, and yolk increased dose-dependently, with significant differences in the SY-supplemented groups (P < 0.001). Increased activities of liver enzymes including alanine transaminase, alkaline phosphatase, and aspartate transaminase, alongside elevated levels of uric acid were notable in the SY60 group (P < 0.05). In addition, histological analysis revealed significant hepatocyte degeneration and a higher liver organ index (P < 0.05), in the SY60 group. All of which suggests potential liver toxicity at higher selenium levels. Antioxidant capacity of the birds were significantly enhanced due to dietary supplementation of SY as indicated by increased serum levels of total antioxidant capacity, and activities of catalase, glutathione peroxidase, and superoxide dismutase (P < 0.05). Analysis of hepatic genes expression revealed that SY15 supplementation significantly upregulated key antioxidant-related genes (Nrf2, HO-1, CAT, and NQO1) and downregulated Keap1 expression (P < 0.05), suggesting strong activation of the antioxidant defense system. In conclusion, SY supplementation at 1.5 mg/kg improved egg quality, increased Se concentrations in eggs, and enhanced antioxidant capacity without affecting laying performance or liver health. This makes it a balanced approach to improving egg quality and poultry health. However, higher supplementation levels (6.0 mg/kg) resulted in liver damage, underscoring the importance of careful dosage consideration.
Collapse
Affiliation(s)
- Longfei Zhang
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture & Rural Affairs, Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards of the Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South St., Haidian District, Beijing 100081, China; College of Animal Science and Technology, Beijing Agricultural University, Beijing 100096, China
| | - Jianmin Zhou
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture & Rural Affairs, Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards of the Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South St., Haidian District, Beijing 100081, China.
| | - Uchechukwu Edna Obianwuna
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture & Rural Affairs, Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards of the Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South St., Haidian District, Beijing 100081, China
| | - Cheng Long
- College of Animal Science and Technology, Beijing Agricultural University, Beijing 100096, China
| | - Kai Qiu
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture & Rural Affairs, Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards of the Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South St., Haidian District, Beijing 100081, China
| | - Haijun Zhang
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture & Rural Affairs, Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards of the Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South St., Haidian District, Beijing 100081, China
| | - Xiaolong Qi
- College of Animal Science and Technology, Beijing Agricultural University, Beijing 100096, China
| | - Shugeng Wu
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture & Rural Affairs, Laboratory of Quality & Safety Risk Assessment for Animal Products on Feed Hazards of the Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, 12 Zhongguancun South St., Haidian District, Beijing 100081, China.
| |
Collapse
|
4
|
Chandra J, Nasir N, Wahab S, Sahebkar A, Kesharwani P. Harnessing the power of targeted metal nanocarriers mediated photodynamic and photothermal therapy. Nanomedicine (Lond) 2024:1-19. [PMID: 39545609 DOI: 10.1080/17435889.2024.2419820] [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: 04/25/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024] Open
Abstract
The treatment of cancer has become a profoundly intricate procedure. Traditional treatment methods, including chemotherapy, surgery and radiotherapy, have been utilized, while notable progress has been achieved in recent years. Among targeted therapies for cancer, folic acid (FA) conjugated metal-based nanoparticles (NP) have emerged as an innovative strategy, namely for photodynamic therapy (PDT) and photothermal therapy (PTT). These NP exploit the strong attraction between FA and folate receptors, which are excessively produced in several cancer cells, in order to enable precise administration and improved effectiveness of treatment. During PDT, metal-based NP functionalized with FA are used as photosensitizers which are activated by light, and produce reactive oxygen species that cause cancer cells to undergo apoptosis. Within the framework of PTT, these NP effectively transform light energy into concentrated heat, specifically targeting and destroying tumor cells. This review examines the fundamental mechanisms by which these NP improve the effectiveness of PDT and PTT while simultaneously presenting important findings that demonstrate the effectiveness of FA-functionalized MNP in laboratory and animal models. In addition, the paper also discusses the problems and potential directions for their clinical translation.
Collapse
Affiliation(s)
- Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazim Nasir
- Department of Basic Medical Sciences, College of Applied Medical Sciences, Khamis Mushait, King Khalid University, Abha, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 61421, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
5
|
Bukowski K, Rogalska A, Marczak A. Folate Receptor Alpha-A Secret Weapon in Ovarian Cancer Treatment? Int J Mol Sci 2024; 25:11927. [PMID: 39595996 PMCID: PMC11593442 DOI: 10.3390/ijms252211927] [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: 10/09/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy worldwide. Due to its nonspecific symptoms and unreliable screening tools, EOC is not diagnosed at an early stage in most cases. Unfortunately, despite achieving initial remission after debulking surgery and platinum-based chemotherapy, most patients experience the recurrence of the disease. The limited therapy approaches have encouraged scientists to search for new detection and therapeutic strategies. In this review, we discuss the role of folate receptor alpha (FRα) in EOC development and its potential application as a biomarker and molecular target in designing new EOC screening and treatment methods. We summarize the mechanisms of the action of various therapeutic strategies based on FRα, including MABs (monoclonal antibodies), ADCs (antibody-drug conjugates), FDCs (folate-drug conjugates), SMDCs (small molecule-drug conjugates), vaccines, and CAR-T (chimeric antigen receptor T) cells, and present the most significant clinical trials of some FRα-based drugs. Furthermore, we discuss the pros and cons of different FR-based therapies, highlighting mirvetuximab soravtansine (MIRV) as the currently most promising EOC-targeting drug.
Collapse
Affiliation(s)
- Karol Bukowski
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland; (A.R.); (A.M.)
| | | | | |
Collapse
|
6
|
Venkatesan J, Murugan D, Lakshminarayanan K, Smith AR, Vasanthakumari Thirumalaiswamy H, Kandhasamy H, Zender B, Zheng G, Rangasamy L. Powering up targeted protein degradation through active and passive tumour-targeting strategies: Current and future scopes. Pharmacol Ther 2024; 263:108725. [PMID: 39322067 DOI: 10.1016/j.pharmthera.2024.108725] [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: 06/01/2024] [Revised: 08/31/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a prominent and vital strategy for therapeutic intervention of cancers and other diseases. One such approach involves the exploration of proteolysis targeting chimeras (PROTACs) for the selective elimination of disease-causing proteins through the innate ubiquitin-proteasome pathway. Due to the unprecedented achievements of various PROTAC molecules in clinical trials, researchers have moved towards other physiological protein degradation approaches for the targeted degradation of abnormal proteins, including lysosome-targeting chimeras (LYTACs), autophagy-targeting chimeras (AUTACs), autophagosome-tethering compounds (ATTECs), molecular glue degraders, and other derivatives for their precise mode of action. Despite numerous advantages, these molecules face challenges in solubility, permeability, bioavailability, and potential off-target or on-target off-tissue effects. Thus, an urgent need arises to direct the action of these degrader molecules specifically against cancer cells, leaving the proteins of non-cancerous cells intact. Recent advancements in TPD have led to innovative delivery methods that ensure the degraders are delivered in a cell- or tissue-specific manner to achieve cell/tissue-selective degradation of target proteins. Such receptor-specific active delivery or nano-based passive delivery of the PROTACs could be achieved by conjugating them with targeting ligands (antibodies, aptamers, peptides, or small molecule ligands) or nano-based carriers. These techniques help to achieve precise delivery of PROTAC payloads to the target sites. Notably, the successful entry of a Degrader Antibody Conjugate (DAC), ORM-5029, into a phase 1 clinical trial underscores the therapeutic potential of these conjugates, including LYTAC-antibody conjugates (LACs) and aptamer-based targeted protein degraders. Further, using bispecific antibody-based degraders (AbTACs) and delivering the PROTAC pre-fused with E3 ligases provides a solution for cell type-specific protein degradation. Here, we highlighted the current advancements and challenges associated with developing new tumour-specific protein degrader approaches and summarized their potential as single agents or combination therapeutics for cancer.
Collapse
Affiliation(s)
- Janarthanan Venkatesan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India; School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Kalaiarasu Lakshminarayanan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Alexis R Smith
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Harashkumar Vasanthakumari Thirumalaiswamy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Hariprasath Kandhasamy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Boutheina Zender
- Department of Biomedical Engineering, Bahçeşehir University, Istanbul 34353, Turkey
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA.
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India.
| |
Collapse
|
7
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [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: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
8
|
Zhou Y, Li C, Chen X, Zhao Y, Liao Y, Huang P, Wu W, Nieto NS, Li L, Tang W. Development of folate receptor targeting chimeras for cancer selective degradation of extracellular proteins. Nat Commun 2024; 15:8695. [PMID: 39379374 PMCID: PMC11461649 DOI: 10.1038/s41467-024-52685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Targeted protein degradation has emerged as a novel therapeutic modality to treat human diseases by utilizing the cell's own disposal systems to remove protein target. Significant clinical benefits have been observed for degrading many intracellular proteins. Recently, the degradation of extracellular proteins in the lysosome has been developed. However, there have been limited successes in selectively degrading protein targets in disease-relevant cells or tissues, which would greatly enhance the development of precision medicine. Additionally, most degraders are not readily available due to their complexity. We report a class of easily accessible Folate Receptor TArgeting Chimeras (FRTACs) to recruit the folate receptor, primarily expressed on malignant cells, to degrade extracellular soluble and membrane cancer-related proteins in vitro and in vivo. Our results indicate that FRTAC is a general platform for developing more precise and effective chemical probes and therapeutics for the study and treatment of cancers.
