Origami millirobots bring health care closer to precision drug delivery – Nanowerk

Jun 15, 2022(Nanowerk News) If youve ever swallowed the same round tablet in hopes of curing everything from stomach cramps to headaches, you already know that medicines arent always designed to treat precise pain points.While over-the-counter pills have cured many ailments for decades, biomedical researchers have only recently begun exploring ways to improve targeted drug delivery when treating more complicated medical conditions, like cardiovascular disease or cancer.A promising innovation within this burgeoning area of biomedicine is the millirobot. These fingertip-sized robots are poised to become medicines future lifesavers to crawl, spin, and swim to enter narrow spaces on their mission to investigate inner workings or dispense medicines.Origami millirobot with spinning-enabled propulsion. (Image: Zhao Lab)Leading research in this field, Stanford University mechanical engineer Renee Zhaois working on many millirobot designs at once including a magnetic crawling robot, which was recently seen worming its way through a stomach on the cover of Science Advances. Powered by magnetic fields which allow for continuous motion and can be instantly applied to generate torque and change the way the robots move her robots can self-select different locomotive states and overcome obstacles in the body. Just by shifting the strength and orientation of the magnetic field, Zhaos team can send the robot sailing across the body at distances in a single leap that are 10 times the robots length.A key aspect of her research, the magnetic actuation also provides untethered control for non-invasive operation and separates the control unit from the device to allow for miniaturization. Zhao said their most recent robot, featured in Nature Communications ("Spinning-enabled wireless amphibious origami millirobot"), is the most robust and multifunctional untethered robot we have ever developed.This new spinning-enabled wireless amphibious origami millirobot is as multifunctional as its name implies. Its an elegantly conceived single unit thats able to speedily travel over an organs slick, uneven surfaces and swim through body fluids, propelling itself wirelessly while transporting liquid medicines. Unlike pills swallowed or liquids injected, this robot withholds medicine until it reaches the target, and then releases a high-concentration drug, said Zhao, who is an assistant professor of mechanical engineering. That is how our robot achieves targeted drug delivery.Reshaping drug deliveryWhats groundbreaking about this particular amphibious robot, according to Zhao, is that it goes beyond the designs of most origami-based robots, which only utilize origamis foldability to control how a robot morphs and moves.On top of looking at how folding could enable the robot to perform certain actions imagine an accordion fold that squeezes out medicine Zhaos team also considered how the dimensions of each folds exact shape influenced the robots rigid motion when it was not folded. As a result, the robots unfolded form inherently lends itself to propulsion through the environment. Such broad-minded considerations allowed the researchers to get more use out of the materials without adding bulk and in Zhaos world, the more functionality achieved from a single structure within the robots design, the less invasive the medical procedure is.Another unique aspect of the design of the robot is the combination of certain geometrical features. A longitudinal hole into the robots center and lateral slits angled up the sides reduced water resistance and helped the robot swim better. This design induces a negative pressure in the robot for fast swimming and meanwhile provides suction for cargo pickup and transportation, Zhao said. We take full advantage of the geometric features of this small robot and explore that single structure for different applications and for different functions.Based on conversations with Stanford Department of Medicine experts, the Zhao Lab is considering how to improve upon current treatments and procedures by building new technologies. If this work goes Zhaos way, her robots wont just provide a handy way to effectively dispense medicine but could also be used to carry instruments or cameras into the body, changing how doctors examine patients. The team is also working on using ultrasound imaging to track where robots go, eliminating any need to cut open organs.The smaller, simpler, the betterWhile we wont see millirobots like Zhaos in real health care settings until more is known about optimal design and imaging best practices, the labs first-of-its-kind swimmer highlighted in Nature Communications is among their robots that are furthest along. Its currently in the trial stages that come before any live animal testing that proceeds human clinical trials.In the meantime, Zhaos team continues combining a variety of novel smart materials and structures into unique designs that ultimately form new biomedical devices. She also plans to continue scaling down her robots to further biomedical research at the microscale.As an engineer, Zhao strives to develop the simplest structures with the most functionality. Her amphibious robot exemplifies that mission, as it inspired her team to more fully consider geometric features not yet commonly prioritized by other origami robot researchers. We started looking at how all these work in parallel, Zhao said. This is a very unique point of this work, and it also has broad potential application in the biomedical field.

Read the original here:
Origami millirobots bring health care closer to precision drug delivery - Nanowerk

Fundamental Knowledge on Nanobots – Bio-IT World

Nanorobots are electromechanical devices comprised of components that are within the nanometer size range. Within medicine, nanorobotic applications have been successfully used for a variety of microbiological, hematological, surgical and dental applications, to name a few.

The nanobots market global size accounted for USD 5.3 billion in 2021 and is expected to reach around USD 21.45 billion by 2030, expected to register growth at a CAGR of 16.8% from 2022 to 2030.

What is Biomedical Nanorobots?

As compared to industrial robots that were originally developed to automate routine and dangerous tasks, biomedical robots are highly specialized and miniature devices that must be capable of performing precise tasks within the human body. Recent advancements in nanotechnology and materials science have therefore promoted the development of both micro- and nanorobots for a wide range of biomedical applications.

Whereas the traditional power sources for industrial robots that require large power supplies and/or battery storage capabilities, both micro- and nanorobots will typically depend on chemically powered motors for their energy needs. To this end, these motors acquire energy by converting locally supplied fuels, such as oxygen or glucose within the body, to propel themselves towards different cellular structures. Nanorobots can also rely on externally powered motors based on either magnetic or ultrasound technology to drive their motion.

One of the most challenges that biomedical researchers have faced during the miniaturization of robotic systems has been the optimization of nanolocomotion. Recent developments in this field have demonstrated the ability of both micro- and nanorobots to efficiently propel themselves through complex biological media or narrow blood vessels. Furthermore, once these microscopic robots have penetrated through these areas, researchers have successfully developed ways in which these devices can collect and remove tissue biopsy samples, obtain detailed images, release active agents at predetermined locations and perform localized diagnoses.

Key market players

Report Scope of theNanobots Market

USD 21.45 Billion

Segments covered in the report

By Type

By Application

By Type of Manufacturing

By End User

Regional Segment

Nanomedicine segment is expected to dominate the application segment of the nanobots market

Based on application, the nanobots market is segmented into nanomedicine, biomedical and other applications. The Nanomedicine segment is expected to dominate the global nanobots market by holding more than 36% of the overall market. Nanobots are widely used in nanomedicine due to the increasing healthcare applications of nanobots. The large market share of this segment is attributed to the large level of commercialization in the healthcare sector for drug delivery, in vivo imaging, active implants, in vitro diagnostic, biomaterial, and drug therapy.

Additionally, increasing innovations in the field of cancer treatment related to the specific target are also contributing to the growth of nanobots market. The biomedical applications segment accounted for the second-largest market share of the overall nanobots marketplace.

Thanks for reading you can also get individual chapter-wise sections or region-wise report versions such as North America, Europe, or the Asia Pacific.

Download a FREE PDF Brochure https://www.precedenceresearch.com/sample/1914

Read the rest here:
Fundamental Knowledge on Nanobots - Bio-IT World

What is medtech and where will it go next? – Verdict

Medtech startups are going from strength to strength. Investors have upped the funding dosage injected into the industry every year over the past decade.

However, that doesnt explain what medtech is or why venture capitalists are so eagerly betting on the sectors future.

But dont worry, weve got you covered.

Medtech stands for medicinal or medical technology. It is shorthand for technologies used within the medical realm.

Now, technically one could argue that things like scalpels, X-rays and stethoscopes would also fall under this category. After all, they were high-tech when they first popped into the scene and they are, undoubtedly, used by doctors around the planet.

However, the term medtech mainly applies to modern technologies with novel applications in healthcare. In other words: the term refers to high-tech solutions like artificial intelligence (AI) systems and robotics being used by doctors, nurses, pharmacologists and other medical practitioners.

And this prospect has clearly caught the imagination of investors

VCs have backed medtech en masse over the past decade. In 2013, investors injected $914m into the global industry across 220 deals, according to data from research firm GlobalData. The number of deals have climbed year on year since. In 2021, VCs bet $38.2bn on the industry across 1,161 deals.

However, there are signs that the flow of investment could be slowing down. As of July 25, only $11.7bn have been injected into the industry across 506 deals. This could be due to industry being caught up in the same macro economic whirlwinds the war in Ukraine, the end of the pandemic, rising inflation, new regulation to mention a few factors that now threatens to pop the tech bubble.

Time will tell whether investment will cool down like it seems to do for the tech industry in general.

Medtech has several applications. The first one worth mentioning is telemedicine. In a push to marry health with convenience, tech companies are developing ways that we can look after ourselves from home. Telemedicine became commonplace during the pandemic, and it has persevered; even now many continue to speak to their GPs over the phone. Following the pandemic, telemedicine services have been repurposed as one of the ways technology can help cities beat the next heatwave.

Beyond the pandemic, handheld and wearable remote patient monitoring devices transmit healthcare data to doctors and further innovations place the power to thrive directly into patients' pockets. For example, wearable blood-glucose monitors remind diabetic patients to take their insulin and specialist mobile games help children with ADHD sustain concentration for longer.

Medtech innovators are also increasingly tapping into the Internet of Things, (IoT), a system of wireless, interrelated and connected digital devices. By connecting medical devices to a server, doctors can, for instance, monitor patients' health remotely.

GlobalData estimates that the global market for IoT platforms for healthcare providers will jump from $10.6bn in 2020 to reach $13.3bn in 2025.

Medtech can also help patients in hospitals in a number of ways.

For instance, tricorders are handheld computers that use sensors, like cameras, to detect a range of health conditions. These time-saving devices, once merely a science-fiction, can now diagnose with at least the accuracy of a physician.

The list of new technologies that will permeate healthcare includes augmented reality (AR). AR headsets will allow surgeons to cycle through different scans of a patient during an operation. Other AR headsets will help to train medical students.

Robotics are already providing help during surgery. Doctors in Seattle already allow robotic appendages to lend a steady hand during minimally-invasive procedures on the brain.

AI has a plethora of applications in healthcare. To mention a few, AI can identify abnormalities on scans that might otherwise go overlooked. AI might also speed up clinical trials. Systems that simulate how a chemical will interact in the body are currently under development.

Interestingly, another AI has even formulated a potential medicine. Supercomputers during Covid-19 also helped to develop the vaccine that enabled us to get back to the new normal, whatever that is. Supercomputers are clusters of interconnected computers with an accumulated processing power that put todays fastest home desktops to shame.

AI have also been combined with robotics in order to improve the length and quality of life of patients suffering from motor neurone disease.

Fascinating developments in nanotechnology continue to broaden the horizons of medicine. Notably, nanomedicine can ensure that chemicals only reach targeted locations in the body. Radioactive medicines are encased in a nanostructure to protect organs, and when the medicine reaches the target location ultrasound is used to break open the casing.

The applications of technology in healthcare are vast and open ended. We have just scratched the surface of the wealth of possibilities in 21st Century medtech.

So it's unsurprising that another few CCs of VC funding for the medtech sector is just what the doctor ordered.

Excerpt from:
What is medtech and where will it go next? - Verdict

Biologys hardest working pigments and MOFs – EurekAlert

image:Illustration of the wide range of electrocatalytic and photocatalytic processes and applications for porphyrin framework materials view more

Credit: Nano Research Energy, Tsinghua University Press

Some of the economic sectors that are the hardest to decarbonize would benefit from the emergence of substantially more efficient catalysts involved in energy conversion chemical reactions. A breakthrough here might depend upon the use of pigments widely deployed in biological processes integrated as a catalyst into novel and highly porous molecular structures that act sort of like sponges.

A paper describing the state of play in this field and the challenges it faces was published in the journal Nano Research Energyon May 29.

In recent years, porphyrins and metalloporphyrins have played an increasingly important role in biomimetic chemistry, solar energy utilization, medicine, and a great many other applications. But use of porphyrins in electrocatalysis and photocatalysis reactions central to many energy conversion processes useful for the clean transition was found to be unstable, deactivate, and difficult to recycle, which has limited the further development of these energy conversion technologies.

So scientists have begun to consider the integration of porphyrins as the organic ligands (the ion that binds to a central metal atom in a complex molecule) into synthetic molecular structures known as metal-organic frameworks (MOFs) and their twin, covalent-organic frameworks (COFs)known as porphyrin-based framework materials.

This should in principle deliver excellent electrocatalysis and photocatalysis performance as the MOF and COF structures are simple to synthesize and highly designed, thus much more controllable and structurally stable, said Yusuke Yamauchi, a co-author of the paper and researcher with the Australian Institute for Bioengineering and Nanotechnology at the University of Queensland.

The researchers, who are themselves involved in porphyrin-based framework materials development, put together a review article describing the state of play in their field. Such review papers are necessary for young fields to advance as they clarify current understanding, discuss advances and challenges, identify research gaps and can even offer guidelines for policy and tips on best practice, Huan Pang, a co-author of the paper and the researcher with the School of Chemistry and Chemical Engineering at the Yangzhou University, China

The paper explores all the current and potential applications of porphyrin-based framework material catalysts, and finds that there remains great potential, but the field confronts several challenges.

In an economy of net-zero greenhouse gas emissions, not everything can be electrifiedparticularly long-haul heavy transportand so some form of clean fuels, such as carbon-neutral synthetic hydrocarbons, ammonia or hydrogen will be necessary. All these fuels involve the conversion of clean energywhether from the sun, wind, water or uraniuminto transportable and stable chemical energy. Part of this process requires the production of clean hydrogen through the use of electricity, light or heat to split water into its constituent elements, hydrogen and oxygen.

Hydrocarbons are composed of differing ratios of carbon and hydrogen, hence the name. Thus the clean, synthetic versions replacing their dirty fossil cousins will require drawing down carbon dioxide from the atmosphere and transforming it into various usable forms of carbon as an input to be married to the clean hydrogen. To draw down atmospheric carbon and make use of it is also known as carbon capture and utilization (CCU).

All these processes, and many others involved in the clean transition (the move from fossil fuels to clean technologies) such as the use of fuel cells and light collection, are in effect chemical reactions that convert energy from one form to another, more usable form. These chemical reactions require addition of substances known as catalysts that speed the reaction up. Some of those catalysts are extremely expensive such as platinum, or are not efficient enough for the end product to compete with fossil fuels, or produce their own environmental challenges.

Thus the hunt is on for more efficient, cheaper and cleaner catalysts such as porphyrin,

The development of efficient non-precious porphyrin-based framework material catalysts to replace precious metal catalysts remains a significant hurdle. The design and construction of porphyrin blocks currently mainly relies on a highly symmetrical design, which limits the diversity of porphyrin framework families and affects their potential catalytic applications. Novel structures that employ porphyrin units with asymmetric design should be considered to extend the substances utility.

The cost of preparing porphyrin framework materials remains high and so it is urgent that engineers develop new synthesis methods if these catalysts are to be taken up in large-scale industrial applications. Reducing the number of steps required in synthesis is an important research, but it is also extremely difficult to do this.

They conclude however that should such challenges be overcome, porphyrin-based framework materials could be a game-changer in the commercialisation of energy conversion processes essential for some of the sectors that are the very hardest to decarbonize.

Porphyrins are some of biologys hardest working substances. This class of pigments is deployed in a wide array of vital processes, from photosynthesis to breathing. Derivatives of these water-soluble, ring-shaped molecules that bind metal ions include chlorophylls in plants and the hemoglobins that carry oxygen in the blood of animals. They also enhance the catalytic activities of enzymes in a range of other life-giving chemical reactions. Metalloporphyrins are of particular interest with respect to the clean transition due to their role as catalysts in water splitting to produce hydrogen and oxygen.

