1
|
Sant’Angelo D, Descamps G, Lecomte V, Stanicki D, Penninckx S, Dragan T, Van Gestel D, Laurent S, Journe F. Therapeutic Approaches with Iron Oxide Nanoparticles to Induce Ferroptosis and Overcome Radioresistance in Cancers. Pharmaceuticals (Basel) 2025; 18:325. [PMID: 40143107 PMCID: PMC11945075 DOI: 10.3390/ph18030325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The emergence of nanotechnology in medicine, particularly using iron oxide nanoparticles (IONPs), may impact cancer treatment strategies. IONPs exhibit unique properties, such as superparamagnetism, biocompatibility, and ease of surface modification, making them ideal candidates for imaging, and therapeutic interventions. Their application in targeted drug delivery, especially with traditional chemotherapeutic agents like cisplatin, has shown potential in overcoming limitations such as low bioavailability and systemic toxicity of chemotherapies. Moreover, IONPs, by releasing iron ions, can induce ferroptosis, a form of iron-dependent cell death, which offers a promising pathway to reverse radio- and chemoresistance in cancer therapy. In particular, IONPs demonstrate significant potential as radiosensitisers, enhancing the effects of radiotherapy by promoting reactive oxygen species (ROS) generation, lipid peroxidation, and modulating the tumour microenvironment to stimulate antitumour immune responses. This review explores the multifunctional roles of IONPs in radiosensitisation through ferroptosis induction, highlighting their promise in advancing treatment for head and neck cancers. Additional research is crucial to fully addressing their potential in clinical settings, offering a novel approach to personalised cancer treatment.
Collapse
Affiliation(s)
- Dorianne Sant’Angelo
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Géraldine Descamps
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Valentin Lecomte
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Sébastien Penninckx
- Department of Medical Physics, Institut Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Tatiana Dragan
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Dirk Van Gestel
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Fabrice Journe
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
2
|
Abdelhamid MS, Wadan AHS, Saad HA, El-Dakroury WA, Hageen AW, Mohammed DH, Mourad S, Mohammed OA, Abdel-Reheim MA, Doghish AS. Nanoparticle innovations in targeted cancer therapy: advancements in antibody-drug conjugates. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03764-7. [PMID: 39825965 DOI: 10.1007/s00210-024-03764-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising strategy in targeted cancer therapy, enabling the precise delivery of cytotoxic agents to tumor sites while minimizing systemic toxicity. However, traditional ADCs face significant limitations, including restricted drug loading capacity, where an optimal drug-to-antibody ratio (DAR) is crucial; low DARs may lead to insufficient potency, while high DARs can cause rapid clearance and increased toxicity. Additionally, ADCs often suffer from instability in circulation due to the potential for premature release of cytotoxic agents, resulting in off-target effects and reduced therapeutic efficacy. Furthermore, their large size can impede adequate penetration into solid tumors, particularly in heterogeneous environments with varying antigen expressions. This review explores the innovative use of nanoparticles as carriers for ADCs, which offers a multifaceted approach to enhance therapeutic efficacy. By leveraging the unique properties of nanoparticles, such as their small size and ability to exploit the enhanced permeability and retention (EPR) effect, researchers can improve drug stability, prolong circulation time, and achieve more effective tumor targeting. Recent studies demonstrate that nanoparticle-encapsulated ADCs can significantly enhance treatment outcomes while reducing off-target effects, as evidenced by improved targeting capabilities and reduced toxicity in preclinical models. Despite the promising advancements, challenges remain, including potential nanoparticle toxicity and manufacturing complexities. This review aims to provide a comprehensive overview of the current research on nanoparticle-encapsulated ADCs. It highlights their potential to transform cancer treatment and offers insights into future directions for optimizing these advanced therapeutic strategies.
Collapse
Affiliation(s)
| | - Al-Hassan Soliman Wadan
- Oral Biology Department, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, 15888, Suez Governorate, Egypt
| | - Hager Adel Saad
- Faculty of Pharmacy, German University in Cairo (GUC), New Cairo, 11835, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed W Hageen
- Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | | | - Sohaila Mourad
- Faculty of Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
3
|
Jin W, Li X, Argandona SM, Ray RM, Lin MKTH, Melle F, Clergeaud G, Lars Andresen T, Nielsen M, Fairen-Jimenez D, Astakhova K, Qvortrup K. Surface engineering of metal-organic framework nanoparticles-based miRNA carrier: Boosting RNA stability, intracellular delivery and synergistic therapy. J Colloid Interface Sci 2025; 677:429-440. [PMID: 39153246 DOI: 10.1016/j.jcis.2024.08.074] [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: 05/09/2024] [Revised: 08/01/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that are critical for the regulation of multiple physiological and pathological processes, thus holding great clinical potential. However, the therapeutic applications of miRNAs are severely limited by their biological instability and poor intracellular delivery. Herein, we describe a dual-layers surface engineering strategy to design an efficient miRNA delivery nanosystem based on metal-organic frameworks (MOFs) incorporating lipid coating. The resulting nanoparticle system was demonstrated to protect miRNA from ribonuclease degradation, enhance cellular uptake and facilitate lysosomal escape. These ensured effective miRNA mediated gene therapy, which synergized with MOF-specific photodynamic therapy and pre-encapsulated doxorubicin (Dox) chemotherapy to provide a multifunctional with therapeutic effectiveness against cencer cells The mechanisms of miRNA binding and Dox loading were revealed, demonstrating the potential of the present MOFs surface-engineered strategy to overcome their inherent pore-size restriction for macromolecular miRNA carrying, enableefficient co-delivery. In vitro studies revealed the potential of our multifunctional system for miRNA delivery and the demonstrated the therapeutic effectiveness against cancer cells, thereby providing a versatile all-in-one MOFs strategy for delivery of nucleic acids and diverse therapeutic molecules in synergistic therapy.
Collapse
Affiliation(s)
- Weiguang Jin
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Xin Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Sergio Mercado Argandona
- The Adsorption and Advanced Laboratory, Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
| | - Roslyn M Ray
- Center for Gene Therapy, City of Hope-Beckman Research Institute, Duarte, CA 91010, USA.
| | - Marie Karen Tracy Hong Lin
- National Center for Nanofabrication and Characterization, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Francesca Melle
- The Adsorption and Advanced Laboratory, Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
| | - Gael Clergeaud
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Martin Nielsen
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - David Fairen-Jimenez
- The Adsorption and Advanced Laboratory, Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
| | - Kira Astakhova
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
4
|
Zhou Z“Z, Si Y, Zhang J, Chen K, George A, Kim S, Zhou L, Liu X“M. A Dual-Payload Antibody-Drug Conjugate Targeting CD276/B7-H3 Elicits Cytotoxicity and Immune Activation in Triple-Negative Breast Cancer. Cancer Res 2024; 84:3848-3863. [PMID: 39186778 PMCID: PMC11565169 DOI: 10.1158/0008-5472.can-23-4099] [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: 12/27/2023] [Revised: 04/30/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and heterogeneous disease that often relapses following treatment with standard radiotherapies and cytotoxic chemotherapies. Combination therapies have potential for treating refractory metastatic TNBC. In this study, we aimed to develop an antibody-drug conjugate with dual payloads (DualADC) as a chemoimmunotherapy for TNBC. The overexpression of an immune checkpoint transmembrane CD276 (also known as B7-H3) was associated with angiogenesis, metastasis, and immune tolerance in more than 60% of patients with TNBC. Development of a mAb capable of targeting the extracellular domain of surface CD276 enabled delivery of payloads to tumors, and a platform was established for concurrent conjugation of a traditional cytotoxic payload and an immunoregulating Toll-like receptor 7/8 agonist to the CD276 mAb. The DualADC effectively killed multiple TNBC subtypes, significantly enhanced immune functions in the tumor microenvironment, and reduced tumor burden by up to 90% to 100% in animal studies. Single-cell RNA sequencing, multiplex cytokine analysis, and histology elucidated the impact of treatment on tumor cells and the immune landscape. This study suggests that the developed DualADC could represent a promising targeted chemoimmunotherapy for TNBC. Significance: An anti-CD276 monoclonal antibody conjugated with both a cytotoxic drug and an immune boosting reagent effectively targets triple-negative breast cancer by inducing tumor cell death and stimulating immune cell infiltration.
Collapse
Affiliation(s)
- Zhuoxin “Zora” Zhou
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Yingnan Si
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jiashuai Zhang
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Kai Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Ashley George
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Seulhee Kim
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Lufang Zhou
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Xiaoguang “Margaret” Liu
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
5
|
Schiavon E, Rezzola S, Filippi E, Turati M, Parrasia S, Bernardotto S, Stocco M, Szabò I, Mattarei A, Ronca R, Morpurgo M. A novel mertansine conjugate for acid-reversible targeted drug delivery validated through the Avidin-Nucleic-Acid-NanoASsembly platform. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102784. [PMID: 39236938 DOI: 10.1016/j.nano.2024.102784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/27/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
In targeted cancer therapy, antibody-drug-conjugates using mertansine (DM1)-based cytotoxic compounds rely on covalent bonds for drug conjugation. Consequently, the cytotoxic DM1 derivative released upon their proteolytic digestion is up to 1000-fold less potent than DM1 and lacks a bystander effect. To overcome these limitations, we developed a DM1 derivative (keto-DM1) suitable for bioconjugation through an acid-reversible hydrazone bond. Its acid-reversible hydrazone conjugate with biotin (B-Hz-DM1) was generated and tested for efficacy using the cetuximab-targeted Avidin-Nucleic-Acid-NanoASsembly (ANANAS) nanoparticle (NP) platform. NP-tethered B-Hz-DM1 is stable at neutral pH and releases its active moiety only in endosome/lysosome mimicking acidic pH. In vitro, the NP/Cetux/B-Hz-DM1 assembly showed high potency on MDA-MB231 breast cancer cells. In vivo both B-Hz-DM1 and NP/Cetux/B-Hz-DM1 reduced tumor growth. A significantly major effect was exerted by the nanoformulation, associated with an increased in situ tumor cell death. Keto-DM1 is a promising acid-reversible mertansine derivative for targeted delivery in cancer therapy.
Collapse
Affiliation(s)
- Elisa Schiavon
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Sara Rezzola
- Molecular and Translational Medicine Dept (DMMT), University of Brescia, v.le Europa 11, 25121 Brescia, Italy
| | - Erica Filippi
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Marta Turati
- Molecular and Translational Medicine Dept (DMMT), University of Brescia, v.le Europa 11, 25121 Brescia, Italy
| | - Sofia Parrasia
- Department of Biology (DiBio), University of Padova, Via U. Bassi, 58/B, Padova, Italy
| | - Simone Bernardotto
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Martina Stocco
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology (DiBio), University of Padova, Via U. Bassi, 58/B, Padova, Italy
| | - Andrea Mattarei
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Roberto Ronca
- Molecular and Translational Medicine Dept (DMMT), University of Brescia, v.le Europa 11, 25121 Brescia, Italy.
| | - Margherita Morpurgo
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy.
| |
Collapse
|
6
|
Llaguno-Munive M, Vazquez-Lopez MI, Garcia-Lopez P. Solid Lipid Nanoparticles, an Alternative for the Treatment of Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:10712. [PMID: 39409041 PMCID: PMC11476567 DOI: 10.3390/ijms251910712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Within the field of nanomedicine, which is revolutionizing cancer treatment, solid lipid nanoparticles (SLNs) have shown advantages over conventional chemotherapy when tested on cancer cells in preclinical studies. SLNs have proven to be an innovative strategy for the treatment of triple-negative breast cancer cells, providing greater efficiency than existing treatments in various studies. The encapsulation of antineoplastic drugs in SLNs has facilitated a sustained, controlled, and targeted release, which enhances therapeutic efficiency and reduces adverse effects. Moreover, the surface of SLNs can be modified to increase efficiency. For instance, the coating of these particles with polyethylene glycol (PEG) decreases their opsonization, resulting in a longer life in the circulatory system. The creation of positively charged cationic SLNs (cSLNs), achieved by the utilization of surfactants or ionic lipids with positively charged structural groups, increases their affinity for cell membranes and plasma proteins. Hyaluronic acid has been added to SLNs so that the distinct pH of tumor cells would stimulate the release of the drug and/or genetic material. The current review summarizes the recent research on SLNs, focusing on the encapsulation and transport of therapeutic agents with a cytotoxic effect on triple-negative breast cancer.
