1
|
Boix-Montesinos P, Medel M, Malfanti A, Đorđević S, Masiá E, Charbonnier D, Carrascosa-Marco P, Armiñán A, Vicent MJ. Rational design of a poly-L-glutamic acid-based combination conjugate for hormone-responsive breast cancer treatment. J Control Release 2024; 375:193-208. [PMID: 39242032 DOI: 10.1016/j.jconrel.2024.09.002] [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: 06/17/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer represents the most prevalent tumor type worldwide, with hormone-responsive breast cancer the most common subtype. Despite the effectiveness of endocrine therapy, advanced disease forms represent an unmet clinical need. While drug combination therapies remain promising, differences in pharmacokinetic profiles result in suboptimal ratios of free drugs reaching tumors. We identified a synergistic combination of bisdemethoxycurcumin and exemestane through drug screening and rationally designed star-shaped poly-L-glutamic acid-based combination conjugates carrying these drugs conjugated through pH-responsive linkers for hormone-responsive breast cancer treatment. We synthesized/characterized single and combination conjugates with synergistic drug ratios/loadings. Physicochemical characterization/drug release kinetics studies suggested that lower drug loading prompted a less compact conjugate conformation that supported optimal release. Screening in monolayer and spheroid breast cancer cell cultures revealed that combination conjugates possessed enhanced cytotoxicity/synergism compared to physical mixtures of single-drug conjugates/free drugs; moreover, a combination conjugate with the lowest drug loading outperformed remaining conjugates. This candidate inhibited proliferation-associated signaling, reduced inflammatory chemokine/exosome levels, and promoted autophagy in spheroids; furthermore, it outperformed a physical mixture of single-drug conjugates/free drugs regarding cytotoxicity in patient-derived breast cancer organoids. Our findings highlight the importance of rational design and advanced in vitro models for the selection of polypeptide-based combination conjugates.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - María Medel
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain
| | - Alessio Malfanti
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Snežana Đorđević
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain
| | - Esther Masiá
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - David Charbonnier
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), IISCIII and CIEMAT, Madrid, Spain
| | - Paula Carrascosa-Marco
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - Ana Armiñán
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain.
| | - María J Vicent
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
2
|
Shayegh F, Türk Z, Armani A, Zarghami N. New insights into polysaccharide-based nanostructured delivery systems in breast cancer: Possible application of antisense oligonucleotides in breast cancer therapy. Int J Biol Macromol 2024; 272:132890. [PMID: 38848829 DOI: 10.1016/j.ijbiomac.2024.132890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 05/27/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
The lack of more effective therapies for breast cancer has enhanced mortality among breast cancer patients. Recent efforts have established efficient treatments to reduce breast cancer-related deaths. The ever-increasing attraction to employing biocompatible polysaccharide-based nanostructures as delivery systems has created interest in various disease therapies, especially breast cancer treatment. A wide range of therapeutic cargo comprising bioactive or chemical drugs, oligonucleotides, peptides, and targeted biomarkers have been considered to comprehend their anti-cancer effects against breast cancer. Some limitations of naked agents or undesired constructs, such as no or low bioavailability, enzymatic digestion, short-range stability, low-cellular uptake, poor solubility, and low surface area, have lessened their effectiveness. However, nanoscale formulations of therapeutic ingredients have provided a promising platform to address the mentioned concerns. For instance, some capable polysaccharides, including cellulose, pectin, chitosan, alginate, and dextran, were developed as breast cancer therapeutics with great nanoparticle structures. This review carefully examines the characteristics of beneficial polysaccharides that are utilized in the formation of nanoparticles (NPs). It also highlights the applications of antisense oligonucleotides (ASOs), and NPs made from polysaccharides in the treatment of breast cancer and suggests ways to enhance these particles for future research.
Collapse
Affiliation(s)
- Fahimeh Shayegh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeynep Türk
- Department of Analytical Chemistry, Faculty of Pharmacy, İstanbul Aydin University, İstanbul, Türkiye
| | - Arta Armani
- Department of Medical Biology and Genetics, Faculty of Medicine, İstanbul Aydin University, İstanbul, Türkiye
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biochemistry, Faculty of Medicine, İstanbul Aydin University, İstanbul, Türkiye.
| |
Collapse
|
3
|
Chellen T, Bausart M, Maus P, Vanvarenberg K, Limaye N, Préat V, Malfanti A. In situ administration of STING-activating hyaluronic acid conjugate primes anti-glioblastoma immune response. Mater Today Bio 2024; 26:101057. [PMID: 38660475 PMCID: PMC11040137 DOI: 10.1016/j.mtbio.2024.101057] [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: 02/07/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor, with a highly immunosuppressive tumor immune microenvironment (TIME). In this work, we investigated the use of the STimulator of INterferon Genes (STING) pathway as an effective means to remodel the GBM TIME through the recruitment of both innate and adaptive immune cell populations. Using hyaluronic acid (HA), we developed a novel polymer-drug conjugate of a non-nucleotide STING agonist (MSA2), called HA-MSA2 for the in situ treatment of GBM. In JAWSII cells, HA-MSA2 exerted a greater increase of STING signaling and upregulation of STING-related downstream cyto-/chemokines in immune cells than the free drug. HA-MSA2 also elicited cancer cell-intrinsic immunostimulatory gene expression and promoted immunogenic cell death of GBM cells. In the SB28 GBM model, local delivery of HA-MSA2 induced a delay in tumor growth and a significant extension of survival. The analysis of the TIME showed a profound shift in the GBM immune landscape after HA-MSA2 treatment, with higher infiltration by innate and adaptive immune cells including dendritic, natural killer (NK) and CD8 T cell populations. The therapeutic potential of this novel polymer conjugate warrants further investigation, particularly with other chemo-immunotherapeutics or cancer vaccines as a promising combinatorial therapeutic approach.
Collapse
Affiliation(s)
- Teenesha Chellen
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Pierre Maus
- UCLouvain, de Duve Institute, Genetics of Autoimmune Diseases and Cancer, Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Nisha Limaye
- UCLouvain, de Duve Institute, Genetics of Autoimmune Diseases and Cancer, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
4
|
Jamshidi Z, Dehghan R, Nejabat M, Abnous K, Taghdisi SM, Hadizadeh F. Dual-targeting CD44 and mucin by hyaluronic acid and 5TR1 aptamer for epirubicin delivery into cancer cells: Synthesis, characterization, in vitro and in vivo evaluation. Heliyon 2024; 10:e24833. [PMID: 38312665 PMCID: PMC10835225 DOI: 10.1016/j.heliyon.2024.e24833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
One of the revolutionized cancer treatment is active targeting nanomedicines. This study aims to create a dual-targeted drug delivery system for Epirubicin (EPI) to cancer cells. Hyaluronic acid (HA) is the first targeting ligand, and 5TR1 aptamer (5TR1) is the second targeting ligand to guide the dual-targeted drug delivery system to the cancer cells. HA is bound to highly expressed receptors like CD44 on cancer cells. 5TR1, DNA aptamer, is capable of recognizing MUC1 glycoprotein, which is overexpressed in cancer cells. The process involved binding EPI and 5TR1 to HA using adipic acid dihydrazide (AA) as a linker. The bond between the components was confirmed using 1H NMR. The binding of 5TR1 to HA-AA-EPI was confirmed using gel electrophoresis. The particle size (132.6 ± 9 nm) and Zeta Potential (-29 ± 4.4 mV) were measured for the final nanoformulation (HA-AA-EPI-5TR1). The release of EPI from the HA-AA-EPI-5TR1 nanoformulation was also studied at different pH levels. In the acidic pH (5.4 and 6.5) release pattern of EPI from the HA-AA-EPI-5TR1 nanoformulation was higher than physiological pH (7.4). The cytotoxicity and cellular uptake of the synthetic nanoformula were evaluated using MTT and flow cytometry analysis. Flow cytometry and cellular cytotoxicity studies were exhibited in a negative MUC1-cell line (CHO) and two positive MUC1+cell lines (MCF-7 and C26). Results confirmed that there is a notable contrast between the dual-targeted (HA-AA-EPI-5TR1) and single-targeted (HA-AA-EPI) nanoformulation in MCF-7 and C26 cell lines (MUC1+). In vivo studies showed that HA-AA-EPI-5TR1 nanoformulation has improved efficiency with limited side effect in C26 tumor-bearing mice. Also, Fluorescence imaging and pathological evaluation showed reduced side effects in the heart tissue of mice receiving HA-AA-EPI-5TR1 than free EPI. So, this targeted approach effectively delivers EPI to cancer cells with reduced side effects.
Collapse
Affiliation(s)
- Zahra Jamshidi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Dehghan
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Nejabat
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Institute, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Lin M, Wang X. Natural Biopolymer-Based Delivery of CRISPR/Cas9 for Cancer Treatment. Pharmaceutics 2023; 16:62. [PMID: 38258073 PMCID: PMC10819213 DOI: 10.3390/pharmaceutics16010062] [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: 11/16/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Over the last decade, the clustered, regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system has become the most promising gene editing tool and is broadly utilized to manipulate the gene for disease treatment, especially for cancer, which involves multiple genetic alterations. Typically, CRISPR/Cas9 machinery is delivered in one of three forms: DNA, mRNA, or ribonucleoprotein. However, the lack of efficient delivery systems for these macromolecules confined the clinical breakthrough of this technique. Therefore, a variety of nanomaterials have been fabricated to improve the stability and delivery efficiency of the CRISPR/Cas9 system. In this context, the natural biopolymer-based carrier is a particularly promising platform for CRISPR/Cas9 delivery due to its great stability, low toxicity, excellent biocompatibility, and biodegradability. Here, we focus on the advances of natural biopolymer-based materials for CRISPR/Cas9 delivery in the cancer field and discuss the challenges for their clinical translation.
Collapse
Affiliation(s)
| | - Xueyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Hajareh Haghighi F, Binaymotlagh R, Fratoddi I, Chronopoulou L, Palocci C. Peptide-Hydrogel Nanocomposites for Anti-Cancer Drug Delivery. Gels 2023; 9:953. [PMID: 38131939 PMCID: PMC10742474 DOI: 10.3390/gels9120953] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer is the second leading cause of death globally, but conventional anticancer drugs have side effects, mainly due to their non-specific distribution in the body in both cancerous and healthy cells. To address this relevant issue and improve the efficiency of anticancer drugs, increasing attention is being devoted to hydrogel drug-delivery systems for different kinds of cancer treatment due to their high biocompatibility and stability, low side effects, and ease of modifications. To improve the therapeutic efficiency and provide multi-functionality, different types of nanoparticles (NPs) can be incorporated within the hydrogels to form smart hydrogel nanocomposites, benefiting the advantages of both counterparts and suitable for advanced anticancer applications. Despite many papers on non-peptide hydrogel nanocomposites, there is limited knowledge about peptide-based nanocomposites, specifically in anti-cancer drug delivery. The aim of this short but comprehensive review is, therefore, to focus attention on the synergies resulting from the combination of NPs with peptide-based hydrogels. This review, which includes a survey of recent advances in this kind of material, does not aim to be an exhaustive review of hydrogel technology, but it instead highlights recent noteworthy publications and discusses novel perspectives to provide valuable insights into the promising synergic combination of peptide hydrogels and NPs for the design of novel anticancer drug delivery systems.
