1
|
Abostait A, Abdelkarim M, Bao Z, Miyake Y, Tse WH, Di Ciano-Oliveir C, Buerki-Thurnherr T, Allen C, Keijzer R, Labouta HI. Optimizing lipid nanoparticles for fetal gene delivery in vitro, ex vivo, and aided with machine learning. J Control Release 2024; 376:678-700. [PMID: 39447842 DOI: 10.1016/j.jconrel.2024.10.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
There is a clinical need to develop lipid nanoparticles (LNPs) to deliver congenital therapies to the fetus during pregnancy. The aim of these therapies is to restore normal fetal development and prevent irreversible conditions after birth. As a first step, LNPs need to be optimized for transplacental transport, safety on the placental barrier and fetal organs and transfection efficiency. We developed and characterized a library of LNPs of varying compositions and used machine learning (ML) models to delineate the determinants of LNP size and zeta potential. Utilizing different in vitro placental models with the help of a Random Forest algorithm, we could identify the top features driving percentage LNP transport and kinetics at 24 h, out of a total of 18 input features represented by 41 LNP formulations and 48 different transport experiments. We further evaluated the LNPs for safety, placental cell uptake, transfection efficiency in placental trophoblasts and fetal lung fibroblasts. To ensure the integrity of the LNPs following transplacental transport, we screened LNPs for transport and transfection using a high-throughput integrated transport-transfection in vitro model. Finally, we assessed toxicity of the LNPs in a tracheal occlusion fetal lung explant model. LNPs showed little to no toxicity to fetal and placental cells. Immunoglobin G (IgG) orientation on the surface of LNPs, PEGylated lipids, and ionizable lipids had significant effects on placental transport. The Random Forest algorithm identified the top features driving LNPs placental transport percentage and kinetics. Zeta potential emerged in the top driving features. Building on the ML model results, we developed new LNP formulations to further optimize the transport leading to 622 % increase in transport at 24 h versus control LNP formulation. To induce preferential siRNA transfection of fetal lung, we further optimized cationic lipid percentage and the lipid-to-siRNA ratio. Studying LNPs in an integrated placental and fetal lung fibroblasts model showed a strong correlation between zeta potential and fetal lung transfection. Finally, we assessed the toxicity of LNPs in a tracheal occlusion lung explant model. The optimized formulations appeared to be safe on ex vivo fetal lungs as indicated by insignificant changes in apoptosis (Caspase-3) and proliferation (Ki67) markers. In conclusion, we have optimized an LNP formulation that is safe, with high transplacental transport and preferential transfection in fetal lung cells. Our research findings represent an important step toward establishing the safety and effectiveness of LNPs for gene delivery to the fetal organs.
Collapse
Affiliation(s)
- Amr Abostait
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Mahmoud Abdelkarim
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Zeqing Bao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Yuichiro Miyake
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo 113-8421, Japan
| | - Wai Hei Tse
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | | | - Tina Buerki-Thurnherr
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen 9014, Switzerland
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Richard Keijzer
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Hagar I Labouta
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada; Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto M5S 3G9, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada.
| |
Collapse
|
2
|
Subramaniam S, Joyce P, Ogunniyi AD, Dube A, Sampson SL, Lehr CM, Prestidge CA. Minimum Information for Conducting and Reporting In Vitro Intracellular Infection Assays. ACS Infect Dis 2024; 10:337-349. [PMID: 38295053 DOI: 10.1021/acsinfecdis.3c00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Bacterial pathogens are constantly evolving to outsmart the host immune system and antibiotics developed to eradicate them. One key strategy involves the ability of bacteria to survive and replicate within host cells, thereby causing intracellular infections. To address this unmet clinical need, researchers are adopting new approaches, such as the development of novel molecules that can penetrate host cells, thus exerting their antimicrobial activity intracellularly, or repurposing existing antibiotics using nanocarriers (i.e., nanoantibiotics) for site-specific delivery. However, inconsistency in information reported across published studies makes it challenging for scientific comparison and judgment of experiments for future direction by researchers. Together with the lack of reproducibility of experiments, these inconsistencies limit the translation of experimental results beyond pre-clinical evaluation. Minimum information guidelines have been instrumental in addressing such challenges in other fields of biomedical research. Guidelines and recommendations provided herein have been designed for researchers as essential parameters to be disclosed when publishing their methodology and results, divided into four main categories: (i) experimental design, (ii) establishing an in vitro model, (iii) assessment of efficacy of novel therapeutics, and (iv) statistical assessment. These guidelines have been designed with the intention to improve the reproducibility and rigor of future studies while enabling quantitative comparisons of published studies, ultimately facilitating translation of emerging antimicrobial technologies into clinically viable therapies that safely and effectively treat intracellular infections.