Collapse
Affiliation(s)
- Monserrat Llaguno-Munive
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Maria Ines Vazquez-Lopez
- Laboratorio de Fármaco-Oncología y Nanomedicina, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Patricia Garcia-Lopez
- Laboratorio de Fármaco-Oncología y Nanomedicina, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| |
Collapse
|
7
|
Wang R, Kumar P, Reda M, Wallstrum AG, Crumrine NA, Ngamcherdtrakul W, Yantasee W. Nanotechnology Applications in Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308639. [PMID: 38126905 PMCID: PMC11493329 DOI: 10.1002/smll.202308639] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Next-generation cancer treatments are expected not only to target cancer cells but also to simultaneously train immune cells to combat cancer while modulating the immune-suppressive environment of tumors and hosts to ensure a robust and lasting response. Achieving this requires carriers that can codeliver multiple therapeutics to the right cancer and/or immune cells while ensuring patient safety. Nanotechnology holds great potential for addressing these challenges. This article highlights the recent advances in nanoimmunotherapeutic development, with a focus on breast cancer. While immune checkpoint inhibitors (ICIs) have achieved remarkable success and lead to cures in some cancers, their response rate in breast cancer is low. The poor response rate in solid tumors is often associated with the low infiltration of anti-cancer T cells and an immunosuppressive tumor microenvironment (TME). To enhance anti-cancer T-cell responses, nanoparticles are employed to deliver ICIs, bispecific antibodies, cytokines, and agents that induce immunogenic cancer cell death (ICD). Additionally, nanoparticles are used to manipulate various components of the TME, such as immunosuppressive myeloid cells, macrophages, dendritic cells, and fibroblasts to improve T-cell activities. Finally, this article discusses the outlook, challenges, and future directions of nanoimmunotherapeutics.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Pramod Kumar
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Noah A. Crumrine
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| |
Collapse
|
8
|
Chen Y, Xiong T, Peng Q, Du J, Sun W, Fan J, Peng X. Self-reporting photodynamic nanobody conjugate for precise and sustainable large-volume tumor treatment. Nat Commun 2024; 15:6935. [PMID: 39138197 PMCID: PMC11322375 DOI: 10.1038/s41467-024-51253-5] [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: 11/12/2023] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Nanobodies (Nbs), the smallest antigen-binding fragments with high stability and affinity derived from the variable domain of naturally occurring heavy-chain-only antibodies in camelids, have been shown as an efficient way to improve the specificity to tumors for photodynamic therapy (PDT). Nonetheless, the rapid clearance of Nbs in vivo restricts the accumulation and retention of the photosensitizer at the tumor site causing insufficient therapeutic outcome, especially in large-volume tumors. Herein, we develop photodynamic conjugates, MNB-Pyra Nbs, through site-specific conjugation between 7D12 Nbs and type I photosensitizer MNB-Pyra (morpholine-modified nile blue structure connected to pyrazolinone) in a 1:2 ratio. The photosensitizers with long-term retention can be released at the tumor site by reactive oxygen species cleavage after illumination, accompanied with fluorescence recovery for self-reporting the occurrence of PDT. Ultimately, a single dose of MNB-Pyra Nbs demonstrate highly effective tumor suppression with high biosafety in the large-volume tumor models after three rounds of PDT. This nanobody conjugate provides a paradigm for the design of precise long-time retention photosensitizers and is expected to promote the development of PDT.
Collapse
Affiliation(s)
- Yingchao Chen
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Tao Xiong
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Qiang Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China.
- Liaoning Binhai Laboratory, Dalian, 116023, China.
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| |
Collapse
|
9
|
Gu Y, Wang Z, Wang Y. Bispecific antibody drug conjugates: Making 1+1>2. Acta Pharm Sin B 2024; 14:1965-1986. [PMID: 38799638 PMCID: PMC11119582 DOI: 10.1016/j.apsb.2024.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 05/29/2024] Open
Abstract
Bispecific antibody‒drug conjugates (BsADCs) represent an innovative therapeutic category amalgamating the merits of antibody‒drug conjugates (ADCs) and bispecific antibodies (BsAbs). Positioned as the next-generation ADC approach, BsADCs hold promise for ameliorating extant clinical challenges associated with ADCs, particularly pertaining to issues such as poor internalization, off-target toxicity, and drug resistance. Presently, ten BsADCs are undergoing clinical trials, and initial findings underscore the imperative for ongoing refinement. This review initially delves into specific design considerations for BsADCs, encompassing target selection, antibody formats, and the linker-payload complex. Subsequent sections delineate the extant progress and challenges encountered by BsADCs, illustrated through pertinent case studies. The amalgamation of BsAbs with ADCs offers a prospective solution to prevailing clinical limitations of ADCs. Nevertheless, the symbiotic interplay among BsAb, linker, and payload necessitates further optimizations and coordination beyond a simplistic "1 + 1" to effectively surmount the extant challenges facing the BsADC domain.
Collapse
Affiliation(s)
- Yilin Gu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhijia Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
10
|
Youssef S, Tsang E, Samanta A, Kumar V, Gothelf KV. Reversible Protection and Targeted Delivery of DNA Origami with a Disulfide-Containing Cationic Polymer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301058. [PMID: 37916910 DOI: 10.1002/smll.202301058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 10/08/2023] [Indexed: 11/03/2023]
Abstract
DNA nanostructures have considerable biomedical potential as intracellular delivery vehicles as they are highly homogeneous and can be functionalized with high spatial resolution. However, challenges like instability under physiological conditions, limited cellular uptake, and lysosomal degradation limit their use. This paper presents a bio-reducible, cationic polymer poly(cystaminebisacrylamide-1,6-diaminohexane) (PCD) as a reversible DNA origami protector. PCD displays a stronger DNA affinity than other cationic polymers. DNA nanostructures with PCD protection are shielded from low salt conditions and DNase I degradation and show a 40-fold increase in cell-association when linked to targeting antibodies. Confocal microscopy reveals a potential secondary cell uptake mechanism, directly delivering the nanostructures to the cytoplasm. Additionally, PCD can be removed by cleaving its backbone disulfides using the intracellular reductant, glutathione. Finally, the application of these constructs is demonstrated for targeted delivery of a cytotoxic agent to cancer cells, which efficiently decreases their viability. The PCD protective agent that is reported here is a simple and efficient method for the stabilization of DNA origami structures. With the ability to deprotect the DNA nanostructures upon entry of the intracellular space, the possibility for the use of DNA origami in pharmaceutical applications is enhanced.
Collapse
Affiliation(s)
- Sarah Youssef
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, 8000, Denmark
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Emily Tsang
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, 8000, Denmark
| | - Anirban Samanta
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, 8000, Denmark
| | - Vipin Kumar
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, 8000, Denmark
| | - Kurt V Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, 8000, Denmark
| |
Collapse
|
11
|
Rad ME, Soylukan C, Kulabhusan PK, Günaydın BN, Yüce M. Material and Design Toolkit for Drug Delivery: State of the Art, Trends, and Challenges. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55201-55231. [PMID: 37994836 DOI: 10.1021/acsami.3c10065] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
The nanomaterial and related toolkit have promising applications for improving human health and well-being. Nanobased drug delivery systems use nanoscale materials as carriers to deliver therapeutic agents in a targeted and controlled manner, and they have shown potential to address issues associated with conventional drug delivery systems. They offer benefits for treating various illnesses by encapsulating or conjugating biological agents, chemotherapeutic drugs, and immunotherapeutic agents. The potential applications of this technology are vast; however, significant challenges exist to overcome such as safety issues, toxicity, efficacy, and insufficient capacity. This article discusses the latest developments in drug delivery systems, including drug release mechanisms, material toolkits, related design molecules, and parameters. The concluding section examines the limitations and provides insights into future possibilities.
Collapse
Affiliation(s)
- Monireh Esmaeili Rad
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Caner Soylukan
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | | | - Beyza Nur Günaydın
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | - Meral Yüce
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
12
|
Juan A, Segrelles C, del Campo-Balguerías A, Bravo I, Silva I, Peral J, Ocaña A, Clemente-Casares P, Alonso-Moreno C, Lorz C. Anti-EGFR conjugated nanoparticles to deliver Alpelisib as targeted therapy for head and neck cancer. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Abstract
Background
Head and neck squamous cell carcinoma (SCC) is one of the most prevalent and deadly cancers worldwide. Even though surgical approaches, radiation therapy, and therapeutic agents are commonly used, the prognosis of this cancer remains poor, with a tendency towards recurrence and metastasis. Current targeted therapeutic options for these patients are limited to monoclonal antibodies against EGFR or PD-1 receptors. Thus, there is an urgent need to introduce new molecularly targeted therapies for treating head and neck SCC. EGFR can be used as a target to improve the ability of nanoparticles to bind to tumor cells and deliver chemotherapeutic agents. In fact, over 90% of head and neck SCCs overexpress EGFR, and other tumor types, such as colorectal and glioblastoma, show EGFR overexpression. The PI3K/mTOR signaling pathway is one of the most commonly altered oncogenic pathways in head and neck SCC. Alpelisib is a specific PI3Kα inhibitor indicated for PIK3CA mutant advanced breast cancer that showed promising activity in clinical trials in head and neck SCC. However, its use is associated with dose-limiting toxicities and treatment-related adverse effects.
Results
We generated polylactide (PLA) polymeric nanoparticles conjugated to anti-EGFR antibodies via chemical cross-linking to a polyethyleneimine (PEI) coating. Antibody-conjugated nanoparticles (ACNP) displayed low polydispersity and high stability. In vivo, ACNP showed increased tropism for EGFR-expressing head and neck tumors in a xenograft model compared to non-conjugated nanoparticles (NP). Alpelisib-loaded nanoparticles were homogeneous, stable, and showed a sustained drug release profile. Encapsulated Alpelisib inhibited PI3K pathway activation in the different cell lines tested that included wild type and altered PIK3CA. Alpelisib-NP and Alpelisib-ACNP decreased by 25 times the half-maximal inhibitory concentration compared to the free drug and increased the bioavailability of the drug in the cells. Herein we propose an efficient strategy to treat head and neck SCC based on nanotechnology.
Conclusions
Anti-EGFR-conjugated polymeric nanoparticles are an effective delivery system to increase drug efficiency and bioavailability in head and neck cancer cells. This strategy can help reduce drug exposure in disease-free organs and decrease drug-associated unwanted side effects.
