1
|
Siatka T, Mát'uš M, Moravcová M, Harčárová P, Lomozová Z, Matoušová K, Suwanvecho C, Krčmová LK, Mladěnka P. Biological, dietetic and pharmacological properties of vitamin B 9. NPJ Sci Food 2025; 9:30. [PMID: 40075081 PMCID: PMC11904035 DOI: 10.1038/s41538-025-00396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Humans must obtain vitamin B9 (folate) from plant-based diet. The sources as well as the effect of food processing are discussed in detail. Industrial production, fortification and biofortification, kinetics, and physiological role in humans are described. As folate deficiency leads to several pathological states, current opinions toward prevention through fortification are discussed. Claimed risks of increased folate intake are mentioned as well as analytical ways for measurement of folate.
Collapse
Affiliation(s)
- Tomáš Siatka
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic
| | - Marek Mát'uš
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232, Bratislava, Slovak Republic
| | - Monika Moravcová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic
| | - Patrícia Harčárová
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic
| | - Zuzana Lomozová
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic
| | - Kateřina Matoušová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05, Hradec Králové, Czech Republic
| | - Chaweewan Suwanvecho
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05, Hradec Králové, Czech Republic
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic
| | - Lenka Kujovská Krčmová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05, Hradec Králové, Czech Republic
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 03, Hradec Králové, Czech Republic.
| |
Collapse
|
2
|
Alexander C, Guo Z, Glover PB, Faulkner S, Pikramenou Z. Luminescent Lanthanides in Biorelated Applications: From Molecules to Nanoparticles and Diagnostic Probes to Therapeutics. Chem Rev 2025; 125:2269-2370. [PMID: 39960048 PMCID: PMC11869165 DOI: 10.1021/acs.chemrev.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Lanthanides are particularly effective in their clinical applications in magnetic resonance imaging and diagnostic assays. They have open-shell 4f electrons that give rise to characteristic narrow, line-like emission which is unique from other fluorescent probes in biological systems. Lanthanide luminescence signal offers selection of detection pathways based on the choice of the ion from the visible to the near-infrared with long luminescence lifetimes that lend themselves to time-resolved measurements for optical multiplexing detection schemes and novel bioimaging applications. The delivery of lanthanide agents in cells allows localized bioresponsive activity for novel therapies. Detection in the near-infrared region of the spectrum coupled with technological advances in microscopies opens new avenues for deep-tissue imaging and surgical interventions. This review focuses on the different ways in which lanthanide luminescence can be exploited in nucleic acid and enzyme detection, anion recognition, cellular imaging, tissue imaging, and photoinduced therapeutic applications. We have focused on the hierarchy of designs that include luminescent lanthanides as probes in biology considering coordination complexes, multimetallic lanthanide systems to metal-organic frameworks and nanoparticles highlighting the different strategies in downshifting, and upconversion revealing some of the opportunities and challenges that offer potential for further development in the field.
Collapse
Affiliation(s)
- Carlson Alexander
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
- Department
of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Zhilin Guo
- Department
of Materials Science and Engineering, Southern
University of Science and Technology, Shenzhen 518055, China
| | - Peter B. Glover
- Defence
Science and Technology Laboratory (DSTL), Porton Down, Salisbury SP4 0JQ, United
Kingdom
| | - Stephen Faulkner
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Zoe Pikramenou
- School
of Chemistry, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
3
|
Jain A, Barge A, Parris CN. Combination strategies with PARP inhibitors in BRCA-mutated triple-negative breast cancer: overcoming resistance mechanisms. Oncogene 2025; 44:193-207. [PMID: 39572842 PMCID: PMC11746151 DOI: 10.1038/s41388-024-03227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 01/22/2025]
Abstract
Triple-negative breast cancer (TNBC) is a particularly aggressive breast cancer subtype, characterised by a higher incidence in younger women, rapid metastasis, and a generally poor prognosis. Patients with TNBC and BRCA mutations face additional therapeutic challenges due to the cancer's intrinsic resistance to conventional therapies. Poly (ADP-ribose) polymerase inhibitors (PARPis) have emerged as a promising targeted treatment for BRCA-mutated TNBC, exploiting vulnerabilities in the homologous recombination repair (HRR) pathway. However, despite initial success, the efficacy of PARPis is often compromised by the development of resistance mechanisms, including HRR restoration, stabilisation of replication forks, reduced PARP1 trapping, and drug efflux. This review explores latest breakthroughs in overcoming PARPi resistance through combination therapies. These strategies include the integration of PARPis with chemotherapy, immunotherapy, antibody-drug conjugates, and PI3K/AKT pathway inhibitors. These combinations aim to enhance the therapeutic efficacy of PARPis by targeting multiple cancer progression pathways. The review also discusses the evolving role of PARPis within the broader treatment paradigm for BRCA-mutated TNBC, emphasising the need for ongoing research and clinical trials to optimise combination strategies. By tackling the challenges associated with PARPi resistance and exploring novel combination therapies, this review sheds light on the future possibilities for improving outcomes for patients with BRCA-mutated TNBC.
Collapse
Affiliation(s)
- Aditi Jain
- Edinburgh Medical School: Biomedical Sciences, The University of Edinburgh, Edinburgh, UK.
| | | | | |
Collapse
|
4
|
Chen Y, Yang SY, Ou X, Wang H, Kong FC, Chow PCY, Wang Y, Jiang Y, Zhao W, Sun J, Kwok RTK, Zheng DW, Yu W, Wang F, Lam JWY, Tang BZ. Engineering a Near-Infrared Spiro-Based Aggregation-Induced Emission Luminogen for DNAzyme-Sensitized Photothermal Therapy with High Efficiency and Accuracy. J Am Chem Soc 2024; 146:35462-35477. [PMID: 39665333 DOI: 10.1021/jacs.4c14818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Aggregation-induced emission luminogen (AIEgens)-based photothermal therapy (PTT) has grown into a sparkling frontier for tumor ablation. However, challenges remain due to the uncoordinated photoluminescence (PL) and photothermal properties of classical AIEgens, along with hyperthermia-induced antiapoptotic responses in tumor cells, hindering satisfactory therapeutic outcomes. Herein, a near-infrared (NIR) spiro-AIEgen TTQ-SA was designed for boosted PTT by auxiliary DNAzyme-regulated tumor cell sensitization. TTQ-SA with a unique molecular structure and packing mode was initially fabricated, endowing it with a strong AIE effect, favorable PL quantum yield, and good photothermal performance. DNAzyme, as a gene silencing tool, could alleviate antiapoptosis response during PTT. By integrating TTQ-SA and DNAzyme into folate-modified poly(lactic-co-glycolic acid) (PLGA) polymer, the as-fabricated nanosystem could promote cell apoptosis and sensitize tumor cells to PTT, thereby maximizing the therapeutic outcomes. With the combination of spiro-AIEgen-based PTT and DNAzyme-based gene silencing, the as-designed nanosystem showed promising NIR and photothermal imaging abilities for tumor targeting and demonstrated significant cell apoptotic, antitumor, and antimetastasis effects against orthotopic breast cancer. Furthermore, a synergistic antitumor effect was realized in spontaneous MMTV-PyMT transgenic mice. These findings offer new insights into AIEgen-based photothermal theranostics and DNAzyme-regulated tumor cell sensitization, paving the way for synergistic gene silencing-PTT nanoplatforms in clinical research.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Sheng-Yi Yang
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Xinwen Ou
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Hui Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Fan-Cheng Kong
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam 999077, Hong Kong SAR, China
| | - Philip C Y Chow
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam 999077, Hong Kong SAR, China
| | - Yifei Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Yuqian Jiang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Wei Zhao
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City 510280, Guangdong, China
| | - Jianwei Sun
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Ryan T K Kwok
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Di-Wei Zheng
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenqian Yu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Fuan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Jacky W Y Lam
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Ben Zhong Tang
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen 518172, Guangdong, China
| |
Collapse
|
5
|
Oliveira I, Rodrigues-Santos P, Ferreira L, Pires das Neves R. Synthetic and biological nanoparticles for cancer immunotherapy. Biomater Sci 2024; 12:5933-5960. [PMID: 39441658 DOI: 10.1039/d4bm00995a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Cancer is becoming the main public health problem globally. Conventional chemotherapy approaches are slowly being replaced or complemented by new therapies that avoid the loss of healthy tissue, limit off-targets, and eradicate cancer cells. Immunotherapy is nowadays an important strategy for cancer treatment, that uses the host's anti-tumor response by activating the immune system and increasing the effector cell number, while, minimizing cancer's immune-suppressor mechanisms. Its efficacy is still limited by poor therapeutic targeting, low immunogenicity, antigen presentation deficiency, impaired T-cell trafficking and infiltration, heterogeneous microenvironment, multiple immune checkpoints and unwanted side effects, which could benefit from improved delivery systems, able to release immunotherapeutic agents to tumor microenvironment and immune cells. Nanoparticles (NPs) for immunotherapy (Nano-IT), have a huge potential to solve these limitations. Natural and/or synthetic, targeted and/or stimuli-responsive nanoparticles can be used to deliver immunotherapeutic agents in their native conformations to the site of interest to enhance their antitumor activity. They can also be used as co-adjuvants that enhance the activity of IT effector cells. These nanoparticles can be engineered in the natural context of cell-derived extracellular vesicles (EVs) or exosomes or can be fully synthetic. In this review, a detailed SWOT analysis is done through the comparison of engineered-synthetic and naturaly-derived nanoparticles in terms of their current and future use in cancer immunotherapy.
