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
|
Ahmad Z, Arshad N, Alsaab HO, Selamoğlu Z, Shah A. Exploration of cellular uptake and endocytosis mechanisms for doxorubicin-loaded poly (amino acid) nanocarriers. Polym Bull (Berl) 2024. [DOI: 10.1007/s00289-024-05413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/30/2024] [Accepted: 07/03/2024] [Indexed: 08/14/2024]
|
3
|
Dharmayanti C, Clulow AJ, Gillam TA, Klingler-Hoffmann M, Albrecht H, Blencowe A. Position Matters: Pyridine Regioisomers Influence Secondary Structure and Micelle Morphology in Polymer-Homopolypeptide Micelles. Biomacromolecules 2024; 25:4095-4109. [PMID: 38850240 DOI: 10.1021/acs.biomac.4c00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2024]
Abstract
Polymer-homopolypeptide block copolymers are a class of bioinspired materials that combine the processability and stability of synthetic polymers with the biocompatibility and unique secondary structures of peptides, such as α-helices and β-sheets. These properties make them ideal candidates for a wide variety of applications, for example, in the pharmaceutical field, where they are frequently explored as building blocks for polymeric micelle drug delivery systems. While homopolypeptide side chains can be furnished with an array of different moieties to impart the copolymers with desirable properties, such as stimulus responsivity, pyridine derivatives represent an underutilized functional group for this purpose. Additionally, the interplay between polypeptide side chain structure, secondary conformation, and micelle morphology is not yet well understood, particularly in the case of structural regioisomers. Therefore, in this work, a series of polymer-homopolypeptide copolymers were prepared from a poly(ethylene glycol)-b-poly(glutamic acid) (PEG-b-PGA) backbone, where the pendant carboxylic acid groups were covalently conjugated to a series of pyridine regioisomers by carbodiimide coupling. These pyridine regioisomers differed only in the position of the nitrogen heteroatom, ortho, meta or para, relative to the linking group, generating a series of PEG-b-poly(pyridinylmethyl glutamate) (PEG-b-PMG) copolymers. Following self-assembly of the copolymers in aqueous solutions, dynamic light scattering (DLS) revealed differences in micelle hydrodynamic diameter (Dh) (ranging from ∼60 to 120 nm), while transmission electron microscopy (TEM) and small-angle X-ray scattering (SAXS) revealed distinctive morphologies ranging from ellipsoidal, to cylindrical, and disc-like, suggesting that subtle changes in positional isomers in the polypeptide block may influence the micelle structure. Analysis of the PEG-b-PMG copolymer micelles by circular dichroism (CD) and attenuated total reflectance Fourier transform infrared (ATR-FTIR) spectroscopy revealed that differences in the morphology were associated with changes in polypeptide secondary structure, which in turn was influenced by the position of the pyridine heteroatom. Overall, these findings contribute to the broader understanding of the relationship between polypeptide structure and micelle morphology and serve as useful insight for the rational design of polymer-polypeptide nanoparticles.
Collapse
Affiliation(s)
- Cintya Dharmayanti
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Andrew J Clulow
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation (ANSTO), Clayton, VIC 3168, Australia
| | - Todd A Gillam
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | | | - Hugo Albrecht
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
4
|
Ge C, He J, Gan M, Qian Y, Zhu J, Wu F, Song Z, Yin L. Conformation-Switchable Polypeptides as Molecular Gates for Controllable Drug Release. Biomacromolecules 2024; 25:3373-3383. [PMID: 38713187 DOI: 10.1021/acs.biomac.4c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The control over secondary structure has been widely studied to regulate the properties of polypeptide materials, which is used to change their functions in situ for various biomedical applications. Herein, we designed and constructed enzyme-responsive polypeptides as gating materials for mesoporous silica nanoparticles (MSNs), which underwent a distorted structure-to-helix transition to promote the release of encapsulated drugs. The polypeptide conjugated on the MSN surface adopted a negatively charged, distorted, flexible conformation, covering the pores of MSN to prevent drug leakage. Upon triggering by alkaline phosphatase (ALP) overproduced by tumor cells, the polypeptide transformed into positively charged, α-helical, rigid conformation with potent membrane-penetrating capabilities, which protruded from the MSN surface to uncover the pores. Such a transition thus enabled cancer-selective drug release and cellular internalization to efficiently kill tumor cells. This study highlights the important role of chain flexibility in modulating the biological function of polypeptides and provides a new application paradigm for synthetic polypeptides with secondary-structure transition.
Collapse
Affiliation(s)
- Chenglong Ge
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jianyin He
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Mudan Gan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yu Qian
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Junliang Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Fan Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Ziyuan Song
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
5
|
Pegoraro C, Domingo-Ortí I, Conejos-Sánchez I, Vicent MJ. Unlocking the Mitochondria for Nanomedicine-based Treatments: Overcoming Biological Barriers, Improving Designs, and Selecting Verification Techniques. Adv Drug Deliv Rev 2024; 207:115195. [PMID: 38325562 DOI: 10.1016/j.addr.2024.115195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Enhanced targeting approaches will support the treatment of diseases associated with dysfunctional mitochondria, which play critical roles in energy generation and cell survival. Obstacles to mitochondria-specific targeting include the presence of distinct biological barriers and the need to pass through (or avoid) various cell internalization mechanisms. A range of studies have reported the design of mitochondrially-targeted nanomedicines that navigate the complex routes required to influence mitochondrial function; nonetheless, a significant journey lies ahead before mitochondrially-targeted nanomedicines become suitable for clinical use. Moving swiftly forward will require safety studies, in vivo assays confirming effectiveness, and methodologies to validate mitochondria-targeted nanomedicines' subcellular location/activity. From a nanomedicine standpoint, we describe the biological routes involved (from administration to arrival within the mitochondria), the features influencing rational design, and the techniques used to identify/validate successful targeting. Overall, rationally-designed mitochondria-targeted-based nanomedicines hold great promise for precise subcellular therapeutic delivery.
Collapse
Affiliation(s)
- Camilla Pegoraro
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inés Domingo-Ortí
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
6
|
Zhao H, Li Y, Chen J, Zhang J, Yang Q, Cui J, Shi A, Wu J. Environmental stimulus-responsive mesoporous silica nanoparticles as anticancer drug delivery platforms. Colloids Surf B Biointerfaces 2024; 234:113758. [PMID: 38241892 DOI: 10.1016/j.colsurfb.2024.113758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/03/2024] [Accepted: 01/13/2024] [Indexed: 01/21/2024]
Abstract
Currently, cancer poses a significant health challenge in the medical community. Traditional chemotherapeutic agents are often accompanied by toxic side effects and limited therapeutic efficacy, restricting their application and advancement in cancer treatment. Therefore, there is an urgent need for developing intelligent drug release systems. Mesoporous silica nanoparticles (MSNs) have many advantages, such as a large specific surface area, substantial pore volume and size, adjustable mesoporous material pore size, excellent biocompatibility, and thermodynamic stability, making them ideal carriers for drug delivery and release. Additionally, they have been widely used to develop novel anticancer drug carriers. Recently, MSNs have been employed to design responsive systems that react to the tumor microenvironment and external stimuli for controlled release of anticancer drugs. This includes factors within the intratumor environment, such as pH, temperature, enzymes, and glutathione as well as external tumor stimuli, such as light, magnetic field, and ultrasound, among others. In this review, we discuss the research progress on environmental stimulus-responsive MSNs in anticancer drug delivery systems, including internal and external environment single stimulus-responsive release and combined stimulus-responsive release. We also summarize the current challenges associated with environmental stimulus-responsive MSNs and elucidate future directions, providing a reference for the functionalization modification and practical application of these MSNs.
Collapse
Affiliation(s)
- Huanhuan Zhao
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Yan Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650034, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Jiaxin Chen
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Jinjia Zhang
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Qiuqiong Yang
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Ji Cui
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Anhua Shi
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China.
| | - Junzi Wu
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China; Department of Geriatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650034, China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China.
| |
Collapse
|
7
|
Zhang Y, Lu Y, Li Y, Xu Y, Song W. Poly(Glutamic Acid)-Engineered Nanoplatforms for Enhanced Cancer Phototherapy. Curr Drug Deliv 2024; 21:326-338. [PMID: 36650626 DOI: 10.2174/1567201820666230116164511] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 01/19/2023]
Abstract
Phototherapies, including photothermal therapy and photodynamic therapy, have gained booming development over the past several decades for their attractive non-invasiveness nature, negligible adverse effects, minimal systemic toxicity, and high spatial selectivity. Phototherapy usually requires three components: light irradiation, photosensitizers, and molecular oxygen. Photosensitizers can convert light energy into heat or reactive oxygen species, which can be used in the tumor-killing process. The direct application of photosensitizers in tumor therapy is restricted by their poor water solubility, fast clearance, severe toxicity, and low cellular uptake. The encapsulation of photosensitizers into nanostructures is an attractive strategy to overcome these critical limitations. Poly(glutamic acid) (PGA) is a kind of poly(amino acid)s containing the repeating units of glutamic acid. PGA has superiority for cancer treatment because of its good biocompatibility, low immunogenicity, and modulated pH responsiveness. The hydrophilicity nature of PGA allows the physical entrapment of photosensitizers and anticancer drugs via the construction of amphiphilic polymers. Moreover, the pendent carboxyl groups of PGA enable chemical conjugation with therapeutic agents. In this mini-review, we highlight the stateof- the-art design and fabrication of PGA-based nanoplatforms for phototherapy. We also discuss the potential challenges and future perspectives of phototherapy, and clinical translation of PGA-based nanomedicines.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Yicong Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai-200093, P. R. China
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Wenliang Song
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai-200093, P. R. China
| |
Collapse
|
8
|
Vicente-Ruiz S, Armiñán A, Maso K, Gallon E, Zagorodko O, Movellan J, Rodríguez-Otormín F, Baues M, May JN, De Lorenzi F, Lammers T, Vicent MJ. Poly-l-glutamic acid modification modulates the bio-nano interface of a therapeutic anti-IGF-1R antibody in prostate cancer. Biomaterials 2023; 301:122280. [PMID: 37598440 DOI: 10.1016/j.biomaterials.2023.122280] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 08/04/2023] [Accepted: 08/12/2023] [Indexed: 08/22/2023]
Abstract
Modifying biological agents with polymers such as polyethylene glycol (PEG) has demonstrated clinical benefits; however, post-market surveillance of PEGylated derivatives has revealed PEG-associated toxicity issues, prompting the search for alternatives. We explore how conjugating a poly-l-glutamic acid (PGA) to an anti-insulin growth factor 1 receptor antibody (AVE1642) modulates the bio-nano interface and anti-tumor activity in preclinical prostate cancer models. Native and PGA-modified AVE1642 display similar anti-tumor activity in vitro; however, AVE1642 prompts IGF-1R internalization while PGA conjugation prompts higher affinity IGF-1R binding, thereby inhibiting IGF-1R internalization and altering cell trafficking. AVE1642 attenuates phosphoinositide 3-kinase signaling, while PGA-AVE1642 inhibits phosphoinositide 3-kinase and mitogen-activated protein kinase signaling. PGA conjugation also enhances AVE1642's anti-tumor activity in an orthotopic prostate cancer mouse model, while PGA-AVE1642 induces more significant suppression of cancer cell proliferation/angiogenesis than AVE1642. These findings demonstrate that PGA conjugation modulates an antibody's bio-nano interface, mechanism of action, and therapeutic activity.