Collapse
|
13
|
Bernardotto S, Frasson I, Faravelli S, Morelli A, Schiavon E, Moscatiello GY, Violatto MB, Pinnola A, Canciani A, Mattarei A, Rossi G, Brini M, Pasetto L, Bonetto V, Bigini P, Forneris F, Richter SN, Morpurgo M. Efficient SARS-CoV-2 infection antagonization by rhACE2 ectodomain multimerized onto the Avidin-Nucleic-Acid-NanoASsembly. Biomaterials 2023; 303:122394. [PMID: 38007919 DOI: 10.1016/j.biomaterials.2023.122394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/18/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Nanodecoy systems based on analogues of viral cellular receptors assembled onto fluid lipid-based membranes of nano/extravescicles are potential new tools to complement classic therapeutic or preventive antiviral approaches. The need for lipid-based membranes for transmembrane receptor anchorage may pose technical challenges along industrial translation, calling for alternative geometries for receptor multimerization. Here we developed a semisynthetic self-assembling SARS-CoV-2 nanodecoy by multimerizing the biotin labelled virus cell receptor -ACE2- ectodomain onto a poly-avidin nanoparticle (NP) based on the Avidin-Nucleic-Acid-NanoASsembly-ANANAS. The ability of the assembly to prevent SARS-CoV-2 infection in human lung cells and the affinity of the ACE2:viral receptor-binding domain (RBD) interaction were measured at different ACE2:NP ratios. At ACE2:NP = 30, 90 % SARS-CoV-2 infection inhibition at ACE2 nanomolar concentration was registered on both Wuhan and Omicron variants, with ten-fold higher potency than the monomeric protein. Lower and higher ACE2 densities were less efficient suggesting that functional recognition between multi-ligand NPs and multi-receptor virus surfaces requires optimal geometrical relationships. In vivo studies in mice showed that the biodistribution and safety profiles of the nanodecoy are potentially suitable for preventing viral infection upon nasal instillation. Viral receptor multimerization using ANANAS is a convenient process which, in principle, could be rapidly adapted to counteract also other viral infections.
Collapse
Affiliation(s)
- Simone Bernardotto
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131, Padova, Italy
| | - Ilaria Frasson
- Department of Molecular Medicine (DMM), University of Padova, Via A. Gabelli, 63, 35121, Padova, Italy
| | - Silvia Faravelli
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9/A, 27100, Pavia, Italy
| | - Annalisa Morelli
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy, Via Mario Negri 2, 20156, Milano, Italy; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Elisa Schiavon
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131, Padova, Italy
| | - Giulia Yuri Moscatiello
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy, Via Mario Negri 2, 20156, Milano, Italy
| | - Martina Bruna Violatto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy, Via Mario Negri 2, 20156, Milano, Italy
| | - Alberta Pinnola
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9/A, 27100, Pavia, Italy
| | - Anselmo Canciani
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9/A, 27100, Pavia, Italy
| | - Andrea Mattarei
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131, Padova, Italy
| | - Gianpaolo Rossi
- Department of Medicine (DIMED), University of Padova, Via Giustiniani, 2, 35131, Padova, Italy
| | - Marisa Brini
- Department of Biology (DIBIO), Viale G. Colombo, 3, 35131, Padova, Italy
| | - Laura Pasetto
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy, Via Mario Negri 2, 20156, Milano, Italy
| | - Valentina Bonetto
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy, Via Mario Negri 2, 20156, Milano, Italy
| | - Paolo Bigini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy, Via Mario Negri 2, 20156, Milano, Italy
| | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Dept. Biology and Biotechnology, University of Pavia, Via Ferrata 9/A, 27100, Pavia, Italy
| | - Sara N Richter
- Department of Molecular Medicine (DMM), University of Padova, Via A. Gabelli, 63, 35121, Padova, Italy; Microbiology and Virology Unit, Padua University Hospital, 35121, Padua, Italy.
| | - Margherita Morpurgo
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131, Padova, Italy.
| |
Collapse
|
14
|
Lee J, Lee J, Sim W, Kim JH, Choi C, Jeon J. Discovering the anti-cancer phytochemical rutin against breast cancer through the methodical platform based on traditional medicinal knowledge. BMB Rep 2023; 56:594-599. [PMID: 37357538 PMCID: PMC10689086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023] Open
Abstract
A number of therapeutic drugs have been developed from functional chemicals found in plants. Knowledge of plants used for medicinal purposes has historically been transmitted by word of mouth or through literature. The aim of the present study is to provide a systemic platform for the development of lead compounds against breast cancer based on a traditional medical text. To verify our systematic approach, integrating processes consisted of text mining of traditional medical texts, 3-D virtual docking screening, and in vitro and in vivo experimental validations were demonstrated. Our text analysis system identified rutin as a specific phytochemical traditionally used for cancer treatment. 3-D virtual screening predicted that rutin could block EGFR signaling. Thus, we validated significant anticancer effects of rutin against breast cancer cells through blockade of EGFR signaling pathway in vitro. We also demonstrated in vivo anti-cancer effects of rutin using the breast cancer recurrence in vivo models. In summary, our innovative approach might be proper for discovering new phytochemical lead compounds designing for blockade of malignant neoplasm including breast cancer. [BMB Reports 2023; 56(11): 594-599].
Collapse
Affiliation(s)
- Jungwhoi Lee
- Department of Biotechnology, College of Applied Life Science, Jeju National University, Jeju 63243, Korea
| | - Jungsul Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Korea, Jeonju 54932, Korea
| | - WooGwang Sim
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Korea, Jeonju 54932, Korea
- Department of Anatomy, University of California San Francisco, California 94143, USA, Jeonju 54932, Korea
| | - Jae-Hoon Kim
- Department of Biotechnology, College of Applied Life Science, Jeju National University, Jeju 63243, Korea
| | - Chulhee Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Korea, Jeonju 54932, Korea
- ILIAS Biologics Inc., Daejeon 34141, Korea
| | - Jongwook Jeon
- Korean Research Institute of Science Technology and Civilization, Jeonbuk National University, Jeonju 54932, Korea
| |
Collapse
|
15
|
Wang G, Cao J, Gui M, Huang P, Zhang L, Qi R, Chen R, Lin L, Han Q, Lin Y, Chen T, He P, Ma J, Fu R, Hong J, Wu Q, Yu H, Chen J, Huang C, Zhang T, Yuan Q, Zhang J, Chen Y, Xia N. The potential of swine pseudorabies virus attenuated vaccine for oncolytic therapy against malignant tumors. J Exp Clin Cancer Res 2023; 42:284. [PMID: 37891570 PMCID: PMC10604416 DOI: 10.1186/s13046-023-02848-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Oncolytic viruses are now well recognized as potential immunotherapeutic agents against cancer. However, the first FDA-approved oncolytic herpes simplex virus 1 (HSV-1), T-VEC, showed limited benefits in some patients in clinical trials. Thus, the identification of novel oncolytic viruses that can strengthen oncolytic virus therapy is warranted. Here, we identified a live-attenuated swine pseudorabies virus (PRV-LAV) as a promising oncolytic agent with broad-spectrum antitumor activity in vitro and in vivo. METHODS PRV cytotoxicity against tumor cells and normal cells was tested in vitro using a CCK8 cell viability assay. A cell kinase inhibitor library was used to screen for key targets that affect the proliferation of PRV-LAV. The potential therapeutic efficacy of PRV-LAV was tested against syngeneic tumors in immunocompetent mice, and against subcutaneous xenografts of human cancer cell lines in nude mice. Cytometry by time of flight (CyTOF) and flow cytometry were used to uncover the immunological mechanism of PRV-LAV treatment in regulating the tumor immune microenvironment. RESULTS Through various tumor-specific analyses, we show that PRV-LAV infects cancer cells via the NRP1/EGFR signaling pathway, which is commonly overexpressed in cancer. Further, we show that PRV-LAV kills cancer cells by inducing endoplasmic reticulum (ER) stress. Moreover, PRV-LAV is responsible for reprogramming the tumor microenvironment from immunologically naïve ("cold") to inflamed ("hot"), thereby increasing immune cell infiltration and restoring CD8+ T cell function against cancer. When delivered in combination with immune checkpoint inhibitors (ICIs), the anti-tumor response is augmented, suggestive of synergistic activity. CONCLUSIONS PRV-LAV can infect cancer cells via NRP1/EGFR signaling and induce cancer cells apoptosis via ER stress. PRV-LAV treatment also restores CD8+ T cell function against cancer. The combination of PRV-LAV and immune checkpoint inhibitors has a significant synergistic effect. Overall, these findings point to PRV-LAV as a serious potential candidate for the treatment of NRP1/EGFR pathway-associated tumors.
Collapse
Affiliation(s)
- Guosong Wang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Jiali Cao
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, People's Republic of China
| | - Mengxuan Gui
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Pengfei Huang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Liang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Ruoyao Qi
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Ruiqi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Lina Lin
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Qiangyuan Han
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Yanhua Lin
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Tian Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Peiqing He
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Jian Ma
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Rao Fu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Junping Hong
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Qian Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Hai Yu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Junyu Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China
| | - Chenghao Huang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China.
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China.
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China.
| | - Yixin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic ProductsNational Innovation Platform for Industry-Education Intergration in Vaccine ResearchSchool of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, People's Republic of China.
| |
Collapse
|
16
|
Bo Y, Zhou J, Cai K, Wang Y, Feng Y, Li W, Jiang Y, Kuo SH, Roy J, Anorma C, Gardner SH, Luu LM, Lau GW, Bao Y, Chan J, Wang H, Cheng J. Leveraging intracellular ALDH1A1 activity for selective cancer stem-like cell labeling and targeted treatment via in vivo click reaction. Proc Natl Acad Sci U S A 2023; 120:e2302342120. [PMID: 37639589 PMCID: PMC10483628 DOI: 10.1073/pnas.2302342120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/09/2023] [Indexed: 08/31/2023] Open
Abstract
Inhibition of overexpressed enzymes is among the most promising approaches for targeted cancer treatment. However, many cancer-expressed enzymes are "nonlethal," in that the inhibition of the enzymes' activity is insufficient to kill cancer cells. Conventional antibody-based therapeutics can mediate efficient treatment by targeting extracellular nonlethal targets but can hardly target intracellular enzymes. Herein, we report a cancer targeting and treatment strategy to utilize intracellular nonlethal enzymes through a combination of selective cancer stem-like cell (CSC) labeling and Click chemistry-mediated drug delivery. A de novo designed compound, AAMCHO [N-(3,4,6-triacetyl- N-azidoacetylmannosamine)-cis-2-ethyl-3-formylacrylamideglycoside], selectively labeled cancer CSCs in vitro and in vivo through enzymatic oxidation by intracellular aldehyde dehydrogenase 1A1. Notably, azide labeling is more efficient in identifying tumorigenic cell populations than endogenous markers such as CD44. A dibenzocyclooctyne (DBCO)-toxin conjugate, DBCO-MMAE (Monomethylauristatin E), could next target the labeled CSCs in vivo via bioorthogonal Click reaction to achieve excellent anticancer efficacy against a series of tumor models, including orthotopic xenograft, drug-resistant tumor, and lung metastasis with low toxicity. A 5/7 complete remission was observed after single-cycle treatment of an advanced triple-negative breast cancer xenograft (~500 mm3).