Collapse
Affiliation(s)
- Yaxian Zhou
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Chunrong Li
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Xuankun Chen
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yuan Zhao
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yaxian Liao
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Penghsuan Huang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Wenxin Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Nicholas S Nieto
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Lingjun Li
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Weiping Tang
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
9
|
Gupta A, O'Cearbhaill RE, Block MS, Hamilton E, Konner JA, Knutson KL, Potts J, Garrett G, Kenney RT, Wenham RM. Vaccination with folate receptor-alpha peptides in patients with ovarian cancer following response to platinum-based therapy: A randomized, multicenter clinical trial. Gynecol Oncol 2024; 189:90-97. [PMID: 39068739 DOI: 10.1016/j.ygyno.2024.07.675] [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: 06/11/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Folate receptor alpha (FRα) is overexpressed on >90% of high-grade epithelial ovarian cancers (EOC). Targeting FRα with antibody-drug conjugates has proven utility in the platinum-resistant setting. It is also a potential therapeutic target for immuno-oncologic agents, such as peptide vaccines that work primarily via adaptive and humoral immunity. We tested the hypothesis that FRα peptide immunization could improve outcomes in patients with EOC following response to platinum-based therapy. METHODS We conducted a randomized, double-blind, multicenter, phase II study to evaluate the safety and efficacy of TPIV200 (a multi-epitope FRα peptide vaccine admixed with GM-CSF) versus GM-CSF alone in 120 women who did not have disease progression after at least 4 cycles of first-line platinum-based therapy. Patients were vaccinated intradermally once every 4 weeks up to 6 times, followed by a boosting period of 6 vaccinations at 12-week intervals. Primary endpoints included safety, tolerability, and progression free survival (PFS). RESULTS At study termination with a median follow-up of 15.2 months (range 1.2-28.4 months), 68 of 119 intention-to-treat patients had disease progression (55% in TPIV200 + GM-CSF arm and 59% in GM-CSF alone arm). The median PFS was 11.1 months (95% CI 8.3-16.6 months) with no significant difference between the treatment groups (10.9 months with TPIV200 + GM-CSF versus 11.1 months with GM-CSF, HR, 0.85; upper 90% CI 1.17]. No patient experienced a ≥ grade 3 drug-related adverse event. CONCLUSION TPIV200 was well tolerated but was not associated with improved PFS. Additional studies are required to uncover potential synergies using multiepitope vaccines targeting FRα. Trial Registration NLM/NCBI Registry, NCT02978222, https://clinicaltrials.gov/search?term=NCT02978222.
Collapse
Affiliation(s)
- Aditi Gupta
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Roisin E O'Cearbhaill
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| | | | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN, United States of America
| | - Jason A Konner
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| | | | - James Potts
- Veristat LLC, Southborough, MA, United States of America
| | - Gerald Garrett
- Marker Therapeutics, Inc., Houston, TX, United States of America
| | - Richard T Kenney
- Marker Therapeutics, Inc., Houston, TX, United States of America
| | - Robert M Wenham
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States of America.
| |
Collapse
|
10
|
Nisha R, Kumar P, Mishra N, Maurya P, Ahmad S, Singh N, Saraf SA. Appraisal of folate functionalized bosutinib cubosomes against hepatic cancer cells: In-vitro, In-silico, and in-vivo pharmacokinetic study. Int J Pharm 2024; 654:123975. [PMID: 38452833 DOI: 10.1016/j.ijpharm.2024.123975] [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: 11/07/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Targeted therapies enhance the efficacy of tumour screening and management while lowering side effects. Multiple tumours, including liver cancer, exhibit elevated levels of folate receptor expression. This research attempted to develop surface-functionalised bosutinib cubosomes against hepatocellular carcinoma. The novelty of this work is the anti-hepatic action of bosutinib (BST) and folic acid-modified bosutinib cubosomes (BSTMF) established through proto-oncogene tyrosine-protein kinase (SrC)/ focal adhesion kinase(FAK), reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and cell cytotoxicity. Later, the in-vivo pharmacokinetics of BSTMF were determined for the first time. The strong affinity of folic acid (FA) for folate receptors allows BSTMF to enter cells via FA receptor-mediated endocytosis. The particle size of the prepared BSTMF was 188.5 ± 2.25 nm, and its zeta potential was -20.19 ± 2.01 mV, an encapsulation efficiency of 90.31 ± 3.15 %, and a drug release rate of 76.70 ± 2.10 % for 48 h. The surface architecture of BSTMF was identified using transmission electron microscopy (TEM) and Atomic force microscopy (AFM). Cell-line studies demonstrated that BSTMF substantially lowered the viability of Hep G2 cells compared to BST and bosutinib-loaded cubosomes (BSTF). BSTMF demonstrated an elevated BST concentration in tumour tissue than in other organs and also displayed superior pharmacokinetics, implying that they hold potential against hepatic cancers. This is the first study to show that BSTMF may be effective against liver cancer by targeting folate receptors and triggering SrC/FAK-dependent apoptotic pathways. Multiple parameters demonstrated that BSTMF enhanced anticancer targeting, therapeutic efficacy, and safety in NDEA-induced hepatocellular carcinoma.
Collapse
Affiliation(s)
- Raquibun Nisha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; Department of Pharmacology, Institute of Pharmaceutical Sciences, University of Lucknow, Lucknow, 226031, India
| | - Nidhi Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Priyanka Maurya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Sector II, Dr Akhilesh Das Nagar, Faizabad Road, Lucknow, 226028, India
| | - Shakir Ahmad
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
| | - Neelu Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Shubhini A Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus: NIPER-Raebareli, Sarojini Nagar, Near CRPF Base Camp, Lucknow 226002, India.
| |
Collapse
|
11
|
Tavares V, Marques IS, Melo IGD, Assis J, Pereira D, Medeiros R. Paradigm Shift: A Comprehensive Review of Ovarian Cancer Management in an Era of Advancements. Int J Mol Sci 2024; 25:1845. [PMID: 38339123 PMCID: PMC10856127 DOI: 10.3390/ijms25031845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Ovarian cancer (OC) is the female genital malignancy with the highest lethality. Patients present a poor prognosis mainly due to the late clinical presentation allied with the common acquisition of chemoresistance and a high rate of tumour recurrence. Effective screening, accurate diagnosis, and personalised multidisciplinary treatments are crucial for improving patients' survival and quality of life. This comprehensive narrative review aims to describe the current knowledge on the aetiology, prevention, diagnosis, and treatment of OC, highlighting the latest significant advancements and future directions. Traditionally, OC treatment involves the combination of cytoreductive surgery and platinum-based chemotherapy. Although more therapeutical approaches have been developed, the lack of established predictive biomarkers to guide disease management has led to only marginal improvements in progression-free survival (PFS) while patients face an increasing level of toxicity. Fortunately, because of a better overall understanding of ovarian tumourigenesis and advancements in the disease's (epi)genetic and molecular profiling, a paradigm shift has emerged with the identification of new disease biomarkers and the proposal of targeted therapeutic approaches to postpone disease recurrence and decrease side effects, while increasing patients' survival. Despite this progress, several challenges in disease management, including disease heterogeneity and drug resistance, still need to be overcome.
Collapse
Affiliation(s)
- Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-072 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-072 Porto, Portugal
| | - Joana Assis
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology of Porto (IPOP), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-072 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
12
|
Kayki-Mutlu G, Aksoyalp ZS, Wojnowski L, Michel MC. A year in pharmacology: new drugs approved by the US Food and Drug Administration in 2022. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1619-1632. [PMID: 36951997 PMCID: PMC10034907 DOI: 10.1007/s00210-023-02465-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
While new drug approvals by the U.S. Food and Drug Administration (FDA) had remained stable or even increased in the first 2 years of the COVID-19 pandemic, the 37 newly approved drugs in 2022 are considerably less than the 53 and 50 new drugs approved in 2020 and 2021, respectively, and less than the rolling 10-year average of 43. As in previous years of this annual review, we assign these new drugs to one of three levels of innovation: first drug against a condition ("first-in-indication"), first drug using a novel molecular mechanism ("first-in-class"), and "next-in-class," i.e., a drug using an already exploited molecular mechanism. We identify two "first-in-indication" (ganaxolon and teplizumab), 20 (54%) "first-in-class," and 17 (46%) "next-in-class" drugs. By treatment area, rare diseases and cancer drugs were once again the most prevalent (partly overlapping) therapeutic areas. Other continuing trends were the use of accelerated regulatory approval pathways and the reliance on biopharmaceuticals (biologics).