Collapse
Affiliation(s)
- Santhni Subramaniam
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Paul Joyce
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Abiodun D Ogunniyi
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, Roseworthy Campus, University of Adelaide, Roseworthy, SA 5371, Australia
| | - Admire Dube
- School of Pharmacy, University of the Western Cape, Bellville, 7535 Cape Town, South Africa
| | - Samantha L Sampson
- South African Medical Research Council Centre for Tuberculosis Research, and Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, 7602 Cape Town, South Africa
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Clive A Prestidge
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
3
|
Wang D, Jiang Q, Dong Z, Meng T, Hu F, Wang J, Yuan H. Nanocarriers transport across the gastrointestinal barriers: The contribution to oral bioavailability via blood circulation and lymphatic pathway. Adv Drug Deliv Rev 2023; 203:115130. [PMID: 37913890 DOI: 10.1016/j.addr.2023.115130] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
Oral administration is the preferred route of drug delivery in clinical practice due to its noninvasiveness, safety, convenience, and high patient compliance. The gastrointestinal tract (GIT) plays a crucial role in facilitating the targeted delivery of oral drugs. However, the GIT presents multiple barriers that impede drug absorption, including the gastric barrier in the stomach and the mucus and epithelial barriers in the intestine. In recent decades, nanotechnology has emerged as a promising approach for overcoming these challenges by utilizing nanocarrier-based drug delivery systems such as liposomes, micelles, polymeric nanoparticles, solid lipid nanoparticles, and inorganic nanoparticles. Encapsulating drugs within nanocarriers not only protects them from degradation but also enhances their transport and absorption across the GIT, ultimately improving oral bioavailability. The aim of this review is to elucidate the mechanisms underlying nanocarrier-mediated transportation across the GIT into systemic circulation via both the blood circulation and lymphatic pathway.
Collapse
Affiliation(s)
- Ding Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Zhefan Dong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Jianwei Wang
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China; China Jinhua Institute of Zhejiang University, Jinhua 321299, PR China.
| |
Collapse
|
4
|
Abostait A, Tyrrell J, Abdelkarim M, Shojaei S, Tse WH, El-Sherbiny IM, Keijzer R, Labouta HI. Placental Nanoparticle Uptake-On-a-Chip: The Impact of Trophoblast Syncytialization and Shear Stress. Mol Pharm 2022; 19:3757-3769. [PMID: 36053057 DOI: 10.1021/acs.molpharmaceut.2c00216] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The placenta is a dynamic and complex organ that plays an essential role in the health and development of the fetus. Placental disorders can affect the health of both the mother and the fetus. There is currently an unmet clinical need to develop nanoparticle-based therapies to target and treat placental disorders. However, little is known about the interaction of nanoparticles (NPs) with the human placenta under biomimetic conditions. Specifically, the impact of shear stress exerted on the trophoblasts (placental epithelial cells) by the maternal blood flow, the gradual fusion of the trophoblasts along the gestation period (syncytialization), and the impact of microvilli formation on the cell uptake of NPs is not known. To this end, we designed dynamic placenta-on-a-chip models using BeWo cells to recapitulate the micro-physiological environment, and we induced different degrees of syncytialization via chemical induction with forskolin. We characterized the degree of syncytialization quantitatively by measuring beta human chorionic gonadotropin (β-hCG) secretion, as well as qualitatively by immunostaining the tight junction protein, ZO-1, and counter nuclear staining. We also characterized microvilli formation under static and dynamic conditions via F-actin staining. We used these models to measure the cell uptake of chondroitin sulfate a binding protein (CSA) conjugated and control liposomes using confocal microscopy, followed by image analysis. Interestingly, exposure of the cells to a dynamic flow of media intrinsically induced syncytialization and microvilli formation compared to static controls. Under dynamic conditions, BeWo cells produced more β-hCG in conditions that increased the cell exposure time to forskolin (p < 0.005). Our cell uptake results clearly show a combined effect of the exerted shear stress and forskolin treatment on the cell uptake of liposomes as uptake increased in forskolin exposed conditions (p < 0.05). Overall, the difference in the extent of cell uptake of liposomes among the different conditions clearly displays a need for the development of dynamic models of the placenta that consider the changes in the placental cell phenotype along the gestation period, including syncytialization, microvilli formation, and the expression of different transport and uptake receptors. Knowledge generated from this work will inform future research aiming at developing drug delivery systems targeting the placenta.
Collapse
Affiliation(s)
- Amr Abostait
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada
| | - Jack Tyrrell
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Mahmoud Abdelkarim
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada.,Biomedical Engineering, University of Manitoba, Winnipeg R3T 5V6, Canada
| | - Shahla Shojaei
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Wai Hei Tse
- Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.,Depts of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg R3A 1R9, Canada
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Richard Keijzer
- Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.,Depts of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg R3A 1R9, Canada
| | - Hagar I Labouta
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.,Biomedical Engineering, University of Manitoba, Winnipeg R3T 5V6, Canada.,Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
5
|
Tse WH, Higgins S, Patel D, Xing M, West AR, Labouta HI, Keijzer R. The maternal-fetal transfer of passive immunity as a mechanism of transplacental nanoparticle drug delivery for prenatal therapies. Biomater Sci 2022; 10:5243-5253. [PMID: 35912636 DOI: 10.1039/d2bm00293k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoparticles administered into the maternal circulation and across the placenta are a potential clinical therapy to treat congenital diseases. The mechanism by which nanoparticles can safely cross the placenta for targeted drug delivery to the fetus remains poorly understood. We demonstrate that the maternal-fetal transfer of passive immunity through the neonatal Fc Receptor (FcRn) can induce the transplacental transfer of chitosan nanoparticles modifed with IgG antibodies (414 ± 27 nm). The transfer of FITC-tagged IgG-modified chitosan nanoparticles was 2.8 times higher (p = 0.0264) compared to similarly-sized unmodified chitosan nanoparticles (375 ± 17 nm). Co-administration of free IgG competitively diminished the transplacental transfer of IgG-modified nanoparticles, yet unmodified nanoparticles remained unaffected. Colocalization of the FcRn and the IgG-modified chitosan nanoparticles were observed with confocal microscopy. Barrier function before and after nanoparticle administration remained intact as determined by TEER (75-79 Ω cm2) and immmunofluorescence of ZO-1 tight junction proteins. The results provide insight into the clinical applications of nanoparticles for prenatal therapies using the mechanism of the maternal-fetal transfer of passive immunity.