Collapse
Affiliation(s)
- Inês Oliveira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Paulo Rodrigues-Santos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
6
|
Marshall RF, Xu H, Berkenstock M. Ocular toxicities associated with antibody drug conjugates. Curr Opin Ophthalmol 2024; 35:494-498. [PMID: 38814581 DOI: 10.1097/icu.0000000000001063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW To review the structure, mechanism of action, and pathophysiology of antibody-drug conjugates (ADCs) used to treat gynecological malignancies associated with ocular adverse effects. RECENT FINDINGS Recent research shows tisotumab vedotin causes ocular toxicity localized to the conjunctiva, with common adverse effects being conjunctivitis, dry eye, blepharitis, and keratitis. Toxicity is likely due to targeting tissue factor (TF) in the conjunctiva, leading to direct delivery of the cytotoxic payload resulting in apoptosis and bystander killing. Mirvetuximab soravtansine causes blurred vision, keratitis, or dry eye with toxicity often localized in the cornea. Off-target inflammation appears to cause ocular adverse effects, with nonreceptor mediated macropinocytosis by corneal stem cells. SUMMARY Collaboration between oncologists and ophthalmologists with adherence to mitigation protocols can decrease the risk of ocular adverse events.
Collapse
Affiliation(s)
- Rayna F Marshall
- Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Hannah Xu
- University of California San Diego, San Diego, California
| | - Meghan Berkenstock
- Division of Ocular Immunology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Kesharwani P, Halwai K, Jha SK, Al Mughram MH, Almujri SS, Almalki WH, Sahebkar A. Folate-engineered chitosan nanoparticles: next-generation anticancer nanocarriers. Mol Cancer 2024; 23:244. [PMID: 39482651 PMCID: PMC11526716 DOI: 10.1186/s12943-024-02163-z] [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: 05/03/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
Chitosan nanoparticles (NPs) are well-recognized as promising vehicles for delivering anticancer drugs due to their distinctive characteristics. They have the potential to enclose hydrophobic anticancer molecules, thereby enhancing their solubilities, permeabilities, and bioavailabilities; without the use of surfactant, i.e., through surfactant-free solubilization. This allows for higher drug concentrations at the tumor sites, prevents excessive toxicity imparted by surfactants, and could circumvent drug resistance. Moreover, biomedical engineers and formulation scientists can also fabricate chitosan NPs to slowly release anticancer agents. This keeps the drugs at the tumor site longer, makes therapy more effective, and lowers the frequency of dosing. Notably, some types of cancer cells (fallopian tube, epithelial tumors of the ovary, and primary peritoneum; lung, kidney, ependymal brain, uterus, breast, colon, and malignant pleural mesothelioma) have overexpression of folate receptors (FRs) on their outer surface, which lets folate-drug conjugate-incorporated NPs to target and kill them more effectively. Strikingly, there is evidence suggesting that the excessively produced FR&αgr (isoforms of the FR) stays consistent throughout treatment in ovarian and endometrial cancer, indicating resistance to conventional treatment; and in this regard, folate-anchored chitosan NPs can overcome it and improve the therapeutic outcomes. Interestingly, overly expressed FRs are present only in certain tumor types, which makes them a promising biomarker for predicting the effectiveness of FR-targeted therapy. On the other hand, the folate-modified chitosan NPs can also enhance the oral absorption of medicines, especially anticancer drugs, and pave the way for effective and long-term low-dose oral metronomic scheduling of poorly soluble and permeable drugs. In this review, we talked briefly about the techniques used to create, characterize, and tailor chitosan-based NPs; and delved deeper into the potential applications of folate-engineered chitosan NPs in treating various cancer types.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Kratika Halwai
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Uttar Pradesh, Kanpur, 208016, India
| | - Mohammed H Al Mughram
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Postal Code 61421, Abha, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Postal Code 61421, Abha, Saudi Arabia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Liu K, Li M, Li Y, Li Y, Chen Z, Tang Y, Yang M, Deng G, Liu H. A review of the clinical efficacy of FDA-approved antibody‒drug conjugates in human cancers. Mol Cancer 2024; 23:62. [PMID: 38519953 PMCID: PMC10960395 DOI: 10.1186/s12943-024-01963-7] [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: 11/24/2023] [Accepted: 02/21/2024] [Indexed: 03/25/2024] Open
Abstract
While strategies such as chemotherapy and immunotherapy have become the first-line standard therapies for patients with advanced or metastatic cancer, acquired resistance is still inevitable in most cases. The introduction of antibody‒drug conjugates (ADCs) provides a novel alternative. ADCs are a new class of anticancer drugs comprising the coupling of antitumor mAbs with cytotoxic drugs. Compared with chemotherapeutic drugs, ADCs have the advantages of good tolerance, accurate target recognition, and small effects on noncancerous cells. ADCs occupy an increasingly important position in the therapeutic field. Currently, there are 13 Food and Drug Administration (FDA)‒approved ADCs and more than 100 ADC drugs at different stages of clinical trials. This review briefly describes the efficacy and safety of FDA-approved ADCs, and discusses the related problems and challenges to provide a reference for clinical work.
Collapse
Affiliation(s)
- Kaifeng Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meijia Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yudong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yutong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Zixin Chen
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yiqi Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meitian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Guoquan Deng
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Hongwei Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China.
| |
Collapse
|
9
|
García-Castro M, Fuentes-Rios D, López-Romero JM, Romero A, Moya-Utrera F, Díaz-Morilla A, Sarabia F. n-Tuples on Scaffold Diversity Inspired by Drug Hybridisation to Enhance Drugability: Application to Cytarabine. Mar Drugs 2023; 21:637. [PMID: 38132958 PMCID: PMC10744741 DOI: 10.3390/md21120637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
A mathematical concept, n-tuples are originally applied to medicinal chemistry, especially with the creation of scaffold diversity inspired by the hybridisation of different commercial drugs with cytarabine, a synthetic arabinonucleoside derived from two marine natural products, spongouridine and spongothymidine. The new methodology explores the virtual chemical-factorial combination of different commercial drugs (immunosuppressant, antibiotic, antiemetic, anti-inflammatory, and anticancer) with the anticancer drug cytarabine. Real chemical combinations were designed and synthesised for 8-duples, obtaining a small representative library of interesting organic molecules to be biologically tested as proof of concept. The synthesised library contains classical molecular properties regarding the Lipinski rules and/or beyond rules of five (bRo5) and is represented by the covalent combination of the anticancer drug cytarabine with ibuprofen, flurbiprofen, folic acid, sulfasalazine, ciprofloxacin, bortezomib, and methotrexate. The insertion of specific nomenclature could be implemented into artificial intelligence algorithms in order to enhance the efficiency of drug-hunting programs. The novel methodology has proven useful for the straightforward synthesis of most of the theoretically proposed duples and, in principle, could be extended to any other central drug.
Collapse
Affiliation(s)
- Miguel García-Castro
- Department of Organic Chemistry, Faculty of Sciences, University of Malaga, Campus de Teatinos s/n, 29071 Malaga, Spain
| | | | | | | | | | | | | |
Collapse
|
10
|
Srivastava N, Chudasama B, Baranwal M. Advancement in magnetic hyperthermia-based targeted therapy for cancer treatment. Biointerphases 2023; 18:060801. [PMID: 38078795 DOI: 10.1116/6.0003079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Magnetic hyperthermia utilizing magnetic nanoparticles (MNPs) and an alternating magnetic field (AMF) represents a promising approach in the field of cancer treatment. Active targeting has emerged as a valuable strategy to enhance the effectiveness and specificity of drug delivery. Active targeting utilizes specific biomarkers that are predominantly found in abundance on cancer cells while being minimally expressed on healthy cells. Current comprehensive review provides an overview of several cancer-specific biomarkers, including human epidermal growth factor, transferrin, folate, luteinizing hormone-releasing hormone, integrin, cluster of differentiation (CD) receptors such as CD90, CD95, CD133, CD20, and CD44 also CXCR4 and vascular endothelial growth factor, these biomarkers bind to ligands present on the surface of MNPs, enabling precise targeting. Additionally, this review touches various combination therapies employed to combat cancer. Magnetic hyperthermia synergistically enhances the efficacy of conventional cancer treatments such as targeted chemotherapy, radiation therapy, gene therapy, and immunotherapy.
Collapse
Affiliation(s)
- Neha Srivastava
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Bhupendra Chudasama
- School of Physics and Materials Science, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| |
Collapse
|
11
|
Zhang Y, Zhang X, Chen J, Jiang S, Han Y, Du H. Maternal Folic Acid Supplementation Improves the Intestinal Health of Offspring Porcine by Promoting the Proliferation and Differentiation of Intestinal Stem Cells. Animals (Basel) 2023; 13:3092. [PMID: 37835698 PMCID: PMC10571947 DOI: 10.3390/ani13193092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Maternal folic acid intake has important effects on offspring growth and development. The mechanism involved in the renewal of intestinal epithelial cells remains unclear. Thus, this study aimed to investigate the potential effect of maternal folic acid supplementation during gestation and lactation on the structural and functional development of the small intestine in piglet offspring. Twenty-four Duroc sows were assigned to a control group (CON) and a folic-acid-supplemented group (CON + FA, supplemented with 15 mg/kg of folic acid). The results showed that maternal folic acid supplementation throughout gestation and lactation significantly increased the body weight, serum folate level, and intestinal folate metabolism in piglets. It also improved the villus length, villus height-to-crypt depth ratio, and transcript levels of nutrient transporters (GLUT4, SNAT2, FABP2, and SLC7A5) in piglets' duodenum and jejunum. In addition, maternal folic acid supplementation increased Ki67-positive cells and the expression of proliferation-related marker genes (C-Myc, CyclinD1, and PCNA) in piglets' intestinal stem cells. It also boosted the expression of genes associated with mature secreted cells (ChrA, Muc2, Lyz, Vil1), indicating enhanced proliferation and differentiation of intestinal stem cells. These findings demonstrate that maternal folic acid supplementation enhances growth performance and gut health in piglet offspring by promoting epithelial cell renewal equilibrium.