Collapse
Affiliation(s)
- Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Zinnet Sevval Aksoyalp
- Department of Pharmacology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Leszek Wojnowski
- Department of Pharmacology, University Medical Center, Universitätsmedizin Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55118 Mainz, Germany
| | - Martin C. Michel
- Department of Pharmacology, University Medical Center, Universitätsmedizin Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55118 Mainz, Germany
| |
Collapse
|
13
|
Gonzalez-Ochoa E, Veneziani AC, Oza AM. Mirvetuximab Soravtansine in Platinum-Resistant Ovarian Cancer. Clin Med Insights Oncol 2023; 17:11795549231187264. [PMID: 37528890 PMCID: PMC10387675 DOI: 10.1177/11795549231187264] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/25/2023] [Indexed: 08/03/2023] Open
Abstract
Ovarian cancer is the second leading cause of death from gynecologic malignancies worldwide. Management of platinum-resistant disease is challenging and clinical outcomes with standard chemotherapy are poor. Over the past decades, significant efforts have been made to understand drug resistance and develop strategies to overcome treatment failure. Antibody drug conjugates (ADCs) are a rapidly growing class of oncologic therapeutics, which combine the ability to target tumor-specific antigens with the cytotoxic effects of chemotherapy. Mirvetuximab soravtansine is an ADC comprising an IgG1 monoclonal antibody against the folate receptor alpha (FRα) conjugated to the cytotoxic maytansinoid effector molecule DM4 that has shown promising clinical activity in patients with FR-α-positive ovarian cancer. This review summarizes current evidence of mirvetuximab soravtansine in platinum-resistant ovarian cancer, focusing on clinical activity, toxicity, and future directions.
Collapse
Affiliation(s)
- Eduardo Gonzalez-Ochoa
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ana C Veneziani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Yusuf A, Almotairy ARZ, Henidi H, Alshehri OY, Aldughaim MS. Nanoparticles as Drug Delivery Systems: A Review of the Implication of Nanoparticles' Physicochemical Properties on Responses in Biological Systems. Polymers (Basel) 2023; 15:polym15071596. [PMID: 37050210 PMCID: PMC10096782 DOI: 10.3390/polym15071596] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
In the last four decades, nanotechnology has gained momentum with no sign of slowing down. The application of inventions or products from nanotechnology has revolutionised all aspects of everyday life ranging from medical applications to its impact on the food industry. Nanoparticles have made it possible to significantly extend the shelf lives of food product, improve intracellular delivery of hydrophobic drugs and improve the efficacy of specific therapeutics such as anticancer agents. As a consequence, nanotechnology has not only impacted the global standard of living but has also impacted the global economy. In this review, the characteristics of nanoparticles that confers them with suitable and potentially toxic biological effects, as well as their applications in different biological fields and nanoparticle-based drugs and delivery systems in biomedicine including nano-based drugs currently approved by the U.S. Food and Drug Administration (FDA) are discussed. The possible consequence of continuous exposure to nanoparticles due to the increased use of nanotechnology and possible solution is also highlighted.
Collapse
Affiliation(s)
- Azeez Yusuf
- Irish Centre for Genetic Lung Disease, Department of Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, D02 YN77 Dublin, Ireland
| | | | - Hanan Henidi
- Research Department, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Ohoud Y Alshehri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11564, Saudi Arabia
| | - Mohammed S Aldughaim
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 11451, Saudi Arabia
| |
Collapse
|
15
|
Das S, Sakhare N, Kumar D, Mathur A, Mirapurkar S, Sheela M, Mohanty B, Chaudhari P, Chakraborty S. Design, characterization and evaluation of a new 99mTc-labeled folate derivative with affinity towards folate receptor. Bioorg Med Chem Lett 2023; 86:129240. [PMID: 36931350 DOI: 10.1016/j.bmcl.2023.129240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/09/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023]
Abstract
Folate receptors (FRs) are known to be over-expressed in several human malignancies and therefore serve as an important target for small radiolabeled folate derivatives for non-invasive imaging of tumor, which is an important tool for future treatment recourse. In the present article, we report the synthesis of a new 99mTc-labeled radiotracer for the aforementioned application following the well-established 99mTc-'4+1' chemistry. Formation of the desired [99mTc]Tc-complex with >95% radiochemical purity was confirmed by radio-HPLC and its structure was ascertained by characterizing a natural rhenium analogue of the said complex. Although the ligand exhibited a weaker affinity towards FRs compared to native folic acid (IC50 8.09 µM vs 29.46 nM), the 99mTc-labeled complex was found to bind folate receptor-positive KB cells with high specificity (∼90%). Similar studies in a folate receptor negative cell line viz. A549 further corroborated the receptor-specificity of the synthesized complex. In vivo studies in KB tumor xenograft showed moderate uptake of ∼2.6% upto 3 h post-injection with high specificity (∼80%). The favorable features observed warrant further screening of the current design towards achieving an improved molecular probe for the said application.
Collapse
Affiliation(s)
- Soumen Das
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology (BRIT), Navi Mumbai 400703, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai 400094, India.
| | - Navin Sakhare
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology (BRIT), Navi Mumbai 400703, India
| | - Dheeraj Kumar
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology (BRIT), Navi Mumbai 400703, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai 400094, India
| | - Anupam Mathur
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology (BRIT), Navi Mumbai 400703, India
| | - Shubhangi Mirapurkar
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology (BRIT), Navi Mumbai 400703, India
| | - M Sheela
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology (BRIT), Navi Mumbai 400703, India
| | - Bhabani Mohanty
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai 410210, India
| | - Pradip Chaudhari
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai 410210, India
| | - Sudipta Chakraborty
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai 400094, India; Radiopharmaceutical Division, Bhabha Atomic Rweesearch Centre (BARC), Trombay, Mumbai 400085, India.
| |
Collapse
|
16
|
de Morais FAP, De Oliveira ACV, Balbinot RB, Lazarin-Bidóia D, Ueda-Nakamura T, de Oliveira Silva S, da Silva Souza Campanholi K, da Silva Junior RC, Gonçalves RS, Caetano W, Nakamura CV. Multifunctional Nanoparticles as High-Efficient Targeted Hypericin System for Theranostic Melanoma. Polymers (Basel) 2022; 15:polym15010179. [PMID: 36616529 PMCID: PMC9824163 DOI: 10.3390/polym15010179] [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: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Biotin, spermine, and folic acid were covalently linked to the F127 copolymer to obtain a new drug delivery system designed for HY-loaded PDT treatment against B16F10 cells. Chemical structures and binders quantification were performed by spectroscopy and spectrophotometric techniques (1NMR, HABA/Avidin reagent, fluorescamine assay). Critical micelle concentration, critical micelle temperature, size, polydispersity, and zeta potential indicate the hydrophobicity of the binders can influence the physicochemical parameters. Spermine-modified micelles showed fewer changes in their physical and chemical parameters than the F127 micelles without modification. Furthermore, zeta potential measurements suggest an increase in the physical stability of these carrier systems. The phototherapeutic potential was demonstrated using hypericin-loaded formulation against B16F10 cells, which shows that the combination of the binders on F127 copolymer micelles enhances the photosensitizer uptake and potentializes the photodynamic activity.
Collapse
Affiliation(s)
- Flávia Amanda Pedroso de Morais
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
- Department of Chemistry, State University of Maringá, Maringá 87020-900, PR, Brazil
- Correspondence: (F.A.P.d.M.); (C.V.N.); Tel.: +55-(44)-3011-3680 (F.A.P.d.M. & C.V.N.)
| | | | - Rodolfo Bento Balbinot
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Danielle Lazarin-Bidóia
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Tânia Ueda-Nakamura
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Sueli de Oliveira Silva
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | | | - Renato Sonchini Gonçalves
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Center for Exact Sciences and Technology, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
| | - Wilker Caetano
- Department of Chemistry, State University of Maringá, Maringá 87020-900, PR, Brazil
| | - Celso Vataru Nakamura
- Technological Innovation Laboratory in the Pharmaceuticals and Cosmetics Development, State University of Maringá, Maringá 87020-900, PR, Brazil
- Correspondence: (F.A.P.d.M.); (C.V.N.); Tel.: +55-(44)-3011-3680 (F.A.P.d.M. & C.V.N.)
| |
Collapse
|
17
|
Fabrication of hesperidin hybrid lecithin-folic acid silver nanoparticles and its evaluation as anti-arthritis formulation in autoimmune arthritic rat model. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
18
|
Wang H, Lin S, Wang S, Jiang Z, Ding T, Wei X, Lu Y, Yang F, Zhan C. Folic Acid Enables Targeting Delivery of Lipodiscs by Circumventing IgM-Mediated Opsonization. NANO LETTERS 2022; 22:6516-6522. [PMID: 35943299 DOI: 10.1021/acs.nanolett.2c01509] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Folic acid (FA) is one of the most widely utilized small-molecule ligands for cancer targeted drug delivery. Natural IgM was recently found to avidly absorb on the surface of FA-functionalized liposomes (FA-sLip), negatively regulating the in vivo performance by efficiently activating complement. Herein, FA-functionalized lipodiscs (FA-Disc) were constructed to successfully circumvent IgM-mediated opsonization and retained binding activity with folate receptors in vivo. The FA moiety along with the bound IgM was restricted to the highly curved rim of lipodiscs, leading to IgM incapability of presenting the membrane-bound conformation to trigger complement activation. The C1q docking, C3 binding, and C5a release were blocked and accelerated blood clearance phenomenon was mitigated of FA-Disc. FA-Disc retained folate binding activity and could effectively target folate receptor positive tumors in vivo. The present study provides a useful solution to avoid the negative regulation by IgM and achieve FA-enabled targeting by exploring disc-shaped nanocarriers.