Collapse
Affiliation(s)
- Wai Hei Tse
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| | - Sean Higgins
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada
| | - Daywin Patel
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, R3T 2N2, Manitoba, Canada
| | - Adrian R West
- Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| | - Hagar I Labouta
- Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Apotex Centre, College of Pharmacy, University of Manitoba, R3E 3T5, Manitoba, Canada
| | - Richard Keijzer
- Departments of Surgery, Division of Pediatric Surgery and Pediatrics & Child Health, University of Manitoba, R3E 0W2, Manitoba, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, R3E 3P4, Manitoba, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, R3E 0W2, Manitoba, Canada
| |
Collapse
|
6
|
Shin S, Kwon S, Yeo Y. Meta-Analysis of Drug Delivery Approaches for Treating Intracellular Infections. Pharm Res 2022; 39:1085-1114. [PMID: 35146592 PMCID: PMC8830998 DOI: 10.1007/s11095-022-03188-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/01/2022] [Indexed: 12/20/2022]
Abstract
This meta-analysis aims to evaluate the trend, methodological quality and completeness of studies on intracellular delivery of antimicrobial agents. PubMed, Embase, and reference lists of related reviews were searched to identify original articles that evaluated carrier-mediated intracellular delivery and pharmacodynamics (PD) of antimicrobial therapeutics against intracellular pathogens in vitro and/or in vivo. A total of 99 studies were included in the analysis. The most commonly targeted intracellular pathogens were bacteria (62.6%), followed by viruses (16.2%) and parasites (15.2%). Twenty-one out of 99 (21.2%) studies performed neither microscopic imaging nor flow cytometric analysis to verify that the carrier particles are present in the infected cells. Only 31.3% of studies provided comparative inhibitory concentrations against a free drug control. Approximately 8% of studies, albeit claimed for intracellular delivery of antimicrobial therapeutics, did not provide any experimental data such as microscopic imaging, flow cytometry, and in vitro PD. Future research on intracellular delivery of antimicrobial agents needs to improve the methodological quality and completeness of supporting data in order to facilitate clinical translation of intracellular delivery platforms for antimicrobial therapeutics.
Collapse
Affiliation(s)
- Sooyoung Shin
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Republic of Korea. .,Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, Gyeonggi-do, 16499, Republic of Korea.
| | - Soonbum Kwon
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47906, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47906, USA. .,Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr., West Lafayette, IN, 47907, USA.
| |
Collapse
|
7
|
Advancedoral vaccine delivery strategies for improving the immunity. Adv Drug Deliv Rev 2021; 177:113928. [PMID: 34411689 DOI: 10.1016/j.addr.2021.113928] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/15/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
Infectious diseases continue to inflict a high global disease burden. The consensus is that vaccination is the most effective option against infectious diseases. Oral vaccines have unique advantages in the prevention of global pandemics due to their ease of use, high compliance, low cost, and the ability to induce both systemic and mucosal immune responses. However, challenges of adapting vaccines for oral administration remain significant. Foremost among these are enzymatic and pH-dependent degradation of antigens in the stomach and intestines, the low permeability of mucus barrier, the nonspecific uptake of antigens at the intestinal mucosal site, and the immune suppression result from the elusive immune tolerance mechanisms. Innovative delivery techniques promise great potential for improving the flexibility and efficiency of oral vaccines. A better understanding of the delivery approaches and the immunological mechanisms of oral vaccine delivery systems may provide new scientific insight and tools for developing the next-generation oral vaccine. Here, an overview of the advanced technologies in the field of oral vaccination is proposed, including mucus-penetrating nanoparticle (NP), mucoadhesive delivery vehicles, targeting antigen-presenting cell (APC) nanocarriers and enhanced paracellular delivery strategies and so on. Meanwhile, the mechanisms of delivery vectors interact with mucosal barriers are discussed.