Collapse
Affiliation(s)
- Yuhui Zhang
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaofeng Zhang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310004, China
| | - Jianjun Chen
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shouchuan Jiang
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yu Han
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huahua Du
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
12
|
Rizzo C, Cancemi P, Buttacavoli M, Di Cara G, D'Amico C, Billeci F, Marullo S, D'Anna F. Insights about the ability of folate based supramolecular gels to act as targeted therapeutic agents. J Mater Chem B 2023; 11:7721-7738. [PMID: 37466082 DOI: 10.1039/d3tb01389h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
With the aim to obtain targeted chemotherapeutic agents, imidazolium and ammonium-based folate salts were synthesized. Their photophysical behavior was investigated both in buffer and buffer/DMSO solution as well as in solid phase, performing UV-vis and fluorescence investigations. Properties of the aggregates were also analyzed by dynamic light scattering. Gelation ability of the salts was analyzed in biocompatible solvents, and gel phases obtained were characterized by determining critical gelation concentrations and gel-solution transition temperatures. Insights about gelator interactions in the tridimensional network were also gained performing ATR-FTIR investigation. Properties of soft materials were further analyzed performing rheology measurements, scanning electron microscopy, fluorescence and resonance light scattering investigations. Antiproliferative activity of organic salts was tested towards two breast cancer cell lines, expressing different levels of folate receptor, namely MDA-MB-231 and MCF-7, and a normal epithelial cell line, like h-TER T-RPE-1, by using MTT assay. Dichlodihydrofluorescein acetate test was performed to verify the role of oxidative stress in cell death. Finally, antiproliferative activity was also evaluated in gel phase, to verify if salts were able to retain biological activity also after the entrapment in the gelatinous network. Results collected evidence that folate based organic salts were able to behave as targeted chemotherapeutic agents both in solution and gel phase, showing uptake mechanism and selectivity indexes that depend on both cancer cell line nature and salt structure.
Collapse
Affiliation(s)
- Carla Rizzo
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| | - Patrizia Cancemi
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Miriam Buttacavoli
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Gianluca Di Cara
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Cesare D'Amico
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Floriana Billeci
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| | - Salvatore Marullo
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| | - Francesca D'Anna
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| |
Collapse
|
13
|
Busslinger SD, Becker AE, Vaccarin C, Deberle LM, Renz ML, Groehn V, Schibli R, Müller C. Investigations Using Albumin Binders to Modify the Tissue Distribution Profile of Radiopharmaceuticals Exemplified with Folate Radioconjugates. Cancers (Basel) 2023; 15:4259. [PMID: 37686538 PMCID: PMC10486429 DOI: 10.3390/cancers15174259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Introducing an albumin-binding entity into otherwise short-lived radiopharmaceuticals can be an effective means to improve their pharmacokinetic properties due to enhanced blood residence time. In the current study, DOTA-derivatized albumin binders based on 4-(p-iodophenyl)butanoate (DOTA-ALB-1 and DOTA-ALB-3) and 5-(p-iodophenyl)pentanoate entities (DOTA-ALB-24 and DOTA-ALB-25) without and with a hydrophobic 4-(aminomethyl)benzoic acid (AMBA) linker unit, respectively, were synthesized and labeled with lutetium-177 for in vitro and in vivo comparison. Overall, [177Lu]Lu-DOTA-ALB-1 demonstrated ~3-fold stronger in vitro albumin-binding affinity and a longer blood residence time (T50%IA ~8 h) than [177Lu]Lu-DOTA-ALB-24 (T50%IA ~0.8 h). Introducing an AMBA linker enhanced the albumin-binding affinity, resulting in a T50%IA of ~24 h for [177Lu]Lu-DOTA-ALB-3 and ~2 h for [177Lu]Lu-DOTA-ALB-25. The same albumin binders without or with the AMBA linker were incorporated into 6R- and 6S-5-methyltetrahydrofolate-based DOTA-conjugates (177Lu-RedFols). Biodistribution studies in mice performed with both diastereoisomers of [177Lu]Lu-RedFol-1 and [177Lu]Lu-RedFol-3, which comprised the 4-(p-iodophenyl)butanoate moiety, demonstrated a slower accumulation in KB tumors than those of [177Lu]Lu-RedFol-24 and [177Lu]Lu-RedFol-25 with the 5-(p-iodophenyl)pentanoate entity. In all cases, the tumor uptake was high (30-45% IA/g) 24 h after injection. Both diastereoisomers of [177Lu]Lu-RedFol-1 and [177Lu]Lu-RedFol-3 demonstrated high blood retention (3.8-8.7% IA/g, 24 h p.i.) and a 2- to 4-fold lower kidney uptake than the corresponding diastereoisomers of [177Lu]Lu-RedFol-24 and [177Lu]Lu-RedFol-25, which were more rapidly cleared from the blood (<0.2% IA/g, 24 h after injection). Kidney retention of the 6S-diastereoisomers of all 177Lu-RedFols was consistently higher than that of the respective 6R-diastereoisomers, irrespective of the albumin binder and linker unit used. It was demonstrated that the blood clearance data obtained with 177Lu-DOTA-ALBs had predictive value for the blood retention times of the respective folate radioconjugates. The use of these albumin-binding entities without or with an AMBA linker may serve for fine-tuning the blood retention of folate radioconjugates and also other radiopharmaceuticals and, hence, optimize their tissue distribution profiles. Dosimetry estimations based on patient data obtained with one of the most promising folate radioconjugates will be crucial to identify the dose-limiting organ, which will allow for selecting the most suitable folate radioconjugate for therapeutic purposes.
Collapse
Affiliation(s)
- Sarah D. Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Anna E. Becker
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Christian Vaccarin
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Luisa M. Deberle
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Marie-Luise Renz
- Merck & Cie KmG, Im Laternenacker 5, 8200 Schaffhausen, Switzerland; (M.-L.R.); (V.G.)
| | - Viola Groehn
- Merck & Cie KmG, Im Laternenacker 5, 8200 Schaffhausen, Switzerland; (M.-L.R.); (V.G.)
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
14
|
Porter RL, Matulonis UA. Mirvetuximab soravtansine for platinum-resistant epithelial ovarian cancer. Expert Rev Anticancer Ther 2023; 23:783-796. [PMID: 37458180 DOI: 10.1080/14737140.2023.2236793] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/20/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
INTRODUCTION Mirvetuximab soravtansine (mirvetuximab) is an antibody drug conjugate (ADC) comprised of a humanized folate receptor alpha (FRα)-binding monoclonal antibody attached via a cleavable linker to the cytotoxic maytansinoid molecule, DM4. FRα is expressed in several epithelial cancers, including high grade serous ovarian cancer (HGSOC). Mirvetuximab received accelerated approval by the United States Food and Drug Administration (FDA) in November 2022 based on the results of the SORAYA trial, which tested mirvetuximab for the treatment of patients with recurrent platinum resistant HGSOC with high FRα expression and showed an overall response rate (ORR) of 32.4% and a median duration of response of 6.9 months. Mirvetuximab toxicities included low grade ocular and gastrointestinal toxicities. The National Comprehensive Cancer Network (NCCN) ovarian cancer 2023 guidelines adopted mirvetuximab as 2A, and mirvetuximab combined with bevacizumab as 2B, recommendations. AREAS COVERED This manuscript will review the preclinical and clinical development of mirvetuximab, the toxicities associated with mirvetuximab and mitigation strategies, and future applications of mirvetuximab. EXPERT OPINION Mirvetuximab represents the first biomarker-directed therapy with an indication specifically for the treatment of PROC. The efficacy and favorable safety profile support further development of mirvetuximab and mirvetuximab combinations in platinum sensitive and newly diagnosed ovarian cancer.
Collapse
Affiliation(s)
- Rebecca L Porter
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, United States of America
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, United States of America
| |
Collapse
|
15
|
Varaganti P, Buddolla V, Lakshmi BA, Kim YJ. Recent advances in using folate receptor 1 (FOLR1) for cancer diagnosis and treatment, with an emphasis on cancers that affect women. Life Sci 2023:121802. [PMID: 37244363 DOI: 10.1016/j.lfs.2023.121802] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
A glycosylphosphatidylinositol (GPI)-anchored glycoprotein called the folate receptor 1 (FOLR1) facilitates the transportation of folate by mediating receptor-mediated endocytosis in response to ligand binding. While FOLR1 expression is typically restricted to the apical surfaces of the epithelium in the lung, kidney, and choroid plexus in healthy people, it is overexpressed in a number of solid tumours, including high-grade osteosarcoma, breast cancer, ovarian cancer, and non-small cell lung cancer. As a result, FOLR1 has become an attractive target for cancer detection and therapy, particularly for cancers that affect women. A number of methods have been developed to target FOLR1 in cancer therapy, including the development of FOLR1-targeted imaging agents for cancer diagnosis and the use of folate conjugates to deliver cytotoxic agents to cancer cells that overexpress FOLR1. Therefore, we focus on the most recent developments in employing FOLR1 for cancer diagnosis and treatment in this review, particularly with regard to cancers that affect women.
Collapse
Affiliation(s)
- Pavitra Varaganti
- Dr. Buddolla's Institute of Life Sciences, Tirupati 517506, Andhra Pradesh, India
| | - Viswanath Buddolla
- Dr. Buddolla's Institute of Life Sciences, Tirupati 517506, Andhra Pradesh, India
| | - Buddolla Anantha Lakshmi
- Department of Electronic Engineering, Gachon University, 1342 Seongnam-Daero, Seongnam, Gyeonggi-Do 13120, Republic of Korea
| | - Young-Joon Kim
- Department of Electronic Engineering, Gachon University, 1342 Seongnam-Daero, Seongnam, Gyeonggi-Do 13120, Republic of Korea.
| |
Collapse
|
16
|
Graván P, Aguilera-Garrido A, Marchal JA, Navarro-Marchal SA, Galisteo-González F. Lipid-core nanoparticles: Classification, preparation methods, routes of administration and recent advances in cancer treatment. Adv Colloid Interface Sci 2023; 314:102871. [PMID: 36958181 DOI: 10.1016/j.cis.2023.102871] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Nanotechnological drug delivery platforms represent a new paradigm for cancer therapeutics as they improve the pharmacokinetic profile and distribution of chemotherapeutic agents over conventional formulations. Among nanoparticles, lipid-based nanoplatforms possessing a lipid core, that is, lipid-core nanoparticles (LCNPs), have gained increasing interest due to lipid properties such as high solubilizing potential, versatility, biocompatibility, and biodegradability. However, due to the wide spectrum of morphologies and types of LCNPs, there is a lack of consensus regarding their terminology and classification. According to the current state-of-the-art in this critical review, LCNPs are defined and classified based on the state of their lipidic components in liquid lipid nanoparticles (LLNs). These include lipid nanoemulsions (LNEs) and lipid nanocapsules (LNCs), solid lipid nanoparticles (SLNs) and nanostructured lipid nanocarriers (NLCs). In addition, we present a comprehensive and comparative description of the methods employed for their preparation, routes of administration and the fundamental role of physicochemical properties of LCNPs for efficient antitumoral drug-delivery application. Market available LCNPs, clinical trials and preclinical in vivo studies of promising LCNPs as potential treatments for different cancer pathologies are summarized.