##

About Nano Research Energy

Nano Research Energy is launched by Tsinghua University Press, aiming at being an international, open-access and interdisciplinary journal. We will publish research on cutting-edge advanced nanomaterials and nanotechnology for energy. It is dedicated to exploring various aspects of energy-related research that utilizes nanomaterials and nanotechnology, including but not limited to energy generation, conversion, storage, conservation, clean energy, etc. Nano Research Energy will publish four types of manuscripts, that is, Communications, Research Articles, Reviews, and Perspectives in an open-access form.

About SciOpen

SciOpen is a professional open access resource for discovery of scientific and technical content published by the Tsinghua University Press and its publishing partners, providing the scholarly publishing community with innovative technology and market-leading capabilities. SciOpen provides end-to-end services across manuscript submission, peer review, content hosting, analytics, and identity management and expert advice to ensure each journals development by offering a range of options across all functions as Journal Layout, Production Services, Editorial Services, Marketing and Promotions, Online Functionality, etc. By digitalizing the publishing process, SciOpen widens the reach, deepens the impact, and accelerates the exchange of ideas.

Nano Research Energy

Porphyrin-based framework materials for energy conversion

29-May-2022

Read the original post:
Biologys hardest working pigments and MOFs - EurekAlert

Singapore makes further advancement in cancer nanomedicine – BSA bureau

Particles released by red blood cells are effective carriers for anti-cancer immunotherapy

A study led by researchers at the Yong Loo Lin School of Medicine, National University of Singapore (NUS Medicine), in collaboration with the Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore (LKCMedicine, NTU Singapore) and A*STARs Genome Institute of Singapore (GIS), has demonstrated that nano-sized vesicles released by red blood cells are a viable platform for delivering immunotherapeutic RNA molecules to suppress breast cancer growth and metastasis.

Published in the Journal of Extracellular Vesicles, the study successfully delivered RIG-I-activating RNAs using small, lipid membrane-bound particles released by red blood cells, called red blood cell extracellular vesicles (RBCEVs), to suppress cancer progression. The team had also discovered in earlier studies that these vesicles are ideal therapeutic carriers with a natural ability to deliver bioactive molecules to many cell types.

To further examine the function of RBCEVs in carrying a broader range of therapeutics to more cancer cell types, the team plans to conduct further research in collaboration with the National University Cancer Institute and Cancer Science Institute of Singapore.

Concurrently, RBCEV technologies are under intensive research at Carmine Therapeutics, an EVX Ventures company which aims to develop the next generation of gene therapy based on RBCEVs for treatments of rare diseases and cancer.

Read the original:
Singapore makes further advancement in cancer nanomedicine - BSA bureau

Nano Therapy Market 2022 Growth Is Expected To See Development Trends and Challenges to 2030 This Is Ardee – This Is Ardee

New York, United States Report Ocean published the latest research report on the Nano Therapy market. In order to comprehend a market holistically, a variety of factors must be evaluated, including demographics, business cycles, and microeconomic requirements that pertain precisely to the market under study. In addition, the Nano Therapy market study demonstrates a detailed examination of the business state, which represents creative ways for company growth, financial factors such as production value, key regions, and growth rate.

Key Companies Covered in theNano TherapyResearch areNanosphere Inc., Cristal Therapeutics, DIM, NanoMedia Solutions Inc., Luna, Nanobiotix, Sirnaomics Inc., Selecta Biosciences Inc., NanoBioMagnetics.n.nu, Nanospectra Biosciences Inc., Tarveda Therapeutics, Parvus Therapeutics, CytImmune Science Inc., Nanoprobes Inc., NanoBio Corporation, Smith and Nephewand other key market players.

TheNano Therapymarket revenue was $$ Million USD in 2016, grew to $$ Million USD in 2022, and will reach $$ Million USD in 2030, with a CAGR of % during 2022-2030.

The Centers for Medicare and Medicaid Services data estimates that the U.S. national healthcare expenditure surpassed US$ 4.1 trillion in 2020 and is forecast to reach US$ 6.2 trillion by 2028. According to the Commonwealth Fund, the U.S. expended nearly 17% of gross domestic product (GDP) on healthcare in 2018. Switzerland was the second-highest-ranking country, expending 12.2%. In addition, New Zealand and Australia devote only 9.3%.Request To Free Sample of This Strategic Report:-https://reportocean.com/industry-verticals/sample-request?report_id=mai284010

According to the U.S. Bureau of Labor Statistics, employment in healthcare fields is forecast to grow 16% from 2020 to 2030, much quicker than the standard for all occupations, counting about 2.6 million new jobs. This estimated growth is mainly due to an elder population, showing to greater demand for healthcare services. The median annual wage for healthcare practitioners and technical fields (such as registered nurses,0020physicians and surgeons, and dental hygienists) was US$ 75,040 in May 2021, which was greater than the median annual wage for all occupations in the economy of US$ 45,760.

Market Overview

Nano therapy is a branch of nanomedicine that involves using nanoparticles to deliver a drug to a given target location in the body so as to treat the disease through a process known as targeting.

GlobalNano TherapyMarket Development Strategy Pre and Post COVID-19, by Corporate Strategy Analysis, Landscape, Type, Application, and Leading 20 Countries covers and analyzes the potential of the global Nano Therapy industry, providing statistical information about market dynamics, growth factors, major challenges, PEST analysis and market entry strategy Analysis, opportunities and forecasts. The biggest highlight of the report is to provide companies in the industry with a strategic analysis of the impact of COVID-19. At the same time, this report analyzed the market of leading 20 countries and introduce the market potential of these countries.

Most important types of Nano Therapy products covered in this report are:Nanomaterial and Biological DeviceNano Electronic BiosensorMolecular NanotechnologyImplantable Cardioverter-Defibrillators

Most widely used downstream fields of Nano Therapy market covered in this report are:Cardiovascular DiseaseCancer TherapyDiabetes TreatmentRheumatoid ArthritisOthers

Top countries data covered in this report:United StatesCanadaGermanyUKFranceItalySpainRussiaChinaJapanSouth KoreaAustraliaThailandBrazilArgentinaChileSouth AfricaEgyptUAESaudi Arabia

SPECIAL OFFER (Avail an Up-to 30% discount on this report) :-https://reportocean.com/industry-verticals/sample-request?report_id=mai284010

Chapter 1 is the basis of the entire report. In this chapter, we define the market concept and market scope of Nano Therapy, including product classification, application areas, and the entire report covered area.

Chapter 2 is the core idea of the whole report. In this chapter, we provide a detailed introduction to our research methods and data sources.

Chapter 3 focuses on analyzing the current competitive situation in the Nano Therapy market and provides basic information, market data, product introductions, etc. of leading companies in the industry. At the same time, Chapter 3 includes the highlighted analysisStrategies for Company to Deal with the Impact of COVID-19.

Chapter 4 provides breakdown data of different types of products, as well as market forecasts.

Different application fields have different usage and development prospects of products. Therefore, Chapter 5 provides subdivision data of different application fields and market forecasts.

Chapter 6 includes detailed data of major regions of the world, including detailed data of major regions of the world. North America, Asia Pacific, Europe, South America, Middle East and Africa.

Chapters 7-26 focus on the regional market. We have selected the most representative 20 countries from 197 countries in the world and conducted a detailed analysis and overview of the market development of these countries.

Chapter 27 focuses on market qualitative analysis, providing market driving factor analysis, market development constraints, PEST analysis, industry trends under COVID-19, market entry strategy analysis, etc.

Access full Report Description, TOC, Table of Figure, Chart, etc. @:-https://reportocean.com/industry-verticals/sample-request?report_id=mai284010

Key Points:Define, describe and forecast Nano Therapy product market by type, application, end user and region.Provide enterprise external environment analysis and PEST analysis.Provide strategies for company to deal with the impact of COVID-19.Provide market dynamic analysis, including market driving factors, market development constraints.Provide market entry strategy analysis for new players or players who are ready to enter the market, including market segment definition, client analysis, distribution model, product messaging and positioning, and price strategy analysis.Keep up with international market trends and provide analysis of the impact of the COVID-19 epidemic on major regions of the world.Analyze the market opportunities of stakeholders and provide market leaders with details of the competitive landscape.

Table of Content:

For More Information or Query or Customization Before Buying, Visit @:https://reportocean.com/industry-verticals/sample-request?report_id=mai284010

Key Benefits for Industry Participants & Stakeholders

Key Questions Answered in the Market Report

Request Full Report :-https://reportocean.com/industry-verticals/sample-request?report_id=mai284010

About Report Ocean:We are the best market research reports provider in the industry. Report Ocean believes in providing quality reports to clients to meet the top line and bottom line goals which will boost your market share in todays competitive environment. Report Ocean is a one-stop solution for individuals, organizations, and industries that are looking for innovative market research reports.

Get in Touch with Us:Report Ocean:Email:sales@reportocean.comAddress: 500 N Michigan Ave, Suite 600, Chicago, Illinois 60611 UNITED STATESTel:+1 888 212 3539 (US TOLL FREE)Website:https://www.reportocean.com

Excerpt from:
Nano Therapy Market 2022 Growth Is Expected To See Development Trends and Challenges to 2030 This Is Ardee - This Is Ardee

New invisibility cloak for therapeutics: Holger Frey receives ERC Advanced Grant to support his innovative research – EurekAlert

image:Professor Dr. Holger Frey view more

Credit: photo/: private

Since the first PEGylated drug was developed in the 1980s, the so-called PEGylation has become a standard procedure in the pharmaceutical sciences. The technique involves concealing active biopharmaceuticals under a kind of "cloak of invisibility" by means of conjugation with the polymer polyethylene glycol (PEG). Consequently, they are not subjected to rapid degradation or undesirable attack by the immune system. The mRNA vaccines designed to protect against infection with the coronavirus are, for example, PEGylated. Unfortunately, problems with the concept are emerging, since an increasing number of individuals is developing antibodies against PEG, which in some instances can trigger severe allergic reactions. Professor Holger Frey of Johannes Gutenberg University Mainz (JGU) is currently developing a novel procedure intended to sidestep the drawbacks of PEGylation, but at the same time preserving its benefits. He has been awarded an ERC Advanced Grant worth EUR 2.5 million to support his research. An Advanced Grant is the EU's most richly endowed funding program, awarded by the European Research Council (ERC) to outstanding researchers. Holger Frey has been Professor of Organic and Macromolecular Chemistry at JGU since 2002 and is an internationally recognized expert in the field of polyether chemistry.

PEGylation a key strategy of current nanomedicine

Polyethylene glycol is a substance that is fairly ubiquitous. It is present in cosmetics, toothpaste, detergents, lithium-ion rechargeable batteries, foodstuffs, and textiles. PEG is employed in pharmaceutical technology and medicine as a carrier medium for active substances and in an extensive range of special applications. On the molecular level, PEG really comes into its own when it is conjugated with biopharmaceuticals and proteins to protect these through the process known as PEGylation. "PEGylation of many commercially available drugs is absolutely indispensable," emphasized Professor Holger Frey. "Without this, our bodies would identify the related active substances, including mRNA vaccines, as dangerous intruders and would rapidly degrade and excrete them. The camouflage effect has worked well for the past 30 years, but it looks like the magic is wearing off."

This is due to our immune system, which in many individuals no longer allows the polymer to circulate undetected in the bloodstream. The results of recent studies indicate that up 70 percent of the population in developed countries has antibodies against PEG; in the early 1980s, the corresponding percentage was just one to two percent. The result is that the immune system often quickly removes drugs conjugated with PEG from the blood circulation so that they are unable to develop their therapeutic effects. Moreover, there can also be intolerance and even severe allergic reactions effects that the technique was originally meant to prevent.

Research group to develop special PEG structures for use with medical drugs

One solution to this problem would be to devise new PEG-derived structures for use in medicine that differ from those exploited in everyday products. The purpose of the ERC-sponsored project RandoPEGMed is thus to create modified polymers for conjugation with medicinal agents. The basis will still be polyethylene glycol, but a PEG supplemented by additional building blocks. "What we are planning to do is break down the uniform structure by the insertion of randomly distributed irregularities," Frey clarified. "This should restore the masking effect, enabling the drugs to reach their intended destinations without being discovered by the immune system." Professor Holger Frey has many years of experience in this particular area. Over the past ten years and with the help of his team of 25 personnel, he has come up with a method that allows to precisely analyze polymer structures on the molecular level.

ERC Advanced Grants: Recognition for international top-level research

Holger Frey studied chemistry at the University of Freiburg and, following study periods in the USA and France, obtained his doctorate at the University of Twente in the Netherlands. He has been Professor of Organic and Macromolecular Chemistry at JGU since 2002. His research field is new polymer materials including polymers for medical and pharmaceutical use as well as bio-based, potentially sustainable materials that could be used as alternatives to plastics made from fossil raw materials. The results of his research have appeared in more than 400 original publications and review articles while he has also obtained more than 40 patents. In addition, he is an associate editor of Polymer Chemistry of the Royal Society of Chemistry, one of the leading journals in the field of polymers. Among the various undertakings he is involved in at JGU, he is also co-project coordinator of Collaborative Research Center 1066 that targets nanodimensional polymer therapeutics for tumor therapy. This is an interdisciplinary research network that involves significant collaboration between the fields of chemistry, the pharmaceutical sciences, and medicine.

ERC Advanced Grants are awarded to outstanding researchers to enable them to work on projects considered to be highly speculative due to their innovative approach, but which, because of this, can open up access to new approaches in the corresponding research field. Only researchers who have already made significant breakthroughs and have been successfully working for at least ten years at the highest levels of international research are eligible for the grant. The only criteria considered in awarding ERC funding are the academic excellence of the researcher in question and the nature of their research project. An ERC grant thus also represents an important acknowledgement of the recipient's individual achievements

Related links:https://erc.europa.eu/news/erc-2021-advanced-grants-results ERC Advanced Grants 2022 ;https://www.ak-frey.chemie.uni-mainz.de/ Research group of Professor Holger Frey ;https://sfb1066.de/ Collaborative Research Center 1066: Nanodimensional polymer therapeutics for tumor therapy

Read more: https://www.uni-mainz.de/presse/aktuell/9265_ENG_HTML.php press release "Nylon as a building block for transparent electronic devices?" (19 Aug. 2019)

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

Here is the original post:
New invisibility cloak for therapeutics: Holger Frey receives ERC Advanced Grant to support his innovative research - EurekAlert

Clinical Trial of Liposomes in Children’s Anticancer Therapy | IJN – Dove Medical Press

Introduction

Recently nanoscale drugs become an area of intense novel drug research.1,2 Several nanocarriers, including liposomes, have been utilized for cancer therapies.3,4 Among these, liposomes have attracted the most attention because of their potency of side effects,5 prolonged the retention-time for encapsulated payloads in cancer cells,6 effectively resolving some of the problems of off-target effects of anticancer drugs by improving the pharmacokinetic profiles and pharmacological properties of several agents.7,8

Clinical trials are the most effective strategy for evaluating the efficacy of a drug on a specific disease9,10 and are a critical step in the successful development of more effective drugs.11 Thus, exploring clinical trials, especially analyzing registered clinical trials, has become an important facet of research to help future clinical practice. ClinicalTrials.gov is a public trial registry provided by the US National Library of Medicine and the US Food and Drug Administration. Zarin et al12 postulated that the number of clinical trials in ClinicalTrials.gov accounted for more than 80% of all studies in the World Health Organizations International Clinical Trials Registry Platform. This proportion will likely expand with further implementation of the Food and Drug Administration Amendments Act (FDAAA 801), which expands the scope of mandatory clinical trial registration.13 Moreover, a joint statement from all International Committee of Medical Journal Editors (ICMJE) member journals indicated that clinical trials must be publicly registered in trials registries before they are considered for publication. Therefore, to better evaluate the breadth of liposome treatments for pediatric cancers, we performed a cross-sectional study to investigate the characteristic of registered trials in ClinicalTrials.gov regarding liposomes in childrens anticancer therapy.

