1
|
Cirillo G, Cappello AR, Curcio M, Fiorillo M, Frattaruolo L, Avena P, Scorzafave L, Dolce V, Nicoletta FP, Iemma F. Novel CD44-Targeted Albumin Nanoparticles: An Innovative Approach to Improve Breast Cancer Treatment. Int J Mol Sci 2024; 25:10560. [PMID: 39408889 PMCID: PMC11477043 DOI: 10.3390/ijms251910560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
This study introduces novel CD44-targeted and redox-responsive nanoparticles (FNPs), proposed as doxorubicin (DOX) delivery devices for breast cancer. A cationized and redox-responsive Human Serum Albumin derivative was synthesized by conjugating Human Serum Albumin with cystamine moieties and then ionically complexing it with HA. The suitability of FNPs for cancer therapy was assessed through physicochemical measurements of size distribution (mean diameter of 240 nm), shape, and zeta potential (15.4 mV). Nanoparticles possessed high DOX loading efficiency (90%) and were able to trigger the drug release under redox conditions of the tumor environment (55% release after 2 h incubation). The use of the carrier increased the cytotoxic effect of DOX by targeting the CD44 protein. It was shown that, upon loading, the cytotoxic effect of DOX was enhanced in relation to CD44 protein expression in both 2D and 3D models. DOX@FNPs significantly decrease cellular metabolism by reducing both oxygen consumption and extracellular acidification rates. Moreover, they decrease the expression of proteins involved in the oxidative phosphorylation pathway, consequently reducing cellular viability and motility, as well as breast cancer stem cells and spheroid formation, compared to free DOX. This new formulation could become pioneering in reducing chemoresistance phenomena and increasing the specificity of DOX in breast cancer patients.
Collapse
Affiliation(s)
| | | | - Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy; (G.C.); (A.R.C.); (L.F.); (P.A.); (L.S.); (F.P.N.); (F.I.)
| | - Marco Fiorillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy; (G.C.); (A.R.C.); (L.F.); (P.A.); (L.S.); (F.P.N.); (F.I.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Kumar K, Fachet M, Hoeschen C. High-Spatial-Resolution Benchtop X-ray Fluorescence Imaging through Bragg-Diffraction-Based Focusing with Bent Mosaic Graphite Crystals: A Simulation Study. Int J Mol Sci 2024; 25:4733. [PMID: 38731956 PMCID: PMC11083219 DOI: 10.3390/ijms25094733] [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: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
X-ray fluorescence imaging (XFI) can localize diagnostic or theranostic entities utilizing nanoparticle (NP)-based probes at high resolution in vivo, in vitro, and ex vivo. However, small-animal benchtop XFI systems demonstrating high spatial resolution (variable from sub-millimeter to millimeter range) in vivo are still limited to lighter elements (i.e., atomic number Z≤45). This study investigates the feasibility of focusing hard X-rays from solid-target tubes using ellipsoidal lens systems composed of mosaic graphite crystals with the aim of enabling high-resolution in vivo XFI applications with mid-Z (42≤Z≤64) elements. Monte Carlo simulations are performed to characterize the proposed focusing-optics concept and provide quantitative predictions of the XFI sensitivity, in silico tumor-bearing mice models loaded with palladium (Pd) and barium (Ba) NPs. Based on simulation results, the minimum detectable total mass of PdNPs per scan position is expected to be on the order of a few hundred nanograms under in vivo conform conditions. PdNP masses as low as 150 ng to 50 ng could be detectable with a resolution of 600 μm when imaging abdominal tumor lesions across a range of low-dose (0.8 μGy) to high-dose (8 μGy) exposure scenarios. The proposed focusing-optics concept presents a potential step toward realizing XFI with conventional X-ray tubes for high-resolution applications involving interesting NP formulations.