Collapse
Affiliation(s)
- Huan Wang
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Shiqi Lin
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Songli Wang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Zhuxuan Jiang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Tianhao Ding
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Xiaoli Wei
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Ying Lu
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
| | - Feng Yang
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
| | - Changyou Zhan
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| |
Collapse
|
19
|
A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
20
|
Salerno A, Seghetti F, Caciolla J, Uliassi E, Testi E, Guardigni M, Roberti M, Milelli A, Bolognesi ML. Enriching Proteolysis Targeting Chimeras with a Second Modality: When Two Are Better Than One. J Med Chem 2022; 65:9507-9530. [PMID: 35816671 PMCID: PMC9340767 DOI: 10.1021/acs.jmedchem.2c00302] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 02/08/2023]
Abstract
Proteolysis targeting chimera (PROTAC)-mediated protein degradation has prompted a radical rethink and is at a crucial stage in driving a drug discovery transition. To fully harness the potential of this technology, a growing paradigm toward enriching PROTACs with other therapeutic modalities has been proposed. Could researchers successfully combine two modalities to yield multifunctional PROTACs with an expanded profile? In this Perspective, we try to answer this question. We discuss how this possibility encompasses different approaches, leading to multitarget PROTACs, light-controllable PROTACs, PROTAC conjugates, and macrocycle- and oligonucleotide-based PROTACs. This possibility promises to further enhance PROTAC efficacy and selectivity, minimize side effects, and hit undruggable targets. While PROTACs have reached the clinical investigation stage, additional steps must be taken toward the translational development of multifunctional PROTACs. A deeper and detailed understanding of the most critical challenges is required to fully exploit these opportunities and decisively enrich the PROTAC toolbox.
Collapse
Affiliation(s)
- Alessandra Salerno
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Francesca Seghetti
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Jessica Caciolla
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Elisa Uliassi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Eleonora Testi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Melissa Guardigni
- Department
for Life Quality Studies, Alma Mater Studiorum
- University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy
| | - Marinella Roberti
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Andrea Milelli
- Department
for Life Quality Studies, Alma Mater Studiorum
- University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy
| | - Maria Laura Bolognesi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
21
|
Zhang A, Liu Q, Huang Z, Zhang Q, Wang R, Cui D. Electrochemical Cytosensor Based on a Gold Nanostar-Decorated Graphene Oxide Platform for Gastric Cancer Cell Detection. SENSORS (BASEL, SWITZERLAND) 2022; 22:s22072783. [PMID: 35408396 PMCID: PMC9003065 DOI: 10.3390/s22072783] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 05/02/2023]
Abstract
Effectively capturing and sensitively detecting cancer cells are critical to clinical diagnosis and cancer therapy. In this work, we prepared gold nanostar-decorated graphene oxide (GO-AuNSs) nanocomposites using a ultraviolet (UV)-induced strategy, and then modified them with a layer of bio-complex rBSA-FA (coupled reduced bovine serum albumin with folic acid) to generate GO-AuNSs@rBSA-FA nanocomposites. Herein, the application of GO and AuNSs not only strengthened the conductivity of the sensing platform but also guaranteed nanocomposites with biocompatible performance. Moreover, the adopted rBSA-FA layer could effectively enhance the stability and specificity towards gastric cancer cells (MGC-803). According to a systemic construction procedure, a novel electrochemical cytosensor based on GO-AuNSs@rBSA-FA was fabricated for MGC-803 cell detection. With the assistance of cyclic voltammetry (CV) and differential pulse voltammetry (DPV), the cytosensor reached a detection limit of 100 cell/mL in a wide linear range of 3 × 102~7 × 106 cell/mL towards MGC-803 cells. The good electrochemical characteristics for the cancer cell analysis indicate a promising prospect of this electrochemical cytosensor in clinical cancer diagnosis.
Collapse
Affiliation(s)
- Amin Zhang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China; (A.Z.); (Q.L.); orange-.- (Z.H.); (Q.Z.); (R.W.)
- National Engineering Research Center for Nanotechnology, 28 Jiangchuan Easternroad, Shanghai 200241, China
| | - Qianwen Liu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China; (A.Z.); (Q.L.); orange-.- (Z.H.); (Q.Z.); (R.W.)
- National Engineering Research Center for Nanotechnology, 28 Jiangchuan Easternroad, Shanghai 200241, China
| | - Zhicheng Huang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China; (A.Z.); (Q.L.); orange-.- (Z.H.); (Q.Z.); (R.W.)
- National Engineering Research Center for Nanotechnology, 28 Jiangchuan Easternroad, Shanghai 200241, China
| | - Qian Zhang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China; (A.Z.); (Q.L.); orange-.- (Z.H.); (Q.Z.); (R.W.)
- National Engineering Research Center for Nanotechnology, 28 Jiangchuan Easternroad, Shanghai 200241, China
| | - Ruhao Wang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China; (A.Z.); (Q.L.); orange-.- (Z.H.); (Q.Z.); (R.W.)
- National Engineering Research Center for Nanotechnology, 28 Jiangchuan Easternroad, Shanghai 200241, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China; (A.Z.); (Q.L.); orange-.- (Z.H.); (Q.Z.); (R.W.)
- National Engineering Research Center for Nanotechnology, 28 Jiangchuan Easternroad, Shanghai 200241, China
- Correspondence:
| |
Collapse
|
22
|
Folic Acid-Modified Fluorescent-Magnetic Nanoparticles for Efficient Isolation and Identification of Circulating Tumor Cells in Ovarian Cancer. BIOSENSORS 2022; 12:bios12030184. [PMID: 35323454 PMCID: PMC8946694 DOI: 10.3390/bios12030184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/16/2022]
Abstract
Ovarian cancer (OC) is a lethal disease occurring in women worldwide. Due to the lack of obvious clinical symptoms and sensitivity biomarkers, OC patients are often diagnosed in advanced stages and suffer a poor prognosis. Circulating tumor cells (CTCs), released from tumor sites into the peripheral blood, have been recognized as promising biomarkers in cancer prognosis, treatment monitoring, and metastasis diagnosis. However, the number of CTCs in peripheral blood is low, and it is a technical challenge to isolate, enrich, and identify CTCs from the blood samples of patients. This work develops a simple, effective, and inexpensive strategy to capture and identify CTCs from OC blood samples using the folic acid (FA) and antifouling-hydrogel-modified fluorescent-magnetic nanoparticles. The hydrogel showed a good antifouling property against peripheral blood mononuclear cells (PBMCs). The FA was coupled to the hydrogel surface as the targeting molecule for the CTC isolation, held a good capture efficiency for SK-OV-3 cells (95.58%), and successfully isolated 2–12 CTCs from 10 OC patients’ blood samples. The FA-modified fluorescent-magnetic nanoparticles were successfully used for the capture and direct identification of CTCs from the blood samples of OC patients.
Collapse
|
23
|
Natural Killer Cells: the Missing Link in Effective Treatment for High-Grade Serous Ovarian Carcinoma. Curr Treat Options Oncol 2022; 23:210-226. [PMID: 35192139 DOI: 10.1007/s11864-021-00929-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/22/2022]
Abstract
OPINION STATEMENT Ovarian cancer (OC), especially high-grade serous cancer (HGSC), is a highly heterogeneous malignancy with limited options for curative treatment and a high frequency of relapse. Interactions between OC and the immune system may permit immunoediting and immune escape, and current standard of care therapies can influence immune cell infiltration and function within the tumor microenvironment. Natural killer (NK) cells are involved in cancer immunosurveillance and immunoediting and can be activated by therapy, but deliberate approaches to maximize NK cell reactivity for treatment of HGSC are in their infancy. NK cells may be the ideal target for immunotherapy of HGSC. The diverse functions of NK cells, and their established roles in immunosurveillance, make them attractive candidates for more precise and effective HGSC treatment. NK cells' functional capabilities differ because of variation in receptor expression and genetics, with meaningful impacts on their anticancer activity. Studying HGSC:NK cell interactions will define the features that predict the best outcomes for patients with the disease, but the highly diverse nature of HGSC will likely require combination therapies or approaches to simultaneously target multiple, co-existing features of the tumor to avoid tumor escape and relapse. We expect that the ideal therapy will enable NK cell infiltration and activity, reverse immunosuppression within the tumor microenvironment, and enable effector functions against the diverse subpopulations that comprise HGSC.