A cross-sectional, descriptive study of clinical trials for LCAT registered on the ClinicalTrials.gov database was conducted. The trials were obtained from ClinicalTrials.gov using the advanced search function with the search term cancer for condition or disease and the term liposome for Other terms on December 30, 2021. All of the identified clinical trials were assessed to obtain records of all studies. Intervention and observation studies were all included. We used the age field as a filter, and we included trials explicitly designed for the child (birth 17 years of age). Next, we manually reviewed all of the trials and selected those using liposomal drugs for childrens anticancer therapy. Trials utilizing non-liposomal drugs were excluded. The following information and data were extracted: registered number, title, study type, conditions, interventions, locations, start date, the status of the trial, study results, study samples, participant ages, primary sponsor, location, primary purpose, phases of each trial, allocation, intervention model, masking and intervention. All trials were then further subclassified according to their study type. We used descriptive statistics to characterize trial categories. Frequencies and percentages were provided for categorical data. All analyses were performed using Microsoft Excel (Microsoft Office Excel 2010, Microsoft Corporation).

The initial search identified 1552 clinical trials on liposomes in cancer therapy registered on the ClinicalTrials.gov database through December 30, 2021. After using the age field (child; birth 17 years of age) as a filter, 352 trials focusing on liposomes in childrens anticancer therapy were included. After carefully reviewing all the information, 278 trials were not liposomal drugs and were excluded. Thus, a total of 74 registered trials focusing on liposomes in childrens anticancer therapy were subsequently included, including four observational studies and 70 intervention trials (Figure 1).

Figure 1 Flowchart of trial selection.

The basic characteristics of the included trials are shown in Table 1. Among the 74 eligible trials, 70 (94.6%) were interventional trials, and the 4 (5.4%) were observational trials. Half of these trials were initiated prior to 2007. Every five years, the number of initiated trials changed a little from 2007 to 2021. Most of the included trials (47.3%) have been completed, although only 23.0% of trials had available results in this database. The sources of funding were indicated for 40.5% of trials. The National Institutes of Health (NIH) was the second-largest contributor, accounting for 36.5% of included trials. North America was the most frequently identified study location (68.9%), followed by Europe (14.9%), Asia (12.2%), and other (4.1%).

Table 1 Characteristics of All Included Trials

Of the four observational trials, two were retrospective, and two trials were prospective. Of the 70 interventional trials, 63 (90.0%) were for treatment, 3 (4.3%) were for supportive care, 2 (2.9%) were for diagnostic, and 2 (2.9%) were for prevention. The allocation concealment was not clear in 48.6% of these studies. 21 (30.0%) trials were randomized, and 15 (21.4%) trials were non-randomized. More than half of the intervention models were single group assignments (52.9%), followed by parallel assignments (22.9%), and unknown (21.4%). Among the 70 interventional trials, the majority of trials (50, 71.4%) were without masking, 13 (18.6%) were with unknown masking, and 7 (10.0%) were with masking (1 single masking, 4 double maskings, and 2 quadruple maskings). 20 (28.6%) were phase 3 trials, 21 (30.0%) were phase 1 trials, and 17 (24.3%) were phase 2 trials. More than half of the trials recruited less than 50 participants, 12 trials (17.1%) recruited 100500 individuals, and 12 trials (17.1%) did not indicate the number of participants. The study design characteristics of interventional trials are displayed in Table 2.

Table 2 Study Design Elements of Interventional Trials (n = 70)

A total of 70 interventional trials investigated 17 liposomal drugs, mainly focused on organic chemicals (43/70, 61.4%). 32 trials (45.7%) investigated liposomal doxorubicin. Of these trials for liposomal drugs, the highest proportion was testing liposomal doxorubicin (45.7%), followed by liposomal vincristine (17.1%) and liposomal cytarabine (5.7%). Three trials investigated liposomal complex compounds, of which two trials were liposomal daunorubicin-cytarabine, and one trial was liposomal doxorubicin-daunorubicin. A summary of studied liposomal drugs for prevention is provided in Table 3.

Table 3 Overview of Drugs for Prevention

A total of 70 interventional trials investigated 17 liposomal drugs for 123 types of cancer. Of these cancers, the highest proportion was leukemia (15.4%), followed by lymphoma (9.8%) and ovarian cancer (8.9%). Detailed data is shown in Figure 2.

Figure 2 Overview cancer types assessed for liposomal treatment for prevention (n = 123). The following cancers appeared only once: advanced cancer, bone cancer, germ cell tumors, glioma, invasive pulmonary aspergillosis, kidney tumor, lung cancer, multiple myeloma, nasopharyngeal carcinoma, pancreatic cancer, pediatric cancer, plasma cell neoplasm, precancerous condition, and prostate cancer.

Liposomes have been extensively investigated for overcoming cancer drug resistance,14 cancer-targeted therapy,15 and as a sustained and controlled release drug delivery system.16 However, liposomes do have limited clinical utility due to properties such as uncontrollable drug release, instability in storage, and insufficient drug loading.17 Specifically, due to their small aqueous internal volumes, liposomes have a relatively low encapsulation efficacy for water-soluble drugs.18 Meanwhile, large-scale liposomes production with low batch-to-batch differences is a challenge for the industry, which ultimately delays the clinical translation of new products.19 In addition, recruitment of children is a persistent challenge for researchers seeking to include these populations in clinical trials.20 First, societal concerns and parental emotional involvement can act to delay or prevent certain types of paediatric research.20,21 Second, medical ethics and clinical trial design for children need further refinement.22 Thus, the number of trials of liposomes in childrens anticancer therapy has not increased significantly over time and clinical trials focusing on liposomes account for only about 4.77% (74/1552) of clinical trials on liposomes in cancer therapy. Liposomally-delivered drugs have predominantly been organic chemicals (43/70, 61.4%). For example, 32 trials (45.7%) investigated liposomal doxorubicin. These results were following previous literature reports on the efficacy of delivering doxorubicin this way. To enhance the solubility of a hydrophobic substance, lipid-based drug delivery systems, especially liposomes, are among the best candidates.2325

In this study, the highest proportion of cancer type for prevention in a children was leukemia (15.4%), and the highest proportion of liposomal drug was in liposomal doxorubicin (45.7%), followed by liposomal vincristine (17.1%) and liposomal cytarabine (5.7%). For decades, the standard of care for treating acute myeloid leukemia (AML) has been the combination of a nucleoside analog with an anthracycline.26,27 Vincristine and cytarabine are nucleoside, and doxorubicin is a type of anthracycline. This indicated that liposomal doxorubicin combined with vincristine or cytarabine for childhood leukemia is an important future direction for liposomes in childrens anticancer therapy.

High quality, adequately powered, masked, appropriately sized, and appropriately sized, and randomized clinical trials represent a critical priority for high-quality clinical trials.2830 However, only 30.0% of trials studied here were randomized, and the majority of trials (71.4%) were without masking. Previously, it has been suggested that efficient trial designs are essential for rare malignancies has randomized trials are less feasible.31 To address this, there are multiple strategies for, such using as a Bayesian posterior predictive approach,32 or using complex innovative design,33 a novel multi-arm, multi-stage (MAMS) design.34 Hearn et al35 discussed in depth this issue highlighting the need for decision-makers to avoid adopting entrenched positions about the nature of the trial design.

The authors declare there are no conflicts of interest regarding the publication of this paper.

1. Jiang X, Zheng Y-W, Bao S, et al. Drug discovery and formulation development for acute pancreatitis. Drug Deliv. 2020;27(1):15621580. doi:10.1080/10717544.2020.1840665

2. Guo S, Liang Y, Liu L, et al. Research on the fate of polymeric nanoparticles in the process of the intestinal absorption based on model nanoparticles with various characteristics: size, surface charge and pro-hydrophobics. J Nanobiotechnol. 2021;19(1):32. doi:10.1186/s12951-021-00770-2

3. Qi -S-S, Sun J-H, Yu H-H, Yu S-Q. Co-delivery nanoparticles of anti-cancer drugs for improving chemotherapy efficacy. Drug Deliv. 2017;24(1):19091926. doi:10.1080/10717544.2017.1410256

4. Kim K, Khang D. Past, present, and future of anticancer nanomedicine. Int J Nanomedicine. 2020;15:57195743. doi:10.2147/IJN.S254774

5. Fenske DB, Cullis PR. Liposomal nanomedicines. Expert Opin Drug Deliv. 2008;5(1):2544. doi:10.1517/17425247.5.1.25

6. Suntres ZE. Liposomal antioxidants for protection against oxidant-induced damage. J Toxicol. 2011;2011:152474. doi:10.1155/2011/152474

7. Landi-Librandi AP, Chrysostomo TN, Caleiro Seixas Azzolini AE, Marzocchi-Machado CM, de Oliveira CA, Lucisano-Valim YM. Study of quercetin-loaded liposomes as potential drug carriers: in vitro evaluation of human complement activation. J Liposome Res. 2012;22(2):8999. doi:10.3109/08982104.2011.615321

8. Mignet N, Seguin J, Chabot GG. Bioavailability of polyphenol liposomes: a challenge ahead. Pharmaceutics. 2013;5(3):457471. doi:10.3390/pharmaceutics5030457

9. Feizabadi M, Fahimnia F, Mosavi Jarrahi A, Naghshineh N, Tofighi S. Iranian clinical trials: an analysis of registered trials in International Clinical Trial Registry Platform (ICTRP). J Evid Based Med. 2017;10(2):9196. doi:10.1111/jebm.12248

10. Chen L, Su Y, Quan L, Zhang Y, Du L. Clinical trials focusing on drug control and prevention of ventilator-associated pneumonia: a comprehensive analysis of trials registered on ClinicalTrials.gov. Original research. Front Pharmacol. 2019;9. doi:10.3389/fphar.2018.01574

11. Jacobsen PB, Wells KJ, Meade CD, et al. Effects of a brief multimedia psychoeducational intervention on the attitudes and interest of patients with cancer regarding clinical trial participation: a multicenter randomized controlled trial. J Clin Oncol. 2012;30(20):25162521. doi:10.1200/JCO.2011.39.5186

12. Zarin DA, Ide NC, Tse T, Harlan WR, West JC, Lindberg DAB. Issues in the registration of clinical trials. JAMA. 2007;297(19):21122120. doi:10.1001/jama.297.19.2112

13. Tse T, Williams RJ, Zarin DA. Reporting basic results in ClinicalTrials.gov. Chest. 2009;136(1):295303. doi:10.1378/chest.08-3022

14. Bai F, Yin Y, Chen T, et al. Development of liposomal pemetrexed for enhanced therapy against multidrug resistance mediated by ABCC5 in breast cancer. Int J Nanomedicine. 2018;13:13271339. doi:10.2147/IJN.S150237

15. Riaz MK, Riaz MA, Zhang X, et al. Surface functionalization and targeting strategies of liposomes in solid tumor therapy: a review. Int J Mol Sci. 2018;19(1):195. doi:10.3390/ijms19010195

16. Yue P-J, He L, Qiu SW, et al. OX26/CTX-conjugated PEGylated liposome as a dual-targeting gene delivery system for brain glioma. Mol Cancer. 2014;13:191. doi:10.1186/1476-4598-13-191

17. Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release. 2015;200:138157. doi:10.1016/j.jconrel.2014.12.030

18. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8(1):102. doi:10.1186/1556-276x-8-102

19. Al-Amin MD, Bellato F, Mastrotto F, et al. Dexamethasone loaded liposomes by thin-film hydration and microfluidic procedures: formulation challenges. Int J Mol Sci. 2020;21(5):1611. doi:10.3390/ijms21051611

20. Cunningham-Erves J, Deakings J, Mayo-Gamble T, Kelly-Taylor K, Miller ST. Factors influencing parental trust in medical researchers for child and adolescent patients clinical trial participation. Psychol Health Med. 2019;24(6):691702. doi:10.1080/13548506.2019.1566623

21. Rentea RM, Oyetunji TA, Peter SDS. Ethics of randomized trials in pediatric surgery. Pediatr Surg Int. 2020;36(8):865867. doi:10.1007/s00383-020-04665-5

22. Nicholl A, Evelegh K, Deering KE, et al. Using a Respectful Approach to Child-centred Healthcare (ReACH) in a paediatric clinical trial: a feasibility study. PLoS One. 2020;15(11):e0241764. doi:10.1371/journal.pone.0241764

23. Nik ME, Malaekeh-Nikouei B, Amin M, et al. Liposomal formulation of Galbanic acid improved therapeutic efficacy of pegylated liposomal Doxorubicin in mouse colon carcinoma. Sci Rep. 2019;9(1):9527. doi:10.1038/s41598-019-45974-7

24. Laverman P, Boerman OC, Storm G, Oyen WJG. (99m)Tc-labelled Stealth liposomal doxorubicin (Caelyx) in glioblastomas and metastatic brain tumours. Br J Cancer. 2002;86(4):659661. doi:10.1038/sj.bjc.6600093

25. Wang G, Wang J, Wu W, Tony To SS, Zhao H, Wang J. Advances in lipid-based drug delivery: enhancing efficiency for hydrophobic drugs. Expert Opin Drug Deliv. 2015;12(9):14751499. doi:10.1517/17425247.2015.1021681

26. Chen EC, Fathi AT, Brunner AM. Reformulating acute myeloid leukemia: liposomal cytarabine and daunorubicin (CPX-351) as an emerging therapy for secondary AML. Onco Targets Ther. 2018;11:34253434. doi:10.2147/OTT.S141212

27. Preisler HD, Anderson K, Rai K, et al. The frequency of long-term remission in patients with acute myelogenous leukaemia treated with conventional maintenance chemotherapy: a study of 760 patients with a minimal follow-up time of 6 years. Br J Haematol. 1989;71(2):189194. doi:10.1111/j.1365-2141.1989.tb04253.x

28. Zwierzyna M, Davies M, Hingorani AD, Hunter J. Clinical trial design and dissemination: comprehensive analysis of ClinicalTrials.gov and PubMed data since 2005. BMJ. 2018;361:k2130. doi:10.1136/bmj.k2130

29. Zhang C, Kwong JSW, Yuan R-X, et al. Effectiveness and tolerability of different recommended doses of PPIs and H(2)RAs in GERD: network meta-analysis and GRADE system. Sci Rep. 2017;7:41021. doi:10.1038/srep41021

30. Oh ES, Fong TG, Hshieh TT, Inouye SK. Delirium in older persons: advances in diagnosis and treatment. JAMA. 2017;318(12):11611174. doi:10.1001/jama.2017.12067

31. Italiano A, Nanda S, Briggs A, et al. Larotrectinib versus prior therapies in tropomyosin receptor kinase fusion cancer: an intra-patient comparative analysis. Cancers. 2020;12(11):3246. doi:10.3390/cancers12113246

32. Dutton P, Love S, Faleti A, Hassan B. The use of Bayesian design in two trials in rare cancers. Trials. 2015;16(Suppl 2):P213. doi:10.1186/1745-6215-16-S2-P213

33. Blagden SP, Billingham L, Brown LC, et al. Effective delivery of Complex Innovative Design (CID) cancer trials-A consensus statement. Br J Cancer. 2020;122(4):473482. doi:10.1038/s41416-019-0653-9

34. Sydes MR, Parmar MKB, Mason MD, et al. Flexible trial design in practice - stopping arms for lack-of-benefit and adding research arms mid-trial in STAMPEDE: a multi-arm multi-stage randomized controlled trial. Trials. 2012;13:168. doi:10.1186/1745-6215-13-168

35. Hearn J, Keat N, Law K, Sharpe R. How cancer research UK is adapting to adaptive designs. Trials. 2011;12(Suppl 1):A6. doi:10.1186/1745-6215-12-S1-A6

See the original post here:
Clinical Trial of Liposomes in Children's Anticancer Therapy | IJN - Dove Medical Press

Global Advanced Functional Materials Market To Be Driven By The Surging Demand From Medical Sector In The Forecast Period Of 2021-2026 …

The new report by Expert Market Research titled, Global Advanced Functional Materials Market Report and Forecast 2021-2026, gives an in-depth analysis of the globaladvanced functional materials market, assessing the market based on its type, end-use, and major regions. The report tracks the latest trends in the industry and studies their impact on the overall market. It also assesses the market dynamics, covering the key demand and price indicators, along with analyzing the market based on the SWOT and Porters Five Forces models.