Collapse
|
8
|
Subramaniam S, Joyce P, Thomas N, Prestidge CA. Bioinspired drug delivery strategies for repurposing conventional antibiotics against intracellular infections. Adv Drug Deliv Rev 2021; 177:113948. [PMID: 34464665 DOI: 10.1016/j.addr.2021.113948] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/04/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022]
Abstract
Bacteria have developed a wealth of strategies to avoid and resist the action of antibiotics, one of which involves pathogens invading and forming reservoirs within host cells. Due to the poor cell membrane permeability, stability and retention of conventional antibiotics, this renders current treatments largely ineffective, since achieving a therapeutically relevant antibiotic concentration at the site of intracellular infection is not possible. To overcome such challenges, current antibiotics are 'repurposed' via reformulation using micro- or nano-carrier systems that effectively encapsulate and deliver therapeutics across cellular membranes of infected cells. Bioinspired materials that imitate the uptake of biological particulates and release antibiotics in response to natural stimuli are recently explored to improve the targeting and specificity of this 'nanoantibiotic' approach. In this review, the mechanisms of internalization and survival of intracellular bacteria are elucidated, effectively accentuating the current treatment challenges for intracellular infections and the implications for repurposing conventional antibiotics. Key case studies of nanoantibiotics that have drawn inspiration from natural biological particles and cellular uptake pathways to effectively eradicate intracellular pathogens are detailed, clearly highlighting the rational for harnessing bioinspired drug delivery strategies.
Collapse
Affiliation(s)
- Santhni Subramaniam
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia
| | - Paul Joyce
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia
| | - Nicky Thomas
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia; The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia
| | - Clive A Prestidge
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
9
|
Abstract
The natural world has provided a host of materials and inspiration for the field of nanomedicine. By taking design cues from naturally occurring systems, the nanoengineering of advanced biomimetic platforms has significantly accelerated over the past decade. In particular, the biomimicry of bacteria, with their motility, taxis, immunomodulation, and overall dynamic host interactions, has elicited substantial interest and opened up exciting avenues of research. More recently, advancements in genetic engineering have given way to more complex and elegant systems with tunable control characteristics. Furthermore, bacterial derivatives such as membrane ghosts, extracellular vesicles, spores, and toxins have proven advantageous for use in nanotherapeutic applications, as they preserve many of the features from the original bacteria while also offering distinct advantages. Overall, bacteria-inspired nanomedicines can be employed in a range of therapeutic settings, from payload delivery to immunotherapy, and have proven successful in combatting both cancer and infectious disease.
Collapse
Affiliation(s)
- Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jessica Pihl
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jiyoung Heo
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Joon Ho Park
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
10
|
Kirtane AR, Verma M, Karandikar P, Furin J, Langer R, Traverso G. Nanotechnology approaches for global infectious diseases. NATURE NANOTECHNOLOGY 2021; 16:369-384. [PMID: 33753915 DOI: 10.1038/s41565-021-00866-8] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 01/26/2021] [Indexed: 05/20/2023]
Abstract
Infectious diseases are a major driver of morbidity and mortality globally. Treatment of malaria, tuberculosis and human immunodeficiency virus infection are particularly challenging, as indicated by the ongoing transmission and high mortality associated with these diseases. The formulation of new and existing drugs in nano-sized carriers promises to overcome several challenges associated with the treatment of these diseases, including low on-target bioavailability, sub-therapeutic drug accumulation in microbial sanctuaries and reservoirs, and low patient adherence due to drug-related toxicities and extended therapeutic regimens. Further, nanocarriers can be used for formulating vaccines, which represent a major weapon in our fight against infectious diseases. Here we review the current burden of infectious diseases with a focus on major drivers of morbidity and mortality. We then highlight how nanotechnology could aid in improving existing treatment modalities. We summarize our progress so far and outline potential future directions to maximize the impact of nanotechnology on the global population.
Collapse
Affiliation(s)
- Ameya R Kirtane
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Malvika Verma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Tata Center for Technology and Design, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paramesh Karandikar
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jennifer Furin
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Tata Center for Technology and Design, Massachusetts Institute of Technology, Cambridge, MA, USA
- Media Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Tata Center for Technology and Design, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
11
|
Shojaei S, Suresh M, Klionsky DJ, Labouta HI, Ghavami S. Autophagy and SARS-CoV-2 infection: Apossible smart targeting of the autophagy pathway. Virulence 2020; 11:805-810. [PMID: 32567972 PMCID: PMC7549903 DOI: 10.1080/21505594.2020.1780088] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/05/2020] [Indexed: 01/04/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak resulted in 5,993,317 confirmed cases worldwide with 365,394 confirmed deaths (as of May 29th, 2020, WHO). The molecular mechanism of virus infection and spread in the body is not yet disclosed, but studies on other betacoronaviruses show that, upon cell infection, these viruses inhibit macroautophagy/autophagy flux and cause the accumulation of autophagosomes. No drug has yet been approved for the treatment of SARS-CoV-2 infection; however, preclinical investigations suggested repurposing of several FDA-approved drugs for clinical trials. Half of these drugs are modulators of the autophagy pathway. Unexpectedly, instead of acting by directly antagonizing the effects of viruses, these drugs appear to function by suppressing autophagy flux. Based on the established cross-talk between autophagy and apoptosis, we speculate that over-accumulation of autophagosomes activates an apoptotic pathway that results in apoptotic death of the infected cells and disrupts the virus replication cycle. However, administration of the suggested drugs are associated with severe adverse effects due to their off-target accumulation. Nanoparticle targeting of autophagy at the sites of interest could be a powerful tool to efficiently overcome SARS-CoV-2 infection while avoiding the common adverse effects of these drugs.