Collapse
Affiliation(s)
- Sonia Vicente-Ruiz
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Ana Armiñán
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Katia Maso
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Elena Gallon
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Oleksandr Zagorodko
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Julie Movellan
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain; Current address: CIDETEC, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Gipuzkoa, Donostia-San Sebastián, Spain
| | | | - Maike Baues
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, Aachen, 52074, Germany
| | - Jan-Niklas May
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, Aachen, 52074, Germany
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, Aachen, 52074, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, Aachen, 52074, Germany
| | - María J Vicent
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
9
|
Fernández-Gómez P, Pérez de la Lastra Aranda C, Tosat-Bitrián C, Bueso de Barrio JA, Thompson S, Sot B, Salas G, Somoza Á, Espinosa A, Castellanos M, Palomo V. Nanomedical research and development in Spain: improving the treatment of diseases from the nanoscale. Front Bioeng Biotechnol 2023; 11:1191327. [PMID: 37545884 PMCID: PMC10401050 DOI: 10.3389/fbioe.2023.1191327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/23/2023] [Indexed: 08/08/2023] Open
Abstract
The new and unique possibilities that nanomaterials offer have greatly impacted biomedicine, from the treatment and diagnosis of diseases, to the specific and optimized delivery of therapeutic agents. Technological advances in the synthesis, characterization, standardization, and therapeutic performance of nanoparticles have enabled the approval of several nanomedicines and novel applications. Discoveries continue to rise exponentially in all disease areas, from cancer to neurodegenerative diseases. In Spain, there is a substantial net of researchers involved in the development of nanodiagnostics and nanomedicines. In this review, we summarize the state of the art of nanotechnology, focusing on nanoparticles, for the treatment of diseases in Spain (2017-2022), and give a perspective on the future trends and direction that nanomedicine research is taking.
Collapse
Affiliation(s)
- Paula Fernández-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
| | - Carmen Pérez de la Lastra Aranda
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Carlota Tosat-Bitrián
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Sebastián Thompson
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
| | - Begoña Sot
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Unidad de Innovación Biomédica, Madrid, Spain
- Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJ UAM), Madrid, Spain
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
- Unidad Asociada al Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
- Unidad Asociada al Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| | - Ana Espinosa
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
- Instituto de Ciencia de Materiales de Madrid, ICMM-CSIC, Madrid, Spain
| | - Milagros Castellanos
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
| | - Valle Palomo
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Unidad Asociada al Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| |
Collapse
|
10
|
Li J, Gong C, Chen X, Guo H, Tai Z, Ding N, Gao S, Gao Y. Biomimetic liposomal nanozymes improve breast cancer chemotherapy with enhanced penetration and alleviated hypoxia. J Nanobiotechnology 2023; 21:123. [PMID: 37038165 PMCID: PMC10084658 DOI: 10.1186/s12951-023-01874-7] [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: 01/14/2023] [Accepted: 03/26/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Doxorubicin (Dox) has been recommended in clinical guidelines for the standard-of-care treatment of breast cancer. However, Dox therapy faces challenges such as hypoxia, acidosis, H2O2-rich conditions and condensed extracellular matrix in TME as well as low targeted ability. METHODS We developed a nanosystem H-MnO2-Dox-Col NPs based on mesoporous manganese dioxide (H-MnO2) in which Dox was loaded in the core and collagenase (Col) was wrapped in the surface. Further the H-MnO2-Dox-Col NPs were covered by a fusion membrane (MP) of inflammation-targeted RAW264.7 cell membrane and pH-sensitive liposomes to form biomimetic MP@H-MnO2-Dox-Col for in vitro and in vivo study. RESULTS Our results shows that MP@H-MnO2-Dox-Col can increase the Dox effect with low cardiotoxicity based on multi-functions of effective penetration in tumor tissue, alleviating hypoxia in TME, pH sensitive drug release as well as targeted delivery of Dox. CONCLUSIONS This multifunctional biomimetic nanodelivery system exhibited antitumor efficacy in vivo and in vitro, thus having potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Juanjuan Li
- School of Pharmacy & Zhong Shan Hospital, Fudan University, Shanghai, 201206, China
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, P. R. China
| | - Xinlu Chen
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Huanhuan Guo
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| | - Nan Ding
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shen Gao
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Yuan Gao
- School of Pharmacy & Zhong Shan Hospital, Fudan University, Shanghai, 201206, China.
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
11
|
Baqeri N, Shahsavari S, Dahouee IA, Shirmard LR. Design of slow-release methotrexate drug delivery system using PHBV magnetic nanoparticles and evaluation of its cytotoxicity. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
12
|
Wang Z, Yang C, Zhang H, Gao Y, Xiao M, Wang Z, Yang L, Zhang J, Ren C, Liu J. In Situ Transformable Supramolecular Nanomedicine Targeted Activating Hippo Pathway for Triple-Negative Breast Cancer Growth and Metastasis Inhibition. ACS NANO 2022; 16:14644-14657. [PMID: 36048539 DOI: 10.1021/acsnano.2c05263] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
As it is closely associated with tumor proliferation, metastasis, and the immunosuppressive microenvironment, the dysfunctional Hippo pathway has become an extremely attractive target for treating multiple cancers. However, to date, the corresponding chemotherapeutic nanomedicines have not been developed. Herein, a supramolecular self-delivery nanomedicine with in situ transforming capacity was tailor-constructed for Hippo-pathway restoration, and its inhibitory effect against tumor growth and metastasis was investigated in a highly aggressive triple-negative breast cancer (TNBC) model. Stimulated by overexpressed glutathione (GSH) and esterase in cancer cells, the self-assembled nanomedicine transformed from inactive nanospheres to active nanofibers conjugating tyrosvaline and spatiotemporally synchronously released the covalently linked flufenamic acid in situ, together activating the maladjusted Hippo pathway by simultaneously acting on different targets upstream and downstream. The transcriptional expression of Yes-associated protein (YAP) and related growth-promoted genes were significantly reduced, finally significantly repressing the proliferation and metastasis of cancer cells. Additionally, the Hippo-pathway restoration showed an excellent radiosensitization effect, making the targeted therapy combined with radiotherapy display a prominent synergistic in vivo anticancer effect against TNBC. This work reports a specifically designed smart nanomedicine to restore the function of the Hippo pathway and sensitize radiotherapy, providing an attractive paradigm for targeted drug delivery and cancer combination therapy.
Collapse
Affiliation(s)
- Zhilong Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Hao Zhang
- Laboratory of Functional and Biomedical Nanomaterials, College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Yang Gao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Meng Xiao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Zhongyan Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Jiamin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Chunhua Ren
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| |
Collapse
|
13
|
Epshtein Y, Blau R, Pisarevsky E, Koshrovski-Michael S, Ben-Shushan D, Pozzi S, Shenbach-Koltin G, Fridrich L, Buzhor M, Krivitsky A, Dey P, Satchi-Fainaro R. Polyglutamate-based nanoconjugates for image-guided surgery and post-operative melanoma metastases prevention. Theranostics 2022; 12:6339-6362. [PMID: 36168618 PMCID: PMC9475454 DOI: 10.7150/thno.72941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/21/2022] [Indexed: 01/01/2023] Open
Abstract
Rationale: Cutaneous melanoma is the most aggressive and deadliest of all skin malignancies. Complete primary tumor removal augmented by advanced imaging tools and effective post-operative treatment is critical in the prevention of tumor recurrence and future metastases formation. Methods: To meet this challenge, we designed novel polymeric imaging and therapeutic systems, implemented in a two-step theranostic approach. Both are composed of the biocompatible and biodegradable poly(α,L-glutamic acid) (PGA) nanocarrier that facilitates extravasation-dependent tumor targeting delivery. The first system is a novel, fluorescent, Turn-ON diagnostic probe evaluated for the precise excision of the primary tumor during image-guided surgery (IGS). The fluorescence activation of the probe occurs via PGA degradation by tumor-overexpressed cathepsins that leads to the separation of closely-packed, quenched FRET pair. This results in the emission of a strong fluorescence signal enabling the delineation of the tumor boundaries. Second, therapeutic step is aimed to prevent metastases formation with minimal side effects and maximal efficacy. To that end, a targeted treatment containing a BRAF (Dabrafenib - mDBF)/MEK (Selumetinib - SLM) inhibitors combined on one polymeric platform (PGA-SLM-mDBF) was evaluated for its anti-metastatic, preventive activity in combination with immune checkpoint inhibitors (ICPi) αPD1 and αCTLA4. Results: IGS in melanoma-bearing mice led to a high tumor-to-background ratio and reduced tumor recurrence in comparison with mice that underwent surgery under white light (23% versus 33%, respectively). Adjuvant therapy with PGA-SLM-mDBF combined with ICPi, was well-tolerated and resulted in prolonged survival and prevention of peritoneal and brain metastases formation in BRAF-mutated melanoma-bearing mice. Conclusions: The results reveal the great clinical potential of our PGA-based nanosystems as a tool for holistic melanoma treatment management.