Collapse
Affiliation(s)
- Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Jingyi Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Kaimin Cai
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Iria Pharma,Champaign, IL61820
| | - Ying Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Yujun Feng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Wenming Li
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Yunjiang Jiang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Shanny Hsuan Kuo
- Department of Pathobiology at College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Jarron Roy
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Chelsea Anorma
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Sarah H. Gardner
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | | | - Gee W. Lau
- Department of Pathobiology at College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Yan Bao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong510120, China
| | - Jefferson Chan
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL61801
- School of Engineering, Westlake University, Hangzhou, Zhejiang Province310024, China
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Iria Pharma,Champaign, IL61820
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL61801
- School of Engineering, Westlake University, Hangzhou, Zhejiang Province310024, China
| |
Collapse
|
17
|
Andrade F, German-Cortés J, Montero S, Carcavilla P, Baranda-Martínez-Abascal D, Moltó-Abad M, Seras-Franzoso J, Díaz-Riascos ZV, Rafael D, Abasolo I. The Nanotechnology-Based Approaches against Kirsten Rat Sarcoma-Mutated Cancers. Pharmaceutics 2023; 15:1686. [PMID: 37376135 DOI: 10.3390/pharmaceutics15061686] [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: 04/15/2023] [Revised: 05/18/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Kirsten rat sarcoma (KRAS) is a small GTPase which acts as a molecular switch to regulate several cell biological processes including cell survival, proliferation, and differentiation. Alterations in KRAS have been found in 25% of all human cancers, with pancreatic cancer (90%), colorectal cancer (45%), and lung cancer (35%) being the types of cancer with the highest mutation rates. KRAS oncogenic mutations are not only responsible for malignant cell transformation and tumor development but also related to poor prognosis, low survival rate, and resistance to chemotherapy. Although different strategies have been developed to specifically target this oncoprotein over the last few decades, almost all of them have failed, relying on the current therapeutic solutions to target proteins involved in the KRAS pathway using chemical or gene therapy. Nanomedicine can certainly bring a solution for the lack of specificity and effectiveness of anti-KRAS therapy. Therefore, nanoparticles of different natures are being developed to improve the therapeutic index of drugs, genetic material, and/or biomolecules and to allow their delivery specifically into the cells of interest. The present work aims to summarize the most recent advances related to the use of nanotechnology for the development of new therapeutic strategies against KRAS-mutated cancers.
Collapse
Affiliation(s)
- Fernanda Andrade
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Departament de Farmàcia i Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Júlia German-Cortés
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Sara Montero
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Pilar Carcavilla
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Diego Baranda-Martínez-Abascal
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Marc Moltó-Abad
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Joaquín Seras-Franzoso
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Zamira Vanessa Díaz-Riascos
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Diana Rafael
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Ibane Abasolo
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| |
Collapse
|
18
|
Rafik ST, Vaidya JS, MacRobert AJ, Yaghini E. Organic Nanodelivery Systems as a New Platform in the Management of Breast Cancer: A Comprehensive Review from Preclinical to Clinical Studies. J Clin Med 2023; 12:jcm12072648. [PMID: 37048731 PMCID: PMC10095028 DOI: 10.3390/jcm12072648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/05/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Breast cancer accounts for approximately 25% of cancer cases and 16.5% of cancer deaths in women, and the World Health Organization predicts that the number of new cases will increase by almost 70% over the next two decades, mainly due to an ageing population. Effective diagnostic and treatment strategies are, therefore, urgently required for improving cure rates among patients since current therapeutic modalities have many limitations and side effects. Nanomedicine is evolving as a promising approach for cancer management, including breast cancer, and various types of organic and inorganic nanomaterials have been investigated for their role in breast cancer diagnosis and treatment. Following an overview on breast cancer characteristics and pathogenesis and challenges of the current treatment strategies, the therapeutic potential of biocompatible organic-based nanoparticles such as liposomes and polymeric micelles that have been tested in breast cancer models are reviewed. The efficacies of different drug delivery and targeting strategies are documented, ranging from synthetic to cell-derived nanoformulations together with a summary of the interaction of nanoparticles with externally applied energy such as radiotherapy. The clinical translation of nanoformulations for breast cancer treatment is summarized including those undergoing clinical trials.
Collapse
Affiliation(s)
- Salma T. Rafik
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria 21516, Egypt
| | - Jayant S. Vaidya
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
| | - Alexander J. MacRobert
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
| | - Elnaz Yaghini
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
| |
Collapse
|
19
|
Violatto MB, Pasetto L, Casarin E, Tondello C, Schiavon E, Talamini L, Marchini G, Cagnotto A, Morelli A, Lanno A, Passoni A, Bigini P, Morpurgo M, Bonetto V. Development of a Nanoparticle-Based Approach for the Blood-Brain Barrier Passage in a Murine Model of Amyotrophic Lateral Sclerosis. Cells 2022; 11:cells11244003. [PMID: 36552768 PMCID: PMC9776960 DOI: 10.3390/cells11244003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The development of nanoparticles (NPs) to enable the passage of drugs across blood-brain barrier (BBB) represents one of the main challenges in neuropharmacology. In recent years, NPs that are able to transport drugs and interact with brain endothelial cells have been tested. Here, we investigated whether the functionalization of avidin-nucleic-acid-nanoassembly (ANANAS) with apolipoprotein E (ApoE) would allow BBB passage in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Our results demonstrated that ANANAS was able to transiently cross BBB to reach the central nervous system (CNS), and ApoE did not enhance this property. Next, we investigated if ANANAS could improve CNS drug delivery. To this aim, the steroid dexamethasone was covalently linked to ANANAS through an acid-reversible hydrazone bond. Our data showed that the steroid levels in CNS tissues of SOD1G93A mice treated with nanoformulation were below the detection limit. This result demonstrates that the passage of BBB is not sufficient to guarantee the release of the cargo in CNS and that a different strategy for drug tethering should be devised. The present study furthermore highlights that NPs can be useful in improving the passage through biological barriers but may limit the interaction of the therapeutic compound with the specific target.
Collapse
Affiliation(s)
- Martina Bruna Violatto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Laura Pasetto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | | | - Camilla Tondello
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy
- Pharmazentrum Frankfurt/ZAFES, Goethe University Hospital Frankfurt, 60596 Frankfurt am Main, Germany
| | - Elisa Schiavon
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy
| | - Laura Talamini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Gloria Marchini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Alfredo Cagnotto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Annalisa Morelli
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Alessia Lanno
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Alice Passoni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Paolo Bigini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
| | - Margherita Morpurgo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy
- Correspondence: (M.M.); (V.B.); Tel.: +39-049-8275330 (M.M.); +39-02-39014548 (V.B.)
| | - Valentina Bonetto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche “Mario Negri” IRCCS, 20133 Milan, Italy
- Correspondence: (M.M.); (V.B.); Tel.: +39-049-8275330 (M.M.); +39-02-39014548 (V.B.)
| |
Collapse
|
20
|
Hamdy NM, Eskander G, Basalious EB. Insights on the Dynamic Innovative Tumor Targeted-Nanoparticles-Based Drug Delivery Systems Activation Techniques. Int J Nanomedicine 2022; 17:6131-6155. [PMID: 36514378 PMCID: PMC9741821 DOI: 10.2147/ijn.s386037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer conventional chemotherapeutic drugs novel formula progress, nowadays, uses nano technology for targeted drug delivery, specifically tailored to overcome therapeutic agents' delivery challenges. Polymer drug delivery systems (DDS) play a crucial role in minimizing off-target side effects arising when using standard cytotoxic drugs. Using nano-formula for targeted localized action, permits using larger effective cytotoxic doses on a single special spot, that can seriously cause harm if it was administered systemically. Therefore, various nanoparticles (NPs) specifically have attached groups for targeting capabilities, not seen in bulk materials, which then need activation. In this review, we will present a simple innovative, illustrative, in a cartoon-way, enumeration of NP anti-cancer drug targeting delivery system activation-types. Area(s) covered in this review are the mechanisms of various NP activation techniques.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Georgette Eskander
- Faculty of Pharmacy, Ain Shams University, Postgraduate Student, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
21
|
Rasel MSI, Mohona FA, Akter W, Kabir S, Chowdhury AA, Chowdhury JA, Hassan MA, Al Mamun A, Ghose DK, Ahmad Z, Khan FS, Bari MF, Rahman MS, Amran MS. Exploration of Site-Specific Drug Targeting-A Review on EPR-, Stimuli-, Chemical-, and Receptor-Based Approaches as Potential Drug Targeting Methods in Cancer Treatment. JOURNAL OF ONCOLOGY 2022; 2022:9396760. [PMID: 36284633 PMCID: PMC9588330 DOI: 10.1155/2022/9396760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
Cancer has been one of the most dominant causes of mortality globally over the last few decades. In cancer treatment, the selective targeting of tumor cells is indispensable, making it a better replacement for conventional chemotherapies by diminishing their adverse side effects. While designing a drug to be delivered selectively in the target organ, the drug development scientists should focus on various factors such as the type of cancer they are dealing with according to which drug, targeting moieties, and pharmaceutical carriers should be targeted. All published articles have been collected regarding cancer and drug-targeting approaches from well reputed databases including MEDLINE, Embase, Cochrane Library, CENTRAL and ClinicalTrials.gov, Science Direct, PubMed, Scopus, Wiley, and Springer. The articles published between January 2010 and December 2020 were considered. Due to the existence of various mechanisms, it is challenging to choose which one is appropriate for a specific case. Moreover, a combination of more than one approach is often utilized to achieve optimal drug effects. In this review, we have summarized and highlighted central mechanisms of how the targeted drug delivery system works in the specific diseased microenvironment, along with the strategies to make an approach more effective. We have also included some pictorial illustrations to have a precise idea about different types of drug targeting. The core contribution of this work includes providing a cancer drug development scientist with a broad preliminary idea to choose the appropriate approach among the various targeted drug delivery mechanisms. Also, the study will contribute to improving anticancer treatment approaches by providing a pathway for lesser side effects observed in conventional chemotherapeutic techniques.
Collapse
Affiliation(s)
- Md. Shamiul Islam Rasel
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Farhana Afrin Mohona
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Wahida Akter
- College of Pharmacy, University of Houston, Houston, USA
| | - Shaila Kabir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Abu Asad Chowdhury
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Md. Abul Hassan
- Department of Science & Technology, Tokushima University Graduate School, Tokushima, Japan
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Dipayon Krisna Ghose
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, King Khalid University, Abha 61413, Saudi Arabia
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Md. Fazlul Bari
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Shah Amran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| |
Collapse
|
22
|
de Araújo RA, da Luz FAC, da Costa Marinho E, Nascimento CP, de Andrade Marques L, Delfino PFR, Antonioli RM, Araújo BJ, da Silva ACAL, Dos Reis Monteiro MLG, Neto MB, Silva MJB. Epidermal growth factor receptor (EGFR) expression in the serum of patients with triple-negative breast carcinoma: prognostic value of this biomarker. Ecancermedicalscience 2022; 16:1431. [PMID: 36158981 PMCID: PMC9458269 DOI: 10.3332/ecancer.2022.1431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) overexpression has been considered a poor prognostic factor in breast cancer. Methodology A prospective study of 206 women with breast cancer analysed by stages (I, II, III and IV) and by immunohistochemical subtype (Luminal A, Luminal B, HER2+ and triple-negative (TN)); 89 healthy controls with normal recent mammography were included. The EGFR measured in the serum (sEGFR) was detected by the Enzyme-Linked Immunosorbent Assay (ELISA) method (R&D Systems kit DY231) collected by blood before any treatment in patients. Kaplan-Meier method and Cox regression were carried out to obtain the prognostic value, considering significance if p < 0.05. Results With a median follow-up of 36.6 months, 47 deaths occurred. Multivariable Cox regression showed difference of overall survival (OS) associated with sEGFR levels (sEGFR ≤ or > 47.8 ng/mL) in patients with TN cancers, but not of Luminal A, Luminal B or HER2+ subtypes; adjusted by stage, the death risk increased by approximately 415% [hazard ratio (HR): 5.149 (1.900-13.955), p = 0.001] for patients with sEGFR > 47.8 ng/mL compared to patients with a lower sEGFR value. There was no significant correlation of sEGFR with staging, histological tumour grade (G1/G2/G3), Ki67 (< or ≥14%) or body mass index. Conclusions Increased sEGFR expression in patients with TN tumours is a significant predictor of lower OS and its quantification is inexpensive and straightforward.