Collapse
Affiliation(s)
- Farid Hajareh Haghighi
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.H.H.); (R.B.); (I.F.)
| | - Roya Binaymotlagh
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.H.H.); (R.B.); (I.F.)
| | - Ilaria Fratoddi
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.H.H.); (R.B.); (I.F.)
| | - Laura Chronopoulou
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.H.H.); (R.B.); (I.F.)
- Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cleofe Palocci
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.H.H.); (R.B.); (I.F.)
- Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
7
|
Junyaprasert VB, Thummarati P. Innovative Design of Targeted Nanoparticles: Polymer-Drug Conjugates for Enhanced Cancer Therapy. Pharmaceutics 2023; 15:2216. [PMID: 37765185 PMCID: PMC10537251 DOI: 10.3390/pharmaceutics15092216] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Polymer-drug conjugates (PDCs) have shown great promise in enhancing the efficacy and safety of cancer therapy. These conjugates combine the advantageous properties of both polymers and drugs, leading to improved pharmacokinetics, controlled drug release, and targeted delivery to tumor tissues. This review provides a comprehensive overview of recent developments in PDCs for cancer therapy. First, various types of polymers used in these conjugates are discussed, including synthetic polymers, such as poly(↋-caprolactone) (PCL), D-α-tocopheryl polyethylene glycol (TPGS), and polyethylene glycol (PEG), as well as natural polymers such as hyaluronic acid (HA). The choice of polymer is crucial to achieving desired properties, such as stability, biocompatibility, and controlled drug release. Subsequently, the strategies for conjugating drugs to polymers are explored, including covalent bonding, which enables a stable linkage between the polymer and the drug, ensuring controlled release and minimizing premature drug release. The use of polymers can extend the circulation time of the drug, facilitating enhanced accumulation within tumor tissues through the enhanced permeability and retention (EPR) effect. This, in turn, results in improved drug efficacy and reduced systemic toxicity. Moreover, the importance of tumor-targeting ligands in PDCs is highlighted. Various ligands, such as antibodies, peptides, aptamers, folic acid, herceptin, and HA, can be incorporated into conjugates to selectively deliver the drug to tumor cells, reducing off-target effects and improving therapeutic outcomes. In conclusion, PDCs have emerged as a versatile and effective approach to cancer therapy. Their ability to combine the advantages of polymers and drugs offers enhanced drug delivery, controlled release, and targeted treatment, thereby improving the overall efficacy and safety of cancer therapies. Further research and development in this field has great potential to advance personalized cancer treatment options.
Collapse
|
8
|
Zhang X, Liu Y, Doungchawee J, Castellanos-García LJ, Sikora KN, Jeon T, Goswami R, Fedeli S, Gupta A, Huang R, Hirschbiegel CM, Cao-Milán R, Majhi PKD, Cicek YA, Liu L, Jerry DJ, Vachet RW, Rotello VM. Bioorthogonal nanozymes for breast cancer imaging and therapy. J Control Release 2023; 357:31-39. [PMID: 36948419 PMCID: PMC10164715 DOI: 10.1016/j.jconrel.2023.03.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/23/2023] [Accepted: 03/18/2023] [Indexed: 03/24/2023]
Abstract
Bioorthogonal catalysis via transition metal catalysts (TMCs) enables the generation of therapeutics locally through chemical reactions not accessible by biological systems. This localization can enhance the efficacy of anticancer treatment while minimizing off-target effects. The encapsulation of TMCs into nanomaterials generates "nanozymes" to activate imaging and therapeutic agents. Here, we report the use of cationic bioorthogonal nanozymes to create localized "drug factories" for cancer therapy in vivo. These nanozymes remained present at the tumor site at least seven days after a single injection due to the interactions between cationic surface ligands and negatively charged cell membranes and tissue components. The prodrug was then administered systemically, and the nanozymes continuously converted the non-toxic molecules into active drugs locally. This strategy substantially reduced the tumor growth in an aggressive breast cancer model, with significantly reduced liver damage compared to traditional chemotherapy.
Collapse
Affiliation(s)
- Xianzhi Zhang
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Jeerapat Doungchawee
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | | | - Kristen N Sikora
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, MA 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | | | - Roberto Cao-Milán
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Prabin K D Majhi
- Department of Veterinary and Animal Science, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, MA 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Liang Liu
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - D Joseph Jerry
- Department of Veterinary and Animal Science, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, MA 01003, USA
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| |
Collapse
|
9
|
Rama B, Ribeiro AJ. Role of nanotechnology in the prolonged release of drugs by the subcutaneous route. Expert Opin Drug Deliv 2023; 20:559-577. [PMID: 37305971 DOI: 10.1080/17425247.2023.2214362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/11/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Subcutaneous physiology is distinct from other parenteral routes that benefit the administration of prolonged-release formulations. A prolonged-release effect is particularly convenient for treating chronic diseases because it is associated with complex and often prolonged posologies. Therefore, drug-delivery systems focused on nanotechnology are proposed as alternatives that can overcome the limitations of current therapeutic regimens and improve therapeutic efficacy. AREAS COVERED This review presents an updated systematization of nanosystems, focusing on their applications in highly prevalent chronic diseases. Subcutaneous-delivered nanosystem-based therapies comprehensively summarize nanosystems, drugs, and diseases and their advantages, limitations, and strategies to increase their translation into clinical applications. An outline of the potential contribution of quality-by-design (QbD) and artificial intelligence (AI) to the pharmaceutical development of nanosystems is presented. EXPERT OPINION Although recent academic research and development (R&D) advances in the subcutaneous delivery of nanosystems have exhibited promising results, pharmaceutical industries and regulatory agencies need to catch up. The lack of standardized methodologies for analyzing in vitro data from nanosystems for subcutaneous administration and subsequent in vivo correlation limits their access to clinical trials. There is an urgent need for regulatory agencies to develop methods that faithfully mimic subcutaneous administration and specific guidelines for evaluating nanosystems.
Collapse
Affiliation(s)
- B Rama
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
| | - A J Ribeiro
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
- Genetics of Cognitive Disfunction, i3S, IBMC, Porto, Portugal
| |
Collapse
|
10
|
Chen TY, Lin NY, Wen CH, Lin CA, Venkatesan P, Wijerathna P, Lin CY, Lai PS. Development of triamcinolone acetonide-hyaluronic acid conjugates with selective targeting and less osteoporosis effect for rheumatoid arthritis treatments. Int J Biol Macromol 2023; 237:124047. [PMID: 36933598 DOI: 10.1016/j.ijbiomac.2023.124047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 03/04/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Rheumatoid arthritis (RA) is a common systemic autoimmune disease in developed countries. In clinical treatment, steroids have been used as bridging and adjunctive therapy after disease-modifying anti-rheumatic drug administration. However, the severe side effects caused by the nonspecific targeting of organs followed by long-term administration have limited their usage in RA. In this study, poorly water-soluble triamcinolone acetonide (TA), a highly potent corticosteroid for intra-articular injection, is conjugated on hyaluronic acid (HA) for intravenous purposes with increased specific drug accumulation in inflamed parts for RA. Our results demonstrate that the designed HA/TA coupling reaction reveals >98 % conjugation efficiency in the dimethyl sulfoxide/water system, and the resulting HA-TA conjugates show lower osteoblastic apoptosis compared with that in free TA-treated osteoblast-like NIH3T3 cells. Furthermore, in a collagen-antibody-induced arthritis animal study, HA-TA conjugates enhanced the initiative targeting ability to inflame tissue and reduce the histopathological arthritic changes (score = 0). Additionally, the level of bone formation marker P1NP in HA-TA-treated ovariectomized mice (303.6 ± 40.6 pg/mL) is significantly higher than that in the free TA-treated group (143.1 ± 3.9 pg/mL), indicating the potential for osteoporotic reduction using an efficient HA conjugation strategy for the long-term administration of steroids against RA.
Collapse
Affiliation(s)
- Tzu-Yang Chen
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; Basic Research Division, Holy Stone Healthcare Co., Ltd., 114 Taipei, Taiwan.
| | - Neng-Yu Lin
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Chih-Hao Wen
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chih-An Lin
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Parthiban Venkatesan
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Prasanna Wijerathna
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chung-Yu Lin
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
11
|
Pastuch-Gawołek G, Szreder J, Domińska M, Pielok M, Cichy P, Grymel M. A Small Sugar Molecule with Huge Potential in Targeted Cancer Therapy. Pharmaceutics 2023; 15:913. [PMID: 36986774 PMCID: PMC10056414 DOI: 10.3390/pharmaceutics15030913] [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: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The number of cancer-related diseases is still growing. Despite the availability of a large number of anticancer drugs, the ideal drug is still being sought that would be effective, selective, and overcome the effect of multidrug resistance. Therefore, researchers are still looking for ways to improve the properties of already-used chemotherapeutics. One of the possibilities is the development of targeted therapies. The use of prodrugs that release the bioactive substance only under the influence of factors characteristic of the tumor microenvironment makes it possible to deliver the drug precisely to the cancer cells. Obtaining such compounds is possible by coupling a therapeutic agent with a ligand targeting receptors, to which the attached ligand shows affinity and is overexpressed in cancer cells. Another way is to encapsulate the drug in a carrier that is stable in physiological conditions and sensitive to conditions of the tumor microenvironment. Such a carrier can be directed by attaching to it a ligand recognized by receptors typical of tumor cells. Sugars seem to be ideal ligands for obtaining prodrugs targeted at receptors overexpressed in cancer cells. They can also be ligands modifying polymers' drug carriers. Furthermore, polysaccharides can act as selective nanocarriers for numerous chemotherapeutics. The proof of this thesis is the huge number of papers devoted to their use for modification or targeted transport of anticancer compounds. In this work, selected examples of broad-defined sugars application for improving the properties of both already-used drugs and substances exhibiting anticancer activity are presented.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Julia Szreder
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Monika Domińska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mateusz Pielok
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Piotr Cichy
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mirosława Grymel
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| |
Collapse
|
12
|
Abbasi YF, Bera H, Cun D, Yang M. Recent advances in pH/enzyme-responsive polysaccharide-small-molecule drug conjugates as nanotherapeutics. Carbohydr Polym 2023; 312:120797. [PMID: 37059536 DOI: 10.1016/j.carbpol.2023.120797] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/26/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Now-a-days, the polysaccharides are extensively employed for the delivery of small-molecule drugs ascribed to their excellent biocompatibility, biodegradability and modifiability. An array of drug molecules is often chemically conjugated with different polysaccharides to augment their bio-performances. As compared to their therapeutic precursors, these conjugates could typically demonstrate an improved intrinsic solubility, stability, bioavailability and pharmacokinetic profiles of the drugs. In current years, various stimuli-responsive particularly pH and enzyme-sensitive linkers or pendants are also exploited to integrate the drug molecules into the polysaccharide backbone. The resulting conjugates could experience a rapid molecular conformational change upon exposure to the microenvironmental pH and enzyme changes of the diseased states, triggering the release of the bioactive cargos at the targeted sites and eventually minimize the systemic side effects. Herein, the recent advances in pH and enzyme -responsive polysaccharide-drug conjugates and their therapeutic benefits are systematically reviewed, following a brief description on the conjugation chemistry of the polysaccharides and drug molecules. The challenges and future perspectives of these conjugates are also precisely discussed.