Collapse
Affiliation(s)
- Pablo Graván
- Department of Applied Physics, Faculty of Science, University of Granada, 18071 Granada, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria de Granada ibs.GRANADA, 18012 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; Excellence Research Unit Modelling Nature (MNat), University of Granada, 18016 Granada, Spain; BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18100 Granada, Spain
| | - Aixa Aguilera-Garrido
- Department of Applied Physics, Faculty of Science, University of Granada, 18071 Granada, Spain
| | - Juan Antonio Marchal
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria de Granada ibs.GRANADA, 18012 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; Excellence Research Unit Modelling Nature (MNat), University of Granada, 18016 Granada, Spain; BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18100 Granada, Spain
| | - Saúl A Navarro-Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; Excellence Research Unit Modelling Nature (MNat), University of Granada, 18016 Granada, Spain; Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, EH4 2XU Edinburgh, UK.
| | | |
Collapse
|
17
|
Young O, Ngo N, Lin L, Stanbery L, Creeden JF, Hamouda D, Nemunaitis J. Folate Receptor as a Biomarker and Therapeutic Target in Solid Tumors. Curr Probl Cancer 2023; 47:100917. [PMID: 36508886 DOI: 10.1016/j.currproblcancer.2022.100917] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022]
Abstract
Folate is a B vitamin necessary for basic biological functions, including rapid cell turnover occurring in cancer cell proliferation. Though the role of folate as a causative versus protective agent in carcinogenesis is debated, several studies have indicated that the folate receptor (FR), notably subtype folate receptor alpha (FRα), could be a viable biomarker for diagnosis, progression, and prognosis. Several cancers, including gastrointestinal, gynecological, breast, lung, and squamous cell head and neck cancers overexpress FR and are currently under investigation to correlate receptor status to disease state. Traditional chemotherapies have included antifolate medications, such as methotrexate and pemetrexed, which generate anticancer activity during the synthesis phase of the cell cycle. Increasingly, the repertoire of pharmacotherapies is expanding to include FR as a target, with a heterogenous pool of directed therapies. Here we discuss the FR, expression and effect in cancer biology, and relevant pharmacologic inhibitors.
Collapse
Affiliation(s)
- Olivia Young
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nealie Ngo
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Leslie Lin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | | - Justin Fortune Creeden
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Danae Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | |
Collapse
|
18
|
Tymon-Rosario J, Gorman M, Richardson DL, Washington C, Santin AD. Advances in antibody-drug conjugates for gynecologic malignancies. Curr Opin Obstet Gynecol 2023; 35:6-14. [PMID: 36484278 DOI: 10.1097/gco.0000000000000838] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) represent a new class of drugs that combine a surface receptor-targeting antibody linked to a cytotoxic molecule delivering the potent cytotoxic payload directly to tumor cells. This review summarizes the current literature demonstrating their use in the treatment of gynecologic malignancies. RECENT FINDINGS Tisotumab vedotin is the first U.S. Food and Drug Administration (FDA) approved ADC for the treatment of gynecologic cancers. While in the phase 3 randomized controlled trial in platinum resistant ovarian cancer patients, FORWARD 1, mirvetuximab did not meet its primary endpoint of progression-free survival. But we await more recent data from the two ongoing phase 3 trials of mirvetuximab in recurrent ovarian cancer patients. HER2/neu, Napi2b, mesothelin, and human trophoblast cell-surface marker (Trop-2) overexpression have also been exploited as excellent targets by novel ADCs in multiple tumors including ovarian, endometrial, and cervical cancers. SUMMARY Current evidence strongly supports the use of ADCs and ongoing clinical trials will provide further information into the potential of making these drugs part of current standard practice allowing patients to be treated with a higher level of personalized cancer care.
Collapse
Affiliation(s)
- Joan Tymon-Rosario
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New Hyde Park, New York 11040
| | - Megan Gorman
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New Hyde Park, New York 11040
| | - Debra L Richardson
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, Connecticut 06520
| | - Christina Washington
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, Connecticut 06520
| | - Alessandro D Santin
- Stephenson Cancer Center at the University of Oklahoma Health Sciences Center, 800 N.E. 10 Street, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
19
|
Sharma A, Shambhwani D, Pandey S, Singh J, Lalhlenmawia H, Kumarasamy M, Singh SK, Chellappan DK, Gupta G, Prasher P, Dua K, Kumar D. Advances in Lung Cancer Treatment Using Nanomedicines. ACS OMEGA 2023; 8:10-41. [PMID: 36643475 PMCID: PMC9835549 DOI: 10.1021/acsomega.2c04078] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/13/2022] [Indexed: 06/01/2023]
Abstract
Carcinoma of the lungs is among the most menacing forms of malignancy and has a poor prognosis, with a low overall survival rate due to delayed detection and ineffectiveness of conventional therapy. Therefore, drug delivery strategies that may overcome undesired damage to healthy cells, boost therapeutic efficacy, and act as imaging tools are currently gaining much attention. Advances in material science have resulted in unique nanoscale-based theranostic agents, which provide renewed hope for patients suffering from lung cancer. Nanotechnology has vastly modified and upgraded the existing techniques, focusing primarily on increasing bioavailability and stability of anti-cancer drugs. Nanocarrier-based imaging systems as theranostic tools in the treatment of lung carcinoma have proven to possess considerable benefits, such as early detection and targeted therapeutic delivery for effectively treating lung cancer. Several variants of nano-drug delivery agents have been successfully studied for therapeutic applications, such as liposomes, dendrimers, polymeric nanoparticles, nanoemulsions, carbon nanotubes, gold nanoparticles, magnetic nanoparticles, solid lipid nanoparticles, hydrogels, and micelles. In this Review, we present a comprehensive outline on the various types of overexpressed receptors in lung cancer, as well as the various targeting approaches of nanoparticles.
Collapse
Affiliation(s)
- Akshansh Sharma
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| | | | - Sadanand Pandey
- Department
of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, Gyeongbuk 38541, South Korea
| | - Jay Singh
- Department
of Chemistry, Institute of Science, Banaras
Hindu University, Varanasi 221005, India
| | - Hauzel Lalhlenmawia
- Department
of Pharmacy, Regional Institute of Paramedical
and Nursing Sciences, Zemabawk, Aizawl, Mizoram 796017, India
| | - Murali Kumarasamy
- Department
of Biotechnology, National Institute of
Pharmaceutical Education and Research, Hajipur 844102, India
| | - Sachin Kumar Singh
- School
of Pharmaceutical Sciences, Lovely Professional
University, Phagwara 144411, India
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department
of Life Sciences, School of Pharmacy, International
Medical University, Kuala Lumpur 57000, Malaysia
| | - Gaurav Gupta
- Department
of Pharmacology, School of Pharmacy, Suresh
Gyan Vihar University, Jaipur 302017, India
- Department
of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical
and Technical Sciences, Saveetha University, Chennai 602117, India
- Uttaranchal
Institute of Pharmaceutical Sciences, Uttaranchal
University, Dehradun 248007, India
| | - Parteek Prasher
- Department
of Chemistry, University of Petroleum &
Energy Studies, Dehradun 248007, India
| | - Kamal Dua
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Discipline
of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Deepak Kumar
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| |
Collapse
|
20
|
Elevated expression of receptors for EGF, PDGF, transferrin and folate within murine and human lupus nephritis kidneys. Clin Immunol 2023; 246:109188. [PMID: 36396012 DOI: 10.1016/j.clim.2022.109188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a chronic autoimmune disease where the body's immune system targets cells and tissue in numerous organs, including the kidneys. Lupus nephritis (LN) is a highly heterogeneous disease, and diagnosis is difficult because clinical manifestations vary widely among patients. Comprehensive proteomic studies reported recently in LN have identified several urinary proteins which are also cell-surface receptors. If indeed these receptor proteins are also hyper-expressed within the kidneys, ligands to these receptors may be useful for drug targeting. METHODS scRNA sequence data analysis and immunohistochemistry were performed on LN kidneys for expression of four implicated receptors, EGFR, FOL2R2, PDGF-RB, and TFRC. RESULTS In reported scRNA sequencing studies from 21 LN patients and 3 healthy control renal biopsies or renal-infiltrating immune cells from 24 LN biopsies, EGFR, FOLR2, PDGF-Rb, and TFRC were all hyper expressed within LN kidneys in comparison to healthy kidneys, either within resident renal cells or infiltrating leukocytes. Immunohistochemistry staining of murine lupus renal biopsies from lupus mice revealed EGFR, FOLR2, TFRC and PDGF-RB were elevated in LN kidneys. Immunohistochemistry staining of human Class II, Class III, and Class IV kidney tissue sections revealed EGFR, TFRC, and PDGF-RB were significantly elevated in proliferative LN kidneys. CONCLUSION These findings underscore the potential of EGFR, TFRC, FOLR2, and PDGF-RB as promising receptors for potential drug-targeting in LN.