Collapse
Affiliation(s)
- Shahla Shojaei
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Madhumita Suresh
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Hagar Ibrahim Labouta
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Biomedical Engineering Program, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Saeid Ghavami
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Research Institute in Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
- Autophagy Research Centre, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Faculty of Medicine, Katowice School of Technology, Katowice, Poland
| |
Collapse
|
12
|
Menina S, Eisenbeis J, Kamal MAM, Koch M, Bischoff M, Gordon S, Loretz B, Lehr C. Bioinspired Liposomes for Oral Delivery of Colistin to Combat Intracellular Infections by Salmonella enterica. Adv Healthc Mater 2019; 8:e1900564. [PMID: 31328434 DOI: 10.1002/adhm.201900564] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/27/2019] [Indexed: 01/07/2023]
Abstract
Bacterial invasion into eukaryotic cells and the establishment of intracellular infection has proven to be an effective means of resisting antibiotic action, as anti-infective agents commonly exhibit a poor permeability across the host cell membrane. Encapsulation of anti-infectives into nanoscaled delivery systems, such as liposomes, is shown to result in an enhancement of intracellular delivery. The aim of the current work is, therefore, to formulate colistin, a poorly permeable anti-infective, into liposomes suitable for oral delivery, and to functionalize these carriers with a bacteria-derived invasive moiety to enhance their intracellular delivery. Different combinations of phospholipids and cholesterol are explored to optimize liposomal drug encapsulation and stability in biorelevant media. These liposomes are then surface-functionalized with extracellular adherence protein (Eap), derived from Staphylococcus aureus. Treatment of HEp-2 and Caco-2 cells infected with Salmonella enterica using colistin-containing, Eap-functionalized liposomes resulted in a significant reduction of intracellular bacteria, in comparison to treatment with nonfunctionalized liposomes as well as colistin alone. This indicates that such bio-invasive carriers are able to facilitate intracellular delivery of colistin, as necessary for intracellular anti-infective activity. The developed Eap-functionalized liposomes, therefore, present a promising strategy for improving the therapy of intracellular infections.
Collapse
Affiliation(s)
- Sara Menina
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection Research (HZI) Saarbrücken 66123 Germany
- Department of PharmacySaarland University Saarbrücken 66123 Germany
| | - Janina Eisenbeis
- Institute of Medical Microbiology and HygieneSaarland University Homburg 66421 Germany
| | - Mohamed Ashraf M. Kamal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection Research (HZI) Saarbrücken 66123 Germany
| | - Marcus Koch
- Institute for New MaterialsSaarland University Saarbrücken 66123 Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and HygieneSaarland University Homburg 66421 Germany
| | - Sarah Gordon
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection Research (HZI) Saarbrücken 66123 Germany
- School of Pharmacy and Biomolecular SciencesJohn Moores University Liverpool L3 3AF UK
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection Research (HZI) Saarbrücken 66123 Germany
| | - Claus‐Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection Research (HZI) Saarbrücken 66123 Germany
- Department of PharmacySaarland University Saarbrücken 66123 Germany
| |
Collapse
|
13
|
Labouta HI, Asgarian N, Rinker K, Cramb DT. Meta-Analysis of Nanoparticle Cytotoxicity via Data-Mining the Literature. ACS NANO 2019; 13:1583-1594. [PMID: 30689359 DOI: 10.1021/acsnano.8b07562] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Developing predictive modeling frameworks of potential cytotoxicity of engineered nanoparticles is critical for environmental and health risk analysis. The complexity and the heterogeneity of available data on potential risks of nanoparticles, in addition to interdependency of relevant influential attributes, makes it challenging to develop a generalization of nanoparticle toxicity behavior. Lack of systematic approaches to investigate these risks further adds uncertainties and variability to the body of literature and limits generalizability of existing studies. Here, we developed a rigorous approach for assembling published evidence on cytotoxicity of several organic and inorganic nanoparticles and unraveled hidden relationships that were not targeted in the original publications. We used a machine learning approach that employs decision trees together with feature selection algorithms ( e.g., Gain ratio) to analyze a set of published nanoparticle cytotoxicity sample data (2896 samples). The specific studies were selected because they specified nanoparticle-, cell-, and screening method-related attributes. The resultant decision-tree classifiers are sufficiently simple, accurate, and with high prediction power and should be widely applicable to a spectrum of nanoparticle cytotoxicity settings. Among several influential attributes, we show that the cytotoxicity of nanoparticles is primarily predicted from the nanoparticle material chemistry, followed by nanoparticle concentration and size, cell type, and cytotoxicity screening indicator. Overall, our study indicates that following rigorous and transparent methodological experimental approaches, in parallel to continuous addition to this data set developed using our approach, will offer higher predictive power and accuracy and uncover hidden relationships. Results obtained in this study help focus future studies to develop nanoparticles that are safe by design.