Collapse
|
13
|
The Chemotherapeutic Efficacy of Hyaluronic Acid Coated Polymeric Nanoparticles against Breast Cancer Metastasis in Female NCr-Nu/Nu Nude Mice. Polymers (Basel) 2023; 15:polym15020284. [PMID: 36679166 PMCID: PMC9863707 DOI: 10.3390/polym15020284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023] Open
Abstract
Polyethylene glycol (PEG) coated Poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) for cancer treatment are biocompatible, nonimmunogenic and accumulate in tumour sites due to the enhanced permeability and retention (EPR). Doxorubicin (DOX) is a potent but cardiotoxic anticancer agent. Hyaluronic acid (HA) occurs naturally in the extra-cellar matrix and binds to CD44 receptors which are overexpressed in cancer metastasis, proven to be characteristic of cancer stem cells and responsible for multidrug resistance. In this study, an athymic mice model of breast cancer metastasis was developed using red fluorescent protein (RFP)-labelled triple negative cancer cells. The animals were divided into four treatment groups (Control, HA-PEG-PLGA nanoparticles, PEG-PLGA nanoparticles, and Free DOX). The tumour size growth was assessed until day 25 when animals were sacrificed. Mice treated with HA-PEG-PLGA NPs inhibited tumour growth. The tumour growth at day 25 (118% ± 13.0) was significantly (p < 0.05) less than PEG-PLGA NPs (376% ± 590 and control (826% ± 970). Fluorescent microscopy revealed that HA-PEG-PLGA NPs had significantly (p < 0.05) less metastasis in liver, spleen, colon, and lungs as compared to control and to Free DOX groups. The efficacy of HA-PEG-PLGA NPs was proven in vivo. Further pharmacokinetic and toxicity studies are required for this formulation to be ready for clinical research.
Collapse
|
14
|
Mansoori-Kermani A, Khalighi S, Akbarzadeh I, Niavol FR, Motasadizadeh H, Mahdieh A, Jahed V, Abdinezhad M, Rahbariasr N, Hosseini M, Ahmadkhani N, Panahi B, Fatahi Y, Mozafari M, Kumar AP, Mostafavi E. Engineered hyaluronic acid-decorated niosomal nanoparticles for controlled and targeted delivery of epirubicin to treat breast cancer. Mater Today Bio 2022; 16:100349. [PMID: 35875198 PMCID: PMC9304880 DOI: 10.1016/j.mtbio.2022.100349] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/17/2022] [Accepted: 07/01/2022] [Indexed: 12/24/2022] Open
Abstract
Targeted drug delivery systems using nanocarriers offer a versatile platform for breast cancer treatment; however, a robust, CD44-targeted niosomal formulation has not been developed and deeply studied (both in vitro and in vivo) yet. Here, an optimized system of epirubicin (Epi)-loaded niosomal nanoparticles (Nio) coated with hyaluronic acid (HA) has been engineered for targeting breast cancer cells. The nanoformulation was first optimized (based on size, polydispersity index, and entrapment efficiency); then, we characterized the morphology, stability, and release behavior of the nanoparticles. Epirubicin release from the HA-coated system (Epi-Nio-HA) showed a 21% (acidic buffer) and 20% (neutral buffer) reduction in comparison with the non-coated group (Epi-Nio). The cytotoxicity and apoptosis results of 4T1 and SkBr3 cells showed an approximately 2-fold increase in the Epi-Nio-HA system over Epi-Nio and free epirubicin, which confirms the superiority of the engineered nanocarriers. Moreover, real-time PCR data demonstrated the down-regulation of the MMP-2, MMP-9, cyclin D, and cyclin E genes expression while caspase-3 and caspase-9 gene expression were up-regulated. Confocal microscopy and flow cytometry studies uncovered the cellular uptake mechanism of the Epi-Nio-HA system, which was CD44-mediated. Furthermore, in vivo studies indicated Epi-Nio-HA decreased mice breast tumor volume by 28% (compared to epirubicin) without side effects on the liver and kidney. Conclusively, our results indicated that the HA-functionalized niosomes provide a promising nanoplatform for efficient and targeted delivery of epirubicin to potentially treat breast cancer.
Collapse
Affiliation(s)
| | - Sadaf Khalighi
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Iman Akbarzadeh
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Fazeleh Ranjbar Niavol
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamidreza Motasadizadeh
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Athar Mahdieh
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Jahed
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St. 3/3, Riga, LV, 1007, Latvia
| | - Masoud Abdinezhad
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Nikoo Rahbariasr
- Polymer Research Laboratory, Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Mahshid Hosseini
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Nima Ahmadkhani
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Behnam Panahi
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
15
|
Ahmadi M, Mahmoodi M, Shoaran M, Nazari-Khanamiri F, Rezaie J. Harnessing Normal and Engineered Mesenchymal Stem Cells Derived Exosomes for Cancer Therapy: Opportunity and Challenges. Int J Mol Sci 2022; 23:ijms232213974. [PMID: 36430452 PMCID: PMC9699149 DOI: 10.3390/ijms232213974] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
There remains a vital necessity for new therapeutic approaches to combat metastatic cancers, which cause globally over 8 million deaths per year. Mesenchymal stem cells (MSCs) display aptitude as new therapeutic choices for cancer treatment. Exosomes, the most important mediator of MSCs, regulate tumor progression. The potential of harnessing exosomes from MSCs (MSCs-Exo) in cancer therapy is now being documented. MSCs-Exo can promote tumor progression by affecting tumor growth, metastasis, immunity, angiogenesis, and drug resistance. However, contradictory evidence has suggested that MSCs-Exo suppress tumors through several mechanisms. Therefore, the exact association between MSCs-Exo and tumors remains controversial. Accordingly, the applications of MSCs-Exo as novel drug delivery systems and standalone therapeutics are being extensively explored. In addition, engineering MSCs-Exo for targeting tumor cells has opened a new avenue for improving the efficiency of antitumor therapy. However, effective implementation in the clinical trials will need the establishment of standards for MSCs-Exo isolation and characterization as well as loading and engineering methods. The studies outlined in this review highlight the pivotal roles of MSCs-Exo in tumor progression and the promising potential of MSCs-Exo as therapeutic drug delivery vehicles for cancer treatment.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5665665811, Iran
| | - Monireh Mahmoodi
- Department of Biology, Faculty of Science, Arak University, Arak 3815688349, Iran
| | - Maryam Shoaran
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz 5665665811, Iran
| | - Fereshteh Nazari-Khanamiri
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia 5714783734, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia 5714783734, Iran
- Correspondence: ; Tel.: +98-9148548503; Fax: +98-4432222010
| |
Collapse
|
16
|
Waheed S, Li Z, Zhang F, Chiarini A, Armato U, Wu J. Engineering nano-drug biointerface to overcome biological barriers toward precision drug delivery. J Nanobiotechnology 2022; 20:395. [PMID: 36045386 PMCID: PMC9428887 DOI: 10.1186/s12951-022-01605-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
The rapid advancement of nanomedicine and nanoparticle (NP) materials presents novel solutions potentially capable of revolutionizing health care by improving efficacy, bioavailability, drug targeting, and safety. NPs are intriguing when considering medical applications because of their essential and unique qualities, including a significantly higher surface to mass ratio, quantum properties, and the potential to adsorb and transport drugs and other compounds. However, NPs must overcome or navigate several biological barriers of the human body to successfully deliver drugs at precise locations. Engineering the drug carrier biointerface can help overcome the main biological barriers and optimize the drug delivery in a more personalized manner. This review discusses the significant heterogeneous biological delivery barriers and how biointerface engineering can promote drug carriers to prevail over hurdles and navigate in a more personalized manner, thus ushering in the era of Precision Medicine. We also summarize the nanomedicines' current advantages and disadvantages in drug administration, from natural/synthetic sources to clinical applications. Additionally, we explore the innovative NP designs used in both non-personalized and customized applications as well as how they can attain a precise therapeutic strategy.
Collapse
Affiliation(s)
- Saquib Waheed
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Zhibin Li
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Fangyingnan Zhang
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Anna Chiarini
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, 37134, Verona, Venetia, Italy
| | - Ubaldo Armato
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, 37134, Verona, Venetia, Italy
| | - Jun Wu
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
- Human Histology & Embryology Section, Department of Surgery, Dentistry, Paediatrics & Gynaecology, University of Verona Medical School, 37134, Verona, Venetia, Italy.
| |
Collapse
|
17
|
Khurana B, Ouk TS, Lucas R, Senge MO, Sol V. Photosensitizer-hyaluronic acid complexes for antimicrobial photodynamic therapy (aPDT). J PORPHYR PHTHALOCYA 2022. [DOI: 10.1142/s1088424622500286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
18
|
Rachmale M, Rajput N, Jadav T, Sahu AK, Tekade RK, Sengupta P. Implication of metabolomics and transporter modulation based strategies to minimize multidrug resistance and enhance site-specific bioavailability: a needful consideration toward modern anticancer drug discovery. Drug Metab Rev 2022; 54:101-119. [PMID: 35254954 DOI: 10.1080/03602532.2022.2048007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Induction of drug-metabolizing enzymes and efflux transporters (DMET) through activation of pregnane x receptor (PXR) is the primary factor involved in almost all bioavailability and drug resistance-related problems of anticancer drugs. PXR is a transcriptional regulator of many metabolizing enzymes and efflux transporters proteins like p-glycoprotein (p-gp), multidrug resistant protein 1 and 2 (MRP 1 and 2), and breast cancer resistant protein (BCRP), etc. Several anticancer drugs are potent activators of PXR receptors and can modulate the gene expression of DMET proteins. Involvement of anticancer drugs in transcriptional regulation of DMET can prompt increased metabolism and efflux of their own or other co-administered drugs, which leads to poor site-specific bioavailability and increased drug resistance. In this review, we have discussed several novel strategies to evade drug-induced PXR activation and p-gp efflux including assessment of PXR ligand and p-gp substrate at early stages of drug discovery. Additionally, we have critically discussed the chemical structure and drug delivery-based approaches to avoid PXR binding and inhibit the p-gp activity of the drugs at their target sites.