Collapse
|
24
|
Wang Z, Zhao S, shi J, Meng F, Yuan J, Zhong Z. Folate-mediated targeted PLK1 inhibition therapy for ovarian cancer: A comparative study of molecular inhibitors and siRNA therapeutics. Acta Biomater 2022; 138:443-452. [PMID: 34757229 DOI: 10.1016/j.actbio.2021.10.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/29/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
PLK1 is a promising target for clinical treatment of diverse malignancies including ovarian cancer (OC), in which PLK1 over-expression is often correlated with poor prognosis and short survival. PLK1 can be blocked with small molecular inhibitors like volasertib (Vol) or silenced with PLK1-specific siRNA (siPLK1), hence effectively suppressing tumor growth. Surprisingly, despite intensive work on molecular inhibitor and siRNA therapeutics, there is no direct comparison between them reported for targeted tumor therapy. Herein, we employing folate as a ligand and polymersomes as a nanovehicle performed a comparative study on Vol and siPLK1 in inhibiting OC in vitro and in vivo. Folate-targeted polymersomal Vol and siPLK1 (termed as FA-Ps-Vol and FA-Ps-siPLK1, respectively) were both nano-sized and stable, and displayed an optimal FA density of 20% for SKOV-3 cells. Notably, FA-Ps-Vol and FA-Ps-siPLK1 exhibited an IC50 of 193 and 770 nM, respectively, to SKOV-3 cells, indicating a greater potency of Vol than siPLK1. The markedly increased uptake for FA-Ps-Vol and FA-Ps-siPLK1 compared with respective non-targeted controls by SKOV-3 tumor xenografts in mice confirmed that FA mediates strong OC-targeting in vivo. Intriguingly, FA-Ps-Vol while greatly lessening toxic effects of Vol potently repressed tumor growth with a remarkable tumor inhibition rate (TIR) of 97% at 20 mg (i.e. 32.4 µmol) Vol equiv./kg. FA-Ps-siPLK1 achieved effective tumor inhibition (TIR = ca. 87% or 90%) at 2 or 4 mg (i.e. 0.15 or 0.3 µmol) siPLK1 equiv./kg without causing adverse effects. This comparative study highlights that molecular inhibitor has the advantage of easy dose escalation and potent protein inhibition at the expense of certain adverse effects while siRNA therapeutics has low toxicity with moderate protein inhibition in vivo. STATEMENT OF SIGNIFICANCE: PLK1 is a promising target for the development of innovative and specific treatments against diverse malignancies. Interestingly, despite intensive work on molecular inhibitors and siRNA against PLK1, little work has been directed to compare their efficacy in targeted tumor therapy. Here, we employed folate as a ligand and polymersomes as a nanovehicle and have performed a comparative study on volasertib and siPLK1 in inhibiting ovarian cancer in vitro and in vivo. Our data show that the dose of volasertib can be easily escalated to induce prominent antitumor efficacy at the expense of certain adverse effects, while siPLK1 brings about moderate protein inhibition and antitumor therapy without causing toxicity at two-orders-of-magnitude lower dose.
Collapse
|
25
|
Wu M, Cui H, Yang Y, Dang B, Li D, Zhang Z, Yang X. Folic‐Acid‐Functionalized Au Nanoclusters with Red Fluorescence Emission for Rapid and Selective Detection of Cancer Cells. ChemistrySelect 2022. [DOI: 10.1002/slct.202103258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Miao Wu
- Changchun University of Technology School of Chemical Engineering Advanced Institute of Materials Science Changchun 130012 P. R. China
| | - Huijing Cui
- Changchun University of Technology School of Chemical Engineering Advanced Institute of Materials Science Changchun 130012 P. R. China
| | - Yizhou Yang
- Changchun University of Technology School of Chemical Engineering Advanced Institute of Materials Science Changchun 130012 P. R. China
| | - Ben Dang
- Changchun University of Technology School of Chemical Engineering Advanced Institute of Materials Science Changchun 130012 P. R. China
| | - Daowei Li
- Jilin University Department of Periodontology School and Hospital of Stomatology Changchun 130012 P. R. China
| | - Zhuqing Zhang
- Changchun University of Technology School of Chemical Engineering Advanced Institute of Materials Science Changchun 130012 P. R. China
| | - Xudong Yang
- Changchun University of Technology School of Chemical Engineering Advanced Institute of Materials Science Changchun 130012 P. R. China
| |
Collapse
|
26
|
Siemiaszko G, Niemirowicz-Laskowska K, Markiewicz KH, Misztalewska-Turkowicz I, Dudź E, Milewska S, Misiak P, Kurowska I, Sadowska A, Car H, Wilczewska AZ. Synergistic effect of folate-conjugated polymers and 5-fluorouracil in the treatment of colon cancer. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00104-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
Background
In recent years, targeted drug delivery strategies have received special attention from the scientific world due to advantages such as more effective therapy and reduction of side effects. The principle of operation is delayed excretion from the bloodstream of the drug delivery system compared to the drug itself, as well as facilitated penetration into diseased cells thanks to the use of ligands recognized by appropriate receptors. Particularly interesting drug carriers are amphiphilic copolymers that form nano-sized micelles with a drug, which can release the drug at a specific place in the body under the influence of appropriate stimuli.
Results
We describe the synthesis of the diblock polymer, poly(2-hydroxyethyl acrylate)-b-poly(N-vinylcaprolactam) using RAFT/MADIX (Reversible Addition-Fragmentation chain Transfer/MAcromolecular Design by Interchange of Xanthate) controlled polymerization affording polymers with good dispersity according to SEC (Size-Exclusion Chromatography). Some post-modifications of the polymer with folic acid were then performed as evidenced by NMR (Nuclear Magnetic Resonance), UV–Vis (UltraViolet–Visible) and FT-IR (Fourier-Transform Infrared) spectroscopy, and TGA (ThermoGravimetric Analysis). The formation of stable micellar systems from polymers with and without the drug, 5-fluorouracil, was confirmed by DLS (Dynamic Light Scattering) and zeta potential measurements, and TEM (Transmission Eelectron Microscopy) imaging. Finally, the cloud point of the polymers was investigated, which turned out to be close to the temperature of the human body. Most importantly, these micellar systems have been explored as a drug delivery system against colon cancer, showing increased cytotoxicity compared to the drug alone. This effect was achieved due to the easier cellular uptake by the interaction of folic acid and its receptors on the surface of cancer cells.
Conclusions
The presented results constitute a solid foundation for the implementation of a nano-sized drug delivery system containing folic acid for practical use in the treatment of drug-resistant cancer, as well as more effective therapy with fewer side effects.
Graphical Abstract
Collapse
|
27
|
Selenium stimulates the antitumour immunity: Insights to future research. Eur J Cancer 2021; 155:256-267. [PMID: 34392068 DOI: 10.1016/j.ejca.2021.07.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/10/2023]
Abstract
Selenium is an essential trace element for regulating immune functions through redox-regulating activity of selenoproteins (e.g. glutathione peroxidase), protecting immune cells from oxidative stress. However, in cancer, selenium has biological bimodal action depending on the concentration. At nutritional low doses, selenium, depending on its form, may act as an antioxidant, protecting against oxidative stress, supporting cell survival and growth, thus, plays a chemo-preventive role; while, at supra-nutritional higher pharmacological doses, selenium acts as pro-oxidant inducing redox signalling and cell death. To date, many studies have been conducted on the benefits of selenium intake in reducing the risk of cancer incidence at the nutritional level, indicating that likely selenium functions as an immunostimulator, i.e. reversing the immunosuppression in tumour microenvironment towards antitumour immunity by activating immune cells (e.g. M1 macrophages and CD8+ T-lymphocytes) and releasing pro-inflammatory cytokines such as interferon-gamma; whereas, fewer studies have explored the effects of supra-nutritional or pharmacological doses of selenium in cancer immunity. This review, thus, systematically analyses the current knowledge about how selenium stimulates the immune system against cancer and lay the groundwork for future research. Such knowledge can be promising to design combinatorial therapies with Selenium-based compounds and other modalities like immunotherapy to lower the adverse effects and increase the efficacy of treatments.
Collapse
|
28
|
Xiao F, Liu J, Zheng Y, Quan Z, Sun W, Fan Y, Luo C, Li H, Wu X. The targeted inhibition of prostate cancer by iron-based nanoparticles based on bioinformatics. J Biomater Appl 2021; 36:3-14. [PMID: 33283584 PMCID: PMC8217887 DOI: 10.1177/0885328220975249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer is an epithelial malignant tumor of the prostate, and it is one of the malignant tumors with a high incidence of urogenital system in men. The local treatment of prostate cancer is mainly radical resection and radical radiotherapy, but they are not applicable to advanced prostate cancer. Systemic therapy mainly includes targeted therapy and immunotherapy which could cause many complications, and will affect the prognosis and quality of life of patients. It is urgent to find new treatments for prostate cancer. Bioinformatics offers hope for us to find reliable therapeutic targets. Bioinformatics can use the tumor informations in database and analyze them to screen out the best differentially expressed genes. Using the selected differentially expressed genes as targets, a gene interference plasmid was designed, and the constructed plasmid was used for targeted gene therapy. There are some problems about gene therapy that need to be solved, such as how to transfer genes to target cells is also an important challenge. Due to their large molecular weight and hydrophilic nature, they cannot enter cells through passive diffusion mechanisms. Here we synthesized a DNA carrier used surface modified iron based nanoparticles, and used it to load plasmid including ShRNA which can inhibit the expression of oncogene SLC4A4 selected by bioinformatics' method. After that we use this iron based nanoparticles/plasmid DNA nanocomposite to treat prostate cancer cells in vitro and in vivo. The target gene SLC4A4 we had selected using bioinformatics had a strong effect on the proliferation of prostate cells; Our nanocomposite could inhibit the expression of SLC4A4 effectively, it had strong inhibitory effects on prostate cancer cells both in vivo and in vitro, and can be used as a potential method for prostate cancer treatment.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Jiayu Liu
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Yongbo Zheng
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Zhen Quan
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Wei Sun
- Fuling Center Hospital of Chongqing City, Chongqing, China
| | - Yao Fan
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Chunli Luo
- Chongqing Medical University, Chongqing, China
| | - Hailiang Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaohou Wu
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| |
Collapse
|
29
|
Razaghi A, Zickler AM, Spallholz J, Kirsch G, Björnstedt M. Selenofolate inhibits the proliferation of IGROV1 cancer cells independently from folate receptor alpha. Heliyon 2021; 7:e07254. [PMID: 34169173 PMCID: PMC8209087 DOI: 10.1016/j.heliyon.2021.e07254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/07/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer is one of the main causes of human mortality worldwide and novel chemotherapeutics are required due to the limitations of conventional cancer therapies. For example, using redox selenium compounds as novel chemotherapeutics seem to be very promising. The objective of this study was to explore if folate could be used as a carrier to deliver a newly synthesised selenium derivative selenofolate into cancer cells. Particularly, the cytotoxic effects of this selenofolate compound were investigated in a variety of cancer cell types including lung, liver, and cervical cancers and specifically IGROV1 cells. Our results showed that selenofolate inhibits the growth of cancer cells in-vitro. However, despite the expectations, folate receptor alpha (FRα) was not involved in the transportation of selenofolate compound into the cells i.e. growth inhibition was independent of FRα, suggesting that multiple transporters (e.g. reduced folate carrier-1) are possibly involved in the delivery and internalisation of folate in IGROV1 cells. Additionally, selenofolate did not exert cell death through apoptosis. Instead, anti-proliferative activity showed to be the main cause of growth inhibition of selenolofate in the IGROV1 cell line. In conclusion, selenofolate inhibits the growth of cancer cells and thus, may be explored further as a potential chemotherapeutic agent.