Collapse
Affiliation(s)
| | - Melanie Fachet
- Chair of Medical Systems Technology, Institute for Medical Technology, Faculty of Electrical Engineering and Information Technology, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.K.)
| | | |
Collapse
|
3
|
Rassomakhina NV, Ryazanova AY, Likhov AR, Bruskin SA, Maloshenok LG, Zherdeva VV. Tumor Organoids: The Era of Personalized Medicine. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S127-S147. [PMID: 38621748 DOI: 10.1134/s0006297924140086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 04/17/2024]
Abstract
The strategies of future medicine are aimed to modernize and integrate quality approaches including early molecular-genetic profiling, identification of new therapeutic targets and adapting design for clinical trials, personalized drug screening (PDS) to help predict and individualize patient treatment regimens. In the past decade, organoid models have emerged as an innovative in vitro platform with the potential to realize the concept of patient-centered medicine. Organoids are spatially restricted three-dimensional clusters of cells ex vivo that self-organize into complex functional structures through genetically programmed determination, which is crucial for reconstructing the architecture of the primary tissue and organs. Currently, there are several strategies to create three-dimensional (3D) tumor systems using (i) surgically resected patient tissue (PDTOs, patient-derived tumor organoids) or (ii) single tumor cells circulating in the patient's blood. Successful application of 3D tumor models obtained by co-culturing autologous tumor organoids (PDTOs) and peripheral blood lymphocytes have been demonstrated in a number of studies. Such models simulate a 3D tumor architecture in vivo and contain all cell types characteristic of this tissue, including immune system cells and stem cells. Components of the tumor microenvironment, such as fibroblasts and immune system cells, affect tumor growth and its drug resistance. In this review, we analyzed the evolution of tumor models from two-dimensional (2D) cell cultures and laboratory animals to 3D tissue-specific tumor organoids, their significance in identifying mechanisms of antitumor response and drug resistance, and use of these models in drug screening and development of precision methods in cancer treatment.
Collapse
Affiliation(s)
- Natalia V Rassomakhina
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Alexandra Yu Ryazanova
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Astemir R Likhov
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Sergey A Bruskin
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Liliya G Maloshenok
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Victoria V Zherdeva
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia.
| |
Collapse
|
4
|
Fernandez JL, Årbogen S, Sadeghinia MJ, Haram M, Snipstad S, Torp SH, Einen C, Mühlenpfordt M, Maardalen M, Vikedal K, Davies CDL. A Comparative Analysis of Orthotopic and Subcutaneous Pancreatic Tumour Models: Tumour Microenvironment and Drug Delivery. Cancers (Basel) 2023; 15:5415. [PMID: 38001675 PMCID: PMC10670202 DOI: 10.3390/cancers15225415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a challenging malignancy, mainly due to its resistance to chemotherapy and its complex tumour microenvironment characterised by stromal desmoplasia. There is a need for new strategies to improve the delivery of drugs and therapeutic response. Relevant preclinical tumour models are needed to test potential treatments. This paper compared orthotopic and subcutaneous PDAC tumour models and their suitability for drug delivery studies. A novel aspect was the broad range of tumour properties that were studied, including tumour growth, histopathology, functional vasculature, perfusion, immune cell infiltration, biomechanical characteristics, and especially the extensive analysis of the structure and the orientation of the collagen fibres in the two tumour models. The study unveiled new insights into how these factors impact the uptake of a fluorescent model drug, the macromolecule called 800CW. While the orthotopic model offered a more clinically relevant microenvironment, the subcutaneous model offered advantages for drug delivery studies, primarily due to its reproducibility, and it was characterised by a more efficient drug uptake facilitated by its collagen organisation and well-perfused vasculature. The tumour uptake seemed to be influenced mainly by the structural organisation and the alignment of the collagen fibres and perfusion. Recognising the diverse characteristics of these models and their multifaceted impacts on drug delivery is crucial for designing clinically relevant experiments and improving our understanding of pancreatic cancer biology.
Collapse
Affiliation(s)
- Jessica Lage Fernandez
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
| | - Sara Årbogen
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
| | - Mohammad Javad Sadeghinia
- Department of Structural Engineering, Norwegian University of Science and Technology, 7491 Trondheim, Norway;
| | - Margrete Haram
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (M.H.); (S.H.T.)