Collapse
Affiliation(s)
- Megha Rachmale
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Niraj Rajput
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Tarang Jadav
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Amit Kumar Sahu
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| |
Collapse
|
19
|
Krishnan V, Dharamdasani V, Bakre S, Dhole V, Wu D, Budnik B, Mitragotri S. Hyaluronic Acid Nanoparticles for Immunogenic Chemotherapy of Leukemia and T-Cell Lymphoma. Pharmaceutics 2022; 14:466. [PMID: 35214193 PMCID: PMC8874923 DOI: 10.3390/pharmaceutics14020466] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Ratiometric delivery of combination chemotherapy can achieve therapeutic efficacy based on synergistic interactions between drugs. It is critical to design such combinations with drugs that complement each other and reduce cancer growth through multiple mechanisms. Using hyaluronic acid (HA) as a carrier, two chemotherapeutic agents-doxorubicin (DOX) and camptothecin (CPT)-were incorporated and tested for their synergistic potency against a broad panel of blood-cancer cell lines. The pair also demonstrated the ability to achieve immunogenic cell death by increasing the surface exposure levels of Calreticulin, thereby highlighting its ability to induce apoptosis via an alternate pathway. Global proteomic profiling of cancer cells treated with HA-DOX-CPT identified pathways that could potentially predict patient sensitivity to HA-DOX-CPT. This lays the foundation for further exploration of integrating drug delivery and proteomics in personalized immunogenic chemotherapy.
Collapse
Affiliation(s)
- Vinu Krishnan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Vimisha Dharamdasani
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Shirin Bakre
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
| | - Ved Dhole
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
| | - Debra Wu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Bogdan Budnik
- Mass Spectrometry Proteomics and Research Laboratory, FAS Division of Science, Harvard University, Cambridge, MA 02138, USA;
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; (V.K.); (V.D.); (S.B.); (V.D.); (D.W.)
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
20
|
Design of Bio-Responsive Hyaluronic Acid-Doxorubicin Conjugates for the Local Treatment of Glioblastoma. Pharmaceutics 2022; 14:pharmaceutics14010124. [PMID: 35057020 PMCID: PMC8781529 DOI: 10.3390/pharmaceutics14010124] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 01/23/2023] Open
Abstract
Glioblastoma is an unmet clinical need. Local treatment strategies offer advantages, such as the possibility to bypass the blood–brain barrier, achieving high drug concentrations at the glioblastoma site, and consequently reducing systemic toxicity. In this study, we evaluated the feasibility of using hyaluronic acid (HA) for the local treatment of glioblastoma. HA was conjugated to doxorubicin (DOX) with distinct bio-responsive linkers (direct amide conjugation HA-NH-DOX), direct hydrazone conjugation (HA-Hz-DOX), and adipic hydrazone (HA-AdpHz-DOX). All HA-DOX conjugates displayed a small size (less than 30 nm), suitable for brain diffusion. HA-Hz-DOX showed the best performance in killing GBM cells in both 2D and 3D in vitro models and displayed superior activity in a subcutaneous GL261 tumor model in vivo compared to free DOX and other HA-DOX conjugates. Altogether, these results demonstrate the feasibility of HA as a polymeric platform for the local treatment of glioblastoma and the importance of rationally designing conjugates.
Collapse
|
21
|
Imiquimod-gemcitabine nanoparticles harness immune cells to suppress breast cancer. Biomaterials 2021; 280:121302. [PMID: 34894584 DOI: 10.1016/j.biomaterials.2021.121302] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 11/21/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
Monotherapy with a single chemotherapeutic regimen has met with significant hurdles in terms of clinical efficacy. The complexity of cancer accentuates the need for an alternative approach with a combination of two or more therapeutic regimens to win the battle. However, it is still a challenge to develop a successful combination of drugs with high efficiency and low toxicity to control cancer growth. While gemcitabine monotherapy remains a choice of standard treatment for advanced breast cancer, the approach has not prolonged the median survival time of metastatic breast cancer patients. Here, we report a hyaluronic acid (HA)-based drug combination of gemcitabine (GEM) with imiquimod (IMQ) to stimulate immune cells for anticancer activity. Treatment of the drug combination (IMQ-HA-GEM) showed enhanced anticancer activity against 4T1 breast tumor cells in vitro. Our study with a microfluidics-based 3D, compartmentalized cancer model showed that infiltration of THP-1 monocytes occurred particularly at the site of cancer cells treated with IMQ-HA-GEM. Moreover, IMQ-HA-GEM significantly suppressed the volume of 4T1 breast tumor of mice in vivo. Flow cytometry study displayed a significantly higher activation of CD11b+ immune cells in the blood of mice treated with IMQ-HA-GEM, whereas immunohistochemistry study revealed greater prevalence of CD68+ tumor-associated macrophages in the tumor. Histological examination of isolated tumors of mice treated with IMQ-HA-GEM further confirmed the efficacy of drug combination on cancer cells. This study supports the conclusion that imiquimod potentiates the effect of gemcitabine by activating immune cells to suppress tumors in the form of combination nanoparticles.
Collapse
|
22
|
Zhang F, Guo J, Zhang Z, Qian Y, Wang G, Duan M, Zhao H, Yang Z, Jiang X. Mesenchymal stem cell-derived exosome: A tumor regulator and carrier for targeted tumor therapy. Cancer Lett 2021; 526:29-40. [PMID: 34800567 DOI: 10.1016/j.canlet.2021.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/02/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that have the ability to differentiate into multiple cell types. Several studies have shown that exosomes secreted by MSCs (MSCs-Exo) play an important role in tumor growth, angiogenesis, invasion, and drug resistance. However, contradictory results have suggested that MSCs-Exo can also suppress tumors through specific mechanisms, such as regulating immune responses and intercellular signaling. Consequently, the relationship between MSCs-Exo and tumors remains controversial. However, it is undeniable that exosomes, as natural vesicles, can be excellent drug carriers and show promise for application in targeted tumor therapy. Here, we review the current knowledge regarding the involvement of MSCs-Exo in tumor progression and their potential as drug delivery systems in targeted therapy. We argue that MSCs-Exo can be used as safe carriers of antitumor drugs.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jinshuai Guo
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhenghou Zhang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yiping Qian
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Guang Wang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Meiqi Duan
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Haiying Zhao
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhi Yang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Xiaofeng Jiang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
23
|
Combination of epigenetic regulation with gene therapy-mediated immune checkpoint blockade induces anti-tumour effects and immune response in vivo. Nat Commun 2021; 12:6742. [PMID: 34795289 PMCID: PMC8602287 DOI: 10.1038/s41467-021-27078-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy has become a powerful cancer treatment, but only a small fraction of patients have achieved durable benefits due to the immune escape mechanism. In this study, epigenetic regulation is combined with gene therapy-mediated immune checkpoint blockade to relieve this immune escape mechanism. PPD (i.e., mPEG-b-PLG/PEI-RT3/DNA) is developed to mediate plasmid-encoding shPD-L1 delivery by introducing multiple interactions (i.e., electrostatic, hydrogen bonding, and hydrophobic interactions) and polyproline II (PPII)-helix conformation, which downregulates PD-L1 expression on tumour cells to relieve the immunosuppression of T cells. Zebularine (abbreviated as Zeb), a DNA methyltransferase inhibitor (DNMTi), is used for the epigenetic regulation of the tumour immune microenvironment, thus inducing DC maturation and MHC I molecule expression to enhance antigen presentation. PPD plus Zeb combination therapy initiates a systemic anti-tumour immune response and effectively prevents tumour relapse and metastasis by generating durable immune memory. This strategy provides a scheme for tumour treatment and the inhibition of relapse and metastasis.
Collapse
|
24
|
Ashrafizadeh M, Mirzaei S, Gholami MH, Hashemi F, Zabolian A, Raei M, Hushmandi K, Zarrabi A, Voelcker NH, Aref AR, Hamblin MR, Varma RS, Samarghandian S, Arostegi IJ, Alzola M, Kumar AP, Thakur VK, Nabavi N, Makvandi P, Tay FR, Orive G. Hyaluronic acid-based nanoplatforms for Doxorubicin: A review of stimuli-responsive carriers, co-delivery and resistance suppression. Carbohydr Polym 2021; 272:118491. [PMID: 34420747 DOI: 10.1016/j.carbpol.2021.118491] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022]
Abstract
An important motivation for the use of nanomaterials and nanoarchitectures in cancer therapy emanates from the widespread emergence of drug resistance. Although doxorubicin (DOX) induces cell cycle arrest and DNA damage by suppressing topoisomerase activity, resistance to DOX has severely restricted its anti-cancer potential. Hyaluronic acid (HA) has been extensively utilized for synthesizing nanoparticles as it interacts with CD44 expressed on the surface of cancer cells. Cancer cells can take up HA-modified nanoparticles through receptor-mediated endocytosis. Various types of nanostructures such as carbon nanomaterials, lipid nanoparticles and polymeric nanocarriers have been modified with HA to enhance the delivery of DOX to cancer cells. Hyaluronic acid-based advanced materials provide a platform for the co-delivery of genes and drugs along with DOX to enhance the efficacy of anti-cancer therapy and overcome chemoresistance. In the present review, the potential methods and application of HA-modified nanostructures for DOX delivery in anti-cancer therapy are discussed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria, 3168, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiobiology Research Center, Iran University of Medical Science, Tehran, Iran
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Palacky University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - I J Arostegi
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - M Alzola
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK; Department of Mechanical Engineering, School of Engineering, Shiv Nadar University, Uttar Pradesh 201314, India
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Center for Materials Interfaces, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy.
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, USA.