Collapse
Affiliation(s)
- Rogério Agenor de Araújo
- Federal University of Uberlândia, Avenida Pará, Bloco 2U, 1720, Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-4653-6786
| | - Felipe Andrés Cordero da Luz
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-9381-4913
| | - Eduarda da Costa Marinho
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-1307-9104
| | - Camila Piqui Nascimento
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-0955-8559
| | - Lara de Andrade Marques
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-2734-8352
| | - Patrícia Ferreira Ribeiro Delfino
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-2196-9318
| | - Rafael Mathias Antonioli
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-3886-1562
| | - Breno Jeha Araújo
- São Paulo State Cancer Institute of the Medical School of the University of São Paulo, São Paulo, SP, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-4892-9911
| | - Ana Cristina Araújo Lemos da Silva
- Federal University of Uberlândia, Avenida Pará, Bloco 2U, 1720, Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil
- https://orcid.org/0000-0002-8220-938X
| | | | - Morun Bernardino Neto
- Department of Basic and Environmental Sciences, University of São Paulo, Lorena, SP, CEP 12602-810, Brazil
- https://orcid.org/0000-0003-4292-7800
| | - Marcelo José Barbosa Silva
- Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, CEP 38405-320, Brazil
- https://orcid.org/0000-0002-5807-4286
| |
Collapse
|
23
|
Targeted Nanocarrier Delivery of RNA Therapeutics to Control HIV Infection. Pharmaceutics 2022; 14:pharmaceutics14071352. [PMID: 35890248 PMCID: PMC9324444 DOI: 10.3390/pharmaceutics14071352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Our understanding of HIV infection has greatly advanced since the discovery of the virus in 1983. Treatment options have improved the quality of life of people living with HIV/AIDS, turning it from a fatal disease into a chronic, manageable infection. Despite all this progress, a cure remains elusive. A major barrier to attaining an HIV cure is the presence of the latent viral reservoir, which is established early in infection and persists for the lifetime of the host, even during prolonged anti-viral therapy. Different cure strategies are currently being explored to eliminate or suppress this reservoir. Several studies have shown that a functional cure may be achieved by preventing infection and also inhibiting reactivation of the virus from the latent reservoir. Here, we briefly describe the main HIV cure strategies, focussing on the use of RNA therapeutics, including small interfering RNA (siRNA) to maintain HIV permanently in a state of super latency, and CRISPR gRNA to excise the latent reservoir. A challenge with progressing RNA therapeutics to the clinic is achieving effective delivery into the host cell. This review covers recent nanotechnological strategies for siRNA delivery using liposomes, N-acetylgalactosamine conjugation, inorganic nanoparticles and polymer-based nanocapsules. We further discuss the opportunities and challenges of those strategies for HIV treatment.
Collapse
|
24
|
Zhang H, Zhang L, Cao Z, Cheong S, Boyer C, Wang Z, Yun SLJ, Amal R, Gu Z. Two-Dimensional Ultra-Thin Nanosheets with Extraordinarily High Drug Loading and Long Blood Circulation for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200299. [PMID: 35521948 DOI: 10.1002/smll.202200299] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Nanoparticle drug delivery is largely restricted by the low drug loading capacity of nanoparticle carriers. To address this critical challenge and maximize the potential of nanoparticle drug delivery, a 2D ultra-thin layered double hydroxide (LDH) nanosheet with exceptionally high drug loading, excellent colloidal stability, and prolonged blood circulation for cancer treatment is constructed. The nanosheet is synthesized via a biocompatible polymer-assisted bottom-up method and exhibits an ultra-thin 2D sheet-like structure that enables a considerable amount of cargo anchoring sites available for drug loading, leading to an extraordinary 734% (doxorubicin/nanoparticle mass ratio) drug loading capacity. Doxorubicin delivered by the nanosheet remains stable on the nanosheet carrier under the physiological pH condition, while showing sustained release in the tumor microenvironment and the intracellular environment, thus demonstrating on-demand drug release as a result of pH-responsive biodegradation of nanosheets. Using in vitro and in vivo 4T1 breast cancer models, the nanosheet-based ultra-high drug-loading system demonstrates even enhanced therapeutic performance compared to the multilayered LDH-based high drug-loading system, in terms of increased cellular uptake efficiency, prolonged blood circulation, superior therapeutic effect, and reduced systemic toxicity.
Collapse
Affiliation(s)
- He Zhang
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Zhenbang Cao
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Soshan Cheong
- Electron Microscope Unit, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Sung Lai Jimmy Yun
- Qingdao International Academician Park Research Institute, Qingdao, Shandong, 266000, P. R. China
| | - Rose Amal
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zi Gu
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
25
|
Pandey M, Choudhury H, Ying JNS, Ling JFS, Ting J, Ting JSS, Zhia Hwen IK, Suen HW, Samsul Kamar HS, Gorain B, Jain N, Mohd Amin MCI. Mucoadhesive Nanocarriers as a Promising Strategy to Enhance Intracellular Delivery against Oral Cavity Carcinoma. Pharmaceutics 2022; 14:pharmaceutics14040795. [PMID: 35456629 PMCID: PMC9025168 DOI: 10.3390/pharmaceutics14040795] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
Oral cancer, particularly squamous cell carcinoma (SCC), has posed a grave challenge to global health due to its high incidence, metastasis, and mortality rates. Despite numerous studies and favorable improvements in the therapeutic strategies over the past few decades, the prognosis of this disease remains dismal. Moreover, several drawbacks are associated with the conventional treatment; including permanent disfigurement and physical impairment that are attributed to surgical intervention, and systemic toxicity that results from aggressive radio- or chemotherapies, which impacts patients’ prognosis and post-treatment quality of life. The highly vascularized, non-keratinized oral mucosa appears as a potential route for cytotoxic drug administration in treating oral cancer. It acts as a non-invasive portal for drug entry targeting the local oral lesions of the early stages of cancer and the systemic metastasis sites of advanced cancer. The absorption of the poorly aqueous-soluble anti-cancer drugs can be enhanced due to the increased permeability of the ulcerous mucosa lining in the disease state and by bypassing the hepatic first-pass metabolism. However, some challenges in oral transmucosal drug delivery include the drugs’ taste, the limited surface area of the membrane lining the oral cavity, and flushing and enzymatic degradation by saliva. Therefore, mucoadhesive nanocarriers have emerged as promising platforms for controlled, targeted drug delivery in the oral cavity. The surface functionalization of nanocarriers with various moieties allows for drug targeting, bioavailability enhancement, and biodistribution at the site of action, while the mucoadhesive feature prolongs the drug’s residence time for preferential accumulation to optimize the therapeutic effect and reduce systemic toxicity. This review has been focused to highlight the potential of various nanocarriers (e.g., nanoparticles, nanoemulsions, nanocapsules, and liposomes) in conferring targeting, solubility and bioavailability enhancement of actives and mucoadhesive properties as novel tumor-targeted drug delivery approaches in oral cancer treatment.
Collapse
Affiliation(s)
- Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Correspondence: (M.P.); (H.C.); Tel.: +60-166-048-589 (M.P.)
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Correspondence: (M.P.); (H.C.); Tel.: +60-166-048-589 (M.P.)
| | - Jenifer Ngu Shao Ying
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Jessica Foo Sze Ling
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Jong Ting
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Jocelyn Su Szhiou Ting
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Ivory Kuek Zhia Hwen
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Ho Wan Suen
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Hazimah Syazwani Samsul Kamar
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia; (J.N.S.Y.); (J.F.S.L.); (J.T.); (J.S.S.T.); (I.K.Z.H.); (H.W.S.); (H.S.S.K.)
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, India;
| | - Neha Jain
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida 201303, India;
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| |
Collapse
|
26
|
Habibi N, Mauser A, Ko Y, Lahann J. Protein Nanoparticles: Uniting the Power of Proteins with Engineering Design Approaches. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104012. [PMID: 35077010 PMCID: PMC8922121 DOI: 10.1002/advs.202104012] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/12/2021] [Indexed: 05/16/2023]
Abstract
Protein nanoparticles, PNPs, have played a long-standing role in food and industrial applications. More recently, their potential in nanomedicine has been more widely pursued. This review summarizes recent trends related to the preparation, application, and chemical construction of nanoparticles that use proteins as major building blocks. A particular focus has been given to emerging trends related to applications in nanomedicine, an area of research where PNPs are poised for major breakthroughs as drug delivery carriers, particle-based therapeutics or for non-viral gene therapy.
Collapse
Affiliation(s)
- Nahal Habibi
- Biointerfaces InstituteDepartment of Chemical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Ava Mauser
- Biointerfaces InstituteDepartment of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Yeongun Ko
- Biointerfaces InstituteDepartment of Chemical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Joerg Lahann
- Biointerfaces InstituteDepartments of Chemical EngineeringMaterial Science and EngineeringBiomedical Engineeringand Macromolecular Science and EngineeringUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
27
|
Ongaro A, Violatto MB, Casarin E, Pellerani I, Marchini G, Ribaudo G, Salmona M, Carbone M, Passoni A, Gnodi E, Schiavon E, Mattarei A, Barisani D, Invernizzi P, Bigini P, Morpurgo M. The mode of dexamethasone decoration influences avidin-nucleic-acid-nano-assembly organ biodistribution and in vivo drug persistence. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102497. [PMID: 34838993 DOI: 10.1016/j.nano.2021.102497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Avidin-Nucleic-Acid-NanoASsemblies (ANANAS) possess natural tropism for the liver and, when loaded with dexamethasone, reduce clinical progression in an autoimmune hepatitis murine model. Here, we investigated the linker chemistry (hydrazide-hydrazone, Hz-Hz, or carbamate hydrazide-hydrazone, Cb-Hz bond) and length (long, 5 kDa PEG, or short, 5-6 carbons) in biotin-dexamethasone conjugates used for nanoparticle decoration through in vitro and in vivo studies. All four newly synthesized conjugates released the drug at acidic pH only. In vitro, the Hz-Hz and the PEG derivatives were less stable than the Cb-Hz and the short chain ones, respectively. Once injected in healthy mice, dexamethasone location in the PEGylated ANANAS outer layer favors liver penetration and resident macrophages uptake, while drug Hz-Hz, but not Cb-Hz, short spacing prolongs drug availability. In conclusion, the tight modulation of ANANAS decoration can significantly influence the host interaction, paving the way for the development of steroid nanoformulations suitable for different pharmacokinetic profiles.
Collapse
Affiliation(s)
- Alberto Ongaro
- Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Martina Bruna Violatto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milano, Italy
| | | | - Isabella Pellerani
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milano, Italy
| | - Gloria Marchini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milano, Italy
| | - Giovanni Ribaudo
- Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Mario Salmona
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milano, Italy
| | - Marco Carbone
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy; International Center for Digestive Diseases
| | - Alice Passoni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milano, Italy
| | - Elisa Gnodi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; International Center for Digestive Diseases
| | - Elisa Schiavon
- Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Andrea Mattarei
- Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Donatella Barisani
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; International Center for Digestive Diseases
| | - Pietro Invernizzi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy; International Center for Digestive Diseases
| | - Paolo Bigini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milano, Italy
| | - Margherita Morpurgo
- Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy; CRIBI Biotechnology Cente, University of Padova, Padova, Italy.
| |
Collapse
|
28
|
Yue S, Zhang Y, Wei Y, Haag R, Sun H, Zhong Z. Cetuximab-Polymersome-Mertansine Nanodrug for Potent and Targeted Therapy of EGFR-Positive Cancers. Biomacromolecules 2021; 23:100-111. [PMID: 34913340 DOI: 10.1021/acs.biomac.1c01065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Targeted nanomedicines particularly armed with monoclonal antibodies are considered to be the most promising advanced chemotherapy for malignant cancers; however, their development is hindered by their instability and drug leakage problems. Herein, we constructed a robust cetuximab-polymersome-mertansine nanodrug (C-P-DM1) for highly potent and targeted therapy of epidermal growth factor receptor (EGFR)-positive solid tumors. C-P-DM1 with a tailored cetuximab surface density of 2 per P-DM1 exhibited a size of ca. 60 nm, high stability with minimum DM1 leakage, glutathione-triggered release of native DM1, and 6.0-11.3-fold stronger cytotoxicity in EGFR-positive human breast (MDA-MB-231), lung (A549), and liver (SMMC-7721) cancer cells (IC50 = 27.1-135.5 nM) than P-DM1 control. Notably, intravenous injection of C-P-DM1 effectively repressed subcutaneous MDA-MB-231 breast cancer and orthotopic A549-Luc lung carcinoma in mice without inducing toxic effects. Strikingly, intratumoral injection of C-P-DM1 completely cured 60% of mice bearing breast tumor without recurrence. This robust cetuximab-polymersome-mertansine nanodrug provides a promising new strategy for targeted treatment of EGFR-positive solid malignancies.