- Cancer Clinic, St. Olavs Hospital, 7006 Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
- Cancer Clinic, St. Olavs Hospital, 7006 Trondheim, Norway
| | - Sverre Helge Torp
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (M.H.); (S.H.T.)
- Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Caroline Einen
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
- EXACT Therapeutics, 0581 Oslo, Norway
| | - Matilde Maardalen
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
- Department of Engineering Science, University of Oxford, Oxford OX1 3NP, UK
| | - Krister Vikedal
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
- Department of Microbiology, Oslo University Hospital, 0424 Oslo, Norway
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway; (S.Å.); (S.S.); (M.M.); (K.V.); (C.d.L.D.)
| |
Collapse
|
5
|
Jiang K, Yu Y, Qiu W, Tian K, Guo Z, Qian J, Lu H, Zhan C. Protein corona on brain targeted nanocarriers: Challenges and prospects. Adv Drug Deliv Rev 2023; 202:115114. [PMID: 37827336 DOI: 10.1016/j.addr.2023.115114] [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: 09/04/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Safe and efficient medical therapy for brain diseases is still an unmet clinical need due to various barriers represented by the blood-brain barrier. Well-designed brain targeted nanocarriers are potential solutions for enhanced brain drug delivery; however, the complicated in vivo process attenuates performance of nanocarriers, which severely hampers clinical translation. The formation of protein corona (PC) is inevitable for nanocarriers circulation and transport in biofluids, acting as an important factor to regulate in vivo performance of nanocarriers. In this review, the reported strategies have been retrospected for better understanding current situation in developing brain targeted nanocarriers. The interplay between brain targeted nanocarriers and plasma proteins is emphasized to comprehend how the nanocarriers adsorb proteins by certain synthetic identity, and following regulations on in vivo performance of nanocarriers. More importantly, the mainstream methods to promote efficiency of nanocarriers by regulating PC, defined as in vitro functionalization and in vivo functionalization strategies, are also discussed. Finally, viewpoints about future development of brain targeted nanocarriers according to the understanding on nanocarriers-PC interaction are proposed.
Collapse
Affiliation(s)
- Kuan Jiang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Yifei Yu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Wei Qiu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Kaisong Tian
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Zhiwei Guo
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Jun Qian
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201399, PR China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201399, PR China.
| | - Changyou Zhan
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China; Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201399, PR China.
| |
Collapse
|
6
|
Domínguez-Oliva A, Hernández-Ávalos I, Martínez-Burnes J, Olmos-Hernández A, Verduzco-Mendoza A, Mota-Rojas D. The Importance of Animal Models in Biomedical Research: Current Insights and Applications. Animals (Basel) 2023; 13:ani13071223. [PMID: 37048478 PMCID: PMC10093480 DOI: 10.3390/ani13071223] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/19/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Animal research is considered a key element in advance of biomedical science. Although its use is controversial and raises ethical challenges, the contribution of animal models in medicine is essential for understanding the physiopathology and novel treatment alternatives for several animal and human diseases. Current pandemics’ pathology, such as the 2019 Coronavirus disease, has been studied in primate, rodent, and porcine models to recognize infection routes and develop therapeutic protocols. Worldwide issues such as diabetes, obesity, neurological disorders, pain, rehabilitation medicine, and surgical techniques require studying the process in different animal species before testing them on humans. Due to their relevance, this article aims to discuss the importance of animal models in diverse lines of biomedical research by analyzing the contributions of the various species utilized in science over the past five years about key topics concerning human and animal health.