Request a free sample copy in PDF or view the report summary@https://www.expertmarketresearch.com/reports/advanced-functional-materials-market/requestsample

The key highlights of the report include:

Market Overview (2016-2026)

The growth in the global advanced functional materials market is induced by the medical device technology which is advancing at a rapid pace. With increased focus on imaging techniques, implantable devices, and regeneration technologyin medicine, drug delivery industrial equipment, and biomedical engineering, the adoption of advanced functional materials is increasing rapidly, that aims to augment growth of the market. Advanced functional materials supersede conventional materials by having superior characteristics such as durability, toughness, durability, and elasticity. The advanced functional material industry for low carbon emissions applications is anticipated to be driven by rising lightweight vehicles demandcombined with improved fuel efficiency.

Industry Definition and Major Segments

Usingeffective power and signaltransmission to every object, advanced functional materials serve to minimise total power usage. Thin conductors or interlinks used within advanced functional material-based mini electronics aid in countering signal propagation and power failure concerns associated with large PCBs and thick interconnects.

Explore the full report with the table of contents@https://www.expertmarketresearch.com/reports/advanced-functional-materials-market

Based on its types, the market is divided into:

Based on end-use, the market is divided into:

On the basis of region, the market is divided into:

Market Trends

In the years ahead, the manufacturing of lighter weight, handy, and adaptable substrate technological tools will boost adoption ofadvanced functional materials. One of the crucial industry trends in the advanced functional materials marketis the strong market for microelectronics andminiaturisation. The healthcare industry has a huge demand for advanced functional materials. In the industry, nanomaterials are the dominant type of material. The use of nano materials in the nanotechnological sector of the healthcare industry is consistently expanding. Nanomedicine is the use of nanotechnology to diagnose, monitor, deliver drugs, treat, and regulate biological systems. Although, an absence of expansion plans and technological innovation is anticipated to stymie the industrys growth over the forecast period.

Key Market Players

The major players in the market are Morgan Advanced Materials plc, KYOCERA Corporation, Hexcel Corporation, Nanophase Technologies Corporation, KURARAY CO., LTD, Murata Manufacturing Co., Ltd., and Henkel AG & Co. KGaA (OTCMKTS: HENKY), among others. The report covers the market shares, capacities, plant turnarounds, expansions, investments and mergers and acquisitions, among other latest developments of these market players.

About Us:

Expert Market Research is a leading business intelligence firm, providing custom and syndicated market reports along with consultancy services for our clients. We serve a wide client base ranging from Fortune 1000 companies to small and medium enterprises. Our reports cover over 100 industries across established and emerging markets researched by our skilled analysts who track the latest economic, demographic, trade and market data globally.

At Expert Market Research, we tailor our approach according to our clients needs and preferences, providing them with valuable, actionable and up-to-date insights into the market, thus, helping them realize their optimum growth potential. We offer market intelligence across a range of industry verticals which include Pharmaceuticals, Food and Beverage, Technology, Retail, Chemical and Materials, Energy and Mining, Packaging and Agriculture.

We also provide state-of-the-art procurement intelligence through our platform,https://www.procurementresource.com. Procurement Resource is a leading platform for digital procurement solutions, offering daily price tracking, market intelligence, supply chain intelligence, procurement analytics, and category insights through our thoroughly researched and infallible market reports, production cost reports, price analysis, and benchmarking.

Informes de Expertos (https://www.informesdeexpertos.com), the Spanish variant of Expert Market Research, is a platform that offers market research and consultancy services to a broad clientele base across Spanish speaking countries. With our primary focus on the Latin America and Spain markets, our research experts provide relevant and actionable insights into the markets and track major trends, economic developments, and global trade data.

Determined to bring client satisfaction, we make sure that our tailored approach meets the clients unique market intelligence requirements. Our syndicated and customized research reports cover a wide spectrum of industries ranging from pharmaceuticals and food and beverage to packaging, logistics, and transportation.

Media Contact

Company Name: EMR Inc.Contact Person: Sofia Williams, Corporate Sales Specialist U.S.A.Email: sales@expertmarketresearch.comToll Free Number: +1-415-325-5166 | +44-702-402-5790Address: 30 North Gould Street, Sheridan, WY 82801, USACity: SheridanState: WyomingCountry: United StatesWebsite:https://www.expertmarketresearch.com

*We at Expert Market Research always thrive to give you the latest information. The numbers in the article are only indicative and may be different from the actual report.

Visit link:
Global Advanced Functional Materials Market To Be Driven By The Surging Demand From Medical Sector In The Forecast Period Of 2021-2026 ...

Engineering nanomedicine for glutathione depletion …

Glutathione (GSH), the main redox buffer, has long been recognized as a pivotal modulator of tumor initiation, progression and metastasis. It is also implicated in the resistance of platinum-based chemotherapy and radiation therapy. Therefore, depleting intracellular GSH was considered a potent solution to combating cancer. However, reducing GSH within cancer cells alone always failed to yield desirable therapeutic effects. In this regard, the convergence of GSH-scavenging agents with therapeutic drugs has thus been pursued in clinical practice. Unfortunately, the therapeutic outcomes are still unsatisfactory due to untargeted drug delivery. Advanced nanomedicine of synergistic GSH depletion and cancer treatment has attracted tremendous interest because they promise to deliver superior therapeutic benefits while alleviating life-threatening side effects. In the past five years, the authors and others have demonstrated that numerous nanomedicines, by simultaneously delivering GSH-depleting agents and therapeutic components, boost not only traditional chemotherapy and radiotherapy but also multifarious emerging treatment modalities, including photodynamic therapy, sonodynamic therapy, chemodynamic therapy, ferroptosis, and immunotherapy, to name a few, and achieved decent treatment outcomes in a large number of rodent tumor models. In this review, we summarize the most recent progress in engineering nanomedicine for GSH depletion-enhanced cancer therapies. Biosynthesis of GSH and various types of GSH-consuming strategies will be briefly introduced. The challenges and perspectives of leveraging nanomedicine for GSH consumption-augmented cancer therapies will be discussed at the end.

See more here:
Engineering nanomedicine for glutathione depletion ...

Lipid-Based Nanoparticles in the Clinic and Clinical …

Vaccines (Basel). 2021 Apr; 9(4): 359.

1Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam; nv.ude.utdt@hnahtiahtihtgnaoh

2Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; ude.hsanom@syus.elletse

3Biomedical Engineering, Malone Engineering Center 402A, Yale University, 55 Prospect St., New Haven, CT 06511, USA; moc.liamg@87eel.koesgnuj

4Vietnam Academy of Science and Technology, Graduate University of Science and Technology, Hanoi 100000, Vietnam; nv.tsav.smai@iahiadneyugn

5Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 01 TL29 District 12, Ho Chi Minh City 700000, Vietnam

6Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; rk.ca.uoja@pdk

2Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; ude.hsanom@syus.elletse

Mariusz Skwarczynski, Academic Editor, Istvan Toth, Academic Editor, and Ralph A. Tripp, Academic Editor

5Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 01 TL29 District 12, Ho Chi Minh City 700000, Vietnam

Received 2021 Mar 23; Accepted 2021 Apr 6.

COVID-19 vaccines have been developed with unprecedented speed which would not have been possible without decades of fundamental research on delivery nanotechnology. Lipid-based nanoparticles have played a pivotal role in the successes of COVID-19 vaccines and many other nanomedicines, such as Doxil and Onpattro, and have therefore been considered as the frontrunner in nanoscale drug delivery systems. In this review, we aim to highlight the progress in the development of these lipid nanoparticles for various applications, ranging from cancer nanomedicines to COVID-19 vaccines. The lipid-based nanoparticles discussed in this review are liposomes, niosomes, transfersomes, solid lipid nanoparticles, and nanostructured lipid carriers. We particularly focus on the innovations that have obtained regulatory approval or that are in clinical trials. We also discuss the physicochemical properties required for specific applications, highlight the differences in requirements for the delivery of different cargos, and introduce current challenges that need further development. This review serves as a useful guideline for designing new lipid nanoparticles for both preventative and therapeutic vaccines including immunotherapies.

Keywords: lipid nanoparticles, liposomes, vaccines, immunotherapy, COVID-19

Nanomedicine is the convergence of nanotechnology, pharmaceutical, and biomedical sciences and has developed rapidly with the design of new nanoformulations for therapeutic purposes, imaging agents and theragnostic applications. Nanoformulation was defined by the Food and Drug Administration (FDA) that is the products in combination with nanoparticles ranging from 1100 nanometers (nm); or other formulations outside of this range showing dimension-dependent properties [1]. These formulations exhibit many advantages over free drug molecules, possessing an enhanced solubility and improved pharmacokinetics, efficacy, and minimal toxicity [1]. More than 50 nanopharmaceuticals have made it to the market consisting of diverse nanoformulations, with lipid nanoparticles being the frontrunner [1,2,3,4]. Lipid nanoparticles are multicomponent lipid systems typically containing a phospholipid, an ionizable lipid, cholesterol, and a PEGylated lipid [5]. The traditional type of lipid nanoparticles is liposomes which was first described in 1961 by the British haematologist, Alec D Bangham [6]. Liposomes were observed under the electron microscope when adding negative stain to dry phospholipids that assembled into spherical shape through a lipid bilayer. Later, in 1980, the first targeted liposomes, modified by active targeting ligands, were developed and led to significantly improve liposome capacity by increasing accumulation at the target tissues/organs/cells without releasing the drug to other sites [7]. As a result, the overall efficacy of these liposomes is improved compared to conventional liposomes. Though liposomes have been explored for 30 years as an effective carrier for a variety of drug molecules, it was only in the 1990s that the first Food and Drug Administration (FDA) approval came. This milestone was reached by Doxil, a stealth liposome encapsulating doxorubicin (a) and used for the clinical treatment of ovarian and metastatic breast cancer as well as various forms of myeloma [1]. Because of the encapsulation of doxorubicin inside PEGylated liposomes, the side effects of free doxorubicin, including chronic cardiomyopathy and congestive heart failure, were significantly mitigated [8]. In addition, the PEGylation of liposomes in Doxil supported prolonging the circulation time of this formula after administration [8]. Thereafter, the passive accumulation into tumours was achieved. Overall, Doxil has significant cardiotoxicity reduction and high anticancer ability compared to free doxorubicin due to the enhanced permeability and retention effect [9].

(a) Structure of FDA approved Doxil and Onpattro (patisiran) nanoparticlesthe first FDA approved liposome and lipid nanoparticle, Created in BioRender.com; (b) chemical structure of the lipids inDoxil and Onpattro.

The start of the 21st century marks the paradigm-shifting development of multi-component formulations for delivering oligonucleotides for gene therapies [2]. These oligonucleotides are macromolecules that exhibit higher therapeutic indexes than conventional chemotherapeutics, especially when the formulation is tailored to reach specific tissues [10]. The main challenge gene delivery facing is the instability of naked nucleic acids in physiological media [11]. The development of suitable formulations that guarantee sufficient in vivo stability as well as tissue targeting ability has therefore been crucial. This advancement was achieved in 2018, with the FDA approval of Onpattro ((Alnylam Pharmaceuticals, Inc., Cambridge, MA, USA) and Sanofi Genzyme (Cambridge, MA, USA)), consisting of siRNA encapsulated in lipid nano-particles (LNPs) (a) for the treatment polyneuropathy in people with hereditary transthyretin-mediated amyloidosis [12,13,14,15]. This LNP is made of (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4-(dimethylamino)-butanoate (DLin-MC3-DMA) lipid, disterarolyphosphatidychloline (DSPC), cholesterol and a PEG-lipid (PEG-DMG) that directs the particle in vivo towards the liver hepatocytes [15,16,17]. The ionizable cationic lipid in the LNP complexes with the nucleic acids in acidic media (pH~4) by electrostatic interaction. At physiological pH of 7.4, this formulation becomes neutrally charged and thereby more stealth which dampens the interaction with blood components. Upon internalization of these LNPs in cells, these structural lipids become positively charged, which promotes complexation with the negatively charged endo/lysosomal membrane. This interaction with cellular compartments then disrupts and releases the nucleic acid in the cytosol, where they can exert their effect. Studies have suggested that the structure and pKa of the ionizable lipids play a crucial role in the delivery efficiency of the cargo to the target cells [5,18,19]. For example, Dlin-MC3-DMA, the ionizable lipid in Onpattro (b), with a pKa of 6.44, has a 10-fold higher potency than (2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane, DLin-KC2-DMA) with a pKa of 6.7 [20,21]. However, it should be noted that subtle differences in the lipid structure can induce variations in the structural packing of LNPs, resulting in a different morphology and delivery profile. To improve the particle stability, DSPC, with two saturated acyl chains and a large head group, is incorporated into the formulation (b). These lipids produce a cylindrical geometry and play the role of helper lipids in the LNP to maintain the outer layer structure and increase the Onpattros formation [22]. The PEG-DMG lipids on the particle surface (a,b) prevent particle aggregation due to stealth properties and prolong the circulation time in vivo. Altogether, this LNP system has a solid core structure, a low surface charge at physiological media, and low immunogenicity [16]. This technology has sparked interest in further developing genetic therapies and novel delivery systems. Lipid-based nanoparticles are a particularly promising vehicle [11,23,24] for gene delivery with their higher biocompatibility than polymeric and inorganic nanoparticles, their inherent penetrating ability, their biodegradability, structural flexibility, and low immunogenicity [25,26]. These nanostructures can also be produced rapidly at large scale, which is a major advantage when moving into clinical trials and commercial applications [25].

Since the success of Doxil, Onpattro, and various other LNPs in clinical trials, a myriad of studies has been conducted to translate R&D efforts into commercial products. This review describes the various FDA-approved lipid-based nanoparticles to provide our perspectives on the opportunities and challenges of future developments. Particularly, we first aim to provide an overview of lipid-based nanoparticles and their structure/property relationship. Next, we survey lipid nanoparticles that have been successfully used for cancer drug delivery. We also discuss the more challenging delivery of genetic materials and, finally, its application in preventative vaccines against viruses as well as therapeutic vaccines against cancers, including immunotherapies.