Collapse
Affiliation(s)
- Hagar I Labouta
- Department of Chemistry, Faculty of Science , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
- College of Pharmacy, Rady Faculty of Health Sciences , University of Manitoba , Winnipeg , Manitoba R3E 0T5 , Canada
- Biomedical Engineering , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
- Department of Pharmaceutics, Faculty of Pharmacy , Alexandria University , Alexandria , 21521 , Egypt
| | - Nasimeh Asgarian
- Department of Chemistry, Faculty of Science , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
| | - Kristina Rinker
- Biomedical Engineering , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
- Department of Chemical and Petroleum Engineering , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
| | - David T Cramb
- Department of Chemistry, Faculty of Science , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 1N4 , Canada
- Department of Chemistry and Biology, Faculty of Science , Ryerson University , Toronto , Ontario M5B 2K3 , Canada
| |
Collapse
|
14
|
Metal Organic Framework (MOF) Particles as Potential Bacteria-Mimicking Delivery Systems for Infectious Diseases: Characterization and Cellular Internalization in Alveolar Macrophages. Pharm Res 2019; 36:53. [PMID: 30790066 DOI: 10.1007/s11095-019-2589-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/10/2019] [Indexed: 02/01/2023]
Abstract
PURPOSE Intramacrophagic bacteria pose a great challenge for the treatment of infectious diseases despite many macrophage targeted drug delivery approaches explored. The use of biomimetic approaches for treating infectious diseases is promising, but not studied extensively. The study purpose is to evaluate iron-based metal-organic frameworks (MOF) as a potential bacteria-mimicking delivery system for infectious diseases. METHODS Two types of carboxylated MOFs, MIL-88A(Fe) and MIL-100(Fe) were developed as "pathogen-like" particles by surface coating with mannose. MOF morphology, cellular uptake kinetics, and endocytic mechanisms in 3D4/21 alveolar macrophages were characterized. RESULTS MIL-88A(Fe) is rod-shape (aspect ratio 1:5) with a long-axis size of 3628 ± 573 nm and MIL-100(Fe) is spherical with diameter of 103.9 ± 7.2 nm. Cellular uptake kinetics of MOFs showed that MIL-100(Fe) nanoparticles were internalized at a faster rate and higher extent compared to MIL-88A(Fe) microparticles. Mannosylation did not improve the uptake of MIL-100(Fe) particles, whereas it highly increased MIL-88A(Fe) cellular uptake and number of cells involved in internalization. Cell uptake inhibition studies indicated that macropinocytosis/phagocytosis was the main endocytic pathway for internalization of MOFs. Accumulation of MOF particles in acidic compartments was clearly observed. CONCLUSIONS The successfully synthesized "pathogen-like" particles provide a novel application of MOF-based particles as biomimetic delivery system for intramacrophagic-based infections.
Collapse
|
15
|
Aspherical and Spherical InvA497-Functionalized Nanocarriers for Intracellular Delivery of Anti-Infective Agents. Pharm Res 2018; 36:22. [PMID: 30519925 PMCID: PMC6290668 DOI: 10.1007/s11095-018-2521-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/08/2018] [Indexed: 01/01/2023]
Abstract
Purpose The objective of this work was to evaluate the potential of polymeric spherical and aspherical invasive nanocarriers, loaded with antibiotic, to access and treat intracellular bacterial infections. Methods Aspherical nanocarriers were prepared by stretching of spherical precursors, and both aspherical and spherical nanocarriers were surface-functionalized with the invasive protein InvA497. The relative uptake of nanocarriers into HEp-2 epithelial cells was then assessed. Nanocarriers were subsequently loaded with a preparation of the non-permeable antibiotic gentamicin, and tested for their ability to treat HEp-2 cells infected with the enteroinvasive bacterium Shigella flexneri. Results InvA497-functionalized nanocarriers of both spherical and aspherical shape showed a significantly improved rate and extent of uptake into HEp-2 cells in comparison to non-functionalized nanocarriers. Functionalized and antibiotic-loaded nanocarriers demonstrated a dose dependent killing of intracellular S. flexneri. A slight but significant enhancement of intracellular bacterial killing was also observed with aspherical as compared to spherical functionalized nanocarriers at the highest tested concentration. Conclusions InvA497-functionalized, polymer-based nanocarriers were able to efficiently deliver a non-permeable antibiotic across host cell membranes to affect killing of intracellular bacteria. Functionalized nanocarriers with an aspherical shape showed an interesting future potential for intracellular infection therapy. Electronic supplementary material The online version of this article (10.1007/s11095-018-2521-3) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Borišev I, Mrđanovic J, Petrovic D, Seke M, Jović D, Srđenović B, Latinovic N, Djordjevic A. Nanoformulations of doxorubicin: how far have we come and where do we go from here? NANOTECHNOLOGY 2018; 29:332002. [PMID: 29798934 DOI: 10.1088/1361-6528/aac7dd] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Nanotechnology, focused on discovery and development of new pharmaceutical products is known as nanopharmacology, and one research area this branch is engaged in are nanopharmaceuticals. The importance of being nano has been particularly emphasized in scientific areas dealing with nanomedicine and nanopharmaceuticals. Nanopharmaceuticals, their routes of administration, obstacles and solutions concerning their improved application and enhanced efficacy have been briefly yet comprehensively described. Cancer is one of the leading causes of death worldwide and evergrowing number of scientific research on the topic only confirms that the needs have not been completed yet and that there is a wide platform for improvement. This is undoubtedly true for nanoformulations of an anticancer drug doxorubicin, where various nanocarrriers were given an important role to reduce the drug toxicity, while the efficacy of the drug was supposed to be retained or preferably enhanced. Therefore, we present an interdisciplinary comprehensive overview of interdisciplinary nature on nanopharmaceuticals based on doxorubicin and its nanoformulations with valuable information concerning trends, obstacles and prospective of nanopharmaceuticals development, mode of activity of sole drug doxorubicin and its nanoformulations based on different nanocarriers, their brief descriptions of biological activity through assessing in vitro and in vivo behavior.