Collapse
Affiliation(s)
- Adriana Domínguez-Oliva
- Master’s Program in Agricultural and Livestock Sciences [Maestría en Ciencias Agropecuarias], Xochimilco Campus, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán 54714, Mexico
| | - Julio Martínez-Burnes
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Victoria City 87000, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology—Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis, Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology—Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis, Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
- Correspondence:
| |
Collapse
|
7
|
Lee T, Kim KS, Na K. Intracellular pH-Regulating Nanoparticles to Improve Anticancer Drug Efficacy for Cancer Treatment. Biomacromolecules 2022; 23:4786-4794. [PMID: 36223489 DOI: 10.1021/acs.biomac.2c00952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Here, we describe an intracellular pH-regulating nanoparticle (IPRN), coencapsulated with chemosensitizers and anticancer agents for effective and safe cancer treatment. IPRN contains a tubulysin derivative (TUB), a hydrophobic anticancer drug, and pantoprazole (PTZ), a hydrophilic proton-pump inhibitor. IPRN with a size of 62 nm has an anionic surface charge and is stable for at least two weeks under storage conditions, though PTZ and TUB encapsulated in IPRN showed different drug release patterns. PTZ was released before TUB, controlling the cancer's intracellular pH, maintaining a pH at which TUB can work well. The encapsulated PTZ increased the pH of endolysosomes and inhibited ion trapping, with TUB ionization, thereby exhibiting increased cytotoxicity compared with free TUB observed in various cancer cell lines, such as human liver adenocarcinoma, human glioblastoma, and human pancreatic carcinoma. IPRN exhibited a 1.9-fold improved tumor growth inhibitory effect in a human liver adenocarcinoma-bearing mouse model, while minimizing the hepatotoxicity of free TUB. Thus, nanomedicines that contain both a chemosensitizer and an anticancer agent, such as IPRN, are expected to be next-generation anticancer agents that reduce the side effects of anticancer drugs and increase the therapeutic effect.
Collapse
Affiliation(s)
- Taebum Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do14662, Republic of Korea.,Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do14662, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do14662, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do14662, Republic of Korea.,Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do14662, Republic of Korea
| |
Collapse
|
8
|
Aboeleneen SB, Scully MA, Harris JC, Sterin EH, Day ES. Membrane-wrapped nanoparticles for photothermal cancer therapy. NANO CONVERGENCE 2022; 9:37. [PMID: 35960404 PMCID: PMC9373884 DOI: 10.1186/s40580-022-00328-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/27/2022] [Indexed: 05/31/2023]
Abstract
Cancer is a global health problem that needs effective treatment strategies. Conventional treatments for solid-tumor cancers are unsatisfactory because they cause unintended harm to healthy tissues and are susceptible to cancer cell resistance. Nanoparticle-mediated photothermal therapy is a minimally invasive treatment for solid-tumor cancers that has immense promise as a standalone therapy or adjuvant to other treatments like chemotherapy, immunotherapy, or radiotherapy. To maximize the success of photothermal therapy, light-responsive nanoparticles can be camouflaged with cell membranes to endow them with unique biointerfacing capabilities that reduce opsonization, prolong systemic circulation, and improve tumor delivery through enhanced passive accumulation or homotypic targeting. This ensures a sufficient dose of photoresponsive nanoparticles arrives at tumor sites to enable their complete thermal ablation. This review summarizes the state-of-the-art in cell membrane camouflaged nanoparticles for photothermal cancer therapy and provides insights to the path forward for clinical translation.
Collapse
Affiliation(s)
| | | | - Jenna C Harris
- Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - Eric H Sterin
- Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Emily S Day
- Biomedical Engineering, University of Delaware, Newark, DE, USA.
- Materials Science and Engineering, University of Delaware, Newark, DE, USA.
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, USA.
| |
Collapse
|
9
|
Cancer nanomedicine: A step towards improving the drug delivery and enhanced efficacy of chemotherapeutic drugs. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Đorđević S, Gonzalez MM, Conejos-Sánchez I, Carreira B, Pozzi S, Acúrcio RC, Satchi-Fainaro R, Florindo HF, Vicent MJ. Current hurdles to the translation of nanomedicines from bench to the clinic. Drug Deliv Transl Res 2022; 12:500-525. [PMID: 34302274 PMCID: PMC8300981 DOI: 10.1007/s13346-021-01024-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
The field of nanomedicine has significantly influenced research areas such as drug delivery, diagnostics, theranostics, and regenerative medicine; however, the further development of this field will face significant challenges at the regulatory level if related guidance remains unclear and unconsolidated. This review describes those features and pathways crucial to the clinical translation of nanomedicine and highlights considerations for early-stage product development. These include identifying those critical quality attributes of the drug product essential for activity and safety, appropriate analytical methods (physical, chemical, biological) for characterization, important process parameters, and adequate pre-clinical models. Additional concerns include the evaluation of batch-to-batch consistency and considerations regarding scaling up that will ensure a successful reproducible manufacturing process. Furthermore, we advise close collaboration with regulatory agencies from the early stages of development to assure an aligned position to accelerate the development of future nanomedicines.