Collapse
Affiliation(s)
- Yana Epshtein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rachel Blau
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, Mail Code 0448, La Jolla, CA 92093-0448
| | - Evgeni Pisarevsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gal Shenbach-Koltin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lidar Fridrich
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Marina Buzhor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
14
|
Ma Y, Lu C, Ji B, Qin J, Cai C, Yang Y, Zhao Y, Liang G, Guo X, Cao G, Li B, Gao P. Integrated Omics Analysis Reveals Alterations in the Intestinal Microbiota and Metabolites of Piglets After Starvation. Front Microbiol 2022; 13:881099. [PMID: 35783381 PMCID: PMC9240708 DOI: 10.3389/fmicb.2022.881099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is a serious public health problem. Short-term starvation is an effective way to lose weight but can also cause harm to the body. However, a systematic assessment of the relationship between the intestinal microbiota and metabolites after complete fasting is lacking. Pigs are the best animal models for exploring the mechanisms of human nutrition digestion and absorption, metabolism, and disease treatment. In this study, 16S rRNA sequencing and liquid chromatography-mass spectrometry were used to analyze the changes in the intestinal microbiota and metabolite profiles in piglets under starvation stress. The results show that the microbial composition was changed significantly in the starvation groups compared with the control group (P < 0.05), suggesting that shifts in the microbial composition were induced by starvation stress. Furthermore, differences in the correlation of the intestinal microbiota and metabolites were observed in the different experimental groups. Starvation may disrupt the homeostasis of the intestinal microbiota and metabolite profile and affect the health of piglets. However, piglets can regulate metabolite production to compensate for the effects of short-term starvation. Our results provide a background to explore the mechanism of diet and short-term hunger for intestinal homeostasis.
Collapse
|
15
|
Lepland A, Malfanti A, Haljasorg U, Asciutto EK, Pickholz M, Bringas M, Đorđević S, Salumäe L, Peterson P, Teesalu T, Vicent MJ, Scodeller P. Depletion of Mannose Receptor-Positive Tumor-associated Macrophages via a Peptide-targeted Star-shaped Polyglutamate Inhibits Breast Cancer Progression in Mice. CANCER RESEARCH COMMUNICATIONS 2022; 2:533-551. [PMID: 36923553 PMCID: PMC10010335 DOI: 10.1158/2767-9764.crc-22-0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/03/2022] [Accepted: 06/03/2022] [Indexed: 12/31/2022]
Abstract
Although many studies have explored the depletion of tumor-associated macrophages (TAM) as a therapeutic strategy for solid tumors, currently available compounds suffer from poor efficacy and dose-limiting side effects. Here, we developed a novel TAM-depleting agent ("OximUNO") that specifically targets CD206+ TAMs and demonstrated efficacy in a triple-negative breast cancer (TNBC) mouse model. OximUNO comprises a star-shaped polyglutamate (St-PGA) decorated with the CD206-targeting peptide mUNO that carries the chemotherapeutic drug doxorubicin (DOX). In the TNBC model, a fluorescently labeled mUNO-decorated St-PGA homed to CD206+ TAMs within primary lesions and metastases. OximUNO exhibited no acute liver or kidney toxicity in vivo. Treatment with OximUNO reduced the progression of primary tumor lesions and pulmonary metastases, significantly diminished the number of CD206+ TAMs and increased the CD8/FOXP3 expression ratio (indicating immunomodulation). Our findings suggest the potential benefit of OximUNO as a TAM-depleting agent for TNBC treatment. Importantly, our studies also represent a novel design of a peptide-targeted St-PGA as a targeted therapeutic nanoconjugate. Significance A peptide-targeted nanoformulation of DOX exclusively eliminates mannose receptor+ TAMs in breast cancer models, generating response without off-target effects (a drawback of many TAM-depleting agents under clinical study).
Collapse
Affiliation(s)
- Anni Lepland
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Alessio Malfanti
- Polymer Therapeutics Laboratory, Prince Felipe Research Centre, Valencia, Spain
| | - Uku Haljasorg
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Eliana K. Asciutto
- School of Science and Technology, National University of San Martin (UNSAM) ICIFI and CONICET, Buenos Aires, Argentina
| | - Monica Pickholz
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Física de Buenos Aires (IFIBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mauro Bringas
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA-CONICET), C1405BWE Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Snežana Đorđević
- Polymer Therapeutics Laboratory, Prince Felipe Research Centre, Valencia, Spain
| | - Liis Salumäe
- Pathology Department, Tartu University Hospital, Tartu, Estonia
| | - Pärt Peterson
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Centre for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, California
| | - María J. Vicent
- Polymer Therapeutics Laboratory, Prince Felipe Research Centre, Valencia, Spain
| | - Pablo Scodeller
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
16
|
Zhang Y, Song W, Lu Y, Xu Y, Wang C, Yu DG, Kim I. Recent Advances in Poly(α- L-glutamic acid)-Based Nanomaterials for Drug Delivery. Biomolecules 2022; 12:636. [PMID: 35625562 PMCID: PMC9138577 DOI: 10.3390/biom12050636] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
Poly(α-L-glutamic acid) (PGA) is a class of synthetic polypeptides composed of the monomeric unit α-L-glutamic acid. Owing to their biocompatibility, biodegradability, and non-immunogenicity, PGA-based nanomaterials have been elaborately designed for drug delivery systems. Relevant studies including the latest research results on PGA-based nanomaterials for drug delivery have been discussed in this work. The following related topics are summarized as: (1) a brief description of the synthetic strategies of PGAs; (2) an elaborated presentation of the evolving applications of PGA in the areas of drug delivery, including the rational design, precise fabrication, and biological evaluation; (3) a profound discussion on the further development of PGA-based nanomaterials in drug delivery. In summary, the unique structures and superior properties enables PGA-based nanomaterials to represent as an enormous potential in biomaterials-related drug delivery areas.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Wenliang Song
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea;
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Changping Wang
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Deng-Guang Yu
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Il Kim
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
17
|
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
|
18
|
Nasrollahpour H, Naseri A, Rashidi MR, Khalilzadeh B. Application of green synthesized WO 3-poly glutamic acid nanobiocomposite for early stage biosensing of breast cancer using electrochemical approach. Sci Rep 2021; 11:23994. [PMID: 34907220 PMCID: PMC8671486 DOI: 10.1038/s41598-021-03209-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/29/2021] [Indexed: 11/09/2022] Open
Abstract
Biopolymer films have drawn growing demand for their application in the point of care domain owing to their biocompatibility, eco-friendly, and eligibility for in vivo analyses. However, their poor conductivity restricts their sensitivity in diagnostics. For high-quality electrochemical biosensor monitoring, two vital factors to be greatly paid attention are the effective merge of amplification modifiers with transducing surface and the superior linking across the recognition interface. Here, we introduce an enzyme-free electrochemical biosensor based on electrosynthesized biocompatible WO3/poly glutamic acid nano-biocomposites to address the hardships specific to the analysis of circulating proteins clinical samples. In addition to its green synthesis route, the poor tendency of both components of the prepared nano-biocomposite to amine groups makes it excellent working in untreated biological samples with high contents of proteins. Several electrochemical and morphological investigations (SEM, EDX, and dot mapping) were fulfilled to gain a reliable and trustful standpoint of the framework. By using this nanobiosensor, the concentration of HER-2 was detectable as low as 1 fg mL-1 with a wide linear response between 1 ng mL-1 and 1 fg mL-1. Meanwhile, the protocol depicted ideal specificity, stability, and reproducibility for the detection of HER-2 protein in untreated serum samples of breast cancer patients.
Collapse
Affiliation(s)
- Hassan Nasrollahpour
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, PO Box 51644-14766, Tabriz, Iran
| | - Abdolhossein Naseri
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, PO Box 51644-14766, Tabriz, Iran.
| | - Mohammad-Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, 51664-14766, Tabriz, Iran.
| |
Collapse
|
19
|
Liu Y, Chen L, Shi Q, Zhao Q, Ma H. Tumor Microenvironment-Responsive Polypeptide Nanogels for Controlled Antitumor Drug Delivery. Front Pharmacol 2021; 12:748102. [PMID: 34776965 PMCID: PMC8578677 DOI: 10.3389/fphar.2021.748102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Tumor microenvironment-responsive polypeptide nanogels belong to a biomaterial with excellent biocompatibility, easily adjustable performance, biodegradability, and non-toxic properties. They are developed for selective delivery of antitumor drugs into target organs to promote tumor cell uptake, which has become an effective measure of tumor treatment. Endogenous (such as reduction, reactive oxygen species, pH, and enzyme) and exogenous (such as light and temperature) responsive nanogels can release drugs in response to tumor tissues or cells to improve drug distribution and reduce drug side effects. This article systematically introduces the research progress in tumor microenvironment-responsive polypeptide nanogels to deliver antitumor drugs and provides a reference for the development of antitumor nanoformulations.