Collapse
Affiliation(s)
- Ivana Borišev
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, Novi Sad, Serbia
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Labouta HI, Sarsons C, Kennard J, Gomez-Garcia MJ, Villar K, Lee H, Cramb DT, Rinker KD. Understanding and improving assays for cytotoxicity of nanoparticles: what really matters? RSC Adv 2018; 8:23027-23039. [PMID: 35540163 PMCID: PMC9081595 DOI: 10.1039/c8ra03849j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 06/16/2018] [Indexed: 11/21/2022] Open
Abstract
Despite years of excellent individual studies, the impact of nanoparticle (NP) cytotoxicity studies remains limited by inconsistent data collection and analysis. It is often unclear how exposure conditions can be used to determine cytotoxicity quantitatively. Discrepancies due to using different measurement conditions, readouts and controls to characterize NP interactions with cells lead to further challenges. To examine which parameters are critical in NP cytotoxicity studies, we have chosen to examine two NP types (liposomes and quantum dots) at different concentrations incubated with two primary vascular endothelial cells, HUVEC and HMVEC-C for a standard time of 24 h. We paid close attention to the effects of positive controls and cell association on interpretation of cytotoxicity data. Various cellular responses (ATP content, oxidative stress, mitochondrial toxicity, and phospholipidosis) were measured in parallel. Interestingly, cell association data varied significantly with the different image analyses. However, cytotoxicity responses could all be correlated with exposure concentration. Cell type did have an effect on cytotoxicity reports. Most significantly, NP cytotoxicity results varied with the inclusion or exclusion of positive controls. In the absence of positive controls, one tends to emphasize small changes in cell responses to NPs.
Collapse
Affiliation(s)
- Hagar I Labouta
- Department of Chemistry, Faculty of Science, University of Calgary Calgary Canada
- Biomedical Engineering, University of Calgary Calgary Canada
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University Alexandria Egypt
| | | | - Jacob Kennard
- Biomedical Engineering, University of Calgary Calgary Canada
| | | | - Kenrick Villar
- Biomedical Engineering, University of Calgary Calgary Canada
| | - Hyungbok Lee
- Biomedical Engineering, University of Calgary Calgary Canada
| | - David T Cramb
- Department of Chemistry, Faculty of Science, University of Calgary Calgary Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary Calgary Canada
| | - Kristina D Rinker
- Biomedical Engineering, University of Calgary Calgary Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary Calgary Canada
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary Calgary Canada
| |
Collapse
|
18
|
Goes A, Fuhrmann G. Biogenic and Biomimetic Carriers as Versatile Transporters To Treat Infections. ACS Infect Dis 2018; 4:881-892. [PMID: 29553240 DOI: 10.1021/acsinfecdis.8b00030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biogenic and biomimetic therapeutics are a relatively new class of systems that are of physiological origin and/or take advantage of natural pathways or aim at mimicking these to improve selective interaction with target tissue. The number of biogenic and bioengineered avenues for drug therapy and diagnostics has multiplied over the past years for many applications, indicating the high expectations associated with this biological route. Nevertheless, the use of "bio"-related approaches for treating or diagnosing infectious diseases is still rare. Given that infectious diseases, in particular bacterial resistances, are seriously on the rise, there is an urgent need to take advantage of biogenic and bioengineered systems to target these challenges. In this manuscript, we first give a definition of the various "bio" terms, including biogenic, biomimetic, bioinspired, and bioengineered and we highlight them using tangible applications in the field of infectious diseases. Our examples cover cell-derived systems, including bioengineered bacteria, virus-like particles, and different cell-mimetics. Moreover, we discuss natural and bioengineered particles such as extracellular vesicles from mammalian and bacterial sources and liposomes. A concluding section outlines the potential for biomaterial-related avenues to overcome challenges associated with difficult-to-treat infections. We critically discuss benefits and risks for these applications and give an outlook on the future of biogenic engineering.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Gregor Fuhrmann
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
19
|
Shahbazi MA, Almeida PV, Correia A, Herranz-Blanco B, Shrestha N, Mäkilä E, Salonen J, Hirvonen J, Santos HA. Intracellular responsive dual delivery by endosomolytic polyplexes carrying DNA anchored porous silicon nanoparticles. J Control Release 2017; 249:111-122. [PMID: 28159519 DOI: 10.1016/j.jconrel.2017.01.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 12/13/2022]
Abstract