Collapse
Affiliation(s)
- Snežana Đorđević
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - María Medel Gonzalez
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel.
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal.
| | - María J Vicent
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain.
| |
Collapse
|
11
|
Ramos TI, Villacis-Aguirre CA, López-Aguilar KV, Santiago Padilla L, Altamirano C, Toledo JR, Santiago Vispo N. The Hitchhiker's Guide to Human Therapeutic Nanoparticle Development. Pharmaceutics 2022; 14:247. [PMID: 35213980 PMCID: PMC8879439 DOI: 10.3390/pharmaceutics14020247] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 02/06/2023] Open
Abstract
Nanomedicine plays an essential role in developing new therapies through novel drug delivery systems, diagnostic and imaging systems, vaccine development, antibacterial tools, and high-throughput screening. One of the most promising drug delivery systems are nanoparticles, which can be designed with various compositions, sizes, shapes, and surface modifications. These nanosystems have improved therapeutic profiles, increased bioavailability, and reduced the toxicity of the product they carry. However, the clinical translation of nanomedicines requires a thorough understanding of their properties to avoid problems with the most questioned aspect of nanosystems: safety. The particular physicochemical properties of nano-drugs lead to the need for additional safety, quality, and efficacy testing. Consequently, challenges arise during the physicochemical characterization, the production process, in vitro characterization, in vivo characterization, and the clinical stages of development of these biopharmaceuticals. The lack of a specific regulatory framework for nanoformulations has caused significant gaps in the requirements needed to be successful during their approval, especially with tests that demonstrate their safety and efficacy. Researchers face many difficulties in establishing evidence to extrapolate results from one level of development to another, for example, from an in vitro demonstration phase to an in vivo demonstration phase. Additional guidance is required to cover the particularities of this type of product, as some challenges in the regulatory framework do not allow for an accurate assessment of NPs with sufficient evidence of clinical success. This work aims to identify current regulatory issues during the implementation of nanoparticle assays and describe the major challenges that researchers have faced when exposing a new formulation. We further reflect on the current regulatory standards required for the approval of these biopharmaceuticals and the requirements demanded by the regulatory agencies. Our work will provide helpful information to improve the success of nanomedicines by compiling the challenges described in the literature that support the development of this novel encapsulation system. We propose a step-by-step approach through the different stages of the development of nanoformulations, from their design to the clinical stage, exemplifying the different challenges and the measures taken by the regulatory agencies to respond to these challenges.
Collapse
Affiliation(s)
- Thelvia I. Ramos
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, Concepción 4070386, Chile; (T.I.R.); (C.A.V.-A.)
- Grupo de Investigación en Sanidad Animal y Humana (GISAH), Carrera Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas—ESPE, Sangolquí 171103, Ecuador
| | - Carlos A. Villacis-Aguirre
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, Concepción 4070386, Chile; (T.I.R.); (C.A.V.-A.)
| | - Katherine V. López-Aguilar
- Carrera Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas—ESPE, Sangolquí 171103, Ecuador;
| | | | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso 2362803, Chile;
- Centro Regional de Estudios en Alimentos Saludables, Av. Universidad 330, Placilla, Sector Curauma, Valparaíso 2340000, Chile
| | - Jorge R. Toledo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, Concepción 4070386, Chile; (T.I.R.); (C.A.V.-A.)