Collapse
|
21
|
Wang AJ, Gao Y, Shi YY, Dai MY, Cai HB. A review of recent advances on single use of antibody-drug conjugates or combination with tumor immunology therapy for gynecologic cancer. Front Pharmacol 2022; 13:1093666. [PMID: 36618922 PMCID: PMC9813853 DOI: 10.3389/fphar.2022.1093666] [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: 11/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint inhibitors have made significant progress in the treatment of various cancers. However, due to the low ICI responsive rate for the gynecologic cancer, ICI two-drug combination therapy tends to be a predominant way for clinical treatment. Antibody-drug conjugates, a promising therapeutic modality for cancer, have been approved by the FDA for breast cancer, lymphoma, multiple myeloma and gastric cancer. On September 2021, the FDA granted accelerated approval to tisotumab vedotin for patients with recurrent or metastatic cervical cancer. Currently, the role of therapy of ADCs on gynecologic tumors was also included in medication regimens. Now more than 30 ADCs targeting for 20 biomarkers are under clinical trials in the field, including monotherapy or combination with others for multiple lines of therapy. Some ADCs have been proved to enhance the antitumor immunity effect on both pre-clinical models and clinical trials. Therefore, combination of ADCs and ICIs are expected in clinical trials. In this review, we discuss current development of ADCs in gynecologic oncology and the combination effects of ICIs and ADCs.
Collapse
Affiliation(s)
- An-Jin Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, Hubei, China,Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Yang Gao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, Hubei, China,Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Yu-Ying Shi
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, Hubei, China,Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Meng-Yuan Dai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, Hubei, China,Hubei Cancer Clinical Study Center, Wuhan, Hubei, China,*Correspondence: Meng-Yuan Dai, ; Hong-Bing Cai,
| | - Hong-Bing Cai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, Hubei, China,Hubei Cancer Clinical Study Center, Wuhan, Hubei, China,*Correspondence: Meng-Yuan Dai, ; Hong-Bing Cai,
| |
Collapse
|
22
|
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]
|
23
|
Nasiri F, Kazemi M, Mirarefin SMJ, Mahboubi Kancha M, Ahmadi Najafabadi M, Salem F, Dashti Shokoohi S, Evazi Bakhshi S, Safarzadeh Kozani P, Safarzadeh Kozani P. CAR-T cell therapy in triple-negative breast cancer: Hunting the invisible devil. Front Immunol 2022; 13. [DOI: https:/doi.org/10.3389/fimmu.2022.1018786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is known as the most intricate and hard-to-treat subtype of breast cancer. TNBC cells do not express the well-known estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expressed by other breast cancer subtypes. This phenomenon leaves no room for novel treatment approaches including endocrine and HER2-specific antibody therapies. To date, surgery, radiotherapy, and systemic chemotherapy remain the principal therapy options for TNBC treatment. However, in numerous cases, these approaches either result in minimal clinical benefit or are nonfunctional, resulting in disease recurrence and poor prognosis. Nowadays, chimeric antigen receptor T cell (CAR-T) therapy is becoming more established as an option for the treatment of various types of hematologic malignancies. CAR-Ts are genetically engineered T lymphocytes that employ the body’s immune system mechanisms to selectively recognize cancer cells expressing tumor-associated antigens (TAAs) of interest and efficiently eliminate them. However, despite the clinical triumph of CAR-T therapy in hematologic neoplasms, CAR-T therapy of solid tumors, including TNBC, has been much more challenging. In this review, we will discuss the success of CAR-T therapy in hematological neoplasms and its caveats in solid tumors, and then we summarize the potential CAR-T targetable TAAs in TNBC studied in different investigational stages.
Collapse
|
24
|
Nasiri F, Kazemi M, Mirarefin SMJ, Mahboubi Kancha M, Ahmadi Najafabadi M, Salem F, Dashti Shokoohi S, Evazi Bakhshi S, Safarzadeh Kozani P, Safarzadeh Kozani P. CAR-T cell therapy in triple-negative breast cancer: Hunting the invisible devil. Front Immunol 2022; 13:1018786. [PMID: 36483567 PMCID: PMC9722775 DOI: 10.3389/fimmu.2022.1018786] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is known as the most intricate and hard-to-treat subtype of breast cancer. TNBC cells do not express the well-known estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expressed by other breast cancer subtypes. This phenomenon leaves no room for novel treatment approaches including endocrine and HER2-specific antibody therapies. To date, surgery, radiotherapy, and systemic chemotherapy remain the principal therapy options for TNBC treatment. However, in numerous cases, these approaches either result in minimal clinical benefit or are nonfunctional, resulting in disease recurrence and poor prognosis. Nowadays, chimeric antigen receptor T cell (CAR-T) therapy is becoming more established as an option for the treatment of various types of hematologic malignancies. CAR-Ts are genetically engineered T lymphocytes that employ the body's immune system mechanisms to selectively recognize cancer cells expressing tumor-associated antigens (TAAs) of interest and efficiently eliminate them. However, despite the clinical triumph of CAR-T therapy in hematologic neoplasms, CAR-T therapy of solid tumors, including TNBC, has been much more challenging. In this review, we will discuss the success of CAR-T therapy in hematological neoplasms and its caveats in solid tumors, and then we summarize the potential CAR-T targetable TAAs in TNBC studied in different investigational stages.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Mehrasa Kazemi
- Department of Laboratory Medicine, Thalassemia Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Maral Mahboubi Kancha
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Milad Ahmadi Najafabadi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Faeze Salem
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Setareh Dashti Shokoohi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Evazi Bakhshi
- Department of Anatomical Sciences, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
25
|
Salek-Maghsoodi M, Golsanamlu Z, Sadeghi-Mohammadi S, Gazizadeh M, Soleymani J, Safaralizadeh R. Simple fluorescence chemosensor for the detection of calcium ions in water samples and its application in bio-imaging of cancer cells. RSC Adv 2022; 12:31535-31545. [PMID: 36380939 PMCID: PMC9631868 DOI: 10.1039/d2ra04815a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/29/2022] [Indexed: 12/27/2023] Open
Abstract
This article describes the design, synthesis and characterization of a sensor suitable for practical measurement of ionized calcium in water samples and cancer cells. Calcium is an important ion in living organs and works as a messenger in several cellular functions. A lack of Ca ions interrupts the immune system and can lead to several diseases. A novel magnetic-polydopamine nanoparticle (PDNP)/rhodamine B (RhB)/folic acid (FA) nanoparticle was developed for the determination of calcium ions in MCF 7 cell lysates and water samples. Furthermore, the produced nanoparticle was employed for bioimaging of folate receptor (FR)-overexpressed cancer cells. This nanoprobe displayed a bright photoluminescence emission at 576 nm under an excitation wavelength of 420 nm. In the presence of calcium ions, the fluorescence emission of the MNPs-PDNPs/RhB/FA probe was proportionally decreased from 20 ng mL-1 to 100 ng mL-1 and 0.5 μg mL-1 to 20 μg mL-1 with a lower limit of quantification (LLOQ) of about 20 ng mL-1. The developed sensor showed a low-interference manner in the presence of possible coexistence interfering ions. In addition, this nanomaterial showed excellent biocompatibility with favorable differentiation ability to attach to the FR-positive cancer cells. The MNPs-PDNPs/RhB/FA nanoparticle has been utilized for bioimaging of the MCF 7 cell with favorable differentiation ability.
Collapse
Affiliation(s)
- Maral Salek-Maghsoodi
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences Tabriz Iran +98 41 3337 5365
| | - Zahra Golsanamlu
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences Tabriz Iran +98 41 3337 5365
| | - Sanam Sadeghi-Mohammadi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Masoud Gazizadeh
- Liver and Gastrointestinal Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Jafar Soleymani
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences Tabriz Iran +98 41 3337 5365
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, Tabriz University Tabriz Iran
| |
Collapse
|
26
|
Inoue M, Muta K, Mohammed AFA, Onodera R, Higashi T, Ouchi K, Ueda M, Ando Y, Arima H, Jono H, Motoyama K. Feasibility Study of Dendrimer-Based TTR-CRISPR pDNA Polyplex for Ocular Amyloidosis <i>in Vitro</i>. Biol Pharm Bull 2022; 45:1660-1668. [DOI: 10.1248/bpb.b22-00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Masamichi Inoue
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Kyosuke Muta
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | | | - Risako Onodera
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Taishi Higashi
- Priority Organization for Innovation and Excellence, Kumamoto University
| | - Kenta Ouchi
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University
| | - Yukio Ando
- Department of Amyloidosis Research, Nagasaki International University
| | - Hidetoshi Arima
- Laboratory of Evidence-Based Pharmacotherapy, Daiichi University of Pharmacy
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Keiichi Motoyama
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
27
|
Géczi Z, Róth I, Kőhidai Z, Kőhidai L, Mukaddam K, Hermann P, Végh D, Zelles T. The use of Trojan-horse drug delivery system in managing periodontitis. Int Dent J 2022; 73:346-353. [PMID: 36175203 DOI: 10.1016/j.identj.2022.08.003] [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: 06/19/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022] Open
Abstract
The aim of this review is to evaluate the possibility of delivering a silver-acid complex via a Trojan-horse mechanism for managing periodontits. We theroised that the complex could be an effective treatment option for bacterial inflammatory processes in the oral cavity. Searches were conducted using MEDLINE, Embase, Web of Science Core Collection, and Google Scholar search engines. We also reviewed several reference lists of the included studies or relevant reviews identified by the search. By using Medical Subject Headings (MeSH) terminology, a comprehensive search was performed for the following keywords: silver, folic acid, periodontitis, macrophages, Trojan-horse mechanism, toxicity, and targeting. Using the keywords mentioned earlier, we selected 110 articles and after appropriate elimination the review was written based on 37 papers. Accordingly the we noted that silver isons were an effective approach to kill oral pathogens. Secondly the Trojan-horse mechanism. could be used by macrophages (as the Trojan horse) to deliver silver ions in large quantities to the inflammatory focus to kill the periodontopathogens. The Trojan-horse mechanism has never been described in the field of dentistry before. The proposed novel approach using the principle of Trojan Horse delivery of drugs/chemicals could be used to manage oral inflammatory conditions. This method can be used to supplement regular treatments.