Collapse
Affiliation(s)
- Yanhong Liu
- Center for Reproductive Medicine, Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Linjiao Chen
- Center for Reproductive Medicine, Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Qingyang Shi
- Center for Reproductive Medicine, Center for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Qing Zhao
- Department of Obstetrics, First Hospital, Jilin University, Changchun, China
| | - Hongshuang Ma
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Zheng B, Bai T, Tao X, Ling J. An Inspection into Multifarious Ways to Synthesize Poly(Amino Acid)s. Macromol Rapid Commun 2021; 42:e2100453. [PMID: 34562289 DOI: 10.1002/marc.202100453] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/09/2021] [Indexed: 12/21/2022]
Abstract
Poly(α-amino acid)s (PAAs) attract growing attention due to their essential role in the application as biomaterials. To synthesize PAAs with desired structures and properties, scientists have developed various synthetic techniques with respective advantages. Here, different approaches to preparing PAAs are inspected. Basic features and recent progresses of these methods are summarized, including polymerizations of amino acid N-carboxyanhydrides (NCAs), amino acid N-thiocarboxyanhydrides (NTAs), and N-phenoxycarbonyl amino acids (NPCs), as well as other synthetic routes. NCA is the most classical monomer to prepare PAAs with high molecular weights (MWs). NTA polymerizations are promising alternative pathways to produce PAAs, which can tolerate nucleophiles including alcohols, mercaptans, carboxyl acids, and water. By various techniques including choosing appropriate solvents or using organic acids as promoters, NTAs polymerize to produce polypeptoids and polypeptides with narrow dispersities and designed MWs up to 55.0 and 57.0 kg mol-1 , respectively. NPC polymerizations are phosgene-free ways to synthesize polypeptides and polypeptoids. For the future prospects, detail investigations into polymerization mechanisms of NTA and NPC are expected. The synthesis of PAAs with designed topologies and assembly structures is another intriguing topic. The advantages and unsettled problems in various synthetic ways are discussed for readers to choose appropriate approaches for PAAs.
Collapse
Affiliation(s)
- Botuo Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.,Fujian Key Laboratory of Polymer Science, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, 350007, China
| | - Tianwen Bai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xinfeng Tao
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jun Ling
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
21
|
Wang Y, Zhang X, Wan K, Zhou N, Wei G, Su Z. Supramolecular peptide nano-assemblies for cancer diagnosis and therapy: from molecular design to material synthesis and function-specific applications. J Nanobiotechnology 2021; 19:253. [PMID: 34425823 PMCID: PMC8381530 DOI: 10.1186/s12951-021-00999-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/15/2021] [Indexed: 01/10/2023] Open
Abstract
Peptide molecule has high bioactivity, good biocompatibility, and excellent biodegradability. In addition, it has adjustable amino acid structure and sequence, which can be flexible designed and tailored to form supramolecular nano-assemblies with specific biomimicking, recognition, and targeting properties via molecular self-assembly. These unique properties of peptide nano-assemblies made it possible for utilizing them for biomedical and tissue engineering applications. In this review, we summarize recent progress on the motif design, self-assembly synthesis, and functional tailoring of peptide nano-assemblies for both cancer diagnosis and therapy. For this aim, firstly we demonstrate the methodologies on the synthesis of various functional pure and hybrid peptide nano-assemblies, by which the structural and functional tailoring of peptide nano-assemblies are introduced and discussed in detail. Secondly, we present the applications of peptide nano-assemblies for cancer diagnosis applications, including optical and magnetic imaging as well as biosensing of cancer cells. Thirdly, the design of peptide nano-assemblies for enzyme-mediated killing, chemo-therapy, photothermal therapy, and multi-therapy of cancer cells are introduced. Finally, the challenges and perspectives in this promising topic are discussed. This work will be useful for readers to understand the methodologies on peptide design and functional tailoring for highly effective, specific, and targeted diagnosis and therapy of cancers, and at the same time it will promote the development of cancer diagnosis and therapy by linking those knowledges in biological science, nanotechnology, biomedicine, tissue engineering, and analytical science.
Collapse
Affiliation(s)
- Yan Wang
- College of Chemistry and Chemical Engineering, Qingdao University, 266071, Qingdao, People's Republic of China
| | - Xiaoyuan Zhang
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing, People's Republic of China
| | - Keming Wan
- College of Chemistry and Chemical Engineering, Qingdao University, 266071, Qingdao, People's Republic of China
| | - Nan Zhou
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing, People's Republic of China
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, 266071, Qingdao, People's Republic of China.
| | - Zhiqiang Su
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing, People's Republic of China.
| |
Collapse
|
22
|
Hao D, Zhang Z, Ji Y. Responsive polymeric drug delivery systems for combination anticancer therapy: experimental design and computational insights. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1960340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Dule Hao
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Zheng Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
23
|
Giraldo E, Nebot VJ, Đorđević S, Requejo-Aguilar R, Alastrue-Agudo A, Zagorodko O, Armiñan A, Martinez-Rojas B, Vicent MJ, Moreno-Manzano V. A rationally designed self-immolative linker enhances the synergism between a polymer-rock inhibitor conjugate and neural progenitor cells in the treatment of spinal cord injury. Biomaterials 2021; 276:121052. [PMID: 34388362 DOI: 10.1016/j.biomaterials.2021.121052] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/04/2021] [Accepted: 07/24/2021] [Indexed: 10/24/2022]
Abstract
Rho/ROCK signaling induced after spinal cord injury (SCI) contributes to secondary damage by promoting apoptosis, inflammation, and axon growth inhibition. The specific Rho-kinase inhibitor fasudil can contribute to functional regeneration after SCI, although inherent low stability has hampered its use. To improve the therapeutic potential of fasudil, we now describe a family of rationally-designed bioresponsive polymer-fasudil conjugates based on an understanding of the conditions after SCI, such as low pH, enhanced expression of specific proteases, and a reductive environment. Fasudil conjugated to poly-l-glutamate via a self-immolative redox-sensitive linker (PGA-SS-F) displays optimal release kinetics and, consequently, treatment with PGA-SS-F significantly induces neurite elongation and axon growth in dorsal root ganglia explants, spinal cord organotypic cultures, and neural precursor cells (NPCs). The intrathecal administration of PGA-SS-F after SCI in a rat model prevents early apoptosis and induces the expression of axonal growth- and neuroplasticity-associated markers to a higher extent than the free form of fasudil. Moreover, a combination treatment comprising the acute transplantation of NPCs pre-treated with PGA-SS-F leads to enhanced cell engraftment and reduced cyst formation after SCI. In chronic SCI, combinatory treatment increases the preservation of neuronal fibers. Overall, this synergistic combinatorial strategy may represent a potentially efficient clinical approach to SCI treatment.
Collapse
Affiliation(s)
- E Giraldo
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain; Department of Biotechnology. Universitat Politècnica de València, Valencia, Spain
| | - V J Nebot
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain; PTS S.L., Valencia, Spain
| | - S Đorđević
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - R Requejo-Aguilar
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain; Dept. Biochemistry and Molecular Biology, University of Cordoba, Cordoba, Spain. Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| | - A Alastrue-Agudo
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain
| | - O Zagorodko
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - A Armiñan
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - B Martinez-Rojas
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain
| | - M J Vicent
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain.
| | - V Moreno-Manzano
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain.