Collapse
Affiliation(s)
- Shujing Yue
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Yaohua Wei
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Rainer Haag
- Department of Biology, Chemistry and Pharmacy, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Huanli Sun
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
29
|
Zhang Y, Li T, Hu Y, Chen J, He Y, Gao X, Zhang Y. Co-delivery of doxorubicin and curcumin via cRGD-peptide modified PEG-PLA self-assembly nanomicelles for lung cancer therapy. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.11.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
30
|
Combes GF, Vučković AM, Perić Bakulić M, Antoine R, Bonačić-Koutecky V, Trajković K. Nanotechnology in Tumor Biomarker Detection: The Potential of Liganded Nanoclusters as Nonlinear Optical Contrast Agents for Molecular Diagnostics of Cancer. Cancers (Basel) 2021; 13:4206. [PMID: 34439360 PMCID: PMC8393257 DOI: 10.3390/cancers13164206] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of premature death, and, as such, it can be prevented by developing strategies for early and accurate diagnosis. Cancer diagnostics has evolved from the macroscopic detection of malignant tissues to the fine analysis of tumor biomarkers using personalized medicine approaches. Recently, various nanomaterials have been introduced into the molecular diagnostics of cancer. This has resulted in a number of tumor biomarkers that have been detected in vitro and in vivo using nanodevices and corresponding imaging techniques. Atomically precise ligand-protected noble metal quantum nanoclusters represent an interesting class of nanomaterials with a great potential for the detection of tumor biomarkers. They are characterized by high biocompatibility, low toxicity, and suitability for controlled functionalization with moieties specifically recognizing tumor biomarkers. Their non-linear optical properties are of particular importance as they enable the visualization of nanocluster-labeled tumor biomarkers using non-linear optical techniques such as two-photon-excited fluorescence and second harmonic generation. This article reviews liganded nanoclusters among the different nanomaterials used for molecular cancer diagnosis and the relevance of this new class of nanomaterials as non-linear optical probe and contrast agents.
Collapse
Affiliation(s)
- Guillaume F. Combes
- Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000 Split, Croatia; (G.F.C.); (A.-M.V.); (M.P.B.); (V.B.-K.)
- Mediterranean Institute for Life Sciences (MedILS), 21000 Split, Croatia
| | - Ana-Marija Vučković
- Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000 Split, Croatia; (G.F.C.); (A.-M.V.); (M.P.B.); (V.B.-K.)
- Mediterranean Institute for Life Sciences (MedILS), 21000 Split, Croatia
| | - Martina Perić Bakulić
- Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000 Split, Croatia; (G.F.C.); (A.-M.V.); (M.P.B.); (V.B.-K.)
| | - Rodolphe Antoine
- UMR 5306, Centre National de la Recherche Scientifique (CNRS), Institute Lumière Matière, Claude Bernard University Lyon 1, F-69622 Villeurbanne, France;
| | - Vlasta Bonačić-Koutecky
- Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000 Split, Croatia; (G.F.C.); (A.-M.V.); (M.P.B.); (V.B.-K.)
- Interdisciplinary Center for Advanced Science and Technology (ICAST), University of Split, 21000 Split, Croatia
- Chemistry Department, Humboldt University of Berlin, 12489 Berlin, Germany
| | - Katarina Trajković
- Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000 Split, Croatia; (G.F.C.); (A.-M.V.); (M.P.B.); (V.B.-K.)
- Mediterranean Institute for Life Sciences (MedILS), 21000 Split, Croatia
| |
Collapse
|
31
|
Gupta N, Yadav V, Patel R. A brief review of the essential role of nanovehicles for improving the therapeutic efficacy of pharmacological agents against tumours. Curr Drug Deliv 2021; 19:301-316. [PMID: 34391379 DOI: 10.2174/1567201818666210813144105] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 11/22/2022]
Abstract
Cancer is the leading cause of death globally. There are several differences between cancer cells and normal cells. From all the therapies, chemotherapy is the most prominent therapy to treat cancer. However, the conventional drug delivery that is used to deliver poorly aqueous soluble chemotherapeutic agents has several obstacles such as whole-body distribution, rapid excretion, degradation before reaching the infected site, side effects, etc. Nanoformulation of these aqueous insoluble agents is the emerging delivery system for targeted and increasing solubility. Among all the three methods (physical, chemical and biological) chemical and biological methods are mostly used for the synthesis of nanovehicles (NVs) of different sizes, shapes and dimensions. A passive targeting delivery system in which NVs supports the pharmacological agents (drugs/genes) is a good way for resolving the obstacles with a conventional delivery system. It enhances the therapeutic efficacy of pharmacological agents (drugs/genes). These NVs have several specific characters like small size, large surface area to volume ratio, surface functionalization, etc. However, this delivery is not able to deliver site-specific delivery of drugs. An active targeting delivery system in which pharmacological agents are loaded on NVs to attack directly on cancer cells and tissues is a superior way for delivering the pharmacological agents compared to a passive targeting delivery system. Various targeting ligands have been investigated and applied for targeting the delivery of drugs such as sugar, vitamin, antibodies, protein, peptides, etc. These targeted ligand supports to guide the NVs accumulated directly on the cancer cells with a higher level of cellular internalization compared to passive targeting and conventional delivery system.
Collapse
Affiliation(s)
- Nitin Gupta
- School of Nano Sciences, Central University of Gujarat, Gandhinagar- 382030, Gujarat, India
| | - Virendra Yadav
- Department of Microbiology, School of Life Sciences, Jaipur National University, Jaipur- 341503, Rajasthan, India
| | - Rakesh Patel
- Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana- 384012, Gujarat, India
| |
Collapse
|
32
|
Kadkhoda J, Akrami-Hasan-Kohal M, Tohidkia MR, Khaledi S, Davaran S, Aghanejad A. Advances in antibody nanoconjugates for diagnosis and therapy: A review of recent studies and trends. Int J Biol Macromol 2021; 185:664-678. [PMID: 34224755 DOI: 10.1016/j.ijbiomac.2021.06.191] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023]
Abstract
Nowadays, the targeted imaging probe and drug delivery systems are the novel breakthrough area in the nanomedicine and treatment of various diseases. Conjugation of monoclonal antibodies and their fragments on nanoparticles (NPs) have a remarkable impact on personalized medicine, such that it provides specific internalization and accumulation in the tumor microenvironment. Targeted imaging and early detection of cancer is presumably the strong participant to a diminution in mortality and recurrence of cancer disease that will be the next generation of the imaging device in clinical application. These intelligent delivery systems can deliver therapeutic agents that target cancerous tissue with minimal side effects and a wide therapeutic window. Overall, the linkage between the antibody and NPs is a critical subject and requires precise design and development. The attachment of antibody nanoconjugates (Ab-NCs) on the antigen surface shouldn't affect the function of the antibody-antigen binding. Also, the stability of the antibody nanoconjugates in blood circulation is concerned to avoid the release of drug in non-targeted regions and the possible for specific toxicity while disposal to the desired site. Here, we update the recent progress of Ab-NCs to improve early detection and cancer therapy.
Collapse
Affiliation(s)
- Jamileh Kadkhoda
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Akrami-Hasan-Kohal
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Khaledi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Vasantha Ramachandran R, Bhat R, Kumar Saini D, Ghosh A. Theragnostic nanomotors: Successes and upcoming challenges. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1736. [PMID: 34173342 DOI: 10.1002/wnan.1736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022]
Abstract
The idea of "fantastic voyagers" carrying out medical tasks within the human body has existed as part of popular culture for many decades. The concept revolved around a miniaturized robot that can travel inside the human body and perform complicated functions such as surgery, navigation of otherwise inaccessible biological environments, and delivery of therapeutics. Since the last decade, significant developments have occurred in this arena that are yet to enter mainstream biomedical practises. Here, we define the challenges to make this fiction into reality. We begin by chalking the journey from pills, nanoparticles, and then to micro-nanomotors. The review describes the principles, physicochemical contexts, and advantages that micro-nanomotors provide. The article then describes micro-nanomotors' obstacles such as maneuverability, in vivo imaging, toxicity, and biodistribution. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
| | - Ramray Bhat
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Deepak Kumar Saini
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India.,Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Ambarish Ghosh
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, India.,Department of Physics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
34
|
Liu R, Poma A. Advances in Molecularly Imprinted Polymers as Drug Delivery Systems. Molecules 2021; 26:3589. [PMID: 34208380 PMCID: PMC8231147 DOI: 10.3390/molecules26123589] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the tremendous efforts made in the past decades, severe side/toxic effects and poor bioavailability still represent the main challenges that hinder the clinical translation of drug molecules. This has turned the attention of investigators towards drug delivery vehicles that provide a localized and controlled drug delivery. Molecularly imprinted polymers (MIPs) as novel and versatile drug delivery vehicles have been widely studied in recent years due to the advantages of selective recognition, enhanced drug loading, sustained release, and robustness in harsh conditions. This review highlights the design and development of strategies undertaken for MIPs used as drug delivery vehicles involving different drug delivery mechanisms, such as rate-programmed, stimuli-responsive and active targeting, published during the course of the past five years.
Collapse
Affiliation(s)
- Rui Liu
- UCL School of Pharmacy, 29–39 Brunswick Square, Bloomsbury, London WC1N 1AX, UK;
| | - Alessandro Poma
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Hospital, UCL Medical School, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
35
|
Zou M, Du Y, Liu R, Zheng Z, Xu J. Nanocarrier-delivered small interfering RNA for chemoresistant ovarian cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1648. [PMID: 33682310 DOI: 10.1002/wrna.1648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States. Because success in early screening is limited, and most patients with advanced disease develop resistance to multiple treatment modalities, the overall prognosis of ovarian cancer is poor. Despite the revolutionary role of surgery and chemotherapy in curing ovarian cancer, recurrence remains a major challenge in treatment. Thus, improving our understanding of the pathogenesis of ovarian cancer is essential for developing more effective treatments. In this review, we analyze the underlying molecular mechanisms leading to chemotherapy resistance. We discuss the clinical benefits and potential challenges of using nanocarrier-delivered small interfering RNA to treat chemotherapy-resistant ovarian cancer. We aim to elicit collaborative studies on nanocarrier-delivered small interfering RNA to improve the long-term survival rate and quality of life of patients with ovarian cancer. This article is categorized under: RNA Methods > RNA Nanotechnology Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Mingyuan Zou
- Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Liu
- The First People's Hospital of Wu'an, Wu'an, Hebei, China
| | - Zeliang Zheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
37
|
Kong X, Cheng R, Wang J, Fang Y, Hwang KC. Nanomedicines inhibiting tumor metastasis and recurrence and their clinical applications. NANO TODAY 2021; 36:101004. [DOI: 10.1016/j.nantod.2020.101004] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Conventional Nanosized Drug Delivery Systems for Cancer Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:3-27. [PMID: 33543453 DOI: 10.1007/978-3-030-58174-9_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clinical responses and tolerability of conventional nanocarriers (NCs) are sometimes different from those expected in anticancer therapy. Thus, new smart drug delivery systems (DDSs) with stimuli-responsive properties and novel materials have been developed. Several clinical trials demonstrated that these DDSs have better clinical therapeutic efficacy in the treatment of many cancers than free drugs. Composition of DDSs and their surface properties increase the specific targeting of therapeutics versus cancer cells, without affecting healthy tissues, and thus limiting their toxicity versus unspecific tissues. Herein, an extensive revision of literature on NCs used as DDSs for cancer applications has been performed using the available bibliographic databases.