Collapse
Affiliation(s)
- Zoltán Géczi
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary.
| | - Ivett Róth
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary
| | - Zsófia Kőhidai
- Department of Oral Diagnostics, Semmelweis University, Budapest, Hungary
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Khaled Mukaddam
- Department of Oral Surgery, University Center for Dental Medicine Basel, University of Basel, Basel, Switzerland
| | - Péter Hermann
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary
| | - Dániel Végh
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary
| | - Tivadar Zelles
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Biopolymeric Prodrug Systems as Potential Antineoplastic Therapy. Pharmaceutics 2022; 14:pharmaceutics14091773. [PMID: 36145522 PMCID: PMC9505808 DOI: 10.3390/pharmaceutics14091773] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Nowadays, cancer represents a major public health issue, a substantial economic issue, and a burden for society. Limited by numerous disadvantages, conventional chemotherapy is being replaced by new strategies targeting tumor cells. In this context, therapies based on biopolymer prodrug systems represent a promising alternative for improving the pharmacokinetic and pharmacologic properties of drugs and reducing their toxicity. The polymer-directed enzyme prodrug therapy is based on tumor cell targeting and release of the drug using polymer–drug and polymer–enzyme conjugates. In addition, current trends are oriented towards natural sources. They are biocompatible, biodegradable, and represent a valuable and renewable source. Therefore, numerous antitumor molecules have been conjugated with natural polymers. The present manuscript highlights the latest research focused on polymer–drug conjugates containing natural polymers such as chitosan, hyaluronic acid, dextran, pullulan, silk fibroin, heparin, and polysaccharides from Auricularia auricula.
Collapse
|
29
|
In vitro Therapeutic Effects of Folate Receptor-Targeted Delivery of Anti-Atherogenic Nanodrug on Macrophage Foam Cells. Macromol Res 2022. [DOI: 10.1007/s13233-022-0082-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
30
|
|
31
|
Chilimoniuk Z, Rocka A, Stefaniak M, Tomczyk Ż, Jasielska F, Madras D, Filip A. Molecular methods for increasing the effectiveness of ovarian cancer treatment: a systematic review. Future Oncol 2022; 18:1627-1650. [PMID: 35129396 DOI: 10.2217/fon-2021-0565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: The aim of the current study is to analyze and summarize the latest research on improving therapy in ovarian cancer. Materials & methods: Data analysis was based on a review of publications from 2011 to 2021 in the PubMed database with use of the search terms including 'EGFR ovarian cancer', 'folate receptor inhibitors ovarian cancer', 'VEGF ovarian cancer', 'PDGF ovarian cancer' and 'CTLA-4 ovarian cancer'. Results: 6643 articles were found; 238 clinical trials and randomized control trials were analyzed; 122 studies were rejected due to inconsistency with the topic of the work. Conclusion: Extensive research on the treatment of ovarian cancer increases the chance of developing the most effective therapy suited to the individual needs of the patient.
Collapse
Affiliation(s)
- Zuzanna Chilimoniuk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Rocka
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Martyna Stefaniak
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Żaklina Tomczyk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Faustyna Jasielska
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Dominika Madras
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Filip
- Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| |
Collapse
|
32
|
OUP accepted manuscript. Nutr Rev 2022; 80:2178-2197. [DOI: 10.1093/nutrit/nuac025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
33
|
Fundamentals and developments in fluorescence-guided cancer surgery. Nat Rev Clin Oncol 2022; 19:9-22. [PMID: 34493858 DOI: 10.1038/s41571-021-00548-3] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
Fluorescence-guided surgery using tumour-targeted imaging agents has emerged over the past decade as a promising and effective method of intraoperative cancer detection. An impressive number of fluorescently labelled antibodies, peptides, particles and other molecules related to cancer hallmarks have been developed for the illumination of target lesions. New approaches are being implemented to translate these imaging agents into the clinic, although only a few have made it past early-phase clinical trials. For this translational process to succeed, target selection, imaging agents and their related detection systems and clinical implementation have to operate in perfect harmony to enable real-time intraoperative visualization that can benefit patients. Herein, we review key aspects of this imaging cascade and focus on imaging approaches and methods that have helped to shed new light onto the field of intraoperative fluorescence-guided cancer surgery with the singular goal of improving patient outcomes.
Collapse
|
34
|
Xu J, Chen L, Shu W, Zhang FZ, Xie BX, Wang HS, Wang YL, Lin RG. Folic acid modified UiO-66 nano drug carrier for combination therapy. NEW J CHEM 2022. [DOI: 10.1039/d2nj02523j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The drug delivery system which called RB-Drug@UiO-66-FA can release anticancer drugs slowly in an acidic environment, and generate 1O2 under light exposure, achieving the effect of chemotherapy–photodynamic therapy.
Collapse
Affiliation(s)
- Jin Xu
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Li Chen
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Wen Shu
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Fang-Zhong Zhang
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Bao-Xuan Xie
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Hai-Shuang Wang
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Yu-Lin Wang
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| | - Rong-Guang Lin
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, P. R. China
| |
Collapse
|
35
|
Ferreira JT, Pina J, Ribeiro CAF, Fernandes R, Tomé JPC, Torres T, Rodríguez-Morgade MS. A ruthenium phthalocyanine functionalized with a folic acid unit as a photosensitizer for photodynamic therapy: Synthesis, characterization and in vitro evaluation. J PORPHYR PHTHALOCYA 2021. [DOI: 10.1142/s1088424621501224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A folate-targeted ruthenium(II) phthalocyanine (Ru(FA-Py)(DMSO)(PEG)[Formula: see text]Pc), endowed with a pyridyl ligand functionalized with one folic acid unit (FA-Py) at one of the two axial coordination sites, and a dimethylsulfoxide (DMSO) ligand coordinated to the other axial position, respectively, is described. In order to enhance its biocompatibility, the RuPc is donated with eight PEG chains attached at the peripheral positions. The observed singlet oxygen quantum yields of the PS measured in DMSO and in water are of 0.74 and 0.36, respectively, in line with those observed for other RuPcs bearing comparable axial and peripheral substitution. In vitro PDT activity of the compound has been evaluated in HT-1376 human bladder cancer cell line. Ru(FA-Py)(DMSO)(PEG)[Formula: see text]Pc revealed a slightly higher cellular uptake than those observed for the corresponding carbohydrate-substituted PSs and a better photodynamic activity compared to the glucose-functionalized RuPc.
Collapse
Affiliation(s)
- Joana T. Ferreira
- Departamento de Química Orgánica, Universidad Autónoma de Madrid (UAM), Cantoblanco, 28049 Madrid, Spain
- LAQV-REQUINTE and Department of Chemistry, University of Aveiro 3810-193 Aveiro, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - João Pina
- CQC, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Carlos A. F. Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rosa Fernandes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
| | - João P. C. Tomé
- LAQV-REQUINTE and Department of Chemistry, University of Aveiro 3810-193 Aveiro, Portugal
- CQE and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tomás Torres
- Departamento de Química Orgánica, Universidad Autónoma de Madrid (UAM), Cantoblanco, 28049 Madrid, Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), UAM, 28049 Madrid, Spain
- Instituto Madrileño de Estudios Avanzados (IMDEA)-Nanociencia, c/ Faraday, 9, Cantoblanco, 28049 Madrid, Spain
| | - M. Salomé Rodríguez-Morgade
- Departamento de Química Orgánica, Universidad Autónoma de Madrid (UAM), Cantoblanco, 28049 Madrid, Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), UAM, 28049 Madrid, Spain
| |
Collapse
|
36
|
Chamseddine AN, Assi T, Mir O, Chouaib S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol Ther 2021; 231:107986. [PMID: 34481812 DOI: 10.1016/j.pharmthera.2021.107986] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) plasticity and diversity are both essential hallmarks of the monocyte-macrophage lineage and the tumor-derived inflammation. TAM exemplify the perfect adaptable cell with dynamic phenotypic modifications that reflect changes in their functional polarization status. Under several tumor microenvironment (TME)-related cues, TAM shift their polarization, hence promoting or halting cancer progression. Immune checkpoint inhibitors (ICI) displayed unprecedented clinical responses in various refractory cancers; but only approximately a third of patients experienced durable responses. It is, therefore, crucial to enhance the response rate of immunotherapy. Several mechanisms of resistance to ICI have been elucidated including TAM role with its essential immunosuppressive functions that reduce both anti-tumor immunity and the subsequent ICI efficacy. In the past few years, thorough research has led to a better understanding of TAM biology and innovative approaches can now be adapted through targeting macrophages' recruitment axis as well as TAM activation and polarization status within the TME. Some of these therapeutic strategies are currently being evaluated in several clinical trials in association with ICI agents. This combination between TAM modulation and ICI allows targeting TAM intrinsic immunosuppressive functions and tumor-promoting factors as well as overcoming ICI resistance. Hence, such strategies, with a better understanding of the mechanisms driving TAM modulation, may have the potential to optimize ICI efficacy.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Biostatistics and Epidemiology, CESP INSERM U1018, OncoStat, Gustave Roussy, F-94805, Villejuif, France.
| | - Tarek Assi
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Pharmacology, Gustave Roussy, F-94805, Villejuif, France; Department of Ambulatory Care, Gustave Roussy, F-94805, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
37
|
Hanley KZ, Horowitz IR, Gordon A, Meisel J, Khanna N. Folate Receptor Alpha Is Preferentially Expressed in the Carcinoma Component of Endometrial Carcinosarcomas: A Potential Target for Adjuvant Therapy. Int J Gynecol Pathol 2021; 40:501-509. [PMID: 33323854 DOI: 10.1097/pgp.0000000000000736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Carcinosarcomas (CSs) of the endometrium are biphasic malignancies, composed of high-grade carcinomatous and sarcomatous components. Surgical stage and pathologic characteristics are the most important prognostic findings, with a 5-yr survival of 15% to 30% in advance stage disease. Folate receptor alpha (FRA) overexpression has been observed in endometrial carcinomas and not yet studied in CSs. This study evaluates semiquantitative expression of FRA in both carcinomatous and sarcomatous components of CSs on whole tissue sections. Immunohistochemistry for FRA expression was performed and extent and intensity of staining were recorded for each case for both histologic components. A total of 46 cases were stained for FRA. The majority of these (40/46, 87%) showed FRA staining at variable intensity in the carcinomatous component, stronger in serous carcinomas and high-grade endometrioid, while only a small subset of tumors demonstrated weak staining in the sarcomatous component (2/46, 4.35%). CS is known to be associated with poor prognosis and adjuvant therapy is recommended even in low stage disease. Serous and high-grade endometrioid carcinomas are the most common carcinomatous components of CSs and are known to show consistently high FRA expression. Folate plays a role in tumor cell migration and loss of cellular adhesion, which are key steps in epithelial-mesenchymal transition, the process by which CS develops from carcinoma cells. Our study shows expression of FRA in the carcinomatous component of almost all CS cases (87%), further favoring FRA as a target for adjuvant treatment. While expression of FRA in the sarcomatous component was rarely observed, the carcinomatous component being associated with metastatic potential underscores the importance of anti-FRA therapy for systemic disease control.