| |
Collapse
|
24
|
Song Y, Ding Y, Dong CM. Stimuli-responsive polypeptide nanoassemblies: Recent progress and applications in cancer nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1742. [PMID: 34310063 DOI: 10.1002/wnan.1742] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
Stimuli-responsive polypeptide nanoassemblies exhibit great potentials for cancer nanomedicines because of desirable biocompatibility and biodegradability, unique secondary conformations, varying functionalities, and especially the stimuli-enhanced therapeutic efficacy and reduced side effect. This review introduces the design and fabrication of stimuli-responsive polypeptide nanoassemblies that exhibit endogenous stimuli (e.g., pH, reduction, reactive oxygen species, adenosine triphosphate and enzyme, etc.) and exogenous light stimuli (e.g., UV and near-infrared light), which are biologically related or applied in the clinic. We also discuss the applications and prospects of those stimuli-responsive polypeptide nanoassemblies that might overcome the biological barriers of cancer nanomedicines for in vivo administration. Much more effort is needed to accelerate the second-generation stimuli-responsive polypeptide nanomedicines for clinical transition and applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yingying Song
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Boix-Montesinos P, Soriano-Teruel PM, Armiñán A, Orzáez M, Vicent MJ. The past, present, and future of breast cancer models for nanomedicine development. Adv Drug Deliv Rev 2021; 173:306-330. [PMID: 33798642 PMCID: PMC8191594 DOI: 10.1016/j.addr.2021.03.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Even given recent advances in nanomedicine development of breast cancer treatment in recent years and promising results in pre-clinical models, cancer nanomedicines often fail at the clinical trial stage. Limitations of conventional in vitro models include the lack of representation of the stromal population, the absence of a three-dimensional (3D) structure, and a poor representation of inter-tumor and intra-tumor heterogeneity. Herein, we review those cell culture strategies that aim to overcome these limitations, including cell co-cultures, advanced 3D cell cultures, patient-derived cells, bioprinting, and microfluidics systems. The in vivo evaluation of nanomedicines must consider critical parameters that include the enhanced permeability and retention effect, the host's immune status, and the site of tumor implantation. Here, we critically discuss the advantages and limitations of current in vivo models and report how the improved selection and application of breast cancer models can improve the clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Paula M Soriano-Teruel
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain; Centro de Investigación Príncipe Felipe, Targeted Therapies on Cancer and Inflammation Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Ana Armiñán
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Mar Orzáez
- Centro de Investigación Príncipe Felipe, Targeted Therapies on Cancer and Inflammation Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
26
|
Brunato S, Mastrotto F, Bellato F, Bastiancich C, Travanut A, Garofalo M, Mantovani G, Alexander C, Preat V, Salmaso S, Caliceti P. PEG-polyaminoacid based micelles for controlled release of doxorubicin: Rational design, safety and efficacy study. J Control Release 2021; 335:21-37. [PMID: 33989691 DOI: 10.1016/j.jconrel.2021.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/11/2022]
Abstract
A library of amphiphilic monomethoxypolyethylene glycol (mPEG) terminating polyaminoacid co-polymers able to self-assemble into colloidal systems was screened for the delivery and controlled release of doxorubicin (Doxo). mPEG-Glu/Leu random co-polymers were generated by Ring Opening Polymerization from 5 kDa mPEG-NH2 macroinitiator using 16:0:1, 8:8:1, 6:10:1, 4:12:1 γ-benzyl glutamic acid carboxy anhydride monomer/leucine N-carboxy anhydride monomer/PEG molar ratios. Glutamic acid was selected for chemical conjugation of Doxo, while leucine units were introduced in the composition of the polyaminoacid block as spacer between adjacent glutamic repeating units to minimize the steric hindrance that could impede the Doxo conjugation and to promote the polymer self-assembly by virtue of the aminoacid hydrophobicity. The benzyl ester protecting the γ-carboxyl group of glutamic acid was quantitatively displaced with hydrazine to yield mPEG5kDa-b-(hydGlum-r-Leun). Doxo was conjugated to the diblock co-polymers through pH-sensitive hydrazone bond. The Doxo derivatized co-polymers obtained with a 16:0:1, 8:8:1, 6:10:1 Glu/Leu/PEG ratios self-assembled into 30-40 nm spherical nanoparticles with neutral zeta-potential and CMC in the range of 4-7 μM. At pH 5.5, mimicking endosome environment, the carriers containing leucine showed a faster Doxo release than at pH 7.4, mimicking the blood conditions. Doxo-loaded colloidal formulations showed a dose dependent cytotoxicity on two cancer cell lines, CT26 murine colorectal carcinoma and 4T1 murine mammary carcinoma with IC50 slightly higher than those of free Doxo. The carrier assembled with the polymer containing 6:10:1 hydGlu/Leu/PEG molar ratio {mPEG5kDa-b-[(Doxo-hydGlu)6-r-Leu10]} was selected for subsequent in vitro and in vivo investigations. Confocal imaging on CT26 cell line showed that intracellular fate of the carrier involves a lysosomal trafficking pathway. The intratumor or intravenous injection to CT26 and 4T1 subcutaneous tumor bearing mice yielded higher antitumor activity compared to free Doxo. Furthermore, mPEG5kDa-b-[(Doxo-hydGlu)6-r-Leu10] displayed a better safety profile when compared to commercially available Caelyx®.
Collapse
Affiliation(s)
- Silvia Brunato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131 Padova, Italy
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131 Padova, Italy
| | - Federica Bellato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131 Padova, Italy
| | - Chiara Bastiancich
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200 Brussels, Belgium
| | - Alessandra Travanut
- Molecular Therapeutics and Formulations Division, School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131 Padova, Italy
| | - Giuseppe Mantovani
- Molecular Therapeutics and Formulations Division, School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Cameron Alexander
- Molecular Therapeutics and Formulations Division, School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Veronique Preat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200 Brussels, Belgium
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131 Padova, Italy.
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
27
|
Tejedor S, Dolz‐Pérez I, Decker CG, Hernándiz A, Diez JL, Álvarez R, Castellano D, García NA, Ontoria‐Oviedo I, Nebot VJ, González‐King H, Igual B, Sepúlveda P, Vicent MJ. Polymer Conjugation of Docosahexaenoic Acid Potentiates Cardioprotective Therapy in Preclinical Models of Myocardial Ischemia/Reperfusion Injury. Adv Healthc Mater 2021; 10:e2002121. [PMID: 33720548 DOI: 10.1002/adhm.202002121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/16/2021] [Indexed: 01/16/2023]
Abstract
While coronary angioplasty represents an effective treatment option following acute myocardial infarction, the reperfusion of the occluded coronary artery can prompt ischemia-reperfusion (I/R) injury that significantly impacts patient outcomes. As ω-3 polyunsaturated fatty acids (PUFAs) have proven, yet limited cardioprotective abilities, an optimized polymer-conjugation approach is reported that improves PUFAs bioavailability to enhance cardioprotection and recovery in animal models of I/R-induced injury. Poly-l-glutamic acid (PGA) conjugation improves the solubility and stability of di-docosahexaenoic acid (diDHA) under physiological conditions and protects rat neonatal ventricular myocytes from I/R injury by reducing apoptosis, attenuating autophagy, inhibiting reactive oxygen species generation, and restoring mitochondrial membrane potential. Enhanced protective abilities are associated with optimized diDHA loading and evidence is provided for the inherent cardioprotective potential of PGA itself. Pretreatment with PGA-diDHA before reperfusion in a small animal I/R model provides for cardioprotection and limits area at risk (AAR). Furthermore, the preliminary findings suggest that PGA-diDHA administration in a swine I/R model may provide cardioprotection, limit edema and decrease AAR. Overall, the evaluation of PGA-diDHA in relevant preclinical models provides evidence for the potential of polymer-conjugated PUFAs in the mitigation of I/R injury associated with coronary angioplasty.
Collapse
Affiliation(s)
- Sandra Tejedor
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Irene Dolz‐Pérez
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Caitlin G. Decker
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Amparo Hernándiz
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Jose L. Diez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Raquel Álvarez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Delia Castellano
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Nahuel A. García
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Imelda Ontoria‐Oviedo
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Vicent J. Nebot
- Polypeptide Therapeutic Solutions S.L. Av. Benjamin Franklin 19, Paterna Valencia 46980 Spain
| | - Hernán González‐King
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Begoña Igual
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| |
Collapse
|
28
|
Wang W, Zhang Q, Li Z, Zhang J, Pan D, Wang B, Zhu H, Zhang H, Gu Z, Luo K. Dendron-Functionalized Polyglutamate-Pyropheophorbide-a Conjugates as Nanomedicines for Breast Cancer Photodynamic Therapy. Macromol Rapid Commun 2021; 42:e2100013. [PMID: 33759304 DOI: 10.1002/marc.202100013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/25/2021] [Indexed: 02/05/2023]
Abstract
Photodynamic therapy (PDT) has shown its promise in the treatment of cancer. Herein, a dendron-functionalized polyglutamic acid (PGA) polymer (PG-L8G-Ppa-Dendron, PGPD) is synthesized and it is conjugated with pyropheophorbide-a (Ppa) for the first time to treat triple negative breast cancer (TNBC), whereas a linear polyglutamate-Ppa conjugate (PGP) is synthesized as a control. Compared to the linear counterpart, the glycosylated polymer-based PGPD with a dendritic structure has excellent solubility and it self-assembles to form uniform-sized nanoparticles. PGPD displays a highly effective PDT effect in the animal model, evidenced with effective induction of reactive oxygen species (ROS) production and cell apoptosis. This may be due to an enhanced efficiency in delivery and accumulation of Ppa by this glycosylated dendritic polymer at tumor sites. Therefore, PGPD can be a highly effective and biosafe nanoagent for PDT of TNBC.
Collapse
Affiliation(s)
- Wenjia Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qianfeng Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.,School of Chemistry and Chemical Engineering, Mianyang Normal University, Mianyang, 621000, China
| | - Zhiqian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.,Department of Applied Chemistry, Faculty of Science, Sichuan Agricultural University, Yaan, 625014, China
| | - Dayi Pan
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bing Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongyan Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
29
|
Dharmayanti C, Gillam TA, Klingler-Hoffmann M, Albrecht H, Blencowe A. Strategies for the Development of pH-Responsive Synthetic Polypeptides and Polymer-Peptide Hybrids: Recent Advancements. Polymers (Basel) 2021; 13:624. [PMID: 33669548 PMCID: PMC7921987 DOI: 10.3390/polym13040624] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Synthetic polypeptides and polymer-peptide hybrid materials have been successfully implemented in an array of biomedical applications owing to their biocompatibility, biodegradability and ability to mimic natural proteins. In addition, these materials have the capacity to form complex supramolecular structures, facilitate specific biological interactions, and incorporate a diverse selection of functional groups that can be used as the basis for further synthetic modification. Like conventional synthetic polymers, polypeptide-based materials can be designed to respond to external stimuli (e.g., light and temperature) or changes in the environmental conditions (e.g., redox reactions and pH). In particular, pH-responsive polypeptide-based systems represent an interesting avenue for the preparation of novel drug delivery systems that can exploit physiological or pathological pH variations within the body, such as those that arise in the extracellular tumour microenvironment, intracellularly within endosomes/lysosomes, or during tissue inflammation. Here, we review the significant progress made in advancing pH-responsive polypeptides and polymer-peptide hybrid materials during the last five years, with a particular emphasis on the manipulation of ionisable functional groups, pH-labile linkages, pH-sensitive changes to secondary structure, and supramolecular interactions.