Lipid-based nanoparticles are classified into 5 categories depending on the fabrication method and on the physicochemical properties of the formulations. These are liposomes, niosomes, transfersomes, solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) [25]. The main properties of each type of particle are described in and . In brief, liposomes, structured by phospholipids and cholesterol, have great drug protection and targeting abilities [27]. These properties lead to liposomes having reduced toxicity with improved efficacy over free drug in the systemic circulation [6]. The application of liposomes in dermal delivery is limited due to their lack of penetrating ability in the stratum corneum. Additional drawbacks include the poor encapsulation of hydrophilic drugs and the weak storage stability due to drug leakage in the media [28]. An alternative to liposomes is niosomes, formed by nonionic surfactants and cholesterol in aqueous conditions, and provide better stability and longer shelf life than their liposomal counterpart [27,29]. Moreover, their neutral shell exhibits compatibility when compared to positively charged liposomes. Despite their advantageous properties, niosomes also suffer from drug leakage and particle aggregation due to a lack of ionic repulsion, which has been a barrier to grant FDA approvals [29,30]. Transfersomes are elastic or deformable nanoparticles composed of phospholipids, edge activators (EA) and cholesterol [28]. The addition of an EA enhances their flexibility which promotes higher tissue permeation. Transfersomes show the highest penetration capability and entrapment efficiency for lipophilic molecules [28]. Their oxidative degradation and high material cost remain a challenge to produce this type of particle in large quantities. All these lipophilic bi/multi-layer nanoparticles with hydrophilic core are able to encapsulate both hydrophilic and hydrophobic drugs without chemical intervention. Hydrophobic drugs are preferentially sandwiched in the external bilayer, while hydrophilic molecules are incorporated in the aqueous cavity (). These particles are therefore considered an effective carrier for a wide spectrum of drugs, with sufficient protection, sustainable drug release, and improved bioavailability.

Schematic representation of the five categories of lipid-based nanoparticles: Liposomes, niosomes, transfersomes, solid lipid nanoparticles (SLNs) and the nanostructured lipid carriers (NLCs). Created in BioRender.com.

Summary of main characteristics of common lipid-based nanoparticles.

The drawbacks related to their limited stability, toxicity, low loading capacity, and convoluted manufacturing has sparked the interest of researchers to develop a new class of particles became [25,29], mainly aiming at improving the encapsulation efficiency [12]. Solid lipid nanoparticles (SLNs, ) are made of solid fats and surfactants to form a spherical nanoparticle with a solid lipid core and a monolayer shell [33,34]. They exhibit greater encapsulation efficiency for hydrophobic drugs than liposomes since they lack an aqueous core. This rigid core also improved the stability of SLNs compared to liposomes and polymeric nanoparticles. Indeed, SLNs in aqueous media could be stored for at least 3 years [35]. The production of SLNs does not require organic solvents, which eliminates the risk of toxicity caused by solvent residues. Additionally, the large-scale production and excellent reproducibility of SLNs are important properties for downstream commercial and clinical applications [29]. Following on SLNs, nanostructured lipid carriers (NLCs) have been developed to enhance drug encapsulation and prevent drug leakage. NLCs are composed of an unstructured lipid interior and a monolayer surfactant periphery (). The core is made of a mixture of solid and liquid lipids that form an imperfect crystal interior to increase the drug loading, while SLNs are saturating the drug loading due to a solid crystal lattice. In addition, this liquid phase in NLC inhibits drug release during storage [36].

Moreover, lipid-polymer hybrid nanoparticles have been studied to create synergies between lipid-based nanoparticles and polymeric particles. Indeed, Zhang et al. made a comparison between lipid-based nanoparticles, polymeric nanoparticles and the hybrid nanoparticles loaded with cisplatin, where the in vivo data suggests that cisplatin-loaded lipid-polymer hybrid nanoparticles inhibited the ovarian carcinoma most effectively [37]. Additionally, these hybrid particles have also been considered in gene delivery applications [24,38]. By optimizing the properties of lipid-based nanoparticles, the safety, encapsulating capacity, stability, pharmacokinetics, bio-distribution, and therapeutic benefit can be controlled as a result [39].

For the delivery of oligonucleotides, nanoparticles need to encapsulate sufficient amounts of nucleic acid and have specific tissues targeting properties [12,13]. Thus, optimization of lipid-based nanoparticle is key for tailoring the delivery to the site of action [39]. Structural determinants, such as the particle size, surface charge, PEGylation, and surface modification by targeting ligands have shown to be critical elements in governing the delivery efficiency of these nanoparticles [13,24,39].

Small size nanoparticles have been shown to facilitate transport in blood and lymphatic capillaries as well as uptake in tissues [13,40]. More specifically, nanoparticles <10 nm are optimal for diffusion into blood capillaries, whilst particles of 10100 nm are favorably uptaken into lymphatic capillaries by convection. When the dimensions are 100200 nm their ability to diffuse rapidly is reduced [40]. A study by Oussoren et al. demonstrated that 40 nm liposomes had higher lymphatic uptake than larger particles after subcutaneous injection [41]. Specifically, 76% of the injected dose of these 40 nm liposomes was taken by the lymph nodes, while larger liposomes remained at the site of subcutaneous injection [41]. For liver targeting, after systemic administration, only particles <100 nm, were able to diffuse through the liver fenestrae to reach hepatocytes and hepatic stellate cells [42,43]. These results indicate that small-sized liposomes enhance lymph node transitivity, and crucially penetrate into liver fenestrae for targeting hepatocytes. For large-sized liposomes (>150 nm), these nanoparticles were taken up by the antigen-presenting cells (APCs) at an injection site, then were carried to the lymph nodes [44]. It was demonstrated that they gained a higher cell affinity within the lymph nodes [44].

In the case of the charge effect of lipid-based nanoparticles, this was a hard aspect to reach a general conclusion. Mai et al. reported that an anionic and cationic liposome exhibited significantly higher association with B cells than uncharged liposome within microvascular network [45]. This phenomenon was explained by the similar complemental proteins of anionic, cationic liposome shells with those being able to interact with B cells. Cationic liposomes showed the highest levels of interaction and internalization by B cell receptor. Cationic liposomes were in situ decorated with opsonins in blood which is recognized by human immune cells. Anionic liposomes were found to mostly attach on the surface of B cells. This finding suggests that charged liposomes might be suitable for vaccine application, while neutral particles might be more suitable for the delivery of chemotherapies [45]. Nakamura et al. found that 30 nm negatively charged LNPs were able to target the lymph node more effectively than positively charged and neutral particles [40]. The results showed that 2030% of the B220+ cells in the LN were DiD-labelled from the neutral and positively charged LNPs, whereas almost 80% of the B220+ cells were labelled with the negatively charged LNPs [40]. Retention of LNPs is at the site of injection was shown to be more pronounced with cationic particles compared to neutral and anionic particles [13,46]. This effect is due to the high electrostatic interaction between the cationic LNPs and negatively charged tissues. Additionally, cationic LNPs have been reported to bind nonspecifically with plasma proteins and have been linked to higher immunogenicity [47]. Taken together, positively charged LNPs have strong cellular affinity but have limited efficacy [47], while negatively charged LNPs show to be transported to lymph node effectively. To take advantage of this charge aspect, charge-reversible LNPs have been designed by Hirai et al. to achieve the best of both worlds in gene delivery [48]. These LNPs, composed of dipalmitoylphosphatidylcholine (DPPC), cholesterol, and dioleoylglycerophosphate-diethylenediamine conjugate (DOP-DEDA) are positively charged at pH of 6.0, neutral at pH of 7.4 and negatively charged at pH of 8.0. This system is neutral in the bloodstream to minimize degradation by plasma proteins and protect the encapsulated cargo. When circulating in the bloodstream, these DOP-DEDA-LNP vehicles bind to apolipoproteins (e.g., apoE3) at their hydrophobic lipid regions, which promotes their uptake by cancerous cells via both clathrin and caveola-mediated endocytosis pathways. In the endo-lysosomal compartment, the pH is low, which leads to DOP-DEDA-lipid nanoparticles being positively charged for enhanced cytosolic penetration in target cells () [48]. These LNPs with pH-dependent charge-invert properties are thought to be a safe and effective vector to induce RNAi-mediated gene-silencing [48]. Another strong determinant for the potency of LNPs is the lipid pKa. A series of studies have shown that a pKa of 6.4 is optimal for maximizing the transfection for siRNA-LNPs [49,50]. This optimal pKa is also changed in the case of mRNA, with an optimal range of 6.66.8 [49].

Impact of pH on the protonation and structure of charge-reversible lipid-based nanoparticles encapsulating siRNA. These lipid nanoparticles become positive charge at pH of 6.0, neutral at pH of 7.4 and a negative charge at pH of 8.0 gained due to an ionizable lipid of di-oleoylglycerophosphate-diethylenediamine conjugate (DOP-DEDA). Used with permission from [48].

As a strategy for improving the targeting capacity, PEGylation of lipid-based nanoparticles has gained much interest in an attempt to reduce the clearance of particles from the bloodstream, and thereby increasing their retention and uptake in tissues into targeting tissues/organs. Studies of Moghimi have revealed that the PEGs modifying anionic liposomes were able to achieve better clearance at the site of subcutaneous administration and higher retention in the lymph nodes compared with bare liposomes. In the case of PEG length, the shorter PEG chains gained lower clearance but higher retention in the lymph nodes compared with the longer PEG modified liposomes [51]. Structurally, reports have shown that using a linear or branched PEG chain can significantly dictate the targeting behaviour and transfection ability of LNPs. Truong group synthesized three LNPs with either Tween 80 (2-[2-[3,5-bis(2-hydroxyethoxy)oxolan-2-yl]-2-(2-hydroxyethoxy)ethoxy]ethyl (E)-octadec-9-enoate), Tween 20 (2-[2-[3,4-bis(2-hydroxyethoxy)oxolan-2-yl]-2-(2-hydroxyethoxy)ethoxy]ethyl dodecanoate) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) (DSPE-PEG) to encapsulate pDNA and investigate their targeting ability, stability and extent of transfection [52]. Tween 20 and Tween 80, both non-ionic surfactants are made of saturated carbon tails with similar branched PEG architecture, compared to the linear structure of DSPE-PEG (). Despite these structural differences, the LNPs exhibited similar stability over a 3-week period. In vivo transfection studies demonstrated that particles with the shorter Tween 20 were able to target the lymph nodes more efficiently. The longer Tween 80, on the other hand, formed LNPs that targeted the spleen but with lower efficiency. LNPs with the linear DSPE-PEG showed predominantly localized transfection at the injection site. Altogether, this study demonstrated that PEGylation of LNPs with branched PEG is a viable approach to target organs with effective transfection selectively [52]. It is hypothesized that, aside from the molecular weight and structure, the PEG density on the particle is also an important factor. Across the three types of LNPs, PEGylation of positively charged lipid nanoparticles showed much difference than bare ones [52]. PEGylation prevented positively charged lipid nanoparticles from high retention at the site of administration. Whilst PEGylation has been proven useful to prevent premature clearance of particle from the systemic circulation, the production of anti-PEG antibodies has emerged as detrimental co-lateral damage. Recently, efforts have been directed towards alternative strategies with biocompatible polymers to prevent the production of anti-PEG antibodies after the first dose that would otherwise lead to a loss of therapeutic efficacy with potential for adverse effects upon subsequent doses [53]. Concurrently, Chen et al. found that incorporation of 4 mol% dexamethasones in lipid-based nanoparticles was able to suppress the immune responses and antibody production after injection [26], which might be promising for the development of pegylated lipid nanocarriers.

Chemical structure of targeting lipids? DSPG (1,2-distearoyl-sn-glycero-3-phospho-rac-glycerol), DMPC (1,2-dimyristoyl-sn-glycero-3-phosphocholine), DMPG (1,2-dimyristoyl-sn-glycero-3-phosphoglycerol), SPH (sphingomyelin), DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine), DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine), EPC (1,2-dioleoyl-sn-glycero-3-ethylphosphocholine), EPG (L--phosphatidylglycerol), DPPC (dipalmitoylphosphatidylcholine), DPPG ([3-[2,3-dihydroxypropoxy(hydroxy)phosphoryl]oxy-2-hexadecanoyloxypropyl]hexadecanoate, cholesterol, Tween 80 (2-[2-[3,5-bis(2-hydroxyethoxy)oxolan-2-yl]-2-(2-hydroxyethoxy)ethoxy]ethyl (E)-octadec-9-enoate), Tween 20 (2-[2-[3,4-bis(2-hydroxyethoxy)oxolan-2-yl]-2-(2-hydroxyethoxy)ethoxy]ethyl dodecanoate), ATX-1 (one of the LUNAR lipids of Arturus Therapeutics, Inc., San Diego, CA, USA) and DSPE-PEG (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol)).

Aside from relying on prolonged circulation and passive cellular uptake of nanoparticles, formulation scientists have realized that, by conjugation of a receptor ligand to the particle surface, uptake into target cells could be enhanced. Modification of lipid-based nanoparticles with targeting ligands is a key strategy for efficiently targeting delivery systems into the lymph nodes or other desired tissues/cells. For example, Vu et al. functionalized liposome surface with Hemagglutinin Antigen (HA) to improve antibody production efficiency [54]. This work showed that HA-functionalized liposome could cross the barrier cells in the lymph node and enhance germinal center formation and follicular helper T Cell Immunity. Monoclonal antibodies (mAbs) are also a promising avenue for conjugation onto nanoparticle surfaces via covalent bonds for leukocyte targeting [39]. Veiga et al. conjugated anti-Ly6C antibodies on lipid-based nanoparticles loaded with siRNA to form the ASSET platform (Anchored Secondary scFv Enabling Targeting) [55]. This formulation was able to selectively target inflammatory leukocytes in vivo, as shown in other studies [56,57]. Similarly, solid tumors often display expressed higher levels of p32 than non-cancerous tissues. To take advantage of this, linTT1 (AKRGARSTA) and LyP-1 peptides have been developed, that can bind to p32 with strong affinity [58]. These peptides were therefore utilized as targeting ligand for tumor homing. Indeed, Hunt et al. used linTT1 as targeting ligand in peritoneal carcinomatosis, and found that these nanoparticles could be absorbed to a greater extent than non-targeted particles in peritoneal tumors in mice [59]. Slik et al. also demonstrated a similar result using linTT1 as a targeting ligand for tumor homing in vitro and in vivo [58]. For the treatment of neurological disorder, targeting the nervous system is a requisite. Kuo et al. designed a formulation with amphiphilic solid lipid nanoparticles decorated with the Ln5-P4 (PPFLMLLKGSTR) peptide (Ln5-P4-ASLNs) for co-delivery of nerve growth factor (NGF) and retinoic acid (RA) [60]. Ln5-P4 bound the 31 integrin and supported cell adhesion and spreading to guide the differentiation of induced pluripotent stem cells toward neurons. This formulation proved to increase the survival rate of induced pluripotent stem cells and the generation of mature neurons as a potential treatment of neurodegenerative diseases and nerve injury in regeneration medicine. In other studies, selectivity for neuronal cells was improved by attachment of the rabies virus glycoprotein (RVG) peptide, a fragment of from the rabies virus glycoprotein, on the surface of siRNA-loaded exosomes [61]. This formulation showed positive effects in a mouse model of Alzheimers disease. To enhance the cellular uptake in dendritic cells for enhanced immune response, lipid-based nanoparticles have been conjugated with cell-penetrating peptides such as R8 and GALA [62]. Many other ligands of dendritic cell receptors (for review see [63]) and of liver cells (for review see [17]) have been developed for targeting purposes and are listed below. Fusogenic peptides such as DOPE have also shown to improve membrane fusion, and thus cell uptake [64]. Transferrin, folic acid, and antibodies can be used as a ligand for lipid-based nanoparticles to target cancer cell receptors [65,66,67,68]. For the objective of improving siRNA transfection, NP3.47, an inhibitor of the Niemann-Pick type C-1 protein (NPC-1), was conjugated to the surface of the lipid-based nanoparticle. NP3.47 promoted the accumulation of siRNA-LNPs in late endosomes/lysosomes up to 3-fold higher than unmodified lipid-based nanoparticles [69]. Due to the increased trapping of NP3.47-LNP-siRNA systems in late endosomes, enhanced opportunities for endosomal escape can be gained from this work for the delivery of siRNA and other oligonucleotide [69].