Bioresponsive cytosolic nanobased multidelivery has been emerging as an enormously challenging novel concept due to the intrinsic protective barriers of the cells and hardly controllable performances of nanomaterials. Here, we present a new paradigm to advance nano-in-nano integration technology amenable to create multifunctional nanovehicles showing considerable promise to overcome restrictions of intracellular delivery, solve impediments of endosomal localization and aid effectual tracking of nanoparticles. A redox responsive intercalator chemistry comprised of cystine and 9-aminoacridine is designed as a cross-linker to cap carboxylated porous silicon nanoparticles with DNA. These intelligent nanocarriers are then encapsulated within novel one-pot electrostatically complexed nano-networks made of a zwitterionic amino acid (cysteine), an anionic bioadhesive polymer (poly(methyl vinyl ether-alt-maleic acid)) and a cationic endosomolytic polymer (polyethyleneimine). This combined nanocomposite is successfully tested for the co-delivery of hydrophobic (sorafenib) or hydrophilic (calcein) molecules loaded within the porous core, and an imaging agent covalently integrated into the polyplex shell by click chemistry. High loading capacity, low cyto- and hemo-toxicity, glutathione responsive on-command drug release, and superior cytosolic delivery are shown as achievable key features of the proposed formulation. Overall, formulating drug molecules, DNA and imaging agents, without any interference, in a physico-chemically optimized carrier may open a path towards broad applicability of these cost-effective multivalent nanocomposites for treating different diseases.
Collapse
Affiliation(s)
- Mohammad-Ali Shahbazi
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland.
| | - Patrick Vingadas Almeida
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland
| | - Alexandra Correia
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland
| | - Barbara Herranz-Blanco
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland
| | - Neha Shrestha
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland
| | - Ermei Mäkilä
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland; Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014, Finland
| | - Jouni Hirvonen
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland.
| |
Collapse
|
20
|
Hadorn M, Boenzli E, Hanczyc MM. Specific and Reversible DNA-Directed Self-Assembly of Modular Vesicle-Droplet Hybrid Materials. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:3561-3566. [PMID: 27010467 DOI: 10.1021/acs.langmuir.5b04003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Modular hybrid structures functionalized to assemble in a controlled manner possess diverse properties necessary for a new generation of complex materials and applications. Here, we functionalized giant unilamellar vesicles and emulsion droplets with biotinylated single-stranded DNA oligonucleotides using streptavidin as an intermediary linker to demonstrate specific and reversible DNA-directed self-assembly into vesicle-droplet hybrid structures. A low molar percentage of PEGylated phospholipids independent of the DNA-based recognition machinery at the supramolecular surface modulated the stability of the system. The reversibility of the aggregation was demonstrated by heating the hybrid structures above the melting temperature of the conjoining double-stranded DNA in the presence of excess biotin. The application of this general assembly control system to diverse multiphase soft materials provides the mechanism to assemble complex modular hybrid systems in a controllable and reversible way, which may provide an advantage where multifunctionality is a target property.
Collapse
Affiliation(s)
- Maik Hadorn
- Laboratory for Artificial Biology, Centre for Integrative Biology (CIBIO), University of Trento , 38122 Trento, Italy
| | - Eva Boenzli
- Laboratory for Artificial Biology, Centre for Integrative Biology (CIBIO), University of Trento , 38122 Trento, Italy
| | - Martin M Hanczyc
- Laboratory for Artificial Biology, Centre for Integrative Biology (CIBIO), University of Trento , 38122 Trento, Italy
| |
Collapse
|
21
|
Menina S, Labouta HI, Geyer R, Krause T, Gordon S, Dersch P, Lehr CM. Invasin-functionalized liposome nanocarriers improve the intracellular delivery of anti-infective drugs. RSC Adv 2016. [DOI: 10.1039/c6ra02988d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Liposomes containing gentamicin and surface-functionalized with InvA497 showed a reduced infection load of both cytosolic and vacuolar intracellular bacteria.
Collapse
Affiliation(s)
- Sara Menina
- Department of Drug Delivery
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)
- Helmholtz Center for Infection Research (HZI)
- Saarbruecken
- Germany
| | - Hagar Ibrahim Labouta
- Department of Drug Delivery
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)
- Helmholtz Center for Infection Research (HZI)
- Saarbruecken
- Germany
| | - Rebecca Geyer
- Department of Molecular Infection Biology
- Helmholtz Center for Infection Research (HZI)
- Braunschweig
- Germany
| | - Tanja Krause
- Department of Molecular Infection Biology
- Helmholtz Center for Infection Research (HZI)
- Braunschweig
- Germany
| | - Sarah Gordon
- Department of Drug Delivery
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)
- Helmholtz Center for Infection Research (HZI)
- Saarbruecken
- Germany
| | - Petra Dersch
- Department of Molecular Infection Biology
- Helmholtz Center for Infection Research (HZI)
- Braunschweig
- Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)
- Helmholtz Center for Infection Research (HZI)
- Saarbruecken
- Germany
| |
Collapse
|