Collapse
Affiliation(s)
- Cintya Dharmayanti
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
| | - Todd A. Gillam
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
- Surface Interactions and Soft Matter Group, Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | | | - Hugo Albrecht
- Drug Discovery and Development Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
| |
Collapse
|
30
|
Polyglutamic acid-based crosslinked doxorubicin nanogels as an anti-metastatic treatment for triple negative breast cancer. J Control Release 2021; 332:10-20. [PMID: 33587988 DOI: 10.1016/j.jconrel.2021.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Treatment of triple negative breast cancer (TNBC)-associated metastasis represents an unmet clinical need, and we lack effective therapeutics for a disease that exhibits high relapse rates and associates with poor patient outcomes. Advanced nanosized drug delivery systems may enhance the efficacy of first-line chemotherapeutics by altering drug pharmacokinetics and enhancing tumor/metastasis targeting to significantly improve efficacy and safety. Herein, we propose the application of injectable poly-amino acid-based nanogels (NGs) as a versatile hydrophilic drug delivery platform for the treatment of TNBC lung metastasis. We prepared biocompatible and biodegradable cross-linked NGs from polyglutamic acid (PGA) loaded with the chemotherapeutic agent doxorubicin (DOX). Our optimized synthetic procedures generated NGs of ~100 nm in size and 25 wt% drug loading content that became rapidly internalized in TNBC cell lines and displayed IC50 values comparable to the free form of DOX. Importantly, PGA-DOX NGs significantly inhibited lung metastases and almost completely suppressed lymph node metastases in a spontaneously metastatic orthotopic mouse TNBC model. Overall, our newly developed PGA-DOX NGs represent a potentially effective therapeutic strategy for the treatment of TNBC metastases.
Collapse
|
31
|
Monahan DS, Almas T, Wyile R, Cheema FH, Duffy GP, Hameed A. Towards the use of localised delivery strategies to counteract cancer therapy-induced cardiotoxicities. Drug Deliv Transl Res 2021; 11:1924-1942. [PMID: 33449342 DOI: 10.1007/s13346-020-00885-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Cancer therapies have significantly improved cancer survival; however, these therapies can often result in undesired side effects to off target organs. Cardiac disease ranging from mild hypertension to heart failure can occur as a result of cancer therapies. This can warrant the discontinuation of cancer treatment in patients which can be detrimental, especially when the treatment is effective. There is an urgent need to mitigate cardiac disease that occurs as a result of cancer therapy. Delivery strategies such as the use of nanoparticles, hydrogels, and medical devices can be used to localise the treatment to the tumour and prevent off target side effects. This review summarises the advancements in localised delivery of anti-cancer therapies to tumours. It also examines the localised delivery of cardioprotectants to the heart for patients with systemic disease such as leukaemia where localised tumour delivery might not be an option.
Collapse
Affiliation(s)
- David S Monahan
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.,Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Talal Almas
- School of Medicine, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Robert Wyile
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland
| | - Faisal H Cheema
- HCA Healthcare, Gulf Coast Division, Houston, TX, USA.,College of Medicine, University of Houston, Houston, TX, USA
| | - Garry P Duffy
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.,Tissue Engineering Research Group (TERG), Department of Anatomy, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland.,Advanced Materials for Biomedical Engineering and Regenerative Medicine (AMBER), National University of Ireland, Trinity College Dublin &, Galway, Ireland.,Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland. .,Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland.
| |
Collapse
|
32
|
Bao Y, Yu H, Zhang Y, Chen L. Comparative study of two poly(amino acid)-based photosensitizer-delivery systems for photodynamic therapy. Int J Biol Macromol 2020; 169:153-160. [PMID: 33326837 DOI: 10.1016/j.ijbiomac.2020.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/29/2020] [Accepted: 12/03/2020] [Indexed: 01/10/2023]
Abstract
The photosensitizers (PS) play a vital role in photodynamic therapy (PDT), but the clinical therapeutic effect is limited by its low solubility, easy aggregation and lack of selective tumor uptake. Hence, some biocompatible materials such as poly(amino acid) have been chosen to deliver PS to solve these problems. In this study, we fabricated two kind of poly(amino acid)-based PS delivery systems by using poly (L-glutamic acid) (PLG) as the backbone material to physically encapsulate (P(T)) and chemically conjugate PS (PT), respectively. Moreover, the anticancer effect of these two PS delivery systems had been comparatively investigated. In vitro experiments verified that the delivery system of PT exhibited more effective treatment effect than that of P(T) because PS was chemically conjugated with PLG in PT which could obviously avoid the π-π stacking effect of PS and induced the aggregation. The less aggregation avoiding the quenching effect could result in an enhanced generation ability of reactive oxygen species to improve PDT treatment effect. Contrarily, the treatment effect of P(T) was lower due to the rigid structure and hydrophobic π-π stacking effect, in which the physically encapsulated PS was easier to aggregate and quench. Hence, the way how to combine carrier with drug is significant for satisfied treatment effect.
Collapse
Affiliation(s)
- Yanli Bao
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Haiyang Yu
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Yu Zhang
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Li Chen
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China.
| |
Collapse
|
33
|
Vanza JD, Patel RB, Patel MR. Nanocarrier centered therapeutic approaches: Recent developments with insight towards the future in the management of lung cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Vicente‐Ruiz S, Serrano‐Martí A, Armiñán A, Vicent MJ. Nanomedicine for the Treatment of Advanced Prostate Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sonia Vicente‐Ruiz
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Antoni Serrano‐Martí
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Ana Armiñán
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| |
Collapse
|
35
|
Wang Y, Jiang L, Zhang Y, Lu Y, Li J, Wang H, Yao D, Wang D. Fibronectin-Targeting and Cathepsin B-Activatable Theranostic Nanoprobe for MR/Fluorescence Imaging and Enhanced Photodynamic Therapy for Triple Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:33564-33574. [PMID: 32633941 DOI: 10.1021/acsami.0c10397] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Because of the lack of specific targets, the highly aggressive triple negative breast cancer (TNBC) is unable to benefit from endocrine therapy or conventional targeting therapy. Even worse, current diagnostic and therapeutic approaches have limited value for TNBC. Therefore, developing TNBC-specific theranostic probes for accurate diagnosis and further selective therapy will build a powerful toolbox for TNBC management. In this contribution, we developed a sequential strategy to enhance the specificity of TNBC theranostics. In this theranostic system, a versatile nanoprobe (Pep-SQ@USPIO) was integrated legitimately for the fibronectin-targeting MR imaging and CTSB-activatable fluorescence imaging, followed with enhanced photodynamic therapy (PDT) of TNBC. First, the fibronectin overexpressed in the extracellular matrix (ECM) of TNBC was used as a biomarker for targeting theranostics using the Cys-Arg-Glu-Lys-Ala (CREKA) peptide. For another, the fluorescence and PDT capacity of self-developed squaraine photosensitizer (SQ) were prequenched by ultrasmall superparamagnetic iron oxide (USPIO), an MR imaging contrast agent. Once the linker, Gly-Phe-Leu-Gly (GFLG) peptide, was selectively cleaved by TNBC-derived CTSB, the liberated SQ photosensitizer allowed light-up fluorescence imaging and enhanced PDT of TNBC. Remarkably, this research demonstrates that tumor-ECM-targeting and endogenous enzyme-activated nanoprobes open a new avenue for TNBC theranostics.
Collapse
Affiliation(s)
- Yanshu Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Liping Jiang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Yimei Lu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jinning Li
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Defan Yao
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
36
|
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
|
37
|
Kemp JA, Keebaugh A, Edson JA, Chow D, Kleinman MT, Chew YC, McCracken AN, Edinger AL, Kwon YJ. Biocompatible Chemotherapy for Leukemia by Acid-Cleavable, PEGylated FTY720. Bioconjug Chem 2020; 31:673-684. [PMID: 31986014 DOI: 10.1021/acs.bioconjchem.9b00822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Targeting the inability of cancerous cells to adapt to metabolic stress is a promising alternative to conventional cancer chemotherapy. FTY720 (Gilenya), an FDA-approved drug for the treatment of multiple sclerosis, has recently been shown to inhibit cancer progression through the down-regulation of essential nutrient transport proteins, selectively starving cancer cells to death. However, the clinical use of FTY720 for cancer therapy is prohibited because of its capability of inducing immunosuppression (lymphopenia) and bradycardia when phosphorylated upon administration. A prodrug to specifically prevent phosphorylation during circulation, hence avoiding bradycardia and lymphopenia, was synthesized by capping its hydroxyl groups with polyethylene glycol (PEG) via an acid-cleavable ketal linkage. Improved aqueous solubility was also accomplished by PEGylation. The prodrug reduces to fully potent FTY720 upon cellular uptake and induces metabolic stress in cancer cells. Enhanced release of FTY720 at a mildly acidic endosomal pH and the ability to substantially down-regulate cell-surface nutrient transporter proteins in leukemia cells only by an acid-cleaved drug were confirmed. Importantly, the prodrug demonstrated nearly identical efficacy to FTY720 in an animal model of BCR-Abl-driven leukemia without inducing bradycardia or lymphopenia in vivo, highlighting its potential clinical value. The prodrug formulation of FTY720 demonstrates the utility of precisely engineering a drug to avoid undesirable effects by tackling specific molecular mechanisms as well as a financially favorable alternative to new drug development. A multitude of existing cancer therapeutics may be explored for prodrug formulation to avoid specific side effects and preserve or enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Yap Ching Chew
- Zymo Research Corporation, Irvine, California 92604, United States
| | | | | | | |
Collapse
|
38
|
Melnyk T, Đorđević S, Conejos-Sánchez I, Vicent MJ. Therapeutic potential of polypeptide-based conjugates: Rational design and analytical tools that can boost clinical translation. Adv Drug Deliv Rev 2020; 160:136-169. [PMID: 33091502 DOI: 10.1016/j.addr.2020.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022]
Abstract
The clinical success of polypeptides as polymeric drugs, covered by the umbrella term "polymer therapeutics," combined with related scientific and technological breakthroughs, explain their exponential growth in the development of polypeptide-drug conjugates as therapeutic agents. A deeper understanding of the biology at relevant pathological sites and the critical biological barriers faced, combined with advances regarding controlled polymerization techniques, material bioresponsiveness, analytical methods, and scale up-manufacture processes, have fostered the development of these nature-mimicking entities. Now, engineered polypeptides have the potential to combat current challenges in the advanced drug delivery field. In this review, we will discuss examples of polypeptide-drug conjugates as single or combination therapies in both preclinical and clinical studies as therapeutics and molecular imaging tools. Importantly, we will critically discuss relevant examples to highlight those parameters relevant to their rational design, such as linking chemistry, the analytical strategies employed, and their physicochemical and biological characterization, that will foster their rapid clinical translation.