In recent years, lipid-based nanoparticles have emerged as the most effective carrier for the delivery of cargo to target cells, which have translated into clinical success. The analytical characterization, the basic technological concepts and highlights have been reviewed extensively before [17,38,39,70]. The current review will focus on the translation of lipid-based nanoparticles into the clinic.

For almost 30 years, liposomes have been blossoming in clinical applications. Twenty-one liposomal products have been approved, encapsulating different small molecule drugs. The clinical success of Doxil has given rise to the approval of many new nanodrugs by the FDA, such as Abelcet, AmBisome, DaunoXome, Depocyt, Inflexal V, Myocet, Visudyne, DepoDur, DepoCyt, Marqibo, Mepact, Exparel, Lipodox, Onivyde, Doxorubicin, Nocita, Vyxeos, Shingrix, LipoplatinTM, and Arikayce [1,2,6,71]. These formulations are not only used in oncology but also in fungal infections and pain management. Each liposome formulation is described in greater detail in , with their respective structural lipids shown in . There are four different liposomal products with doxorubicin (Doxil, Myocet, Lipodox, and Liposomal doxorubicin) that are indicated for breast neoplasms. Of these products, Myocet is a conventional formulation, while Doxil, Lipodox, and Doxorubicin are stealth, pegylated liposomes. Aside from these three stealth liposomes, there is only one other product, Onivyde that contains stealth properties by PEGylation. For anaesthetic applications, there are two different liposomes approved that encapsulate bupivacaine. For life-threatening fungal infections, there are also two liposomal formulations on the market that encapsulate amphotericin B. Marqibo, a conventional liposome loaded with vincristine, was approved in 2009 for the treatment of acute lymphoblastic leukaemia and in 2012 for hematologic malignancy and solid tumor treatment. In addition, other drugs including daunorubicin, cytarabine, verteporfin, morphine, mifamurtide, irinotecan, cytarabine, and amikacin were formulated in liposomes. Among them, only Curosurf encapsulated surfactant protein B and C (SP-B and SP-C) in a conventional liposome for the treatment of respiratory distress syndrome (RDS) in premature infants. An atypical formulation is Vyxeos, which has two drugs incorporated into the liposomes, thereby exploiting the synergistic effect of daunorubicin and cytarabine to treat acute myeloid leukaemia more effectively. In summary, within this period, there were 13 chemotherapeutics and one protein formulated in conventional and stealth liposomes successfully commercialized.

Overview of the approved liposomes in EU and US.

Aside from those formulations, there are many other liposomes indicated for chemotherapy in currently in clinical trials (). MBP-426 of Mebiopharm Co., Ltd. (Tokyo, Japan) is an oxaliplatin-encapsulated transferrin-conjugated N-glutaryl phosphatidylethanolamine-liposome and is indicated for gastric, oesophageal and gastro-oesophageal adenocarcinoma. The phase I clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT00355888","term_id":"NCT00355888"}}NCT00355888) of MBP-426 was completed [7], with phase IIa/b starting ({"type":"clinical-trial","attrs":{"text":"NCT00964080","term_id":"NCT00964080"}}NCT00964080) for characterization of the safety profile in combination with leucovorin and fluorouracil. Mebiopharm Co., Ltd. has also developed other products including MBP-Y003, MBP-Y004, and MBP-Y005 in preclinical stages which are transferrin-conjugated liposomes loaded with methotrexate, docetaxel, and gemcitabine, respectively [73]. These four products are designed with a transferrin ligand for targeting receptors that are overexpressed in cancerous tissues.

Liposomal formulations in clinical trials.

ThermoDox of Celsion is a heat-sensitive liposome loaded with doxorubicin for the treatment of hepatocellular carcinoma. The thermosensitive lipid is able to change structure at 4045 C to release doxorubicin rapidly in the tumor through radiofrequency ablation. Although four commercialized liposomes loading doxorubicin were already launched into the market successfully, ThermoDox is a new product with advanced characteristics, that showed a 5-fold release in doxorubicin concentration at the tumor site when compared to Doxil [74]. Phase III clinical trials of ThermoDox in combination with standardized radiofrequency ablation ({"type":"clinical-trial","attrs":{"text":"NCT02112656","term_id":"NCT02112656"}}NCT02112656) have been completed [7,74].

MM-302 of Merrimack Pharmaceuticals, is a stealth liposome modified with antibodies targeting the human epidermal growth factor receptor 2 (HER2) and loaded with doxorubicin [75], which has applied for phase 1 clinical trial in 2011. MM-302 aimed at overcoming the limitations of doxorubicin related to cardiac toxicity and to the ineffective targeting of cancerous cells. MM-302 was assessed in combination with trastuzumab or trastuzumab plus cyclophosphamide to treat advanced HER2-positive breast cancer. The promising data of phase 1 clinical trials inspired MM-302 to move to phase 2. However, the efficacy results did not show significant benefit compared to comparator treatments, which led to Merrimack discontinuing further trials with MM-302 in 2016 [76].

SPI-77 developed by Sequus Pharmaceuticals (Johnson & Johnson) is a cisplatin-encapsulated stealth liposome, developed for the treatment of recurrent ovarian cancer [77] and stage IV non-small cell lung cancer (NSCLC) [78]. SPI-77 is hypothesized to mitigate the systemic toxicity of cisplatin and to achieve a high delivery capacity. However, due to lack of significant data, in phase 1 and phase 2 clinical trials, the manufacturer decided to halt further trials [77,78,79].

The liposome-encapsulated mitoxantrone (LEM) from INSYS Therapeutics Inc is made of lyophilized lipids mixed with mitoxantrone salt (under the commercial name of Novantrone). This formulation was generated to improve the safety and efficacy of free Novantrone. LEM entered phase 1 clinical trials in 2001 with an identifier of {"type":"clinical-trial","attrs":{"text":"NCT00024492","term_id":"NCT00024492"}}NCT00024492. Patients with advanced solid tumors (40 participants) were recruited for intravenous injection of LEM. The results from the blood pharmacokinetics and tumor observation were completed in 2004, but no results were posted.

OSI-211, a liposome encapsulating lurtotecan, was developed for treatment of recurrent small-cell lung cancer. OSI-211 was clinically tested ({"type":"clinical-trial","attrs":{"text":"NCT00046787","term_id":"NCT00046787"}}NCT00046787) by Astellas Pharma Inc. (Chuo City, Tokyo, Japan) The University of Pittsburgh and ALZA company developed S-CKD602 and completed phase 1 clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT00177281","term_id":"NCT00177281"}}NCT00177281) to determine the maximum tolerated dose as well as the safety in patients with advanced tumors. S-CKD602 is a PEGylated liposome encapsulating CKD-602, a camptothecin analogue inhibiting topoisomerase I, with liposomal formulation consisting of N-(carbonyl-methoxypolyethylene glycol 2000)-DSPE and DSPC [80]. Another liposome named LEP-ETU also entered phase 1 clinical trials ({"type":"clinical-trial","attrs":{"text":"NCT00080418","term_id":"NCT00080418"}}NCT00080418). LEP-ETU is a liposome formed by DOPC, cholesterol and cardiolipin that encapsulates paclitaxel to treat ovarian, breast and lung cancers [81]. Topotecan Liposomes Injection (TLI) also entered Phase 1 clinical trials ({"type":"clinical-trial","attrs":{"text":"NCT00765973","term_id":"NCT00765973"}}NCT00765973) to test its safety and efficacy. Up till now, these four products have completed their clinical trial phase, but no updated information has been released.

Three products, including LiPlaCis, INX-0076 and TLD-1 have been in progress of clinical trials. INX-0076 was formulated into a liposome with topotecan for advanced solid tumors. LiPlaCis, developed for treatment of advanced solid tumours, is a liposomal formulation, incorporating cisplatin, which is composed of lipids with degradation properties controlled by the sPLA2 enzyme for a tumour-triggered release mechanism [82]. TLD-1 is a novel liposome encapsulating doxorubicin, indicated for advanced solid tumors [83].

Recently, nucleic acid therapeutics, such as small interfering RNAs (siRNA), small activating RNAs (saRNA), and messenger RNA (mRNA), have gained much traction and have been at the forefront of medicine with their potential in delivery efficiency and treatment of a wide range of diseases [87,88]. However, these genetic drugs are prone to rapid degradation by serum endonucleases. To protect them, liposomes or lipid nanoparticles (LNPs, including SLNs and/or NLCs) have been increasingly utilized as a delivery system which have sufficient encapsulating capacity and which are capable of targeting tissues and cells [89]. A series of ionizable lipids have been designed for gene delivery, each with their own pKa and structural properties, such as 1,2-dioleoyl-3-dimethylaminopropane (DODAP, pKa of 6.6), 1,2-dilinoleoyl-3-dimethylaminopropane, 1,2- dilinoleyloxy-3-dimethylaminopropane (DLin-DMA, pKa of 6.8), 2,2-dilinoleyl- 4-dimethylaminomethyl-[1,3]-dioxolane (DLin-K-DMA, pKa of 5.94), 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane (DLin-KC2-DMA, pKa of 6.68) () [18], and (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4-(dimethylamino)-butanoate (Dlin-MC3-DMA, pKa of 6.44) (). Aside from the pKa value, the linker between the head group and alkyl chains is also a determinant for delivery efficiency. The ketal linker was demonstrated to be the best candidates when compared to ester- and alkoxy-linkers [18]. Dlin-K-DMA with significant in vivo silencing of factor VII was compared to other lipids without ketal linkers [18]. A series of linker optimization, resulted in DLin-MC3-DMA, which is now considered as a promising candidate for genetic drug delivery and able to launch in the clinic. Dlin-MC3-DMA exhibited a 10-fold higher potency than Dlin-KC2-DMA for hepatic gene silencing in vivo [21]. An illustration of this lipids potency is provided with Onpattro (). The second siRNA therapeutic to receive FDA approval was GIVLAARITM (givosiran, ALN-AS1) in November 2019 [90] and a market authorization from the European Committeea synthetic siRNA targeting the ALAS1 gene in hepatocytes. This therapy is prescribed for adult patients with acute hepatic porphyria, a genetic disorder resulting in the buildup of toxic porphyrin molecules which are formed during the production of heme. This RNA therapy, however, is composed naked nucleotide-modified siRNA without a carrier, but in the presence of a targeting ligand that directs the drug towards the liver.

Chemical structures of the most common ionizable cationic lipids: 1,2-dioleoyl-3-dimethylaminopropane (DODAP), 1,2-dilinoleoyl-3-dimethylaminopropane, 1,2- dilinoleyloxy-3-dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl- 4-dimethylaminomethyl-[1,3]-dioxolane (DLin-K-DMA) and 2,2-dilinoleyl-4-(2- dimethylaminoethyl)-[1,3]-dioxolane (DLin-KC2-DMA).

An LNP platform developed by Arcturus Therapeutics, Inc. (San Diego, CA, USA) for RNA delivery was named as lipid-enabled and unlocked nucleomonomer agent modified RNA (LUNAR). LUNAR is made of a proprietary ionizable amino lipids (58 molar %, ATX, ), a phospholipid 1,2-distearoyl-sn-glycero-3-phosphocholine (7 molar %, DSPC), cholesterol (33.5 molar %) and DMG-PEG2000 (1.5 molar %) [91]. The ATX lipids are similar to a lipid family developed by Alnylam/Acuitas that can be modified to target specific cells type or tissues for a variety of indications. Unlike the conventional cationic lipids, the ATX lipids are degraded under physiological conditions through the breaking of the ester linkages, thereby facilitating rapid degradation for faster metabolism and better safety profile [92]. It was shown that LUNAR was employed to encapsulate the human FIX (hFIX) mRNA for treatment of hemophilia B in a preclinical setting [93]. The efficacy of LUNAR in mRNA delivery was 5-fold higher than other lipid carriers with DLin-MC3-DMA or heptatriaconta-6,9,28,31-tetraen-19-yl-4-(dimethylamino)butanoate) (MC3). Additionally, the LUNAR formulation did not elicit an adverse immune response, such as an increase of liver enzymes that are markers for acute liver toxicity [93].

Aside from Onpattro as the first commercial RNA product, GIVLAARITM and mRNA-LUNAR in preclinical studies, a variety of RNA-LNP have now entered clinical trials (). TKM-080301 was studied and produced by Arbutus Biopharma Corporation for the treatment of solid tumors, such as gastrointestinal neuroendocrine tumors or adrenocortical carcinoma (ACC) [94]. TKM-080301 is composed of a siRNA encapsulated in LNPs that can target polo-like kinase 1 (PLK1), which regulates critical aspects of tumor progression. A phase I/II clinical study was conducted ({"type":"clinical-trial","attrs":{"text":"NCT01262235","term_id":"NCT01262235"}}NCT01262235) with promising safety and anti-tumor efficacy data [94,95]. After testing on 16 patients at 0.6 or 0.75 mg/kg/week for 18 cycles, it was confirmed that TKM-080301 could increase PLK1 expression and inactivate the target in ACC. Later, in 2018, TKM-080301 was clinically evaluated for safety, pharmacokinetics and preliminary anti-tumor activity in patients with advanced hepatocellular carcinoma ({"type":"clinical-trial","attrs":{"text":"NCT02191878","term_id":"NCT02191878"}}NCT02191878) [96,97]. In this early-phase study with 43 patients, the antitumor effect of TKM-080301 was limited. Consequently, TKM-080301 did not continue for further evaluation as a single agent for the treatment of HCC, and clinical studies have been terminated [97].

List of RNA-encapsulated lipid nano-particles (LNPs) with indication and clinical trial information.

Dicerna Pharmaceuticals (Lexington, Massachusetts MA, USA), a company specializing in RNA medicines that silence genes has developed DCR-MYC, a lipid particle that incorporates synthetic double-stranded RNA to target the MYC oncogene and suppress cancer progression [26,98]. This therapy has been evaluated in a dose escalation study in patients with multiple myeloma, lymphoma or solid tumors ({"type":"clinical-trial","attrs":{"text":"NCT02110563","term_id":"NCT02110563"}}NCT02110563). Additionally, DCR-MYC was also evaluated in clinical trials for hepatocellular carcinoma ({"type":"clinical-trial","attrs":{"text":"NCT02314052","term_id":"NCT02314052"}}NCT02314052) [99]. However, all clinical studies related to DCR-MYC have been stopped by Dicerna, as the early efficacy results did not meet the companys expectations to warrant further development [99,100].