Collapse
|
38
|
Shulpekova Y, Nechaev V, Kardasheva S, Sedova A, Kurbatova A, Bueverova E, Kopylov A, Malsagova K, Dlamini JC, Ivashkin V. The Concept of Folic Acid in Health and Disease. Molecules 2021; 26:molecules26123731. [PMID: 34207319 PMCID: PMC8235569 DOI: 10.3390/molecules26123731] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022] Open
Abstract
Folates have a pterine core structure and high metabolic activity due to their ability to accept electrons and react with O-, S-, N-, C-bounds. Folates play a role as cofactors in essential one-carbon pathways donating methyl-groups to choline phospholipids, creatine, epinephrine, DNA. Compounds similar to folates are ubiquitous and have been found in different animals, plants, and microorganisms. Folates enter the body from the diet and are also synthesized by intestinal bacteria with consequent adsorption from the colon. Three types of folate and antifolate cellular transporters have been found, differing in tissue localization, substrate affinity, type of transferring, and optimal pH for function. Laboratory criteria of folate deficiency are accepted by WHO. Severe folate deficiencies, manifesting in early life, are seen in hereditary folate malabsorption and cerebral folate deficiency. Acquired folate deficiency is quite common and is associated with poor diet and malabsorption, alcohol consumption, obesity, and kidney failure. Given the observational data that folates have a protective effect against neural tube defects, ischemic events, and cancer, food folic acid fortification was introduced in many countries. However, high physiological folate concentrations and folate overload may increase the risk of impaired brain development in embryogenesis and possess a growth advantage for precancerous altered cells.
Collapse
Affiliation(s)
- Yulia Shulpekova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Vladimir Nechaev
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Svetlana Kardasheva
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Alla Sedova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Anastasia Kurbatova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Elena Bueverova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Arthur Kopylov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119121 Moscow, Russia;
| | - Kristina Malsagova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119121 Moscow, Russia;
- Correspondence: ; Tel.: +7-499-764-9878
| | | | - Vladimir Ivashkin
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| |
Collapse
|
39
|
Hwang HS, Kim H, Han G, Lee JW, Kim K, Kwon IC, Yang Y, Kim SH. Extracellular Vesicles as Potential Therapeutics for Inflammatory Diseases. Int J Mol Sci 2021; 22:5487. [PMID: 34067503 PMCID: PMC8196952 DOI: 10.3390/ijms22115487] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EV) deliver cargoes such as nucleic acids, proteins, and lipids between cells and serve as an intercellular communicator. As it is revealed that most of the functions associated to EVs are closely related to the immune response, the important role of EVs in inflammatory diseases is emerging. EVs can be functionalized through EV surface engineering and endow targeting moiety that allows for the target specificity for therapeutic applications in inflammatory diseases. Moreover, engineered EVs are considered as promising nanoparticles to develop personalized therapeutic carriers. In this review, we highlight the role of EVs in various inflammatory diseases, the application of EV as anti-inflammatory therapeutics, and the current state of the art in EV engineering techniques.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Korea
| | - Hyosuk Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
| | - Geonhee Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Jong Won Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.S.H.); (H.K.); (G.H.); (J.W.L.); (K.K.); (I.C.K.)
| |
Collapse
|
40
|
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
|
41
|
Petrone I, Bernardo PS, dos Santos EC, Abdelhay E. MTHFR C677T and A1298C Polymorphisms in Breast Cancer, Gliomas and Gastric Cancer: A Review. Genes (Basel) 2021; 12:587. [PMID: 33920562 PMCID: PMC8073588 DOI: 10.3390/genes12040587] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 02/07/2023] Open
Abstract
Folate (vitamin B9) is found in some water-soluble foods or as a synthetic form of folic acid and is involved in many essential biochemical processes. Dietary folate is converted into tetrahydrofolate, a vital methyl donor for most methylation reactions, including DNA methylation. 5,10-methylene tetrahydrofolate reductase (MTHFR) is a critical enzyme in the folate metabolism pathway that converts 5,10-methylenetetrahydrofolate into 5-methyltetrahydrofolate, which produces a methyl donor for the remethylation of homocysteine to methionine. MTHFR polymorphisms result in reduced enzyme activity and altered levels of DNA methylation and synthesis. MTHFR polymorphisms have been linked to increased risks of several pathologies, including cancer. Breast cancer, gliomas and gastric cancer are highly heterogeneous and aggressive diseases associated with high mortality rates. The impact of MTHFR polymorphisms on these tumors remains controversial in the literature. This review discusses the relationship between the MTHFR C677T and A1298C polymorphisms and the increased risk of breast cancer, gliomas, and gastric cancer. Additionally, we highlight the relevance of ethnic and dietary aspects of population-based studies and histological stratification of highly heterogeneous tumors. Finally, this review discusses these aspects as potential factors responsible for the controversial literature concerning MTHFR polymorphisms.
Collapse
Affiliation(s)
- Igor Petrone
- Stem Cell Laboratory, Center for Bone Marrow Transplants, Brazilian National Cancer Institute—INCA, Rio de Janeiro 20230-240, Brazil; (E.C.d.S.); (E.A.)
- Stricto Sensu Graduate Program in Oncology, INCA, Rio de Janeiro 20230-240, Brazil;
| | - Paula Sabbo Bernardo
- Stricto Sensu Graduate Program in Oncology, INCA, Rio de Janeiro 20230-240, Brazil;
- Laboratory of Cellular and Molecular Hemato-Oncology, Molecular Hemato-Oncology Program, Brazilian National Cancer Institute—INCA, Rio de Janeiro 20230-240, Brazil
| | - Everton Cruz dos Santos
- Stem Cell Laboratory, Center for Bone Marrow Transplants, Brazilian National Cancer Institute—INCA, Rio de Janeiro 20230-240, Brazil; (E.C.d.S.); (E.A.)
- Stricto Sensu Graduate Program in Oncology, INCA, Rio de Janeiro 20230-240, Brazil;
| | - Eliana Abdelhay
- Stem Cell Laboratory, Center for Bone Marrow Transplants, Brazilian National Cancer Institute—INCA, Rio de Janeiro 20230-240, Brazil; (E.C.d.S.); (E.A.)
- Stricto Sensu Graduate Program in Oncology, INCA, Rio de Janeiro 20230-240, Brazil;
| |
Collapse
|
42
|
Mansur AAP, Mansur HS, Leonel AG, Carvalho IC, Lage MCG, Carvalho SM, Krambrock K, Lobato ZIP. Supramolecular magnetonanohybrids for multimodal targeted therapy of triple-negative breast cancer cells. J Mater Chem B 2021; 8:7166-7188. [PMID: 32614035 DOI: 10.1039/d0tb01175d] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the undeniable advances in recent decades, cancer remains one of the deadliest diseases of the current millennium, where the triple-negative breast cancer (TNBC) is very aggressive, extremely metastatic, and resistant to conventional chemotherapy. The nanotheranostic approach focusing on targeting membrane receptors often expressed at abnormal levels by cancer cells can be a strategic weapon for fighting malignant tumors. Herein, we introduced a novel "all-in-one nanosoldier" made of colloidal hybrid nanostructures, which were designed for simultaneously targeting, imaging, and killing TNBC cells. These nanohybrids comprised four distinct components: (a) superparamagnetic iron oxide nanoparticles, as bi-functional nanomaterials for inducing ferroptosis via inorganic nanozyme-mediated catalysis and magnetotherapy by hyperthermia treatment; (b) carboxymethyl cellulose biopolymer, as a water-soluble capping macromolecule; (c) folic acid, as the membranotopic vector for targeting folate receptors; (d) and doxorubicin (DOX) drug for chemotherapy. The results demonstrated that this novel strategy was highly effective for targeting and killing TNBC cells in vitro, expressing high levels of folate membrane-receptors. The results evidenced that three integrated mechanisms triggered the deaths of the cancer cells in vitro: (a) ferroptosis, by magnetite nanoparticles inducing a Fenton-like reaction; (b) magneto-hyperthermia effect by generating heat under an alternate magnetic field; and (c) chemotherapy, through the DOX intracellular release causing DNA dysfunction. This "all-in-one nanosoldier" strategy offers a vast realm of prospective alternatives for attacking cancer cells, combining multimodal therapy and the delivery of therapeutic agents to diseased sites and preserving healthy cells, which is one of the most critical clinical challenges faced in fighting drug-resistant breast cancers.
Collapse
Affiliation(s)
- Alexandra A P Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Herman S Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Alice G Leonel
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Isadora C Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Manuela C G Lage
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Sandhra M Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Klaus Krambrock
- Department of Physics, Federal University of Minas Gerais - UFMG, Brazil
| | - Zelia I P Lobato
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais - UFMG, Brazil
| |
Collapse
|
43
|
Distaffen HE, Jones CW, Abraham BL, Nilsson BL. Multivalent display of chemical signals on
self‐assembled
peptide scaffolds. Pept Sci (Hoboken) 2021. [DOI: 10.1002/pep2.24224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
44
|
Tymon-Rosario J, Zeybek B, Santin AD. Novel antibody-drug conjugates: current and future roles in gynecologic oncology. Curr Opin Obstet Gynecol 2021; 33:26-33. [PMID: 32618744 PMCID: PMC8253558 DOI: 10.1097/gco.0000000000000642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) represent a new class of drugs that combine a surface receptor-targeting antibody linked to a cytotoxic molecule. This review summarizes the current literature demonstrating their tremendous promise as therapeutic agents in the treatment of aggressive gynecologic malignancies. RECENT FINDINGS Several antigens have proven to be differentially overexpressed in a variety of gynecologic tumors when compared with normal surrounding tissue and serve as novel targets for ADC therapy. In the last few years HER2/neu, folic acid-alpha (FRα) and Trop-2 overexpression have been exploited as excellent targets by novel ADCs such as Trastuzumab emtansine (T-DM1), SYD985, IMGN853 (Mirvetuximab soravtansine) and Sacituzumab govitecan (SG, IMMU-132) in multiple tumors including ovarian, endometrial and cervical cancers. Although the selectivity of ADCs with noncleavable linkers (i.e. T-DM1) has shown negligible effect on surrounding antigen negative cells, those ADCs with cleavable linkers (i.e. SYD985, IMGN853 and SG) may kill both antigen-positive target cells and surrounding antigen-negative cells via the bystander effect. SUMMARY Preclinical data strongly supports these ADCs and ongoing clinical trials will shed further light into the potential of making these drugs part of current standard practice and providing our patients with a higher level of personalized cancer care.