Collapse
Affiliation(s)
- Ali Razaghi
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University-Hospital, SE-14152, Stockholm, Sweden
| | - Antje Maria Zickler
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University-Hospital, SE-14152, Stockholm, Sweden
| | - Julian Spallholz
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University-Hospital, SE-14152, Stockholm, Sweden
| | - Gilbert Kirsch
- Université de Lorraine, CNRS, L2CM, F-57000, Metz, France
| | - Mikael Björnstedt
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University-Hospital, SE-14152, Stockholm, Sweden
- Corresponding author.
| |
Collapse
|
30
|
|
31
|
Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorg Chem 2021; 112:104946. [PMID: 33989916 DOI: 10.1016/j.bioorg.2021.104946] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| |
Collapse
|
32
|
Ma Y, Yu S, Ni S, Zhang B, Kung ACF, Gao J, Lu A, Zhang G. Targeting Strategies for Enhancing Paclitaxel Specificity in Chemotherapy. Front Cell Dev Biol 2021; 9:626910. [PMID: 33855017 PMCID: PMC8039396 DOI: 10.3389/fcell.2021.626910] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/25/2021] [Indexed: 11/15/2022] Open
Abstract
Paclitaxel (PTX) has been used for cancer treatment for decades and has become one of the most successful chemotherapeutics in the clinic and financially. However, serious problems with its use still exist, owing to its poor solubility and non-selective toxicity. With respect to these issues, recent advances have addressed the water solubility and tumor specificity related to PTX application. Many measures have been proposed to remedy these limitations by enhancing tumor recognition via ligand-receptor-mediated targeting as well as other associated strategies. In this review, we investigated various kinds of ligands that have emerged as PTX tumor-targeting tools. In particular, this article highlights small molecule-, protein-, and aptamer-functionalized conjugates and nanoparticles (NPs), providing a promising approach for PTX-based individualized treatment prospects.
Collapse
Affiliation(s)
- Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China.,Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Sifan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Shuaijian Ni
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China.,Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Baoxian Zhang
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong.,Increasepharm (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong
| | - Angela Chun Fai Kung
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong.,Increasepharm (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong
| | - Jin Gao
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong.,Increasepharm (Hengqin) Institute Co. Limited, Zhuhai, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China.,Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China.,Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| |
Collapse
|
33
|
Morey J, Llinás P, Bueno-Costa A, León AJ, Piña MN. Raltitrexed-Modified Gold and Silver Nanoparticles for Targeted Cancer Therapy: Cytotoxicity Behavior In Vitro on A549 and HCT-116 Human Cancer Cells. MATERIALS 2021; 14:ma14030534. [PMID: 33499297 PMCID: PMC7866044 DOI: 10.3390/ma14030534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
Two different raltitrexed gold and silver nanoparticles for the delivery of an antitumoral drug into cancer cells were synthesized and characterized. A cysteine linker was used for the covalent bonding of raltitrexed to the surface of nanoparticles. To evaluate the efficacy of the antifolate-derivative nanoparticles, their cytotoxicity was assayed in vitro with A549 human lung adenocarcinoma and HCT-116 colorectal carcinoma human cells. Modified nanoparticles are a biocompatible material, and administration of silver raltitrexed nanoparticles strongly inhibited the viability of the cancer cells; gold raltitrexed nanoparticles do not show any type of cytotoxic effect. The results suggest that silver raltitrexed nanoparticles could be a potential delivery system for certain cancer cells.
Collapse
Affiliation(s)
- Jeroni Morey
- Department of Chemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (J.M.); (A.J.L.)
| | - Pere Llinás
- Department of Biochemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (P.L.); (A.B.-C.)
| | - Alberto Bueno-Costa
- Department of Biochemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (P.L.); (A.B.-C.)
| | - Alberto J. León
- Department of Chemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (J.M.); (A.J.L.)
| | - M. Nieves Piña
- Department of Chemistry, University of the Balearic Islands, Crta. de Valldemossa, Km. 7.5, 07122 Palma de Mallorca, Balearic Islands, Spain; (J.M.); (A.J.L.)
- Correspondence: ; Tel.: +34-971-172847
| |
Collapse
|
34
|
Liu Z, Xie L, Yan J, Liu P, Wen H, Liu H. Folic Acid-Targeted MXene Nanoparticles for Doxorubicin Loaded Drug Delivery. Aust J Chem 2021. [DOI: 10.1071/ch21216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MXenes are two-dimensional (2D) materials with a large specific surface area and abundant surface functional groups. A folate receptors-targeted drug carrier was constructed based on the rich surface functional groups and high biocompatibility of MXenes. This drug carrier possesses as high as 69.9 % drug-loading capability and as long as 48 h drug release time. Tumour targeting and a pH-responsive mechanism can make MXene nanoparticles quickly accumulate in tumour sites and slowly release loads. The results showed that DOX was released in a large amount in a PBS solution at pH 4.5. Compared with the naked drug, MXenes-FA-SP@DOX has a higher cell inhibition rate and a longer drug action time at a lower concentration (less than 10 μg mg−1). This drug delivery system exhibited potential applications for the treatment of malignant tumour and this work extends the biomedical applications of MXenes in nanomedicine.
Collapse
|
35
|
Ma Y, Yu S, Ni S, Zhang B, Kung ACF, Gao J, Lu A, Zhang G. Targeting Strategies for Enhancing Paclitaxel Specificity in Chemotherapy. Front Cell Dev Biol 2021. [PMID: 33855017 DOI: 10.3389/fcell.2021.626910/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Paclitaxel (PTX) has been used for cancer treatment for decades and has become one of the most successful chemotherapeutics in the clinic and financially. However, serious problems with its use still exist, owing to its poor solubility and non-selective toxicity. With respect to these issues, recent advances have addressed the water solubility and tumor specificity related to PTX application. Many measures have been proposed to remedy these limitations by enhancing tumor recognition via ligand-receptor-mediated targeting as well as other associated strategies. In this review, we investigated various kinds of ligands that have emerged as PTX tumor-targeting tools. In particular, this article highlights small molecule-, protein-, and aptamer-functionalized conjugates and nanoparticles (NPs), providing a promising approach for PTX-based individualized treatment prospects.
Collapse
Affiliation(s)
- Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Sifan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Shuaijian Ni
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Baoxian Zhang
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
- Increasepharm (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong
| | - Angela Chun Fai Kung
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
- Increasepharm (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong
| | - Jin Gao
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
- Increasepharm (Hengqin) Institute Co. Limited, Zhuhai, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
- Increasepharm and Hong Kong Baptist University Joint Centre for Nucleic Acid Drug Discovery, Hong Kong Science Park, New Territories, Hong Kong
| |
Collapse
|
36
|
Hashemkhani M, Muti A, Sennaroğlu A, Yagci Acar H. Multimodal image-guided folic acid targeted Ag-based quantum dots for the combination of selective methotrexate delivery and photothermal therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 213:112082. [PMID: 33221627 DOI: 10.1016/j.jphotobiol.2020.112082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/13/2020] [Accepted: 11/08/2020] [Indexed: 01/06/2023]
Abstract
Multifunctional quantum dots (QDs) with photothermal therapy (PTT) potential loaded with an anticancer drug and labelled with a targeting agent can be highly effective nano-agents for tumour specific, image-guided PTT/chemo combination therapy of cancer. Ag-chalcogenides are promising QDs with good biocompatibility. Ag2S QDs are popular theranostic agents for imaging in near-infrared with PTT potential. However, theranostic applications of AgInS2 QDs emitting in the visible region and its PTT potential need to be explored. Here, we first present a simple synthesis of small, glutathione (GSH) coated AgInS2 QDs with peak emission at 634 nm, 21% quantum yield, and excellent long-term stability without an inorganic shell. Ag2S-GSH QDs emitting in the near-infrared region (peak emission = 822 nm) were also produced. Both QDs were tagged with folic acid (FA) and conjugated with methotrexate (MTX). About 3-fold higher internalization of FA-tagged QDs by folate-receptor (FR) overexpressing HeLa cells than HT29 and A549 cells was observed. Delivery of MTX by QD-FA-MTX reduced the IC50 of the drug from 10 μg/mL to 2.5-5 μg/mL. MTX release was triggered at acidic pH, which was further enhanced with local temperature increase created by laser irradiation. Irradiation of AgInS2-GSH QDs at 640 nm (300 mW) for 10 min, caused about 10 °C temperature increase but did not cause any thermal ablation of cells. On the other hand, Ag2S-GSH-FA based PTT effectively and selectively killed HeLa cells with 10 min 808 nm laser irradiation via mostly necrosis with an IC50 of 5 μg Ag/mL. Under the same conditions, IC50 of MTX was reduced to 0.21 μg/mL if Ag2S-GSH-FA.