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
25
|
Della Sala F, Fabozzi A, di Gennaro M, Nuzzo S, Makvandi P, Solimando N, Pagliuca M, Borzacchiello A. Advances in Hyaluronic-Acid-Based (Nano)Devices for Cancer Therapy. Macromol Biosci 2021; 22:e2100304. [PMID: 34657388 DOI: 10.1002/mabi.202100304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the main cause of fatality all over the world with a considerable growth rate. Many biologically active nanoplatforms are exploited for tumor treatment. Of nanodevices, hyaluronic acid (HA)-based systems have shown to be promising candidates for cancer therapy due to their high biocompatibility and cell internalization. Herein, surface functionalization of different nanoparticles (NPs), e.g., organic- and inorganic-based NPs, is highlighted. Subsequently, HA-based nanostructures and their applications in cancer therapy are presented.
Collapse
Affiliation(s)
- Francesca Della Sala
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Antonio Fabozzi
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Mario di Gennaro
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Stefano Nuzzo
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Pooyan Makvandi
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Nicola Solimando
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Maurizio Pagliuca
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| |
Collapse
|
26
|
Triple negative breast cancer and non-small cell lung cancer: Clinical challenges and nano-formulation approaches. J Control Release 2021; 337:27-58. [PMID: 34273417 DOI: 10.1016/j.jconrel.2021.07.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Abstract
Triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC) are amongst the most aggressive forms of solid tumors. TNBC is highlighted by absence of genetic components of progesterone receptor, HER2/neu and estrogen receptor in breast cancer. NSCLC is characterized by integration of malignant carcinoma into respiratory system. Both cancers are associated with poor median and overall survival rates with low progression free survival with high incidences of relapse. These cancers are characterized by tumor heterogeneity, genetic mutations, generation of cancer-stem cells, immune-resistance and chemoresistance. Further, these neoplasms have been reported for tumor cross-talk into second primary cancers for each other. Current chemotherapeutic regimens include usage of multiple agents in tandem to affect tumor cells through multiple mechanisms with various such combinations being clinically tested. However, lack of controlled delivery and effective temporospatial presence of chemotherapeutics has resulted in suboptimal therapeutic response. Consequently, passive targeted albumin bound paclitaxel and PEGylated liposomal doxorubicin have been clinically used and tested with newer drugs for improved therapeutic efficacy in these cancers. Active targeting of nanocarriers against surface overexpressed proteins in both neoplasms have been explored. However, use of single agent nanoparticulate formulations against both cancers have failed to elicit desired outcomes. This review aims to identify clinical unmet need in these cancers while establishing a correlation with tested nano-formulation approaches and issues with preclinical to clinical translation. Lipid and polymer-based drug-drug and drug-gene combinatorial nanocarriers delivering multiple chemotherapeutics simultaneously to desired site of action have been detailed. Finally, emerging opportunities such as pharmacological targets (immune check point and epigentic modulators) as well as gene-based modulation (siRNA/CRISPR/Cas9) and the nano-formulation challenges for effective treatment of both cancers have been explored.
Collapse
|
27
|
Gao M, Deng H, Zhang W. Hyaluronan-based Multifunctional Nano-carriers for Combination Cancer Therapy. Curr Top Med Chem 2021; 21:126-139. [PMID: 32962617 DOI: 10.2174/1568026620666200922113846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/30/2022]
Abstract
Hyaluronan (HA) is a natural linear polysaccharide that has excellent hydrophilicity, biocompatibility, biodegradability, and low immunogenicity, making it one of the most attractive biopolymers used for biomedical researches and applications. Due to the multiple functional sites on HA and its intrinsic affinity for CD44, a receptor highly expressed on various cancer cells, HA has been widely engineered to construct different drug-loading nanoparticles (NPs) for CD44-targeted anti-tumor therapy. When a cocktail of drugs is co-loaded in HA NP, a multifunctional nano-carriers could be obtained, which features as a highly effective and self-targeting strategy to combat cancers with CD44 overexpression. The HA-based multidrug nano-carriers can be a combination of different drugs, various therapeutic modalities, or the integration of therapy and diagnostics (theranostics). Up to now, there are many types of HA-based multidrug nano-carriers constructed by different formulation strategies, including drug co-conjugates, micelles, nano-gels and hybrid NP of HA and so on. This multidrug nano-carrier takes the full advantages of HA as an NP matrix, drug carriers and targeting ligand, representing a simplified and biocompatible platform to realize the targeted and synergistic combination therapy against the cancers. In this review, recent progress of HA-based multidrug nano-carriers for combination cancer therapy is summarized and the potential challenges for translational applications have been discussed.
Collapse
Affiliation(s)
- Menghan Gao
- State Key Laboratory of Medical Molecular Biology & Department of Biomedical Engineering, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Hong Deng
- State Key Laboratory of Medical Molecular Biology & Department of Biomedical Engineering, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Weiqi Zhang
- State Key Laboratory of Medical Molecular Biology & Department of Biomedical Engineering, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
28
|
Krishnan V, Peng K, Sarode A, Prakash S, Zhao Z, Filippov SK, Todorova K, Sell BR, Lujano O, Bakre S, Pusuluri A, Vogus D, Tsai KY, Mandinova A, Mitragotri S. Hyaluronic acid conjugates for topical treatment of skin cancer lesions. SCIENCE ADVANCES 2021; 7:7/24/eabe6627. [PMID: 34117055 PMCID: PMC8195472 DOI: 10.1126/sciadv.abe6627] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 04/23/2021] [Indexed: 05/31/2023]
Abstract
Skin cancer is one of the most common types of cancer in the United States and worldwide. Topical products are effective for treating cancerous skin lesions when surgery is not feasible. However, current topical products induce severe irritation, light-sensitivity, burning, scaling, and inflammation. Using hyaluronic acid (HA), we engineered clinically translatable polymer-drug conjugates of doxorubicin and camptothecin termed, DOxorubicin and Camptothecin Tailored at Optimal Ratios (DOCTOR) for topical treatment of skin cancers. When compared to the clinical standard, Efudex, DOCTOR exhibited high cancer-cell killing specificity with superior safety to healthy skin cells. In vivo studies confirmed its efficacy in treating cancerous lesions without irritation or systemic absorption. When tested on patient-derived primary cells and live-skin explants, DOCTOR killed the cancer with a selectivity as high as 21-fold over healthy skin tissue from the same donor. Collectively, DOCTOR provides a safe and potent option for treating skin cancer in the clinic.
Collapse
Affiliation(s)
- Vinu Krishnan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Kevin Peng
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Apoorva Sarode
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Supriya Prakash
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Zongmin Zhao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Sergey K Filippov
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Kristina Todorova
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Brittney R Sell
- Departments of Anatomic Pathology and Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Omar Lujano
- Department of Molecular, Cellular, and Developmental Biology (MCDB), University of California, Santa Barbara, Santa Barbara, CA 93117, USA
| | - Shirin Bakre
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Anusha Pusuluri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Douglas Vogus
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Kenneth Y Tsai
- Departments of Anatomic Pathology and Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Anna Mandinova
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
- Broad Institute of Harvard and MIT, 7 Cambridge Center, MA 02142, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| |
Collapse
|
29
|
Sallam MA, Wyatt Shields Iv C, Prakash S, Kim J, Pan DC, Mitragotri S. A dual macrophage polarizer conjugate for synergistic melanoma therapy. J Control Release 2021; 335:333-344. [PMID: 34048840 DOI: 10.1016/j.jconrel.2021.05.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/08/2021] [Accepted: 05/23/2021] [Indexed: 12/16/2022]
Abstract
Tumor associated macrophages (TAMs) play a paradoxical role in the fate of aggressive tumors like melanoma. Immune modulation of TAMs from the tumor-permissive M2 phenotype to antitumoral M1 phenotype is an emerging attractive approach in melanoma therapy. Resiquimod is a TLR7/8 agonist that shifts the polarization of macrophages towards M1 phenotype. Bexarotene (BEX) is a retinoid that induce the expression of phagocytic receptors in macrophages besides its ability to downregulate the M2 polarization. However, the clinical use of both agents is hindered by poor pharmacokinetic properties. Here, for the first time we repurposed BEX based on its immunomodulatory properties and combined it with RES by designing hyaluronic acid (HA) conjugates of both drugs that act synergistically as a dual macrophage polarizer to promote the M1 phenotype and suppress the M2 phenotype. This combination enhanced the macrophage secretion of proinflammatory cytokines (IL-6 and TNF-α), while suppressing the production of tumor promoting cytokine CCL22. It enhanced the macrophage phagocytic ability and showed superior inhibitory effects against B16F10 cells. In vivo studies on a mouse melanoma model confirmed the superiority of the dual conjugate compared to the single HA-drug conjugates in suppressing the tumor growth. Immunoprofiling of the excised tumors revealed a significant increase in the M1/M2 ratio of TAMs in mice treated with the dual conjugate. Our intravenously injectable HA conjugate of RES and BEX provides a promising immunotherapeutic combination strategy for resetting the M1/M2 ratio, supporting the tumoricidal activity of TAMs for effective melanoma treatment.
Collapse
Affiliation(s)
- Marwa A Sallam
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Industrial pharmacy, Faculty of pharmacy, Alexandria University, 21521, Egypt
| | - C Wyatt Shields Iv
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Supriya Prakash
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jayoung Kim
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Daniel C Pan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
30
|
Pak PJ, Lee DG, Sung JH, Jung SH, Han TY, Park SH, Chung N. Synergistic effect of the herbal mixture C5E on gemcitabine treatment in PANC‑1 cells. Mol Med Rep 2021; 23:315. [PMID: 33760105 PMCID: PMC7974510 DOI: 10.3892/mmr.2021.11954] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
The present study aimed to determine the anticancer effect of the herbal mixture extract C5E in the pancreatic cancer cell line, PANC-1, in the absence or presence of gemcitabine treatment, a chemotherapeutic drug used for the treatment of pancreatic cancer. The anticancer effects of C5E, gemcitabine and C5E plus gemcitabine in PANC-1 cells following 72 h of treatment were investigated. The effect of each treatment on cell cycle arrest, apoptosis and the proportion of side population (SP) cells was determined using flow cytometric analysis following propidium iodide (PI), Annexin V-FITC/PI double staining and Hoechst 33342 staining, respectively. SP cells share similar characteristics to cancer stem-like cells, and a reduction in the SP is considered to be indicative of an anticancer effect. The percentage of SP cells and the cell viability of general PANC-1 cells were significantly decreased in response to all treatments. The percentage of SP cells was reduced from 8.2% (control) to 3.9, 7.2 and 5.1% following the treatment with C5E, gemcitabine and the co-treatment, respectively. All three treatments were discovered to inhibit cell viability by arresting the cell cycle at the S phase and promoted cell death by inducing early apoptosis, with the levels of apoptosis being increased from 1.9% (control) to 7.3, 2.5 and 12.0% following the treatment with C5E, gemcitabine and the co-treatment, respectively. The mRNA expression levels of sonic hedgehog, which is implicated in the development of certain types of cancer, were downregulated to a greater extent following the co-treatment with C5E and gemcitabine compared with the treatment with either C5E or gemcitabine alone. As the co-treatment with gemcitabine and C5E was more effective than each individual treatment, the present study suggested that the combined treatment may exhibit synergistic effects in PANC-1 cells.