Collapse
Affiliation(s)
- Tetiana Melnyk
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Snežana Đorđević
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
39
|
Zagorodko O, Nebot VJ, Vicent MJ. The generation of stabilized supramolecular nanorods from star-shaped polyglutamates. Polym Chem 2020. [DOI: 10.1039/c9py01442j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We developed a new strategy of polyglutamate nanorod preparation based on supramolecular polymers stabilized with hydrophobic drugs.
Collapse
Affiliation(s)
- O. Zagorodko
- Polymer Therapeutics Lab. Prince Felipe Research Center
- 46012 Valencia
- Spain
| | - V. J. Nebot
- Polymer Therapeutics Lab. Prince Felipe Research Center
- 46012 Valencia
- Spain
| | - M. J. Vicent
- Polymer Therapeutics Lab. Prince Felipe Research Center
- 46012 Valencia
- Spain
| |
Collapse
|
40
|
Dolz-Pérez I, Sallam MA, Masiá E, Morelló-Bolumar D, Pérez Del Caz MD, Graff P, Abdelmonsif D, Hedtrich S, Nebot VJ, Vicent MJ. Polypeptide-corticosteroid conjugates as a topical treatment approach to psoriasis. J Control Release 2019; 318:210-222. [PMID: 31843640 DOI: 10.1016/j.jconrel.2019.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/18/2019] [Accepted: 12/11/2019] [Indexed: 12/19/2022]
Abstract
Topical treatment of mild-to-moderate psoriasis with corticosteroids suffers from challenges that include reduced drug bioavailability at the desired site of action. The retention of therapeutics within the epidermis can safely treat skin inflammation, scaling, and erythema associated with psoriasis while avoiding possible side effects associated with systemic treatments. We successfully synthesized and characterized a pH-responsive biodegradable poly-L-glutamic acid (PGA)-fluocinolone acetonide (FLUO) conjugate that allows the controlled release of the FLUO to reduce skin inflammation. Additionally, the application of a hyaluronic acid (HA)-poly-L-glutamate cross polymer (HA-CP) vehicle boosted skin permeation. During in vitro and ex vivo analyses, we discovered that PGA-FLUO inhibited pro-inflammatory cytokine release, suggesting that polypeptidic conjugation fails to affect the anti-inflammatory activity of FLUO. Additionally, ex vivo human skin permeation studies using confocal microscopy revealed the presence of PGA-FLUO within the epidermis, but a minimal presence in the dermis, thereby reducing the likelihood of FLUO entering the systemic circulation. Finally, we demonstrated that PGA-FLUO applied within HA-CP effectively reduced psoriasis-associated phenotypes in an in vivo mouse model of human psoriasis while also lowering levels of pro-inflammatory cytokines in tissue and serum. Overall, our experimental results demonstrate that PGA-FLUO within an HA-CP penetration enhancer represents an effective topical treatment for psoriasis.
Collapse
Affiliation(s)
- Irene Dolz-Pérez
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Marwa A Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, El Sultan Hussein St Azarita, Egypt; John A. Paulson school of engineering and applied sciences and Wyss institute for biologically inspired engineering, Harvard University, 52 Oxford St, Cambridge, MA 02138, USA
| | - Esther Masiá
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Screening Platform, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Daniel Morelló-Bolumar
- Polypeptide Therapeutic Solutions S.L. C/ Benjamin Franklin 19 (Paterna), Valencia 46980, Spain
| | - M Dolores Pérez Del Caz
- Servicio de cirugía plástica, Hospital Universitario y Politécnico La Fe, Av. de Fernando Abril Martorell 106, Valencia 46026, Spain
| | - Patrick Graff
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Königin-Luise Str. 2+4, Berlin 14195, Germany
| | - Doaa Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, El Sultan Hussein St Azarita, Egypt
| | - Sarah Hedtrich
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Königin-Luise Str. 2+4, Berlin 14195, Germany; Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada
| | - Vicent J Nebot
- Polypeptide Therapeutic Solutions S.L. C/ Benjamin Franklin 19 (Paterna), Valencia 46980, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Screening Platform, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| |
Collapse
|
41
|
Jiang X, Fan X, Xu W, Zhao C, Wu H, Zhang R, Wu G. Self-assembled peptide nanoparticles responsive to multiple tumor microenvironment triggers provide highly efficient targeted delivery and release of antitumor drug. J Control Release 2019; 316:196-207. [PMID: 31682910 DOI: 10.1016/j.jconrel.2019.10.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/29/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
Stimuli-responsive drug delivery systems based on tumor microenvironment conditions show tremendous promise to enhance tumor-targeted delivery and drug release. Herein, a multifunctional peptide (P51) was developed for programmed delivery of the hydrophobic chemotherapeutic agent pirarubicin. P51 was prepared with a ligand-specific targeting for the cancer biomarker Arg-Gly-Asp (RGD), and three tumor microenvironment-sensitive release triggers, acid environment, reducing agent, and a specific enzyme. The peptides Cys-s-s-Cys (disulfide linkage) and Pro-Val-Gly-Leu-Ile-Gly correspond to the cleavage sites of a reducing agent (DTT) and an enzyme (MMP-2). The peptides act as a junction between Ser-Glu-Glu-Asp-Pro (a negatively charged sequence) and a 41-residue peptide containing an α-helix that has the capacity to encapsulate pirarubicin via electrostatic and hydrophobic interactions. These interactions can be disrupted by the acidic tumor microenvironment. Self-assembly of P51 and pirarubicin (P51-THP NPs) results into stable spherical nanoparticles in a single step. We have demonstrated that the acid environment, DTT, and MMP-2 stimulate the release of pirarubicin from P51-THP NPs and, more importantly, the efficiency of drug release is markedly increased when all three release triggers are present. In addition, more effective tumor targeting, antitumor effect, and reduced systemic toxicity of P51-THP NPs have been confirmed by in vitro and in vivo results.
Collapse
Affiliation(s)
- Xinglu Jiang
- Medical School of Southeast University, Nanjing 210009, People's Republic of China; Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, People's Republic of China
| | - Xiaobo Fan
- Medical School of Southeast University, Nanjing 210009, People's Republic of China
| | - Wei Xu
- Medical School of Southeast University, Nanjing 210009, People's Republic of China
| | - Chenggui Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, People's Republic of China
| | - Hailu Wu
- Medical School of Southeast University, Nanjing 210009, People's Republic of China; Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, People's Republic of China
| | - Rui Zhang
- Medical School of Southeast University, Nanjing 210009, People's Republic of China
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
42
|
Dong Y, Liao H, Fu H, Yu J, Guo Q, Wang Q, Duan Y. pH-Sensitive Shell-Core Platform Block DNA Repair Pathway To Amplify Irreversible DNA Damage of Triple Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2019; 11:38417-38428. [PMID: 31556584 DOI: 10.1021/acsami.9b12140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Triple negative breast cancer (TNBC) is insensitive to either chemotherapy or endocrine therapy because of the powerful DNA reparation and the negative expression of surface antigens, which urgently claims for an effective approach to improve the prognosis. Herein, DNA repair blocker BRCA1 small interfering RNA (siRNA) was introduced with cisplatin (Pt) into the elaborately designed pH-sensitive shell-core platform to enhance the chemotherapeutic treatment effect by silencing the DNA repair related gene. In this platform, BRCA1 siRNA and Pt prodrug (Pro-Pt) were separately encapsulated in the porous outer shell and hydrophobic inner core with extremely high encapsulation efficiency and stability effectively preventing them from degradation during circulation. Suitable size and urokinase plasminogen activator analogues (uPA) with high affinity for the uPA receptor (uPAR) realized an excellent dual passive and active tumor targeting ability. Moreover, the exposed PEG hydrophilic chain prevented the nanoparticles (NPs) from precipitating by serum protein or inactivating by nuclease in the blood cycle. Most importantly, the degradable CaP (calcium ions and phosphate ions) shell with smart pH sensitivity would dissipate from NPs in the lysosomes to burst the lysosome membranes so as to guarantee the lysosomal escape and the sequential release of the siRNA and Pro-Pt where the BRCA1 siRNA blocked the DNA repairing pathway followed by reducing Pro-Pt to Pt for irreversible DNA damage. Hence, the uPA-SP@CaP NPs provided a promising strategy for high-efficiency treatment of TNBC along with bringing new hope for more patients.