Collapse
Affiliation(s)
- Mahshid Hashemkhani
- Koç University, Graduate School of Materials Science and Engineering, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey
| | - Abdullah Muti
- Laser Research Laboratory, Departments of Physics and Electrical-Electronics Engineering, Koç University, Istanbul 34450, Turkey
| | - Alphan Sennaroğlu
- Koç University, Graduate School of Materials Science and Engineering, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey; Laser Research Laboratory, Departments of Physics and Electrical-Electronics Engineering, Koç University, Istanbul 34450, Turkey; Koç University Surface Science and Technology Center (KUYTAM)Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey
| | - Havva Yagci Acar
- Koç University, Graduate School of Materials Science and Engineering, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey; Koç University, Department of Chemistry, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey.
| |
Collapse
|
37
|
Folate Modified Long Circulating Nano-Emulsion as a Promising Approach for Improving the Efficiency of Chemotherapy Drugs in Cancer Treatment. Pharm Res 2020; 37:242. [PMID: 33188481 DOI: 10.1007/s11095-020-02811-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE In order to improve the therapeutic efficiency of the chemotherapeutic drug paclitaxel in tumors, a folate-based Paclitaxel nanoemulsion (FNEs) was developed for tumor targeted treatment. METHODS In this study, we designed a folate-targeted nanoemulsion (folate/PEG-DSPE/nanoemulsion, FNEs) based on the traditional nanoemulsion using the principle of long-circulation targeting receptor mediated. The nanoemulsion (folate/PEG-DSPE/nanoemulsion, FNEs) was fabricated using high-pressure homogenization with a microfluidizer. RESULTS The nanoemulsion (folate/PEG-DSPE/nanoemulsion, FNEs) can improve the delivery efficiency of nanocarriers at the tumor site by virtue of the high expression of folate receptors on the tumor surface. Malvern Nanoseries device and transmission electron microscopy (TEM) analyses showed that the nanoemulsions were spherical with an average diameter of 140 nm. The nanoemulsions can effectively carry paclitaxel (PTX) with an encapsulation rate of about 95%. And in vitro experiments have shown that it can efficiently increase the uptake of PTX in 4 T1 breast cancer cells and FNEs had a targeting capability hundredfold higher than that of PTX-loaded nanoemulsions (PTX-NEs) without folate. In vivo experiments have shown that the pharmacokinetic parameters of FNEs were better than those of other PTX groups and FNEs can significantly enhance circulation time in the body of the subcutaneously implanted 4 T1 breast cancer in mice, increase the accumulation of chemotherapy drugs at tumor sites and effectively inhibit tumor growth with lower system toxicity. CONCLUSIONS This study can effectively improve the therapeutic efficiency of chemotherapy drugs for tumors, and provide an useful reference for solving the problem of low efficacy of chemotherapy drugs in clinical treatment of tumors. Graphical Abstract Schematic representation of Folic acid/PEG-DSPE/nano-emulsion (FNEs) specifically target tumor cells and enhanced anti-tumor effects.
Collapse
|
38
|
Wang S, Zhou D, Xu Z, Song J, Qian X, Lv X, Luan J. Anti-tumor Drug Targets Analysis: Current Insight and Future Prospect. Curr Drug Targets 2020; 20:1180-1202. [PMID: 30947670 DOI: 10.2174/1389450120666190402145325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/13/2022]
Abstract
The incidence and mortality of malignant tumors are on the rise, which has become the second leading cause of death in the world. At present, anti-tumor drugs are one of the most common methods for treating cancer. In recent years, with the in-depth study of tumor biology and related disciplines, it has been gradually discovered that the essence of cell carcinogenesis is the infinite proliferation of cells caused by the disorder of cell signal transduction pathways, followed by a major shift in the concept of anti-tumor drugs research and development. The focus of research and development is shifting from traditional cytotoxic drugs to a new generation of anti-tumor drugs targeted at abnormal signaling system targets in tumor cells. In this review, we summarize the targets of anti-tumor drugs and analyse the molecular mechanisms of their effects, which lay a foundation for subsequent treatment, research and development.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Dexi Zhou
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Zhenyu Xu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jing Song
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xueyi Qian
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiongwen Lv
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Institute for Liver Disease of Anhui Medical University, Hefei, Anhui Province, China
| | - Jiajie Luan
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| |
Collapse
|
39
|
Kavand A, Anton N, Vandamme T, Serra CA, Chan-Seng D. Synthesis and functionalization of hyperbranched polymers for targeted drug delivery. J Control Release 2020; 321:285-311. [DOI: 10.1016/j.jconrel.2020.02.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
|
40
|
Phase Ib study of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), in combination with bevacizumab in patients with platinum-resistant ovarian cancer. Gynecol Oncol 2020; 157:379-385. [DOI: 10.1016/j.ygyno.2020.01.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 11/20/2022]
|
41
|
Synthesis and characterization of folic acid-chitosan nanoparticles loaded with thymoquinone to target ovarian cancer cells. J Radioanal Nucl Chem 2020. [DOI: 10.1007/s10967-020-07058-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Abstract
PURPOSE OF REVIEW Antibody drug conjugates (ADC) are a novel class of cancer therapeutics, delivering cytotoxic therapy directly to cancer cells, and show promise in the management of platinum-resistant ovarian cancer. Herein we summarize the ADC landscape currently in clinical study. RECENT FINDINGS Mirvetuximab Soravtansine, IMGN853, is an ADC targeting the folate receptor alpha (FRα) and has demonstrated promising single agent activity and a favorable toxicity profile in FRα-positive, platinum-resistant, epithelial ovarian cancer (EOC). The antitumor effect is seen primarily in less heavily pretreated EOC patients with moderate-to-high FRα tumor expression. A phase III study, randomizing patients to either IMGN853 or the physician's choice of single-agent chemotherapy has completed accrual. Additional ADC are being evaluated in ovarian cancer including agents that target NaPiB2, Trop2, mesothelin, and MUC16 are in phase 1 clinical trials. SUMMARY ADC bind antigens overexpressed on cancer cells and provide site-selective drug delivery, with the goal to increase therapeutic efficacy of cytotoxics while decreasing the off-target toxicity of the payloads. With appropriate antigen selection and adequate, measurable antigen threshold targets, these new agents may provide an improved strategy for overcoming resistance to standard chemotherapy in ovarian cancer.
Collapse
|
43
|
Singh R. Nanotechnology based therapeutic application in cancer diagnosis and therapy. 3 Biotech 2019; 9:415. [PMID: 31696020 PMCID: PMC6811486 DOI: 10.1007/s13205-019-1940-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Due to the lack of early diagnosis, cancer remains as one of the leading cause of human mortality. Inability to translate research into clinical trials and also inability of chemotherapeutics delivery to targeted tumor sites are major drawbacks in cancer therapeutics. With the emergence of nanomedicine, several nanoprobes (conjugated with targeting ligands and chemotherapeutic drugs) are developed. It can interact with biological system and thus sense and monitor the biological events with high efficiency and accuracy along with therapy application. Nanoparticles like gold and iron oxide are frequently used in the computed tomography and magnetic resonance imaging applications, respectively. Moreover, enzymatic activity of gold and iron oxide nanoparticles enables the visible colorimetric diagnostic of cancer cells, whereas, fluorescence property of quantum dots and upconversion nanoparticles helps in in vivo imaging application. Other than this, drug conjugation with nanoparticles also reduces the systemic toxic effect of chemotherapeutic drugs. Due to their several unique intrinsic properties, nanoparticles itself can also be employed as therapeutics in cancer treatment by photothermal therapy (PTT) and photodynamic therapy (PDT). Thus, the main focus of this review is to emphasize on current progress in diagnostic and therapeutic application of nanoprobes in cancer.