Collapse
Affiliation(s)
- Pyo June Pak
- Department of Biosystems and Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Dong Gun Lee
- Department of Biosystems and Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Ji Hyun Sung
- Flow Cytometry Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Republic of Korea
| | - Seung Hyun Jung
- School of Oriental Medicine, Dongguk University, Ilsan 10326, Republic of Korea
| | - Tae-Young Han
- BanryongInsu Herb Clinic, Seoul 06099, Republic of Korea
| | - Sung Hyo Park
- Department of Biosystems and Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Namhyun Chung
- Department of Biosystems and Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
31
|
Hao W, Zheng Z, Zhu L, Pang L, Ma J, Zhu S, Du L, Jin Y. 3D printing-based drug-loaded implanted prosthesis to prevent breast cancer recurrence post-conserving surgery. Asian J Pharm Sci 2021; 16:86-96. [PMID: 33613732 PMCID: PMC7878459 DOI: 10.1016/j.ajps.2020.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/25/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Systemic chemotherapy of breast cancer is commonly delivered as a large dose and has toxic side effects. Local chemotherapy would overcome the shortcomings of systemic reconstruction and could play an important role in breast cancer surgery according to personalized demand. The application of three-dimensional (3D) printing technology makes personalized customization possible. We designed and prepared a prosthesis containing paclitaxel (PTX) and doxorubicin (DOX) microspheres (PPDM) based on 3D printing to prevent tumor recurrence and metastasis after breast conserving surgery. Polydimethysiloxane has good biocompatibility and was used as a drug carrier in this study. The average particle size of the PTX and DOX microspheres were approximately 3.1 µm and 2.2 µm, respectively. The drug loading of PTX and DOX microspheres was 4.2% and 2.1%, respectively. In vitro drug release studies demonstrated that the 3D-printed prosthesis loaded with PTX and DOX microspheres could release the drugs continuously for more than 3 weeks and thereby suppress cancer recurrence with reduced side effects. The PTX and DOX microspheres not only exerted a synergistic effect, but also achieved a good sustained release effect. In vivo evaluation showed that the PPDM could effectively inhibit breast cancer recurrence and metastasis in mice with breast cancer. PPDM are expected to achieve postoperative chemotherapy for breast cancer and be highly efficient to prevent local breast cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Wenyan Hao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Weifang Medical University, Weifang 261000, China
| | | | - Lin Zhu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lulu Pang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jinqiu Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Siqing Zhu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Anhui Medical University, Hefei 230032, China
| | - Lina Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Weifang Medical University, Weifang 261000, China
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Anhui Medical University, Hefei 230032, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Anhui Medical University, Hefei 230032, China
| |
Collapse
|
32
|
Paroha S, Verma J, Dubey RD, Dewangan RP, Molugulu N, Bapat RA, Sahoo PK, Kesharwani P. Recent advances and prospects in gemcitabine drug delivery systems. Int J Pharm 2021; 592:120043. [DOI: 10.1016/j.ijpharm.2020.120043] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/17/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
|
33
|
Alven S, Aderibigbe BA. The Therapeutic Efficacy of Dendrimer and Micelle Formulations for Breast Cancer Treatment. Pharmaceutics 2020; 12:E1212. [PMID: 33333778 PMCID: PMC7765183 DOI: 10.3390/pharmaceutics12121212] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is among the most common types of cancer in women and it is the cause of a high rate of mortality globally. The use of anticancer drugs is the standard treatment approach used for this type of cancer. However, most of these drugs are limited by multi-drug resistance, drug toxicity, poor drug bioavailability, low water solubility, poor pharmacokinetics, etc. To overcome multi-drug resistance, combinations of two or more anticancer drugs are used. However, the combination of two or more anticancer drugs produce toxic side effects. Micelles and dendrimers are promising drug delivery systems that can overcome the limitations associated with the currently used anticancer drugs. They have the capability to overcome drug resistance, reduce drug toxicity, improve the drug solubility and bioavailability. Different classes of anticancer drugs have been loaded into micelles and dendrimers, resulting in targeted drug delivery, sustained drug release mechanism, increased cellular uptake, reduced toxic side effects of the loaded drugs with enhanced anticancer activity in vitro and in vivo. This review article reports the biological outcomes of dendrimers and micelles loaded with different known anticancer agents on breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| |
Collapse
|
34
|
Sallam MA, Prakash S, Krishnan V, Todorova K, Mandinova A, Mitragotri S. Hyaluronic Acid Conjugates of Vorinostat and Bexarotene for Treatment of Cutaneous Malignancies. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Marwa A. Sallam
- John A. Paulson School of Engineering and Applied Sciences, and Wyss Institute of Biologically Inspired Engineering Harvard University Cambridge MA 02138 USA
- Faculty of pharmacy Alexandria University Egypt
| | - Supriya Prakash
- John A. Paulson School of Engineering and Applied Sciences, and Wyss Institute of Biologically Inspired Engineering Harvard University Cambridge MA 02138 USA
| | - Vinu Krishnan
- John A. Paulson School of Engineering and Applied Sciences, and Wyss Institute of Biologically Inspired Engineering Harvard University Cambridge MA 02138 USA
| | - Kristina Todorova
- Cutaneous Biology Research Center Massachusetts General Hospital and Harvard Medical School Building 149 13th Street Charlestown MA 02129 USA
| | - Anna Mandinova
- Cutaneous Biology Research Center Massachusetts General Hospital and Harvard Medical School Building 149 13th Street Charlestown MA 02129 USA
- Broad Institute of Harvard and MIT 7 Cambridge Center Cambridge MA 02142 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, and Wyss Institute of Biologically Inspired Engineering Harvard University Cambridge MA 02138 USA
| |
Collapse
|
35
|
Lactoferrin-dual drug nanoconjugate: Synergistic anti-tumor efficacy of docetaxel and the NF-κB inhibitor celastrol. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111422. [PMID: 33255023 DOI: 10.1016/j.msec.2020.111422] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/27/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Despite the progress in cancer nanotherapeutics, some obstacles still impede the success of nanocarriers and hinder their clinical translation. Low drug loading, premature drug release, off-target toxicity and multi-drug resistance are among the most difficult challenges. Lactoferrin (LF) has demonstrated a great tumor targeting capacity via its high binding affinity to low density lipoprotein (LDL) and transferrin (Tf) receptors overexpressed by various cancer cells. Herein, docetaxel (DTX) and celastrol (CST) could be successfully conjugated to LF backbone for synergistic breast cancer therapy. Most importantly, the conjugate self-assembled forming nanoparticles of 157.8 nm with elevated loading for both drugs (6.94 and 5.98% for DTX and CST, respectively) without risk of nanocarrier instability. Moreover, the nanoconjugate demonstrated enhanced in vivo anti-tumor efficacy in breast cancer-bearing mice, as reflected by a reduction in tumor volume, prolonged survival rate and significant suppression of NF-κB p65, TNF-α, COX-2 and Ki-67 expression levels compared to the group given free combined DTX/CST therapy and to positive control. This study demonstrated the proof-of-principle for dual drug coupling to LF as a versatile nanoplatform that could augment their synergistic anticancer efficacy.
Collapse
|
36
|
Alven S, Aderibigbe BA. Nanoparticles Formulations of Artemisinin and Derivatives as Potential Therapeutics for the Treatment of Cancer, Leishmaniasis and Malaria. Pharmaceutics 2020; 12:E748. [PMID: 32784933 PMCID: PMC7466127 DOI: 10.3390/pharmaceutics12080748] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer, malaria, and leishmaniasis remain the deadly diseases around the world although several strategies of treatment have been developed. However, most of the drugs used to treat the aforementioned diseases suffer from several pharmacological limitations such as poor pharmacokinetics, toxicity, drug resistance, poor bioavailability and water solubility. Artemisinin and its derivatives are antimalarial drugs. However, they also exhibit anticancer and antileishmanial activity. They have been evaluated as potential anticancer and antileishmanial drugs but their use is also limited by their poor water solubility and poor bioavailability. To overcome the aforementioned limitations associated with artemisinin and its derivatives used for the treatment of these diseases, they have been incorporated into nanoparticles. Several researchers incorporated this class of drugs into nanoparticles resulting in enhanced therapeutic outcomes. Their potential efficacy for the treatment of parasitic infections such as malaria and leishmaniasis and chronic diseases such as cancer has been reported. This review article will be focused on the nanoparticles formulations of artemisinin and derivatives for the treatment of cancer, malaria, and leishmaniasis and the biological outcomes (in vitro and in vivo).
Collapse
|
37
|
Schneible JD, Young AT, Daniele MA, Menegatti S. Chitosan Hydrogels for Synergistic Delivery of Chemotherapeutics to Triple Negative Breast Cancer Cells and Spheroids. Pharm Res 2020; 37:142. [PMID: 32661774 PMCID: PMC7983306 DOI: 10.1007/s11095-020-02864-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE This study aimed to develop a hydrogel system for treating aggressive triple negative breast cancer (TNBC) via kinetically-controlled delivery of the synergistic drug pair doxorubicin (DOX) and gemcitabine (GEM). A 2D assay was adopted to evaluate therapeutic efficacy by determining combination index (CI), and a 3D assay using cancer spheroids was implemented to assess the potential for translation in vivo. METHODS The release of DOX and GEM from an acetylated-chitosan (ACS, degree of acetylation χAc = 40 ± 5%) was characterized to identify a combined drug loading that affords release kinetics and dose that are therapeutically synergistic. The selected DOX/GEM-ACS formulation was evaluated in vitro with 2-D and 3-D models of TNBC to determine the combination index (CI) and the tumor volume reduction, respectively. RESULTS Therapeutically desired release dosages and kinetics of GEM and DOX were achieved. When evaluated with a 2-D model of TNBC, the hydrogel afforded a CI of 0.14, indicating a stronger synergism than concurrent administration of DOX and GEM (CI = 0.23). Finally, the therapeutic hydrogel accomplished a notable volume reduction of the cancer spheroids (up to 30%), whereas the corresponding dosages of free drugs only reduced growth rate. CONCLUSIONS The ACS hydrogel delivery system accomplishes drug release kinetics and molar ratio that affords strong therapeutically synergism. These results, in combination with the choice of ACS as affordable and highly abundant source material, provide a strong pre-clinical demonstration of the potential of the proposed system for complementing surgical resection of aggressive solid tumors.