Collapse
Affiliation(s)
- Yang Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200032 , China
| | - Hongze Liao
- Marine Drugs Research Center, Department of Pharmacy, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200127 , China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200032 , China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200032 , China
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200032 , China
| | - Qi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200032 , China
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering , East China University of Science and Technology , Shanghai 200237 , China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai 200032 , China
| |
Collapse
|
43
|
Arroyo‐Crespo JJ, Armiñán A, Charbonnier D, Deladriere C, Palomino‐Schätzlein M, Lamas‐Domingo R, Forteza J, Pineda‐Lucena A, Vicent MJ. Characterization of triple-negative breast cancer preclinical models provides functional evidence of metastatic progression. Int J Cancer 2019; 145:2267-2281. [PMID: 30860605 PMCID: PMC6767480 DOI: 10.1002/ijc.32270] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/25/2019] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer (TNBC), an aggressive, metastatic and recurrent breast cancer (BC) subtype, currently suffers from a lack of adequately described spontaneously metastatic preclinical models that faithfully reproduce the clinical scenario. We describe two preclinical spontaneously metastatic TNBC orthotopic murine models for the development of advanced therapeutics: an immunodeficient human MDA-MB-231-Luc model and an immunocompetent mouse 4T1 model. Furthermore, we provide a broad range of multifactorial analysis for both models that could provide relevant information for the development of new therapies and diagnostic tools. Our comparisons uncovered differential growth rates, stromal arrangements and metabolic profiles in primary tumors, and the presence of cancer-associated adipocyte infiltration in the MDA-MB-231-Luc model. Histopathological studies highlighted the more rapid metastatic spread to the lungs in the 4T1 model following a lymphatic route, while we observed both homogeneous (MDA-MB-231-Luc) and heterogeneous (4T1) metastatic spread to axillary lymph nodes. We encountered unique metabolomic signatures in each model, including crucial amino acids and cell membrane components. Hematological analysis demonstrated severe leukemoid and lymphoid reactions in the 4T1 model with the partial reestablishment of immune responses in the immunocompromised MDA-MB-231-Luc model. Additionally, we discovered β-immunoglobulinemia and increased basal levels of G-CSF correlating with a metastatic switch, with G-CSF also promoting extramedullary hematopoiesis (both models) and causing hepatosplenomegaly (4T1 model). Overall, we believe that the characterization of these preclinical models will foster the development of advanced therapeutic strategies for TNBC treatment, especially for the treatment of patients presenting both, primary tumors and metastatic spread.
Collapse
Affiliation(s)
- Juan J. Arroyo‐Crespo
- Polymer Therapeutics LaboratoryCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - Ana Armiñán
- Polymer Therapeutics LaboratoryCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - David Charbonnier
- Polymer Therapeutics LaboratoryCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
- Screening Platform, Centro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - Coralie Deladriere
- Polymer Therapeutics LaboratoryCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - Martina Palomino‐Schätzlein
- Joint Research Unit in Clinical MetabolomicsCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - Rubén Lamas‐Domingo
- Joint Research Unit in Clinical MetabolomicsCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - Jerónimo Forteza
- Unidad Mixta Centro de Investigación Príncipe Felipe‐Instituto Valenciano de PatologíaCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| | - Antonio Pineda‐Lucena
- Joint Research Unit in Clinical MetabolomicsCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
- Drug Discovery UnitInstituto de Investigación Sanitaria La FeAvda. Fernando Abril Martorell, 106, 46026ValenciaSpain
| | - María J. Vicent
- Polymer Therapeutics LaboratoryCentro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
- Screening Platform, Centro de Investigación Príncipe FelipeAv. Eduardo Primo Yúfera 3Valencia, 46012Spain
| |
Collapse
|
44
|
Jacobs J, Pavlović D, Prydderch H, Moradi MA, Ibarboure E, Heuts JPA, Lecommandoux S, Heise A. Polypeptide Nanoparticles Obtained from Emulsion Polymerization of Amino Acid N-Carboxyanhydrides. J Am Chem Soc 2019; 141:12522-12526. [PMID: 31348858 DOI: 10.1021/jacs.9b06750] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Polypeptide nanoparticles were obtained by the miniemulsion polymerization of S-(o-nitrobenzyl)-l-cysteine (NBC) N-carboxyanhydride (NCA). Through process optimization, reaction conditions were identified that allowed the polymerization of the water sensitive NCA to yield nanoparticles of about 220 nm size. Subsequent UV-irradiation of the nanoparticle emulsions caused the in situ removal of the nitrobenzyl group and particle cross-linking through disulfide bond formation accompanied by the shrinkage of the particles.
Collapse
Affiliation(s)
- Jaco Jacobs
- School of Chemical Sciences , Dublin City University , Glasnevin, Dublin 9 , Ireland
| | - Dražen Pavlović
- Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| | - Hannah Prydderch
- Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| | - Mohammad-Amin Moradi
- Department of Chemical Engineering & Chemistry and Institute for Complex Molecular Systems , Eindhoven University of Technology , PO Box 513, 5600 MB Eindhoven , The Netherlands
| | - Emmanuel Ibarboure
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629 , F-33600 Pessac , France
| | - Johan P A Heuts
- Department of Chemical Engineering & Chemistry and Institute for Complex Molecular Systems , Eindhoven University of Technology , PO Box 513, 5600 MB Eindhoven , The Netherlands
| | | | - Andreas Heise
- Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| |
Collapse
|
45
|
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]
|
46
|
Zhang H, Zhang X, Ren Y, Cao F, Hou L, Zhang Z. An in situ microenvironmental nano-regulator to inhibit the proliferation and metastasis of 4T1 tumor. Theranostics 2019; 9:3580-3594. [PMID: 31281499 PMCID: PMC6587164 DOI: 10.7150/thno.33141] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/19/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor microenvironment, such as hypoxia and presence of immune cells, plays a critical role in cancer initiation, growth as well as progression, and seriously affects antitumor effect. Accordingly, we constructed a kind of multifunctional nanoparticles (NPs) with macrophage transformation and oxygen (O2) generation characteristics, to regulate the tumor microenvironment. Methods: In this study, we synthesized mesoporous Prussian blue (MPB) NPs with low molecular weight hyaluronic acid (LMWHA) surface modification (LMWHA-MPB), and discovered that LMWHA-MPB could be used as an in situ macrophages converter and O2 generator. Results:In vitro results showed after uptake by M2 macrophages, LMWHA-MPB displayed the potential in remodeling tumor-associated macrophages (TAMs) phenotype (pro-tumor M2→anti-tumor M1), and anti-metastatic effect on 4T1 cells. Furthermore, in vivo visualized near-infrared (NIR) imaging data proved IR783 labeled LMWHA-MPB NPs could selectively accumulate in tumor sites. Then plenty of O2 generated to alleviate tumor hypoxia via catalytic decomposition of endogenous hydrogen peroxide (H2O2). Based on these outstanding characteristics, LMWHA-MPB NPs were adopted as multifunctional nanocarriers to load sonosensitizer hematoporphyrin monomethyl ether (HMME) for O2 self-provided sonodynamic therapy (SDT). In vivo anti-tumor results showed LMWHA-MPB/HMME could effectively inhibit the proliferation and metastasis of 4T1 tumors by improving tumor microenvironment. Conclusion: The multifunctional NPs can be used as in situ microenvironmental nano-regulators to inhibit the proliferation and metastasis of 4T1 tumor.
Collapse
Affiliation(s)
- Huijuan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China
| | - Xiaoge Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanping Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fang Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, China
| |
Collapse
|
47
|
Yang XL, Xing X, Li J, Liu YH, Wang N, Yu XQ. Enzymatic synthesis of selenium-containing amphiphilic aliphatic polycarbonate as an oxidation-responsive drug delivery vehicle. RSC Adv 2019; 9:6003-6010. [PMID: 35517302 PMCID: PMC9060885 DOI: 10.1039/c8ra10282a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/04/2019] [Indexed: 11/30/2022] Open
Abstract
Although functional aliphatic polycarbonates (APCs) have attracted prominent research interest as stimuli-responsive biomaterials, the majority of functional APCs are fabricated by detrimental organometallic catalysts or organo-catalysts. Herein, a facile synthetic strategy based on enzymatic polymerization was developed to construct a selenium-containing amphiphilic aliphatic polycarbonate (mPEG-b-CMP45). Specifically, the selenium in its backbone framework underwent a hydrophobic–hydrophilic transition upon exposure to the abnormal ROS level of the tumor, thus providing a promising platform for ROS-triggered drug release. This amphiphilic mPEG-b-CMP45 efficiently encapsulated doxorubicin (DOX) via self-assembly in aqueous solution and showed an excellent ability to regulate the release of DOX in response to H2O2 at biologically relevant concentrations (100 μM). These DOX-loaded nanoparticles could easily be internalized into U87 cells and possess the inherent antitumor properties of DOX, while they exhibited much lower cytotoxicity in normal cells HL-7702. Moreover, in many cases, the introduction of selenium caused high cytotoxicity of the materials, but the cytotoxicity results in HL-7702 cells demonstrated the good biocompatibility of mPEG-b-CMP45. These collective data suggested the potential use of mPEG-b-CMP45 as a biocompatible and smart drug delivery vehicle. A facile synthetic strategy based on enzymatic polymerization was developed to construct a ROS-responsive polycarbonate served as biocompatible drug vehicle.![]()
Collapse
Affiliation(s)
- Xian-Ling Yang
- Key Laboratory of Green Chemistry & Technology
- Ministry of Education
- College of Chemistry
- Sichuan University
- Chengdu 610064
| | - Xiu Xing
- Key Laboratory of Green Chemistry & Technology
- Ministry of Education
- College of Chemistry
- Sichuan University
- Chengdu 610064
| | - Jun Li
- Key Laboratory of Green Chemistry & Technology
- Ministry of Education
- College of Chemistry
- Sichuan University
- Chengdu 610064
| | - Yan-Hong Liu
- Key Laboratory of Green Chemistry & Technology
- Ministry of Education
- College of Chemistry
- Sichuan University
- Chengdu 610064
| | - Na Wang
- Key Laboratory of Green Chemistry & Technology
- Ministry of Education
- College of Chemistry
- Sichuan University
- Chengdu 610064
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry & Technology
- Ministry of Education
- College of Chemistry
- Sichuan University
- Chengdu 610064
| |
Collapse
|
48
|
Maso K, Grigoletto A, Vicent MJ, Pasut G. Molecular platforms for targeted drug delivery. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:1-50. [DOI: 10.1016/bs.ircmb.2019.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|