Collapse
Affiliation(s)
- Joan Tymon-Rosario
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | |
Collapse
|
45
|
Novel Anti-FOLR1 Antibody-Drug Conjugate MORAb-202 in Breast Cancer and Non-Small Cell Lung Cancer Cells. Antibodies (Basel) 2021; 10:antib10010006. [PMID: 33535554 PMCID: PMC7930947 DOI: 10.3390/antib10010006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/26/2020] [Accepted: 01/19/2021] [Indexed: 12/28/2022] Open
Abstract
Antibody-drug conjugates (ADCs), which are currently being developed, may become promising cancer therapeutics. Folate receptor α (FOLR1), a glycosylphosphatidylinositol-anchored membrane protein, is an attractive target of ADCs, as it is largely absent from normal tissues but is overexpressed in malignant tumors of epithelial origin, including ovarian, lung, and breast cancer. In this study, we tested the effects of novel anti-FOLR1 antibody-eribulin conjugate MORAb-202 in breast cancer and non-small cell lung cancer (NSCLC) cell lines. FOLR1 expression, cell proliferation, bystander killing effects, and apoptosis were evaluated in seven breast cancer and nine NSCLC cell lines treated with MORAb-202. Tumor growth and FOLR1 expression were assessed in T47D and MCF7 orthotopic xenograft mouse models after a single intravenous administration of MORAb-202 (5 mg/kg). MORAb-202 was associated with inhibited cell proliferation, with specific selectivity toward FOLR1-expressing breast cancer cell lines. Eribulin, the payload of MORAb-202, was unleashed in HCC1954 cells, diffused into intercellular spaces, and then killed the non-FOLR1-expressing MCF7 cells in co-culture systems. In orthotopic xenograft mouse models, FOLR1-expressing T47D tumors and non-FOLR1-expressing MCF7 tumors were suppressed upon MORAb-202 administration. The novel anti-FOLR1 antibody-eribulin conjugate MORAb-202 has potential antitumor effects in breast cancer.
Collapse
|
46
|
Hawner M, Ducho C. Cellular Targeting of Oligonucleotides by Conjugation with Small Molecules. Molecules 2020; 25:E5963. [PMID: 33339365 PMCID: PMC7766908 DOI: 10.3390/molecules25245963] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022] Open
Abstract
Drug candidates derived from oligonucleotides (ON) are receiving increased attention that is supported by the clinical approval of several ON drugs. Such therapeutic ON are designed to alter the expression levels of specific disease-related proteins, e.g., by displaying antigene, antisense, and RNA interference mechanisms. However, the high polarity of the polyanionic ON and their relatively rapid nuclease-mediated cleavage represent two major pharmacokinetic hurdles for their application in vivo. This has led to a range of non-natural modifications of ON structures that are routinely applied in the design of therapeutic ON. The polyanionic architecture of ON often hampers their penetration of target cells or tissues, and ON usually show no inherent specificity for certain cell types. These limitations can be overcome by conjugation of ON with molecular entities mediating cellular 'targeting', i.e., enhanced accumulation at and/or penetration of a specific cell type. In this context, the use of small molecules as targeting units appears particularly attractive and promising. This review provides an overview of advances in the emerging field of cellular targeting of ON via their conjugation with small-molecule targeting structures.
Collapse
Affiliation(s)
| | - Christian Ducho
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2 3, 66 123 Saarbrücken, Germany;
| |
Collapse
|
47
|
Vanza JD, Patel RB, Patel MR. Nanocarrier centered therapeutic approaches: Recent developments with insight towards the future in the management of lung cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Zhao Z, Ukidve A, Kim J, Mitragotri S. Targeting Strategies for Tissue-Specific Drug Delivery. Cell 2020; 181:151-167. [PMID: 32243788 DOI: 10.1016/j.cell.2020.02.001] [Citation(s) in RCA: 548] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/19/2022]
Abstract
Off-target effects of systemically administered drugs have been a major hurdle in designing therapies with desired efficacy and acceptable toxicity. Developing targeting strategies to enable site-specific drug delivery holds promise in reducing off-target effects, decreasing unwanted toxicities, and thereby enhancing a drug's therapeutic efficacy. Over the past three decades, a large body of literature has focused on understanding the biological barriers that hinder tissue-specific drug delivery and strategies to overcome them. These efforts have led to several targeting strategies that modulate drug delivery in both the preclinical and clinical settings, including small molecule-, nucleic acid-, peptide-, antibody-, and cell-based strategies. Here, we discuss key advances and emerging concepts for tissue-specific drug delivery approaches and their clinical translation.
Collapse
Affiliation(s)
- Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Anvay Ukidve
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Jayoung Kim
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Meng X, Sun P, Xu H, Wang Z. Folic acid-functionalized magnetic nanoprobes via a PAMAM dendrimer/SA-biotin mediated cascade-amplifying system for the efficient enrichment of circulating tumor cells. Biomater Sci 2020; 8:6395-6403. [PMID: 33034317 DOI: 10.1039/d0bm01212b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
As a liquid biopsy, circulating tumor cells (CTCs) have great significance for the early diagnosis, timely treatment, and practical evaluation of metastasis or recurrence of cancer. However, the enrichment of rare CTCs in complex blood samples is still a significant challenge. Here, unique and highly sensitive folic acid (FA)-functionalized cascade amplification system-modified magnetic nanoparticles (MNPs) were constructed to effectively capture CTCs in whole blood. In this system, as a targeted molecule, numerous FA molecules were conjugated on the surface of PAMAM dendrimers (PAMAM-FA) (first amplification) through a polyethylene glycol (PEG) linker, which could promote the more facile binding of folate receptors (FR) on the surface of ovarian cancer cells (SKOV3 cells). Then, PAMAM-FA was further modified with biotin to fabricate biotin-PAMAM-FA (BPF), which could combine with streptavidin (SA)-modified MNPs (SMs) via the SA-biotin system to efficiently target and separate CTCs. The capture efficiency of the constructed MNPs-SA ∼ biotin-PAMAM-FA (SM@BPF) nanoprobes was 90.3% with high cell viability (∼93.2%) and minimal non-specific adsorption (∼25%). Moreover, fewer nanoprobes were absorbed on the surface of the SM@BPF-captured SKOV3 cells (one-step method) compared with the SM/BPF-captured SKOV3 cells (two-step method), which was beneficial for further biological analysis. We expect that this recognition molecule-based cascade amplification system will provide an innovative CTCs enrichment platform for the early-stage diagnosis of ovarian cancer.
Collapse
Affiliation(s)
- Xiangyu Meng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, Jiangsu, PR China.
| | | | | | | |
Collapse
|
50
|
Prieto-Montero R, Katsumiti A, Cajaraville MP, López-Arbeloa I, Martínez-Martínez V. Functionalized Fluorescent Silica Nanoparticles for Bioimaging of Cancer Cells. SENSORS (BASEL, SWITZERLAND) 2020; 20:E5590. [PMID: 33003513 PMCID: PMC7582890 DOI: 10.3390/s20195590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/16/2020] [Accepted: 09/26/2020] [Indexed: 12/12/2022]
Abstract
Functionalized fluorescent silica nanoparticles were designed and synthesized to selectively target cancer cells for bioimaging analysis. The synthesis method and characterization of functionalized fluorescent silica nanoparticles (50-60 nm), as well as internalization and subcellular localization in HeLa cells is reported here. The dye, rhodamine 101 (R101) was physically embedded during the sol-gel synthesis. The dye loading was optimized by varying the synthesis conditions (temperature and dye concentration added to the gel) and by the use of different organotriethoxysilanes as a second silica precursor. Additionally, R101, was also covalently bound to the functionalized external surface of the silica nanoparticles. The quantum yields of the dye-doped silica nanoparticles range from 0.25 to 0.50 and demonstrated an enhanced brightness of 230-260 fold respect to the free dye in solution. The shell of the nanoparticles was further decorated with PEG of 2000 Da and folic acid (FA) to ensure good stability in water and to enhance selectivity to cancer cells, respectively. In vitro assays with HeLa cells showed that fluorescent nanoparticles were internalized by cells accumulating exclusively into lysosomes. Quantitative analysis showed a significantly higher accumulation of FA functionalized fluorescent silica nanoparticles compared to nanoparticles without FA, proving that the former may represent good candidates for targeting cancer cells.
Collapse
Affiliation(s)
- Ruth Prieto-Montero
- Departamento de Química Física, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48080 Bilbao, Spain; (R.P.-M.); (I.L.-A.)
| | - Alberto Katsumiti
- Departamento de Zoología y Biología Celular Animal, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48080 Bilbao, Spain; (A.K.); (M.P.C.)
| | - Miren Pilare Cajaraville
- Departamento de Zoología y Biología Celular Animal, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48080 Bilbao, Spain; (A.K.); (M.P.C.)
| | - Iñigo López-Arbeloa
- Departamento de Química Física, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48080 Bilbao, Spain; (R.P.-M.); (I.L.-A.)
| | - Virginia Martínez-Martínez
- Departamento de Química Física, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48080 Bilbao, Spain; (R.P.-M.); (I.L.-A.)
| |
Collapse
|