Collapse
Affiliation(s)
- John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA
| | - Ashlyn T Young
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, North Carolina, USA
| | - M A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, North Carolina, USA.
- Department of Electrical and Computer Engineering, North Carolina State University, 890 Oval Drive, Raleigh, North Carolina, USA.
| | - S Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
- Biomanufacturing Training and Education Center, North Carolina State University, 850 Oval Dr, Raleigh, North Carolina, USA.
| |
Collapse
|
38
|
Tumor-targeted and self-assembled mixed micelles as carriers for enhanced anticancer efficacy of gemcitabine. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
39
|
Schneible JD, Shi K, Young AT, Ramesh S, He N, Dowdey CE, Dubnansky JM, Lilova RL, Gao W, Santiso E, Daniele M, Menegatti S. Modified gaphene oxide (GO) particles in peptide hydrogels: a hybrid system enabling scheduled delivery of synergistic combinations of chemotherapeutics. J Mater Chem B 2020; 8:3852-3868. [PMID: 32219269 PMCID: PMC7945679 DOI: 10.1039/d0tb00064g] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The scheduled delivery of synergistic drug combinations is increasingly recognized as highly effective against advanced solid tumors. Of particular interest are composite systems that release a sequence of drugs with defined kinetics and molar ratios to enhance therapeutic effect, while minimizing the dose to patients. In this work, we developed a homogeneous composite comprising modified graphene oxide (GO) nanoparticles embedded in a Max8 peptide hydrogel, which provides controlled kinetics and molar ratios of release of doxorubicin (DOX) and gemcitabine (GEM). First, modified GO nanoparticles (tGO) were designed to afford high DOX loading and sustained release (18.9% over 72 h and 31.4% over 4 weeks). Molecular dynamics simulations were utilized to model the mechanism of DOX loading as a function of surface modification. In parallel, a Max8 hydrogel was developed to release GEM with faster kinetics and achieve a 10-fold molar ratio to DOX. The selected DOX/tGO nanoparticles were suspended in a GEM/Max8 hydrogel matrix, and the resulting composite was tested against a triple negative breast cancer cell line, MDA-MB-231. Notably, the composite formulation afforded a combination index of 0.093 ± 0.001, indicating a much stronger synergism compared to the DOX-GEM combination co-administered in solution (CI = 0.396 ± 0.034).
Collapse
Affiliation(s)
- John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Kaihang Shi
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Ashlyn T Young
- Joint Department of Biomedical Engineering, North Carolina State University - University of North Carolina Chapel Hill, North Carolina, USA
| | - Srivatsan Ramesh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Nanfei He
- Department of Textile Engineering, Chemistry, and Science, 1020 Main Campus Drive, Raleigh, North Carolina, USA
| | - Clay E Dowdey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Jean Marie Dubnansky
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Radina L Lilova
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Wei Gao
- Department of Textile Engineering, Chemistry, and Science, 1020 Main Campus Drive, Raleigh, North Carolina, USA
| | - Erik Santiso
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University - University of North Carolina Chapel Hill, North Carolina, USA and Department of Electrical and Computer Engineering, North Carolina State University, 890 Oval Drive, Raleigh, North Carolina, USA.
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| |
Collapse
|
40
|
Alven S, Nqoro X, Buyana B, Aderibigbe BA. Polymer-Drug Conjugate, a Potential Therapeutic to Combat Breast and Lung Cancer. Pharmaceutics 2020; 12:E406. [PMID: 32365495 PMCID: PMC7284459 DOI: 10.3390/pharmaceutics12050406] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 12/19/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer is a chronic disease that is responsible for the high death rate, globally. The administration of anticancer drugs is one crucial approach that is employed for the treatment of cancer, although its therapeutic status is not presently satisfactory. The anticancer drugs are limited pharmacologically, resulting from the serious side effects, which could be life-threatening. Polymer drug conjugates, nano-based drug delivery systems can be utilized to protect normal body tissues from the adverse side effects of anticancer drugs and also to overcome drug resistance. They transport therapeutic agents to the target cell/tissue. This review article is based on the therapeutic outcomes of polymer-drug conjugates against breast and lung cancer.
Collapse
|
41
|
Russell LM, Liu CH, Grodzinski P. Nanomaterials innovation as an enabler for effective cancer interventions. Biomaterials 2020; 242:119926. [PMID: 32169771 DOI: 10.1016/j.biomaterials.2020.119926] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Nanomedicines have been developing very rapidly and have started to play a significant role in several cancer therapeutic modalities. Early on, the nanomedicine field focused on optimizing pharmacokinetics, toxicity, and/or biodistribution of an agent through nanoparticle formulation. In other cases, where materials science is employed more decisively, nanomedicine can include the creation of new agents that take advantage of nanoscale materials properties to enhance treatment efficacy through unique mode of action, molecular targeting, or controlled drug release. Both current and future nanomedicines will seek to contribute to the therapeutic and diagnostic landscape through creative leveraging of mechanical, electrical, optical, magnetic, and biological nanomaterial properties. In this work, we discuss how by modulating these material properties, one can design more diverse and more effective cancer interventions. We focus on six areas in cancer management, including in vitro diagnostics, clinical imaging, theranostics, combination therapy, immunotherapy, and gene therapy.
Collapse
Affiliation(s)
- Luisa M Russell
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina H Liu
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
42
|
Dallavalle S, Dobričić V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, Tsakovska I, Zidar N, Fruttero R. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat 2020; 50:100682. [PMID: 32087558 DOI: 10.1016/j.drup.2020.100682] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is the dominant cause of the failure of cancer chemotherapy. The design of antitumor drugs that are able to evade MDR is rapidly evolving, showing that this area of biomedical research attracts great interest in the scientific community. The current review explores promising recent approaches that have been developed with the aim of circumventing or overcoming MDR. Encouraging results have been obtained in the investigation of the MDR-modulating properties of various classes of natural compounds and their analogues. Inhibition of P-gp or downregulation of its expression have proven to be the main mechanisms by which MDR can be surmounted. The use of hybrid molecules that are able to simultaneously interact with two or more cancer cell targets is currently being explored as a means to circumvent drug resistance. This strategy is based on the design of hybrid compounds that are obtained either by merging the structural features of separate drugs, or by conjugating two drugs or pharmacophores via cleavable/non-cleavable linkers. The approach is highly promising due to the pharmacokinetic and pharmacodynamic advantages that can be achieved over the independent administration of the two individual components. However, it should be stressed that the task of obtaining successful multivalent drugs is a very challenging one. The conjugation of anticancer agents with nitric oxide (NO) donors has recently been developed, creating a particular class of hybrid that can combat tumor drug resistance. Appropriate NO donors have been shown to reverse drug resistance via nitration of ABC transporters and by interfering with a number of metabolic enzymes and signaling pathways. In fact, hybrid compounds that are produced by covalently attaching NO-donors and antitumor drugs have been shown to elicit a synergistic cytotoxic effect in a variety of drug resistant cancer cell lines. Another strategy to circumvent MDR is based on nanocarrier-mediated transport and the controlled release of chemotherapeutic drugs and P-gp inhibitors. Their pharmacokinetics are governed by the nanoparticle or polymer carrier and make use of the enhanced permeation and retention (EPR) effect, which can increase selective delivery to cancer cells. These systems are usually internalized by cancer cells via endocytosis and accumulate in endosomes and lysosomes, thus preventing rapid efflux. Other modalities to combat MDR are described in this review, including the pharmaco-modulation of acridine, which is a well-known scaffold in the development of bioactive compounds, the use of natural compounds as means to reverse MDR, and the conjugation of anticancer drugs with carriers that target specific tumor-cell components. Finally, the outstanding potential of in silico structure-based methods as a means to evaluate the ability of antitumor drugs to interact with drug transporters is also highlighted in this review. Structure-based design methods, which utilize 3D structural data of proteins and their complexes with ligands, are the most effective of the in silico methods available, as they provide a prediction regarding the interaction between transport proteins and their substrates and inhibitors. The recently resolved X-ray structure of human P-gp can help predict the interaction sites of designed compounds, providing insight into their binding mode and directing possible rational modifications to prevent them from becoming P-gp drug substrates. In summary, although major efforts were invested in the search for new tools to combat drug resistant tumors, they all require further implementation and methodological development. Further investigation and progress in the abovementioned strategies will provide significant advances in the rational combat against cancer MDR.
Collapse
Affiliation(s)
- Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Loretta Lazzarato
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Elena Gazzano
- Department of Oncology, Università degli Studi di Torino, Via Santena 5/bis, 10126 Turin, Italy
| | - Miguel Machuqueiro
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, C8 Building, Campo Grande, 1749-016, Lisbon, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| | - Ilza Pajeva
- QSAR and Molecular Modelling Department, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 105, 1113 Sofia, Bulgaria
| | - Ivanka Tsakovska
- QSAR and Molecular Modelling Department, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 105, 1113 Sofia, Bulgaria
| | - Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Roberta Fruttero
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Turin, Italy.
| |
Collapse
|
43
|
Zhuang B, Chen T, Xiao Z, Jin Y. Drug-loaded implantable surgical cavity-adaptive hydrogels for prevention of local tumor recurrence. Int J Pharm 2020; 577:119048. [PMID: 31978462 DOI: 10.1016/j.ijpharm.2020.119048] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
High local post-surgical cancer recurrence severely impairs the patients' prognosis and survival rates. Here, an injectable in situ forming hydrogel was designed to locally controlled release gemcitabine (GEM) and doxorubicin (DOX) to prevent local cancer recurrence. The hydrogel was rapidly formed at the post-surgical cavity after the aldehyde hyaluronic acid (HA-CHO) and the carboxymethyl chitosan (CM-CS) were mixed and immediately injected. Meanwhile, DOX was conjugated to HA-CHO and GEM was doped in CM-CS to obtain GD-HA/CS-Gel. The drug-free hydrogels showed low cytotoxicity on L929 cells and good in vivo biocompatibility. The hydrogels had appropriate viscoelasticity and rapid self-healing ability, favoring long-term local residence at the injected site where GEM quickly released and DOX slowly released. GEM and DOX showed the synergistic anticancer effect on 4T1 cells. Breast cancer 4T1-cell xenograft models were established and the tumors were surgically resected. GD-HA/CS-Gel was implanted in the post-surgical cavity and cancer recurrence and distant lung metastasis were completely prevented in comparison with the single drug-loaded hydrogel or drug solutions. The locally implanted dual drug-loaded cavity-adaptive hydrogel is a promising medication for prevention of post-surgical tumor recurrence.