In the line of using genetic therapies for cancer applications, Wagner et al. developed a nanoliposomal EphA2-targeted therapeutic (EphA2 siRNA), which has a neutral charge and is aimed at reducing organ toxicity associated with charged particles [101]. EphA2 belongs to a subfamily of the tyrosine kinase receptors, and is overexpressed in breast, lung, prostate, ovarian, pancreatic, and endometrial cancer [101]. The encapsulated siRNA therefore disturbs the cancer cell proliferation and slows down tumor growth. In preclinical studies, this formulation exhibited a significant anti-cancer effect with mild responses of hemolytic reaction, inflammation and mononuclear cell infiltration in gastrointestinal tract, heart and kidney [102]. These promising results have led EphA2 siRNA to entering phase 1 clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT01591356","term_id":"NCT01591356"}}NCT01591356), where it will be used in patients with advanced or recurrent solid tumors [103].

ModernaTX, Inc. and AstraZeneca have developed the mRNA-2752 encapsulated LNP and have applied for a phase 1 clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT03739931","term_id":"NCT03739931"}}NCT03739931). This mRNA encodes for OX40L, a T-cell co-stimulator, IL-23 and IL-36 pro-inflammatory cytokines. Patients with solid tumors were treated individually with mRNA-2752 or in combination with durvalumab. Tumor shrinkage in monotherapy or dual-therapy with durvalumab was observed in pre-clinical studies in patients with advanced solid malignancy or lymphoma, which supports the move of mRNA-2752 towards phase 1 studies. Using similar technology, ModernaTX, Inc. studied another LNP candidate (mRNA 2416) for treatment of patients with advanced malignancies [110]. mRNA-2416 is delivered directly to tumors that over-express OX40 which then activate strongly T cell responses that kill the tumor. Currently, this candidate has been recruiting for phase 12 clinical trials with identifier of {"type":"clinical-trial","attrs":{"text":"NCT03323398","term_id":"NCT03323398"}}NCT03323398 [110].

Alnylam Pharmaceuticals also completed a Phase 1 clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT01158079","term_id":"NCT01158079"}}NCT01158079) for ALN-VSP02 in 2012 [107], which is an LNP with siRNA that targets the expression of vascular endothelial growth factor (VEGF) and kinesin spindle protein (KSP) [106]. These factors are overexpressed in many tumors and contribute to tumor proliferation and survival. Therefore, ALN-VSP02 silences these two mRNAs and prevents the translation of the KSP and VEGF proteins, which inhibits tumor growth in these patients.

The LNP-RNA system has proven to be a versatile platform with uses beyond cancer treatments. From 2009 to 2019, there were 6 similar products approved for clinical trials (). Arbutus Biopharma Corporation completed phase 2 clinical trials ({"type":"clinical-trial","attrs":{"text":"NCT02631096","term_id":"NCT02631096"}}NCT02631096) for ARB-001467 [17]. This formula contained siRNAs against the four hepatitis B virus transcripts and was indicated for patients with hepatitis B. Bristol-Myers Squibb and Nitto Denko Corporation completed a phase 1b/2 clinical trial of ND-L02-s0201, which was indicated for patients with moderate to extensive hepatic fibrosis ({"type":"clinical-trial","attrs":{"text":"NCT02227459","term_id":"NCT02227459"}}NCT02227459). ND-L02-s0201, a heat shock protein 47 siRNA, moderates collagen synthesis and secretion to prevent the fibrosis. This clinical study has started in Japan after being initiated in Europe and the United States. Arrowhead Research had also generated the ARC-520 to treat hepatitis B virus and applied phase 2 clinical trials ({"type":"clinical-trial","attrs":{"text":"NCT02065336","term_id":"NCT02065336"}}NCT02065336). ARC-520 formulated from interference RNA, which can reduce all RNA transcripts derived from covalently closed circular DNA that leads to reduce viral antigens and hepatitis B virus DNA. However, ARC-520 was terminated due to delivery-associated toxicity [112]. Currently, Dicerna Pharmaceuticals, Inc. has developed LNPs with siRNA (named as DCR-HBVS) targeting the mRNA for the hepatitis B surface antigen (HbsAg) for treatment of chronic hepatitis B. Phase 1 clinical trial for DCR-HBVS has been requested ({"type":"clinical-trial","attrs":{"text":"NCT03772249","term_id":"NCT03772249"}}NCT03772249) when promising preclinical data was obtained, in which a mouse model of hepatitis B, treated by DCR-HBVS was significantly reduced.

Alnylam Pharmaceuticals also completed a phase 1 clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT01437059","term_id":"NCT01437059"}}NCT01437059) for ALN-PCS02 for the treatment of patients with hypercholesterolemia. ALN-PCS02 is formulated from siRNA encapsulated in lipid LNPs, in which siRNA reduces the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme of plasma cholesterol metabolism leading to lower levels of low-density lipoprotein [102].

Tekmira registered a phase 1 clinical trial with identifier of {"type":"clinical-trial","attrs":{"text":"NCT00927459","term_id":"NCT00927459"}}NCT00927459 for PRO-040201 in 2009, which contains siRNA loaded in a stable nucleic acid LNP. PRO-040201 can target ApoB produced by hepatocytes to control the level of cholesterol in blood. Although the preliminary clinical trial demonstrated that PRO-040201 delivered siRNA effectively to liver and reduce significantly low density lipoprotein, there were flu-like symptoms at the highest dose [102]. Thus, Tekmira decided to terminate the study in 2010.

At the ending of 2019, Wuhan city, Hubei province of China appeared pneumonia patients. After that, in January 2020, a novel coronavirus causing pneumonia was confirmed by Chinese under the name of 2019 nCoV. World Health Organization (WHO), Chinese authorities and other partners have worked to understanding about properties, sources, prevention of virus spread, and treatments [113]. However, COVID-19 pandemic is spreading all over the world unprecedentedly that impacts on global economy individual and community health seriously. As a result, urgent demand is not only to save COVID-19 patients but also to develop vaccines.

Nucleic acid-based vaccines have gained much attention and the first candidates have entered clinical trials [114]. Nucleic acid vaccines possess many advantages over conventional protein-based vaccines such as ease of synthesis, safety, effective antigen manipulation, cost, and scale-up ability [115,116]. However, nucleic acids display some inherent disadvantages [114,116]. DNA has low immunogenicity and might integrate with human genome [116]. RNA is rapidly degraded in physiological media and efficiently excreted by glomerular filtration within less than 10 min [14]. In spite of that, RNA is still considered the best platform technology for developing vaccines against various diseases, both non-infectious diseases and infectious [117]. In vitro transcribed RNA vaccines exhibit efficient antigen expression and self-adjuvancy [118]. Adjuvants are often added to vaccines in order to enhance and prolong the immune response [119], but they also have the potential to cause inflammatory side effects [120]. Therefore, RNA vaccines are becoming a promising candidate as self-adjuvant vaccines with minimal inflammatory side effects [121]. Particles for vaccine delivery have to achieve a high loading capacity, sustainable release, no leakage, and simple manufacturing [122]. To maximize the efficacy of those vaccine candidates as well as to achieve a favourable index for human applications, scaffolds carrying those agents play an important role in vaccine efficacy. Careful consideration of appropriate scaffolds for a specific vaccine plays a key role in vaccine fabrication. Related to liposome scaffolds, four other approved liposomes Inflexal V, Epaxal, Mosquirix and Shingrix were commercialized successfully as vaccines [6,7,71]. Those liposomes are incorporating virosomal influenza vaccine, inactivated hepatitis A virus, RTS,S antigen-based vaccine, and glycoprotein E based vaccine [6,7]. LNPs have become ideal candidates for vaccine design, they are similar to viral structures with virus-like dimensions, and able to carry antigens and adjuvants [123].

Along with the history of vaccine development, adjuvants are equally as important and have led to significant advances in vaccine formulations and efficacy thereof. Especially Alum (insoluble aluminium salts) is incorporated in many childhood vaccines such as DTaP (diphtheria, tetanus, pertussis) vaccines, the pneumococcal conjugate vaccine, and hepatitis B vaccines [124,125]. Of other approved vaccines in 2015, there were many formulations that included adjuvants. For example, AS01 (MPL (a naturally derived TLR4 ligand)and QS21 saponin), MF59, and immunostimulatory oligonucleotides are presented in Shingrix, Fluad and Heplisav, respectively [124]. Besides, various other vaccine adjuvants including AS04, RC-529, CpG ODN, TLR9 agonist, TLR4 agonist, and virosomes have been included in licensed products [124,125]. Those vaccines confirm that incorporation of adjuvants can progress the vaccine into the clinic more rapidly and successfully. Also, a broad range of lipids has been reported to possess the strong adjuvant activity. Especially, cationic lipid, dimethyldioctadecylammonium bromide (DDA), showed the deposition of the antigen at the injection site, the enhancement of a cellular antigen internalization, and an antigen association [126]. As a result, LNPs made from DDA were forecasted to possess self-adjuvant activity. Indeed, Anderluzzi et al. reported that an emulsion of polymeric nanoparticles, liposomes, and solid lipid nanoparticles structured by DDA gained high antigen adsorption efficiency, in vitro antigen trafficking, in vivo distribution and high antibody response [126]. However, the immunogenicity level was strongly dependent on the type of formulation. The nanoparticle system exhibited high cell uptake and antigen processing, while the emulsion showed high antibody responses [126].

In another case, LNPs were used as a carrier system for adjuvants and mRNA to achieve their synergistic effects in immune stimulation. Lee et al. fabricated the LNPs with an adjuvant of Pam3 (tri-palmitoyl-S-glyceryl cysteine linked to the penta- peptide) to carry mRNA for cancer immunotherapy. Their results indicated that this formulation triggered different TLRs to increase the population of CD8+ T cells, thereby preventing tumor growth. So the combination of an adjuvant and mRNA in an LNP carrier could be a promising avenue in mRNA-based cancer therapeutics [118].

LNPs and liposomes showed their best suitability for RNA-based vaccines in protective ability, pharmacokinetics, tissue distribution and targeted delivery (dendritic cells and macrophages) [12,114,122,127,128]. With optimization of over 1000 candidates, it was concluded that LNPs are suitable delivery vehicles for mRNA coding antigens for anti-cancerous vaccines [13]. Such an RNA-LNPs have been designed to reverse M2-like macrophages or other immunosuppressive phenotypes, to activate the innate immunity, to inhibit other soluble immunosuppressive factors, and to induce tumor-infiltrating lymphocytes for immunomodulation strategies to treat malignant tumors (). In fact, there are a lot of studies related to this topic that have been summarized in the selected reviews [12,13,14]. Herein, we will focus on the clinical trials of developed vaccines, summarized in .

Immunomodulation strategies to improve cancer immunotherapy in nanomedicines: Nanomedicine was designed to induce immunogenic cell death, to promote antitumor immunity (cancer vaccination), to modulate immune cells, to activate innate immunity, to inhibit soluble immunosuppressive factors, to alternate tumor matrix, to engineer lymphocyte and normalize vessel [129].

Clinical trial information of LNPs for cancer vaccines.

Lipo-MERIT is a cancer vaccine [133] that is made of four mRNAs encoding for NY-ESO-1, MAGE-A3, tyrosinase and TPTE which are encapsulated in liposomes without any modification with molecular ligands [134]. Lipo-MERIT travels to spleen and is taken up by splenic dendritic cells and macrophages to activate NK, B, CD4+, and CD8+ T cells. For the progress of commercial preparation, Lipo-MERIT is currently in Phase 1 clinical trials [130].

Another cancer vaccine in clinical trials is the mRNA-LNP personalized cancer vaccine, mRNA-4157, developed by ModernaTX, Inc. and Merck Sharp & Dohme Corp. This vaccine is indicated for patients with resected solid tumors including bladder carcinoma, melanoma and non-small cell lung carcinoma (NSCLC). In addition, mRNA-4157 is also used in combination with pembrolizumab for patients with advanced or metastatic cancers. After administration, this LNP is uptaken and translated by antigen presenting cells, thereby inducing both cytotoxic T-lymphocyte and memory T-cell-dependent immune responses to destroy the cancer cells. Currently, mRNA-4157 is being clinically evaluated for safety and efficacy in Phase 1 ({"type":"clinical-trial","attrs":{"text":"NCT03313778","term_id":"NCT03313778"}}NCT03313778) [131] and Phase 2 ({"type":"clinical-trial","attrs":{"text":"NCT03897881","term_id":"NCT03897881"}}NCT03897881) [132].

Stimuvax (L-BLP-25, BLP25 liposome) of EMD Serono & Merck KgaA, Darmstadt, Germany was also entered phase 3 clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT00409188","term_id":"NCT00409188"}}NCT00409188). This therapeutic vaccine provides immunity to kill the cancer cells expressing a glycoprotein antigen of Mucin 1 (MUC-1). The trial for patients with advanced non-small cell lung cancer (NSCLC), but the results were disappointing and did not meet the primary endpoint [135]. As a result, L-BLP-25 was terminated.

In March 2020, the World Health Organization (WHO) declared that the Coronavirus Infectious Disease (COVID-19) was a global pandemic. In March 2021, more than 129.4 million cases and 2.8 million deaths were reported all over the world [123]. Concurrently, COVID-19 has a high probability of becoming a seasonal disease with high infection rates and a long incubation period [123]. Consequently, developing COVID-19 vaccines has been a necessity for the global population [136], for which numerous platforms have been investigated. There are 12 vaccines approved by the FDA. Others are in clinical trials, 93 vaccines are listed with 257 trials [127,137], of which, 29 vaccines in Phase 1, 39 vaccines in Phase 2, and 25 vaccines in Phase 3 [137]. The activating mechanisms in most of those vaccine candidates are based on the induction of neutralizing antibodies against the spike (S) protein to prevent the uptake into human cells via the human angiotensin-converting enzyme-2 (ACE2) receptor [127]. Thus far, for mRNA-LNPs only mRNA-1273 and BNT162 have been successfully developed and others are progressing in clinical trials () [12,128,138,139,140].

List of the COVID-19 vaccines using LNPs.

LNPs encapsulating mRNA developed by BioNTech SE and Pfizer for the SARS-CoV-2 vaccine had four candidates (BNT162a1, BNT162b1, BNT162b2, and BNT162c2). They were designed from two types of a nucleoside-modified mRNA, a uridine containing mRNA and a self-amplifying RNA [138,141]. They have been tested in Phase 2 clinical trials ({"type":"clinical-trial","attrs":{"text":"NCT04380701","term_id":"NCT04380701"}}NCT04380701)in healthy volunteers aged from 18 years to 85 years [142], and in Phase 3 ({"type":"clinical-trial","attrs":{"text":"NCT04368728","term_id":"NCT04368728"}}NCT04368728) [143]. The BNT162b2 candidate has finished phase 3 clinical trials with promising results of a safe and effective vaccine. In front of the urgent need of Covid-19 vaccine, Pfizer and BioNTech submitted their BNT162b2 to the FDA to request an emergency use authorization. On 11 December 2020, the FDA approved Pfizer-BioNTech COVID-19 Vaccine distributed in the United States [144].