Collapse
Affiliation(s)
- Ragini Singh
- School of Agriculture Science, Liaocheng University, No. 1 Hunan Road, Liaocheng, Shandong China
| |
Collapse
|
44
|
Zhu W, Niu J, He M, Zhang L, Lv X, Liu F, Jiang L, Zhang J, Yu Z, Zhao L, Bi J, Yan Y, Wei Q, Huo H, Fan Y, Chen Y, Ding J, Wei M. SNORD89 promotes stemness phenotype of ovarian cancer cells by regulating Notch1-c-Myc pathway. J Transl Med 2019; 17:259. [PMID: 31395064 PMCID: PMC6686521 DOI: 10.1186/s12967-019-2005-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/31/2019] [Indexed: 12/29/2022] Open
Abstract
Background Ovarian cancer is the leading cause of death in gynecological cancer. Cancer stem cells (CSCs) contribute to the occurrence, progression and resistance. Small nucleolar RNAs (SnoRNAs), a class of small molecule non-coding RNA, involve in the cancer cell stemness and tumorigenesis. Methods In this study, we screened out SNORNAs related to ovarian patient’s prognosis by analyzing the data of 379 cases of ovarian cancer patients in the TCGA database, and analyzed the difference of SNORNAs expression between OVCAR-3 (OV) sphere-forming (OS) cells and OV cells. After overexpression or knockdown SNORD89, the expression of Nanog, CD44, and CD133 was measured by qRT-PCR or flow cytometry analysis in OV, CAOV-3 (CA) and OS cells, respectively. CCK-8 assays, plate clone formation assay and soft agar colony formation assay were carried out to evaluate the changes of cell proliferation and self-renewal ability. Scratch migration assay and trans-well invasion analysis were used for assessing the changes of migration and invasion ability. Results High expression of SNORD89 indicates the poor prognosis of ovarian cancer patients and was associated with patients’ age, therapy outcome. SNORD89 highly expressed in ovarian cancer stem cells. The overexpression of SNORD89 resulted in the increased stemness markers, S phase cell cycle, cell proliferation, invasion and migration ability in OV and CA cells. Conversely, these phenomena were reversed after SNORD89 silencing in OS cells. Further, we found that SNORD89 could upregulate c-Myc and Notch1 expression in mRNA and protein levels. SNORD89 deteriorates the prognosis of ovarian cancer patients by regulating Notch1-c-Myc pathway to promote cell stemness and acts as an oncogene in ovarian tumorigenesis. Consequently, SNORD89 can be a novel prognostic biomarker and therapeutic target for ovarian cancer. Electronic supplementary material The online version of this article (10.1186/s12967-019-2005-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenjing Zhu
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Department of Pharmacy, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Jumin Niu
- Shenyang Women's and Children's Hospital, Shenyang, Liaoning, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Liwen Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Xuemei Lv
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Fangxiao Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Jing Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Jia Bi
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Yuanyuan Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Hong Huo
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Yue Fan
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China
| | - Yuzong Chen
- Bioinformatics and Drug Design Group, Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Jian Ding
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China. .,Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China. .,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
45
|
Geersing A, de Vries RH, Jansen G, Rots MG, Roelfes G. Folic acid conjugates of a bleomycin mimic for selective targeting of folate receptor positive cancer cells. Bioorg Med Chem Lett 2019; 29:1922-1927. [DOI: 10.1016/j.bmcl.2019.05.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
|
46
|
Kim HJ, Lee S, Oh YS, Chang HK, Kim YS, Hong SH, Kim JY, Park YW, Lee SJ, Song SW, Kim JJ, Heo K. Humanized Anti-hepatocyte Growth Factor Monoclonal Antibody (YYB-101) Inhibits Ovarian Cancer Progression. Front Oncol 2019; 9:571. [PMID: 31355133 PMCID: PMC6631954 DOI: 10.3389/fonc.2019.00571] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/12/2019] [Indexed: 11/29/2022] Open
Abstract
Current chemotherapy regimens have certain limitations in improving the survival rates of patients with advanced ovarian cancer. Hepatocyte growth factor (HGF) is important in ovarian cancer cell migration and invasion. This study assessed the effects of YYB-101, a humanized monoclonal anti-HGF antibody, on the growth and metastasis of ovarian cancer cells. YYB-101 suppressed the phosphorylation of the HGF receptor c-MET and inhibited the migration and invasion of SKOV3 and A2780 ovarian cancer cells. Moreover, the combination of YYB-101 and paclitaxel synergistically inhibited tumor growth in an in vivo ovarian cancer mouse xenograft model and significantly increased the overall survival (OS) rate compared with either paclitaxel or YYB-101 alone. Taken together, these findings suggest that YYB-101 has therapeutic potential in ovarian cancer when combined with conventional chemotherapy agents.
Collapse
Affiliation(s)
- Hyun Jung Kim
- Research Institute, National Cancer Center, Goyang-si, South Korea.,Department of Bioinspired Science, Ewha Womans University, Seoul, South Korea
| | - Sukmook Lee
- Department of Applied Chemistry, Kookmin University, Seoul, South Korea
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Ha Kyun Chang
- Center for Uterine Cancer, National Cancer Center, Research Institute and Hospital, Goyang-si, South Korea
| | - Young Sang Kim
- National OncoVenture, National Cancer Center, Goyang-si, South Korea
| | - Sung Hee Hong
- National OncoVenture, National Cancer Center, Goyang-si, South Korea.,Clinical Research Team, Hanmi Pharm. Co., Ltd., Seoul, South Korea
| | - Jung Yong Kim
- National OncoVenture, National Cancer Center, Goyang-si, South Korea
| | - Young-Whan Park
- National OncoVenture, National Cancer Center, Goyang-si, South Korea
| | - Song-Jae Lee
- Yooyoung Central Research Institute, Yooyoung Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Seong-Won Song
- Yooyoung Central Research Institute, Yooyoung Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Jung Ju Kim
- Yooyoung Central Research Institute, Yooyoung Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang-si, South Korea
| |
Collapse
|
47
|
Li X, Chen J, Liu H, Deng Z, Li J, Ren T, Huang L, Chen W, Yang Y, Zhong S. β-Cyclodextrin coated and folic acid conjugated magnetic halloysite nanotubes for targeting and isolating of cancer cells. Colloids Surf B Biointerfaces 2019; 181:379-388. [PMID: 31170644 DOI: 10.1016/j.colsurfb.2019.05.068] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/11/2019] [Accepted: 05/27/2019] [Indexed: 02/08/2023]
Abstract
The study developed a simple, effective and inexpensive strategy for capturing, enriching and detecting circulating tumor cells (CTCs) by using folic acid (FA) as the targeting molecule instead of antibodies. This work constructed magnetic halloysite nanotubes (MHNTs) coated with biocompatible β-cyclodextrin (CD), and conjugated to FA via a PEG-Ad linker, to specifically capture the FA receptor (FR)-overexpressing cancer cells. The capture efficiencies of MHNTs@β-CD@Ad-PEG-FA for the Skov3, Hela and A549 cancer cells were 96.3%, 97.0% and 95.6% respectively. In addition, the nanoparticles were able to capture very low numbers of the cancer cells (25-500 cells/mL) from PBS and whole blood, as well as selectively capture the cancer cells over normal HEK 293 T cells. Furthermore, the captured cells were viable and grew normally in vitro, indicating the future potential of downstream analyses. This approach can be adapted for different CTCs, once the tumor-specific surface markers are identified and the efficacy of targeting ligands is established. Taken together, FA-conjugated MHNTs nanoparticles are a highly promising tool for isolating CTCs for the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Xiufang Li
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Jian Chen
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Hui Liu
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Zhiwei Deng
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Jianbing Li
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Tao Ren
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Ling Huang
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Wenqing Chen
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Yanjing Yang
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China.
| | - Shian Zhong
- School of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China.
| |
Collapse
|
48
|
Targeted Delivery of Nanoparticulate Cytochrome C into Glioma Cells Through the Proton-Coupled Folate Transporter. Biomolecules 2019; 9:biom9040154. [PMID: 31003476 PMCID: PMC6523331 DOI: 10.3390/biom9040154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 01/14/2023] Open
Abstract
In this study, we identified the proton-coupled folate transporter (PCFT) as a route for targeted delivery of drugs to some gliomas. Using the techniques of confocal imaging, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and small interfering (siRNA) knockdown against the PCFT, we demonstrated that Gl261 and A172 glioma cells, but not U87 and primary cultured astrocytes, express the PCFT, which provides selective internalization of folic acid (FA)-conjugated cytochrome c-containing nanoparticles (FA-Cyt c NPs), followed by cell death. The FA-Cyt c NPs (100 µg/mL), had no cytotoxic effects in astrocytes but caused death in glioma cells, according to their level of expression of PCFT. Whole-cell patch clamp recording revealed FA-induced membrane currents in FA-Cyt c NPs-sensitive gliomas, that were reduced by siRNA PCFT knockdown in a similar manner as by application of FA-Cyt c NPs, indicating that the PCFT is a route for internalization of FA-conjugated NPs in these glioma cells. Analysis of human glioblastoma specimens revealed that at least 25% of glioblastomas express elevated level of either PCFT or folate receptor (FOLR1). We conclude that the PCFT provides a mechanism for targeted delivery of drugs to some gliomas as a starting point for the development of efficient methods for treating gliomas with high expression of PCFT and/or FOLR1.
Collapse
|
49
|
Birrer MJ, Betella I, Martin LP, Moore KN. Is Targeting the Folate Receptor in Ovarian Cancer Coming of Age? Oncologist 2019; 24:425-429. [PMID: 30635448 DOI: 10.1634/theoncologist.2018-0459] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/21/2018] [Indexed: 11/17/2022] Open
Abstract
Prognosis for women with epithelial ovarian cancer remains poor. One new molecular target in epithelial ovarian cancer is folate receptor alpha (FRα). This commentary discusses the characteristics that contribute to its attractiveness as a candidate for therapeutic intervention.
Collapse
Affiliation(s)
- Michael J Birrer
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ilaria Betella
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lainie P Martin
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathleen N Moore
- Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
50
|
Li S, Jiang J, Yan Y, Wang P, Huang G, Kim NH, Lee JH, He D. Red, green, and blue fluorescent folate-receptor-targeting carbon dots for cervical cancer cellular and tissue imaging. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:1054-1063. [DOI: 10.1016/j.msec.2018.08.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/31/2017] [Accepted: 08/29/2018] [Indexed: 10/28/2022]
|