| | - Nelson Santiago Vispo
- School of Biological Sciences and Engineering, Yachay Tech University, Hda. San José s/n y Proyecto Yachay, Urcuquí 100119, Ecuador
| |
Collapse
|
12
|
Miatmoko A, Mianing EA, Sari R, Hendradi E. Nanoparticles use for Delivering Ursolic Acid in Cancer Therapy: A Scoping Review. Front Pharmacol 2022; 12:787226. [PMID: 35002719 PMCID: PMC8740088 DOI: 10.3389/fphar.2021.787226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Ursolic acid is a natural pentacyclic triterpenoid that exerts a potent anticancer effect. Furthermore, it is classified as a BCS class IV compound possessing low permeability and water solubility, consequently demonstrating limited bioavailability in addition to low therapeutic effectiveness. Nanoparticles are developed to modify the physical characteristics of drug and can often be produced in the range of 30–200 nm, providing highly effective cancer therapy due to the Enhanced Permeation and Retention (EPR) Effect. This study aims to provide a review of the efficacy and safety of various types of Ursolic Acid-loading nanoparticles within the setting of preclinical and clinical anticancer studies. This literature study used scoping review method, where the extracted data must comply with the journal inclusion criteria of within years of 2010–2020. The identification stage produced 237 suitable articles. Duplicate screening was then conducted followed by the initial selection of 18 articles that had been reviewed and extracted for data analysis. Based on this review, the use of nanoparticles can be seen to increase the anticancer efficacy of Ursolic Acid in terms of several parameters including pharmacokinetic data, survival rates and inhibition rates, as well as the absence of serious toxicity in preclinical and clinical trials in terms of several parameters including body weight, blood clinical chemistry, and organ histipathology. Based on this review, the use of nanoparticles has been able to increase the anticancer efficacy of Ursolic Acid, as well as show the absence of serious toxicity in preclinical and clinical trials. Evenmore, the liposome carrier provides development data that has reached the clinical trial phase I. The use of nanoparticle provides high potential for Ursolic Acid delivery in cancer therapy.
Collapse
Affiliation(s)
- Andang Miatmoko
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia.,Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, Indonesia
| | - Ester Adelia Mianing
- Study Program of Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Retno Sari
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Esti Hendradi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
13
|
Diagnostic Ability of Methods Depicting Distress of Tumor-Bearing Mice. Animals (Basel) 2021; 11:ani11082155. [PMID: 34438613 PMCID: PMC8388504 DOI: 10.3390/ani11082155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Experiments on animals can provide important information for improving the life expectancy and life quality of patients. At the same time, the welfare of these animals is a growing public concern. Therefore, many laws and international guidelines were established with the goal of minimizing the harm inflicted on these animals. A prerequisite of improving animal welfare is to correctly measure how much distress the experiments cause to these animals. However, it is often unknown as to which methods are appropriate to assess distress. Mice bearing subcutaneous tumors are the most frequently used animal model to study the therapeutic effects of drugs. We evaluated if body weight, faecal corticosterone metabolites concentration, burrowing activity and a distress score were capable of differentiating between mice before cancer cell injection and mice bearing large tumors. We observed that only adjusted body weight change and faecal corticosterone metabolites concentration were capable of measuring distress caused by large subcutaneous tumors. Therefore, these two methods are appropriate to assess the welfare of mice with subcutaneous tumors. This knowledge provides a solid basis to optimize animal welfare in future studies. For example, both methods can define the ideal time point when an experiment should end by finding a good compromise between minimal distress for the animals and maximal knowledge gain for mankind. Abstract Subcutaneous tumor models in mice are the most commonly used experimental animal models in cancer research. To improve animal welfare and the quality of scientific studies, the distress of experimental animals needs to be minimized. For this purpose, one must assess the diagnostic ability of readout parameters to evaluate distress. In this study, we evaluated different noninvasive readout parameters such as body weight change, adjusted body weight change, faecal corticosterone metabolites concentration, burrowing activity and a distress score by utilising receiver operating characteristic curves. Eighteen immunocompromised NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice were used for this study; half were subcutaneously injected with A-375 cells (human malignant melanoma cells) that resulted in large tumors. The remaining mice were inoculated with SCL-2 cells (cutaneous squamous cell carcinoma cells), which resulted in small tumors. The adjusted body weight and faecal corticosterone metabolites concentration had a high diagnostic ability in distinguishing between mice before cancer cell injection and mice bearing large tumors. All other readout parameters had a low diagnostic ability. These results suggest that adjusted body weight and faecal corticosterone metabolites are useful to depict the distress of mice bearing large subcutaneous tumors.