Collapse
Affiliation(s)
- Bo Zhuang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Ting Chen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhimei Xiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng 475004, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; Guangdong Pharmaceutical University, Guangzhou 510006, China; Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng 475004, China.
| |
Collapse
|
44
|
Li Y, Zhao Z, Liu H, Fetse JP, Jain A, Lin CY, Cheng K. Development of a Tumor-Responsive Nanopolyplex Targeting Pancreatic Cancer Cells and Stroma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:45390-45403. [PMID: 31769963 PMCID: PMC7372733 DOI: 10.1021/acsami.9b15116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Desmoplasia plays a pivotal role in promoting pancreatic cancer progression and is associated with poor clinical outcome. Targeting the desmoplastic tumor microenvironment in combination with chemotherapy is therefore a promising strategy for pancreatic cancer therapy. Here, we report a novel biodegradable copolymer to codeliver LY2109761 (a TGF-β receptor I/II inhibitor) and CPI-613 (a novel chemotherapy agent) to desmoplastic stroma and tumor cells, respectively, in the tumor microenvironment. Hydrophobic CPI-613 is conjugated to the hydrophilic copolymer via a newly designed MMP-2-responsive linker to form a trigger-responsive nanopolyplex. LY2109761 is hydrophobic and encapsulated into the hydrophobic core of the nanopolyplex. The resulting nanopolyplex is modified with a plectin-1-targeting peptide to enhance the accumulation of the nanopolyplex in pancreatic tumors. The nanopolyplex aims to normalize the stroma by blocking the interaction between tumor cells and pancreatic stellate cells to inhibit the activation of pancreatic stellate cells and subsequently reduce the dense extracellular matrix. Normalized stroma increases the penetration of the nanopolyplex into the tumor. The nanopolyplex shows enhanced accumulation in xenograft pancreatic tumors in a biodistribution study. Moreover, the targeted nanopolyplex markedly inhibits tumor growth in an orthotopic pancreatic cancer mouse model by dual-targeting tumor cells and stroma. Overall, the multifunctional nanopolyplex is a promising platform for pancreatic cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kun Cheng
- Corresponding author: Kun Cheng, Ph.D., University of Missouri Curators’ Distinguished Professor, Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, Phone: (816) 235-2425, Fax: (816) 235-5779,
| |
Collapse
|
45
|
Luo Z, Dai Y, Gao H. Development and application of hyaluronic acid in tumor targeting drug delivery. Acta Pharm Sin B 2019; 9:1099-1112. [PMID: 31867159 PMCID: PMC6900560 DOI: 10.1016/j.apsb.2019.06.004] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/08/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Hyaluronic acid (HA) is a natural polysaccharide that has gained much attention due to its biocompatibility, enzyme degradation capacity and active tumor targeting capacity. Its receptor, CD44, is overexpressed in many kinds of cancers and is associated with tumor progress, infiltration and metastasis. Therefore, many researchers have developed various HA-based drug delivery systems for CD44-mediated tumor targeting. In this review, we systemically overview the basic theory of HA, its receptor and hyaluronidase, then we categorize the studies in HA-based drug delivery systems according to the functions of HA, including tumor-targeting materials, enzyme-sensitive biodegradable modality, pH-sensitive component, reduction-sensitive component, and the gel backbone. Finally, the perspective is discussed.
Collapse
Affiliation(s)
- Zhijian Luo
- Ultrasound Diagnosis Department of the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou 646000, China
| | - Yan Dai
- Department of Pharmacy of the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou 646000, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Liu Y, Li K, Wu Y, Ma J, Tang P, Liu Y, Wu D. PVA reinforced gossypolone and doxorubicin π-π stacking nanoparticles towards tumor targeting and ultralow dose synergistic chemotherapy. Biomater Sci 2019; 7:3662-3674. [PMID: 31179466 DOI: 10.1039/c9bm00674e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To improve the tumor synergistic therapeutic effects of carrier-free dual-drug delivery systems and realize ultralow dose administration, we developed a tumor targeting and high-efficiency synergistic chemotherapy system (HA-Gn@DPGn NPs) based on polyvinyl alcohol (PVA) reinforced gossypolone (Gn) and doxorubicin (DOX) π-π stacking nanoparticles (DPGn NPs), in which PVA filled the gaps between Gn and DOX and bridged Gn and DOX tightly. Hyaluronic acid modifier hyaluronic acid-gossypolone (HA-Gn) was covered on the surface of DPGn NPs to form HA-Gn@DPGn NPs that procured active targeting properties. This system presented a spherical shape with a uniform hydrodynamic size of 87 ± 6.8 nm, a high drug loading of 80.31%, and high stability. FTIR and UV spectra demonstrated that HA-Gn was covered on the surface of the system and showed significant π-π stacking properties. A considerably low combination index of Gn and DOX (0.1862) was determined at an ultra-low dose of DOX under a Gn : DOX ratio of 50 : 1. HA-Gn@DPGn NPs also demonstrated excellent tumor synergistic therapeutic efficacy (TIR > 87%) at an ultralow dose of DOX and Gn. This system demonstrates high tumor comprehensive synergistic therapeutic efficacy at an ultralow drug dose with multiple favorable therapeutic characteristics, including negligible side effects, tumor targeting ability and thermal-responsive drug release, and thus has considerable potential for tumor synergistic therapy.
Collapse
Affiliation(s)
- Yiming Liu
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | | | | | | | | | | | | |
Collapse
|
47
|
Rosch JG, Landry MR, Thomas CR, Sun C. Enhancing chemoradiation of colorectal cancer through targeted delivery of raltitrexed by hyaluronic acid coated nanoparticles. NANOSCALE 2019; 11:13947-13960. [PMID: 31305836 PMCID: PMC7213297 DOI: 10.1039/c9nr04320a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Combined modality therapy incorporating raltitrexed (RTX), a thymidylate synthase inhibitor, and radiation can lead to improved outcome for rectal cancer patients. To increase delivery and treatment efficacy, we formulated a hyaluronic acid (HA) coated nanoparticle encapsulating RTX (HARPs) through layer-by-layer assembly. These particles were determined to have a diameter of ∼115 nm, with a polydispersity index of 0.112 and a zeta potential of -22 mV. Cell uptake in CT26 cells determined through flow cytometry showed a ∼5-fold increase between untargeted and HA-coated particles. Through viability and DNA damage assays, we assessed the potency of the free RTX and HARPs, and found increased DNA damage in cells treated with the RTX-loaded nanoparticles administered concurrently with radiation. In vivo efficacy through tumor growth inhibition was investigated in a syngeneic murine colorectal cancer model. Nanoparticle treatment showed no acute toxicity in vivo, and all treatments showed survival benefits for their respective groups compared to controls. HARPs alone slowed tumor growth, although not significantly. Radiation alone and in combination with the HARPs showed significant growth delay. Notably, the combination treatment significantly hindered tumor progression relative to the HARPs highlighting the benefit of this multipronged treatment. These results provide a foundation for loading RTX in a nanoparticle formulation, and establish a combined radiation and drug dosing schedule to determine optimal tumor growth delay and subsequent treatment efficacy.
Collapse
Affiliation(s)
- Justin G Rosch
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Madeleine R Landry
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Charles R Thomas
- Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA and Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
48
|
Polymeric nanoparticles as carrier for targeted and controlled delivery of anticancer agents. Ther Deliv 2019; 10:527-550. [DOI: 10.4155/tde-2019-0044] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent decades, many novel methods by using nanoparticles (NPs) have been investigated for diagnosis, drug delivery and treatment of cancer. Accordingly, the potential of NPs as carriers is very significant for the delivery of anticancer drugs, because cancer treatment with NPs has led to the improvement of some of the drug delivery limitations such as low blood circulation time and bioavailability, lack of water solubility, drug adverse effect. In addition, the NPs protect drugs against enzymatic degradation and can lead to the targeted and/or controlled release of the drug. The present review focuses on the potential of NPs that can help the targeted and/or controlled delivery of anticancer agents for cancer therapy.
Collapse
|
49
|
Schneible JD, Singhal A, Lilova RL, Hall CK, Grafmüller A, Menegatti S. Tailoring the Chemical Modification of Chitosan Hydrogels to Fine-Tune the Release of a Synergistic Combination of Chemotherapeutics. Biomacromolecules 2019; 20:3126-3141. [PMID: 31310515 DOI: 10.1021/acs.biomac.9b00707] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Combination chemotherapy with a defined ratio and sequence of drug release is a clinically established and effective route to treat advanced solid tumors. In this context, a growing body of literature demonstrates the potential of hydrogels constructed with chemically modified polysaccharides as depots for controlled release of chemotherapeutics. Identifying the appropriate modification in terms of physicochemical properties of the functional group and its degree of substitution (χ) to achieve the desired release profile for multiple drugs is, however, a complex multivariate problem. To address this issue, we have developed a computational toolbox that models the migration of a drug pair through a hydrated network of polysaccharide chains modified with hydrophobic moieties. In this study, we chose doxorubicin (DOX) and Gemcitabine (GEM) as model drugs, as their synergistic effect against breast cancer has been thoroughly investigated, and chitosan as the model polymer. Our model describes how the modification of chitosan chains with acetyl, butanoyl, and heptanoyl moieties at different values χ governs both the structure of the hydrogel network and drug migration through it. Our experimental data confirm the in silico predictions for both single- and dual-drug release and, most notably, the counterintuitive inversion of release vs χ that occurs when switching from a single- to a dual-drug system. Consensus between predicted and experimental data indicates that acetyl modifications (χ = 32-42%) and butanoyl modifications (χ = 19-24%) provide synergistic GEM/DOX release molar ratios (i.e., 5-10). Collectively, these results demonstrate the potential of this model in guiding the design of chemotherapeutic hydrogels to combat cancer.
Collapse
Affiliation(s)
- John D Schneible
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Ankush Singhal
- Department of Theory and Biosystems , Max Planck Institute for Colloids and Interfaces , Potsdam 14476 , Germany
| | - Radina L Lilova
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Carol K Hall
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Andrea Grafmüller
- Department of Theory and Biosystems , Max Planck Institute for Colloids and Interfaces , Potsdam 14476 , Germany
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| |
Collapse
|
50
|
Immunological consequences of chemotherapy: Single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305:130-154. [DOI: 10.1016/j.jconrel.2019.04.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023]
|