Collapse
|
39
|
Akhter MH, Beg S, Tarique M, Malik A, Afaq S, Choudhry H, Hosawi S. Receptor-based targeting of engineered nanocarrier against solid tumors: Recent progress and challenges ahead. Biochim Biophys Acta Gen Subj 2020; 1865:129777. [PMID: 33130062 DOI: 10.1016/j.bbagen.2020.129777] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Background In past few decades, the research on engineered nanocarriers (NCs) has gained significant attention in cancer therapy due to selective delivery of drug molecules on the diseased cells thereby preventing unwanted uptake into healthy cells to cause toxicity. Scope of review The applicability of enhanced permeability and retention (EPR) effect for the delivery of nanomedicines in cancer therapy has gained limited success due to poor accessibility of the drugs to the target cells where non-specific payload delivery to the off target region lack substantial reward over the conventional therapeutic systems. Major conclusions In spite of the fact, nanomedicines fabricated from the biocompatible nanocarriers have reduced targeting potential for meaningful clinical benefits. However, over expression of receptors on the tumor cells provides opportunity to design functional nanomedicine to bind substantially and deliver therapeutics to the cells or tissues of interest by alleviating the bio-toxicity and unwanted effects. This critique will give insight into the over expressed receptor in various tumor and targeting potential of functional nanomedicine as new therapeutic avenues for effective treatment. General significance This review shortly shed light on EPR-based drug targeting using nanomedicinal strategies, their limitation, and advances in therapeutic targeting to the tumor cells.
Collapse
Affiliation(s)
- Md Habban Akhter
- Department of Pharmaceutics, Faculty of Pharmacy, DIT University, Dehradun, India
| | - Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Mohammed Tarique
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Arshi Malik
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Biochemistry, Cancer Metabolism & Epigenetic Unit, Faculty of Science, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
40
|
Akahane T, Kanomata N, Harada O, Yamashita T, Kurebayashi J, Tanimoto A, Moriya T. Targeted next-generation sequencing assays using triplet samples of normal breast tissue, primary breast cancer, and recurrent/metastatic lesions. BMC Cancer 2020; 20:944. [PMID: 33004031 PMCID: PMC7528467 DOI: 10.1186/s12885-020-07432-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 09/16/2020] [Indexed: 02/08/2023] Open
Abstract
Background Next-generation sequencing (NGS) has shown that recurrent/metastatic breast cancer lesions may have additional genetic changes compared with the primary tumor. These additional changes may be related to tumor progression and/or drug resistance. However, breast cancer-targeted NGS is not still widely used in clinical practice to compare the genomic profiles of primary breast cancer and recurrent/metastatic lesions. Methods Triplet samples of genomic DNA were extracted from each patient’s normal breast tissue, primary breast cancer, and recurrent/metastatic lesion(s). A DNA library was constructed using the QIAseq Human Breast Cancer Panel (93 genes, Qiagen) and then sequenced using MiSeq (Illumina). The Qiagen web portal was utilized for data analysis. Results Successful results for three or four samples (normal breast tissue, primary tumor, and at least one metastatic/recurrent lesion) were obtained for 11 of 35 breast cancer patients with recurrence/metastases (36 samples). We detected shared somatic mutations in all but one patient, who had a germline mutation in TP53. Additional mutations that were detected in recurrent/metastatic lesions compared with primary tumor were in genes including TP53 (three patients) and one case each of ATR, BLM, CBFB, EP300, ERBB2, MUC16, PBRM1, and PIK3CA. Actionable mutations and/or copy number variations (CNVs) were detected in 73% (8/11) of recurrent/metastatic breast cancer lesions. Conclusions The QIAseq Human Breast Cancer Panel assay showed that recurrent/metastatic breast cancers sometimes acquired additional mutations and CNV. Such additional genomic changes could provide therapeutic target.
Collapse
Affiliation(s)
- Toshiaki Akahane
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Department of Pathology, Hokuto Hospital, Obihiro, Hokkaido, Japan
| | - Naoki Kanomata
- Department of Pathology, St. Luke's International Hospital, Akashi-cho 9-1, Chuoku, Tokyo, 104-8560, Japan. .,Department of Pathology, Kawasaki Medical School, Kurashiki, Japan.
| | - Oi Harada
- Department of Pathology, Hokuto Hospital, Obihiro, Hokkaido, Japan
| | - Tetsumasa Yamashita
- Department of Breast and Thyroid Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Junichi Kurebayashi
- Department of Breast and Thyroid Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Akihide Tanimoto
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takuya Moriya
- Department of Pathology, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
41
|
Sorolla A, Sorolla MA, Wang E, Ceña V. Peptides, proteins and nanotechnology: a promising synergy for breast cancer targeting and treatment. Expert Opin Drug Deliv 2020; 17:1597-1613. [PMID: 32835538 DOI: 10.1080/17425247.2020.1814733] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The use of nanoparticles for breast cancer targeting and treatment has become a reality. They are safe and possess interesting peculiarities such as the unspecific accumulation into the tumor site and the possibility to activate controlled drug release as compared to free drugs. However, there are still many areas of improvement which can certainly be addressed with the use of peptide-based elements. AREAS COVERED The article reviews different preclinical strategies employing peptides and proteins in combination with nanoparticles for breast cancer targeting and treatment as well as peptide and protein-targeted encapsulated drugs, and it lists the current clinical status of therapies using peptides and proteins for breast cancer. EXPERT OPINION The conjugation of protein and peptides can improve tumor homing of nanoparticles, increase cellular penetration and attack specific drivers and vulnerabilities of the breast cancer cell to promote tumor cytotoxicity while reducing secondary effects in healthy tissues. Examples are the use of antibodies, arginylglycylaspartic acid (RGD) peptides, membrane disruptive peptides, interference peptides, and peptide vaccines. Although their implementation in the clinic has been relatively slow up to now, we anticipate great progress in the field which will translate into more efficacious and selective nanotherapies for breast cancer.
Collapse
Affiliation(s)
- Anabel Sorolla
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Maria Alba Sorolla
- Biomedical Research Institute (IRB Lleida), Research Group of Cancer Biomarkers , Lleida, Spain
| | - Edina Wang
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad De Castilla-La Mancha , Albacete, Spain.,Centro De Investigación En Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII , Madrid, Spain
| |
Collapse
|
42
|
Juan A, Cimas FJ, Bravo I, Pandiella A, Ocaña A, Alonso-Moreno C. An Overview of Antibody Conjugated Polymeric Nanoparticles for Breast Cancer Therapy. Pharmaceutics 2020; 12:pharmaceutics12090802. [PMID: 32854255 PMCID: PMC7558516 DOI: 10.3390/pharmaceutics12090802] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/09/2023] Open
Abstract
Nanoparticles (NPs) are promising drug delivery systems (DDS) for identifying and treating cancer. Active targeting NPs can be generated by conjugation with ligands that bind overexpressed or mutant cell surface receptors on target cells that are poorly or not even expressed on normal cells. Receptor-mediated endocytosis of the NPs occurs and the drug is released inside the cell or in the surrounding tissue due to the bystander effect. Antibodies are the most frequently used ligands to actively target tumor cells. In this context, antibody-based therapies have been extensively used in HER2+ breast cancer. However, some patients inherently display resistance and in advanced stages, almost all eventually progress. Functionalized NPs through conjugation with antibodies appear to be a promising strategy to optimize targeted therapies due to properties related to biocompatibility, suitable delivery control and efficiency of functionalization. This review is focused on the different strategies to conjugate antibodies into polymeric NPs. Recent antibody conjugation approaches applied to the improvement of breast cancer therapy are highlighted in this review.
Collapse
Affiliation(s)
- Alberto Juan
- Oncología traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Francisco J. Cimas
- Oncología traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer-CSIC, IBSAL- Salamanca and CIBERONC, 37007 Salamanca, Spain;
| | - Alberto Ocaña
- Oncología traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Experimental Therapeutics Unit, Hospital clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain
- Correspondence: (A.O.); (C.A.-M.); Tel.: +34-635-681806 (A.O.); +34-9675-99200 (C.A.-M)
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
- Correspondence: (A.O.); (C.A.-M.); Tel.: +34-635-681806 (A.O.); +34-9675-99200 (C.A.-M)
| |
Collapse
|
43
|
Ju Y, Guo H, Edman M, Hamm-Alvarez SF. Application of advances in endocytosis and membrane trafficking to drug delivery. Adv Drug Deliv Rev 2020; 157:118-141. [PMID: 32758615 PMCID: PMC7853512 DOI: 10.1016/j.addr.2020.07.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022]
Abstract
Multidisciplinary research efforts in the field of drug delivery have led to the development of a variety of drug delivery systems (DDS) designed for site-specific delivery of diagnostic and therapeutic agents. Since efficient uptake of drug carriers into target cells is central to effective drug delivery, a comprehensive understanding of the biological pathways for cellular internalization of DDS can facilitate the development of DDS capable of precise tissue targeting and enhanced therapeutic outcomes. Diverse methods have been applied to study the internalization mechanisms responsible for endocytotic uptake of extracellular materials, which are also the principal pathways exploited by many DDS. Chemical inhibitors remain the most commonly used method to explore endocytotic internalization mechanisms, although genetic methods are increasingly accessible and may constitute more specific approaches. This review highlights the molecular basis of internalization pathways most relevant to internalization of DDS, and the principal methods used to study each route. This review also showcases examples of DDS that are internalized by each route, and reviews the general effects of biophysical properties of DDS on the internalization efficiency. Finally, options for intracellular trafficking and targeting of internalized DDS are briefly reviewed, representing an additional opportunity for multi-level targeting to achieve further specificity and therapeutic efficacy.
Collapse
Affiliation(s)
- Yaping Ju
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, USA
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, USA
| | - Maria Edman
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, USA
| | - Sarah F Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, USA; Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
44
|
Zhang H, Dong S, Li Z, Feng X, Xu W, Tulinao CMS, Jiang Y, Ding J. Biointerface engineering nanoplatforms for cancer-targeted drug delivery. Asian J Pharm Sci 2020; 15:397-415. [PMID: 32952666 PMCID: PMC7486517 DOI: 10.1016/j.ajps.2019.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/22/2019] [Accepted: 11/18/2019] [Indexed: 12/30/2022] Open
Abstract
Over the past decade, nanoparticle-based therapeutic modalities have become promising strategies in cancer therapy. Selective delivery of anticancer drugs to the lesion sites is critical for elimination of the tumor and an improved prognosis. Innovative design and advanced biointerface engineering have promoted the development of various nanocarriers for optimized drug delivery. Keeping in mind the biological framework of the tumor microenvironment, biomembrane-camouflaged nanoplatforms have been a research focus, reflecting their superiority in cancer targeting. In this review, we summarize the development of various biomimetic cell membrane-camouflaged nanoplatforms for cancer-targeted drug delivery, which are classified according to the membranes from different cells. The challenges and opportunities of the advanced biointerface engineering drug delivery nanosystems in cancer therapy are discussed.