Collapse
|
14
|
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
|
15
|
Torresan V, Guadagnini A, Badocco D, Pastore P, Muñoz Medina GA, Fernàndez van Raap MB, Postuma I, Bortolussi S, Bekić M, Čolić M, Gerosa M, Busato A, Marzola P, Amendola V. Biocompatible Iron-Boron Nanoparticles Designed for Neutron Capture Therapy Guided by Magnetic Resonance Imaging. Adv Healthc Mater 2021; 10:e2001632. [PMID: 33369251 DOI: 10.1002/adhm.202001632] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/04/2020] [Indexed: 12/13/2022]
Abstract
The combination of multiple functions in a single nanoparticle (NP) represents a key advantage of nanomedicine compared to traditional medical approaches. This is well represented by radiotherapy in which the dose of ionizing radiation should be calibrated on sensitizers biodistribution. Ideally, this is possible when the drug acts both as radiation enhancer and imaging contrast agent. Here, an easy, one-step, laser-assisted synthetic procedure is used to generate iron-boron (Fe-B) NPs featuring the set of functions required to assist neutron capture therapy (NCT) with magnetic resonance imaging. The Fe-B NPs exceed by three orders of magnitude the payload of boron isotopes contained in clinical sensitizers. The Fe-B NPs have magnetic properties of interest also for magnetophoretic accumulation in tissues and magnetic hyperthermia to assist drug permeation in tissues. Besides, Fe-B NPs are biocompatible and undergo slow degradation in the lysosomal environment that facilitates in vivo clearance through the liver-spleen-kidneys pathway. Overall, the Fe-B NPs represent a new promising tool for future exploitation in magnetic resonance imaging-guided boron NCT at higher levels of efficacy and tolerability.
Collapse
Affiliation(s)
- Veronica Torresan
- Department of Chemical Sciences University of Padova Via Marzolo 1 Padova I‐35131 Italy
| | - Andrea Guadagnini
- Department of Chemical Sciences University of Padova Via Marzolo 1 Padova I‐35131 Italy
| | - Denis Badocco
- Department of Chemical Sciences University of Padova Via Marzolo 1 Padova I‐35131 Italy
| | - Paolo Pastore
- Department of Chemical Sciences University of Padova Via Marzolo 1 Padova I‐35131 Italy
| | - Guillermo Arturo Muñoz Medina
- Physics Institute of La Plata (IFLP‐CONICET) Physics Department, Faculty of Exact Sciences National University of La Plata La Plata 1900 Argentina
| | - Marcela B. Fernàndez van Raap
- Physics Institute of La Plata (IFLP‐CONICET) Physics Department, Faculty of Exact Sciences National University of La Plata La Plata 1900 Argentina
| | - Ian Postuma
- INFN (National Institute of Nuclear Physics) Pavia Via Bassi 6 Pavia 27100 Italy
| | - Silva Bortolussi
- INFN (National Institute of Nuclear Physics) Pavia Via Bassi 6 Pavia 27100 Italy
- Department of Physics University of Pavia Pavia 27100 Italy
| | - Marina Bekić
- Institute for the Application of Nuclear Energy University of Belgrade Belgrade 11080 Serbia
| | - Miodrag Čolić
- Institute for the Application of Nuclear Energy University of Belgrade Belgrade 11080 Serbia
- Medical Faculty Foča University of East Sarajevo Republika Srpska Foča 73300 Bosnia and Herzegovina
| | - Marco Gerosa
- Department of Computer Science University of Verona Verona 37134 Italy
| | - Alice Busato
- Department of Computer Science University of Verona Verona 37134 Italy
| | - Pasquina Marzola
- Department of Computer Science University of Verona Verona 37134 Italy
| | - Vincenzo Amendola
- Department of Chemical Sciences University of Padova Via Marzolo 1 Padova I‐35131 Italy
| |
Collapse
|