Collapse
Affiliation(s)
- Huaiyu Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Shujun Dong
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | | | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
45
|
Mainini F, Eccles MR. Lipid and Polymer-Based Nanoparticle siRNA Delivery Systems for Cancer Therapy. Molecules 2020; 25:E2692. [PMID: 32532030 PMCID: PMC7321291 DOI: 10.3390/molecules25112692] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 01/08/2023] Open
Abstract
RNA interference (RNAi) uses small interfering RNAs (siRNAs) to mediate gene-silencing in cells and represents an emerging strategy for cancer therapy. Successful RNAi-mediated gene silencing requires overcoming multiple physiological barriers to achieve efficient delivery of siRNAs into cells in vivo, including into tumor and/or host cells in the tumor micro-environment (TME). Consequently, lipid and polymer-based nanoparticle siRNA delivery systems have been developed to surmount these physiological barriers. In this article, we review the strategies that have been developed to facilitate siRNA survival in the circulatory system, siRNA movement from the blood into tissues and the TME, targeted siRNA delivery to the tumor or specific cell types, cellular uptake, and escape from endosomal degradation. We also discuss the use of various types of lipid and polymer-based carriers for cancer therapy, including a section on anti-tumor nanovaccines enhanced by siRNAs. Finally, we review current and recent clinical trials using NPs loaded with siRNAs for cancer therapy. The siRNA cancer therapeutics field is rapidly evolving, and it is conceivable that precision cancer therapy could, in the relatively near future, benefit from the combined use of cancer therapies, for example immune checkpoint blockade together with gene-targeting siRNAs, personalized for enhancing and fine-tuning a patient's therapeutic response.
Collapse
Affiliation(s)
| | - Michael R. Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand;
| |
Collapse
|
46
|
Dynamic covalent chemistry-regulated stimuli-activatable drug delivery systems for improved cancer therapy. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2019.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
47
|
Muley H, Fadó R, Rodríguez-Rodríguez R, Casals N. Drug uptake-based chemoresistance in breast cancer treatment. Biochem Pharmacol 2020; 177:113959. [PMID: 32272110 DOI: 10.1016/j.bcp.2020.113959] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most prevalent type of tumor and the second leading cause of death due to cancer among women. Although screening methods, diagnosis and therapeutic options have improved in the last decade, chemoresistance remains an important challenge. There is evidence relating breast cancer resistance with signaling pathways involving hormone and growth receptors, survival, apoptosis and the activation of efflux pumps. However, the resistance mechanisms linked to drug uptake are poorly understood, despite it often being observed that the drug content is lower in resistant cancer cells and that the entry of the drug into these cells is a limiting process for the subsequent therapeutic effect.In this review, we provide an overview of drug uptake-based resistance mechanisms developed by cancer cells in the four main types of chemotherapy used in breast cancer: anthracyclines, taxanes, oxazaphosphorines and platinum-based drugs. The contribution of tumor microenvironment to reduced drug-uptake and multidrug resistance is also analyzed. As a developing field, nanomedicine-based approaches provide promising opportunities to improve drug specific targeting, cell interaction and uptake into cancer cells. The endocytic-mediated pathways attributed to the different types of nanoformulations as well as the contribution of nanotherapeutics to overcoming chemoresistance affecting drug uptake in breast cancer will be described. New approaches focusing on drug uptake mechanisms could improve breast cancer chemotherapy, obtaining better dose-response outcomes and reducing toxic side effects.
Collapse
Affiliation(s)
- Helena Muley
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Rosalía Rodríguez-Rodríguez
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain.
| |
Collapse
|
48
|
Falgàs A, Pallarès V, Serna N, Sánchez-García L, Sierra J, Gallardo A, Alba-Castellón L, Álamo P, Unzueta U, Villaverde A, Vázquez E, Mangues R, Casanova I. Selective delivery of T22-PE24-H6 to CXCR4 + diffuse large B-cell lymphoma cells leads to wide therapeutic index in a disseminated mouse model. Theranostics 2020; 10:5169-5180. [PMID: 32373205 PMCID: PMC7196303 DOI: 10.7150/thno.43231] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Novel therapeutic strategies are urgently needed to reduce relapse rates and enhance survival in Diffuse Large B-Cell Lymphoma (DLBCL) patients. CXCR4-overexpressing cancer cells are good targets for therapy because of their association with dissemination and relapse in R-CHOP treated DLBCL patients. Immunotoxins that incorporate bacterial toxins are potentially effective in treating haematological neoplasias, but show a narrow therapeutic index due to the induction of severe side effects. Therefore, when considering the delivery of these toxins as cancer therapeutics, there is a need not only to increase their uptake in the target cancer cells, and their stability in blood, but also to reduce their systemic toxicity. We have developed a therapeutic nanostructured protein T22-PE24-H6 that incorporates exotoxin A from Pseudomonas aeruginosa, which selectively targets lymphoma cells because of its specific interaction with a highly overexpressed CXCR4 receptor (CXCR4+) in DLBCL. Methods: T22-PE24-H6 cytotoxicity and its dependence on the CXCR4 receptor were evaluated in DLBCL cell lines using cell viability assays. Different in vitro experiments (mitochondrial membrane potential, Western Blot, Annexin V and DAPI staining) were conducted to determine T22-PE24-H6 cell death mechanisms. In vivo imaging and therapeutic effect studies were performed in a disseminated DLBCL mouse model that mimics organ infiltration in DLBCL patients. Finally, immunohistochemistry and histopathology analyses were used to evaluate the antineoplastic effect and systemic toxicity. Results: In vitro, T22-PE24-H6 induced selective cell death of CXCR4+ DLBCL cells by activating the apoptotic pathway. In addition, repeated T22-PE24-H6 intravenous administration in a CXCR4+ DLBCL-disseminated mouse model showed a significant reduction of lymphoma burden in organs clinically affected by DLBCL cells (lymph nodes and bone marrow). Finally, we did not observe systemic toxicity associated to the nanoparticle treatment in non-DLBCL-infiltrated organs. Conclusion: We have demonstrated here a potent T22-PE24-H6 antineoplastic effect, especially in blocking dissemination in a CXCR4+ DLBCL model without associated toxicity. Thereby, T22-PE24-H6 promises to become an effective alternative to treat CXCR4+ disseminated refractory or relapsed DLBCL patients.
Collapse
|
49
|
Marques AC, Costa PJ, Velho S, Amaral MH. Functionalizing nanoparticles with cancer-targeting antibodies: A comparison of strategies. J Control Release 2020; 320:180-200. [PMID: 31978444 DOI: 10.1016/j.jconrel.2020.01.035] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 01/07/2023]
Abstract
Standard cancer therapies sometimes fail to deliver chemotherapeutic drugs to tumor cells in a safe and effective manner. Nanotechnology takes the lead in providing new therapeutic options for cancer due to major potential for selective targeting and controlled drug release. Antibodies and antibody fragments are attracting much attention as a source of targeting ligands to bind specific receptors that are overexpressed on cancer cells. Therefore, researchers are devoting time and effort to develop targeting strategies based on nanoparticles functionalized with antibodies, which hold great promise to enhance therapeutic efficacy and circumvent severe side effects. Several methods have been described to immobilize antibodies on the surface of nanoparticles. However, selecting the most appropriate for each application is challenging but also imperative to preserve antigen binding ability and yield stable antibody-conjugated nanoparticles. From this perspective, we aim to provide considerable knowledge on the most widely used methods of functionalization that can be helpful for decision-making and design of conjugation protocols as well. This review summarizes adsorption, covalent conjugation (carbodiimide, maleimide and "click" chemistries) and biotin-avidin interaction, while discussing the advantages, limitations and relevant therapeutic approaches currently under investigation.
Collapse
Affiliation(s)
- A C Marques
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - P J Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - S Velho
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
50
|
Morais-Rodrigues F, Silv Erio-Machado R, Kato RB, Rodrigues DLN, Valdez-Baez J, Fonseca V, San EJ, Gomes LGR, Dos Santos RG, Vinicius Canário Viana M, da Cruz Ferraz Dutra J, Teixeira Dornelles Parise M, Parise D, Campos FF, de Souza SJ, Ortega JM, Barh D, Ghosh P, Azevedo VAC, Dos Santos MA. Analysis of the microarray gene expression for breast cancer progression after the application modified logistic regression. Gene 2019; 726:144168. [PMID: 31759986 DOI: 10.1016/j.gene.2019.144168] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/21/2019] [Accepted: 10/11/2019] [Indexed: 01/02/2023]
Abstract
Methods based around statistics and linear algebra have been increasingly used in attempts to address emerging questions in microarray literature. Microarray technology is a long-used tool in the global analysis of gene expression, allowing for the simultaneous investigation of hundreds or thousands of genes in a sample. It is characterized by a low sample size and a large feature number created a non-square matrix, and by the incomplete rank, that can generate countless more solution in classifiers. To avoid the problem of the 'curse of dimensionality' many authors have performed feature selection or reduced the size of data matrix. In this work, we introduce a new logistic regression-based model to classify breast cancer tumor samples based on microarray expression data, including all features of gene expression and without reducing the microarray data matrix. If the user still deems it necessary to perform feature reduction, it can be done after the application of the methodology, still maintaining a good classification. This methodology allowed the correct classification of breast cancer sample data sets from Gene Expression Omnibus (GEO) data series GSE65194, GSE20711, and GSE25055, which contain the microarray data of said breast cancer samples. Classification had a minimum performance of 80% (sensitivity and specificity), and explored all possible data combinations, including breast cancer subtypes. This methodology highlighted genes not yet studied in breast cancer, some of which have been observed in Gene Regulatory Networks (GRNs). In this work we examine the patterns and features of a GRN composed of transcription factors (TFs) in MCF-7 breast cancer cell lines, providing valuable information regarding breast cancer. In particular, some genes whose αi ∗ associated parameter values revealed extreme positive and negative values, and, as such, can be identified as breast cancer prediction genes. We indicate that the PKN2, MKL1, MED23, CUL5 and GLI genes demonstrate a tumor suppressor profile, and that the MTR, ITGA2B, TELO2, MRPL9, MTTL1, WIPI1, KLHL20, PI4KB, FOLR1 and SHC1 genes demonstrate an oncogenic profile. We propose that these may serve as potential breast cancer prediction genes, and should be prioritized for further clinical studies on breast cancer. This new model allows for the assignment of values to the αi ∗ parameters associated with gene expression. It was noted that some αi ∗ parameters are associated with genes previously described as breast cancer biomarkers, as well as other genes not yet studied in relation to this disease.
Collapse
Affiliation(s)
- Francielly Morais-Rodrigues
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil.
| | - Rita Silv Erio-Machado
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Rodrigo Bentes Kato
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Diego Lucas Neres Rodrigues
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Juan Valdez-Baez
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Vagner Fonseca
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil; KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Emmanuel James San
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Lucas Gabriel Rodrigues Gomes
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Roselane Gonçalves Dos Santos
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Marcus Vinicius Canário Viana
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil; Federal University of Pará, UFPA, Brazil
| | - Joyce da Cruz Ferraz Dutra
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Mariana Teixeira Dornelles Parise
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Doglas Parise
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Frederico F Campos
- Department of Computer Science, Federal University of Minas Gerais, Brazil Av Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | | | - José Miguel Ortega
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal 721172, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Vasco A C Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| | - Marcos A Dos Santos
- Department of Computer Science, Federal University of Minas Gerais, Brazil Av Antônio Carlos, 6627, Belo Horizonte, MG 31270-901, Brazil
| |
Collapse
|