Using a similar strategy, mRNA-1273 COVID-19 is fabricated from synthetic mRNA inside LNPs [128,145]. This synthetic mRNA encodes for the spike, S protein of SARS-CoV-2 viruses [12,138,139] which is a key factor on viral surfaces binding to the host cell through ACE2 receptor. The S protein of SARS-CoV-2 viruses mediates cell attachment, receptor recognition, and fusion for viral penetration and infection [146]. This formula has been developed by Moderna TX, Inc, and in the process of FDA approval (phase 1) with reference number of {"type":"clinical-trial","attrs":{"text":"NCT04283461","term_id":"NCT04283461"}}NCT04283461 [127,128,147]. It also moved to phase 2 ({"type":"clinical-trial","attrs":{"text":"NCT04405076","term_id":"NCT04405076"}}NCT04405076) to be assessed for reactogenicity, immunogenicity and safety in healthy male and non-pregnant females from 18 years old [140,148]. This candidate has entered Phase 3 ({"type":"clinical-trial","attrs":{"text":"NCT04470427","term_id":"NCT04470427"}}NCT04470427) to be evaluated for safety, efficacy and immunogenicity to prevent COVID-19 for up to two years [140,149]. On 17 December 2020, Moderna TX, Inc gained the emergency-use authorization of the FDA for mRNA-1273 [150].

McKay et al. studied LNPs encapsulated with self-amplifying RNA (saRNA) as the new SARS-CoV-2 vaccine named LNP-nCoVsaRNA or COVAC1 [136,151]. These self-amplifying RNA constructs have been proposed because any antigen of interest can be encoded and formulated at a lower dose than conventional mRNA. For in vitro and in vivo experiments, the LNP-nCoVsaRNA vaccine for SARS-CoV2 exhibited robust antibody and cellular responses. These outstanding results induced the strong belief that the LNP-nCoVsaRNA vaccine would promote immunogenicity in humans. Currently, Imperial College London applied the LNP-nCoVsaRNA vaccine for the clinical trial, currently in Phase 1 (ISRCTN17072692) [140,152].

Duke-NUS Medical School and Arcturus Therapeutics Inc. have collaborated for the development of a COVID-19 vaccine named ARCT-021. They used the LUNAR lipid-mediated delivery system to encapsulate RNA (STARRTM). In the preclinical stage, ARCT-021 has demonstrated to be a safe and efficient vaccine for COVID-19 at low dose. It was able to induce CD8+ T-cell and T-helper cellular immune responses without adjuvants and viral vector. Moving to Phase 1/2 clinical studies (NCT4480957), ARCT-021 has been evaluated with safety, tolerability and immunogenicity at multiple dose levels from 110 g. The age groups for this study are healthy people from 1880 years, with the exception of pregnant and breast-feeding women. Preliminary data for ARCT-021 showed a favourable safety profile at a relatively low dose [153]. In addition, other similar vaccine platforms including ChulaCov19 mRNA vaccine (Chulalongkorn University) and SARS-CoV-2 mRNA vaccine (Shulan, HangzhouHospital) have applied for clinical trials. The ChulaCov19 mRNA vaccine has been registered for phase 1 clinical trials ({"type":"clinical-trial","attrs":{"text":"NCT04566276","term_id":"NCT04566276"}}NCT04566276) with no study recruitment yet [151]. The SARS-CoV-2 mRNA vaccine was applied to Chinese Clinical Trial Registry for Phase 1 clinical trial (ChiCTR2000034112). This trial will evaluate the safety, tolerance and immunogenicity of multiple doses in the population above 18 year olds [151].

The LNPs under development for the COVID-19 vaccines do not only include the aforementioned approved products or those still in the clinical trials, but also include numerous others that are continuously being developed in pre-clinical stage () [154]. Many companies, research institutes, and universities worldwide have been listed on the WHO list with RNA-encapsulated LNP vaccines. Globe Biotech Ltd. had SARS-CoV-2 D614G variant LNP-encapsulated mRNA. Max-Planck-Institute of Colloids and Interfaces had LNPs encapsulating mRNA targeting Langerhans cell. Sanofi Pasteur and Translate Bio studied the mRNA-based vaccine MRT5500 and are going to apply for clinical trial at the ending of 2020 [155]. Now, MRT5500 has started phase1/2 clinical trial. It was expected to get interim results in the third quarter of 2021 [156]. CanSino Biologics and Precision NanoSystems had a collaboration to develop a COVID-19 RNA vaccine composed of mRNA and lipid nanoparticle carrier [157]. Daiichi-Sankyo Co. has developed mRNA-based COVID-19 vaccine named as DS-5670. For next clinical studies, Daiichi-Sankyo Co. is going to collaborate with the University of Tokyo [140]. IMV Inc (Dartmouth, Canada), formulated their DPX-COVID-19 vaccine candidate. DPX is the lipid-based delivery system in which peptide antigens are dissolved in lipids with the final formulation stored in dry form [158]. After dissolution, this vaccine is injected intramuscularly. There is no releasing mechanism at the injection site, but the peptide antigens act as adjuvant and the formulated DPX can then recruit the antigen presenting cells that induce an immune response from the lymph nodes [158]. Na-Na Zhang et al. studied a thermostable mRNA vaccine candidate for preventing COVID-19 infection [159]. They used LNPs to encapsulate mRNA targeting the receptor-binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In the mouse model they used, the this LNP induced neutralizing antibodies and T-cell responses with high protective immunity against SARS-CoV-2. Additionally, this vaccine candidate can be stored at room temperature for at least one week. With these advantages, this LNP-mRNA candidate (RQ3011-RBD) is moving into phase 1 clinical trial evaluation by Fudan University, Shanghai JiaoTong University and RNACure Biopharma [140,159]. Moreover, they designed two other vaccine candidates, RQ3013-VLP and RQ3012-Spike, that include a cocktail of mRNA constructs. The RQ3013-VLP candidate contains a cocktail of mRNAs encoding 3 viral structural proteins of S (spike), M (membrane), and E (envelope). The RQ3012-Spike vaccine carries mRNA encoding the full-length wild-type S. In mice model, RQ3013-VLP exhibited the best immune response, across three these candidates, while RQ3011-RBD induced insufficient immunity at a low concentration of 2 g RNA/dose. Depending on these preliminary tests, it can be concluded that mRNA vaccines can act as a flexible platform to design effective candidates [159].

Besides, the developing strategy of vaccines has still studied for other diseases. Shirai et al. demonstrated that LNPs could act as an adjuvant for influenza vaccines [162]. This was confirmed through the results about the immune-stimulatory effects on dendritic cells in mice and the protection ability of LNPs encapsulating the conventional seasonal split vaccine (SV) in comparison with bare SVs and SVs combined Alum. The LNP was made of 1,2-dioleoyl-3-trimethylammonium- propane, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, N-(carbonyl-methoxypolyethyleneglycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine and cholesterol. It was found that the LNPs with SVs achieved a similar efficiency as SVs combined Alum. Alum induced a high inflammatory response, which is considered a limitation of traditional adjuvants. In contrary, LNPs were able to induce SV-specific immune responses without inflammation [162]. Swaminathan et al. also assessed the adjuvant activity of LNPs alone and of LNPs incorporating the synthetic TLR9 agonist, IMO-2125 adjuvant, in a mouse model [163]. This LNP formulation was composed of DSPC, cholesterol, DMG-PEG2000 and an asymmetric ionizable amino lipid. Surprisingly, the LNP without adjuvant was able to induce B-cell responses against HbsAg (hepatitis B virus surface antigen) and ovalbumin sub-unit antigens at a comparable level than in the presence of other adjuvants including IMO-2125, 3-O-deactytaled monophosphoryl lipid and aluminum-based adjuvants. The LNP not only induced a significant enhancement of immune responses but also elicited a higher Th1-type response compared to IMO-2125 alone. So the combination of LNPs and immune-modulatory oligonucleotide adjuvants led to have the synergistic effects for immune responses and to manipulate those immune qualities that is quite different from the inorganic adjuvants.

The clinical development of lipid-based nanoparticle technologies with chemo- and nucleic acid therapeutics have demonstrated the potential of lipid-based carriers in the treatment of a range of diseases. However, the number of successful products that have reached the market does not accurately represent the number of formulations in (pre)clinical trials, indicating that the development of these nanoparticles still suffers from difficulties and challenges in the translation from animals to humans. Recently, several strategies have been developed to overcome these limitations. To improve the stability of nanoparticles and prevent drug leakage, lipid structures have been designed that efficiently complex by ionic attraction with the encapsulated therapeutic. Ionizable lipids, such as DOP-DEDA, have shown to be favourable for gene encapsulation. Cholesterol, on the other hand, is essential in providing stability to the liposomal structures resulting in tight packing of the drugs. Stability of the LNPs in physiological media and systemic circulation is achieved by modifying the particle surface with a PEG-lipid, thereby reducing the recognization by the reticuloendothelial system. However, the production of anti-PEG antibodies following administration of the first dose has been reported to reduce the therapeutic efficacy and/or cause adverse reactions upon the following doses. The quest for PEG alternatives has, therefore, become necessary to enable repeated injections. Besides the prolonged circulation, LNPs must target the specific tissues/cells/organs and then internalize through cell membranes to release drugs at the site of action. To achieve this requirement, LNPs have been designed as smart materials with selective ligands with degradation being triggered by changes in pH, temperature or oxidation/reduction.

The selective association of LNPs with target cells remains a challenge. Nucleic acid vaccines for example cannot be injected directly into our lymph nodes or spleen, which are tissues that are home to immune cells responsible for making antibodies and killing cancer cells. The targeted delivery of nucleic acids from injection sites to immune cells in lymph nodes or the spleen is therefore critical to maximising the production of antibody or long-lived antigen-specific cytotoxic T cells. Such delivery systems need the ability to target lymph nodes but also cross the barrier cells in the lymph nodes to interact with immune cells. Additionally, after internalization, the controlled release of therapeutics to aberrant cells has to be initiated effectively. These mechanisms that facilitate cell uptake, internalization, and payload release have not yet achieved the expected results. The success of LNPs with selective ligands in the market have not been completed yet. Therefore, with various cell type-specific ligands and stimulus agents, it is expected that studies related efficacy of modified LNPs for different diseases will enter clinical trials soon. The co-delivery or drug co-encapsulating with adjuvants are of interest in this field to improve efficacy and immune modulation. Additionally, the manufacturing and scaling up process of LNPs, has been challenging. New methodologies of LNP preparation based on microfluidics have been considered as the most robust to date, but it exhibits limitations in formulating multifunctional LNPs.

Concurrently, to combat the COVID-19 pandemic, the fast development of RNA and LNP-based vaccines have gained emergency FDA approval, which demonstrates the rapid and effective response of this approach against complex diseases. These vaccines are not limited to infectious diseases but have also been developed for other disease types such as cancer and hyperlipidemia. Some clinical trials are almost at the final phase. Numerous studies are at early stages and will increase in the future, which forecasts a range of products to be launched on the market.

In conclusion, we believe that the success of mRNA-LNP vaccines opens an exciting chapter for LNP technology. A long road to optimizing LNP formulations for small molecule drugs and nucleic acid delivery has been paid off, and LNPs have become, once again, a frontrunner in nano drug delivery system. This review highlights key lessons learnt from this long road and serves as a reference for designing LNPs. Further development of LNPs is still urgently needed to address current global health challenges, which requires collaborative efforts of scientists in different fields.

Conceptualization, N.P.T.; writingoriginal draft preparation, T.T.H.T. and E.J.A.S.; writingediting, E.J.A.S. and N.P.T.; writingreview, J.S.L., D.H.N., and K.D.P.; funding acquisition, N.P.T. All authors have read and agreed to the published version of the manuscript.

This research was funded by the Australian Research Council and the National Health and Medical Research Council, grant number DE180100076, DP200100231 and GNT2002827.

Not applicable.

Not applicable.

Not applicable.

The authors declare no conflict of interest.

Publishers Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Originally posted here:
Lipid-Based Nanoparticles in the Clinic and Clinical ...

What is Nanosafety and Why is it Important? – AZoNano

In the last two decades, the field of nanotechnology has been rapidly expanding and has been experimented with in various applications, such as consumer products, nanomedicine, medical imaging, tissue engineering, textile fabrics.

Image Credit:Natale Zanardi/Shutterstock.com

The field is applied in designing, synthesizing, characterizing, and applying materials as well as devices that are functionally organized in one or more dimensions on the nanometer scale between 1 to 100 nm (Chhantyal, 2020).

However, as nanoparticles are manipulated, they are categorized into certain types, sizes, and concentrations, some of which pose a great risk to human health and the environment. These nanoparticles are labeled toxic and require a trained person in the well-ventilated lab when being used. After use, the nanoparticles should be disposed of in a designated place.

The growing concern of potential health and environmental risks associated with nanoparticles has triggered various safety regulations around the world as well as the general concern of awareness among the public and the field experts.

Due to their small size, nanoparticles can easily surpass biological membranes and harm cells, tissues, and organs. The particles can also get into the body by various routes, such as inhalation, ingestion, or contact through the skin (Asmatulu, 2011).

However, the benefit of nanoparticles should also be well appreciated as many notable examples are seen in the medical field as drug delivery in cancer research (Yao, et al., 2020). Due to their improved advantage of stability and biocompatibility over conventional drugs, nanoparticles are widely being used to precisely target the imperfect cells and release drugs.

The use of engineered nanoparticles as a possible cure for many life-threatening diseases has seen the effect of toxic nanoparticles as, when used intravenously, react with blood and its components, affecting the characteristics of their interactions with tissues and cells (Boraschi, Costantino, & Italiani, 2012).

However, the understanding of the toxicity level of these nanoparticles is still underestimated as the affiliation between the properties of nanoparticles on the human body has not gathered enough clinical data (Yuvaraj, Yuvaraj, Arunkumar, Pandiyan, & Subramanian, 2020).

Even with the current advanced system, their odd shapes and high reactivity make nanoparticles effect on the metabolism hard to predict. In practice, they can fail to trigger the body's defensive mechanisms and harm tissues.

Various studies indicate that different nanoparticles cause inflammation(Yazdi, et al., 2010), and result in severe sickness asthma, bronchitis, lung and liver cancer, Parkinson, Alzheimer, heart disease, and colon cancer (Karakoti, Hench, & Seal , 2006).

As valuable as the field of nanotechnology is, it raises a huge amount of public interest in nanotoxicity.

Professionals engaged with nanoparticles are more concerned about the toxicity of the materials as they handle them during the fabrications, transportation, handling, usage, recycling, and waste disposal (D & Rao, 2011).

Lack of regulation of the use and commercialization of nanomaterials poses a direct threat to public health and the environment (Allan, Belz, Hoeveler, & Hugas, 2021). In many countries, current legislation does not expressively address nanoparticles or nanomaterials in their laws and regulations.

In addition, the nomenclature and classification of nanomaterials as novel substances among the scientific community and governments is disputed, and several definitions do not even include the safety considerations of these materials (Boverhof, Bramante, Butala, Clancy, & Gordon, 2015).

The importance of nanosafety is to communicate accurate information regarding the origins and processes of nanotoxicity in humans and the environment. It means developing precaution measures at the workplace, researching treatment for its medical conditions, improvising safe material-handling techniques in the field. Doing so will promote broader societal acceptance of nanotechnology.

The field of nanotechnology has proven applicable to problem-solving in interdisciplinary fields and has greatly impacted our lives with diverse potential benefits.

However, safety from its materials to humans or the environment should not be taken lightly. The continuous exposure of humans to nanoparticles brings significant concern about their potential risks.

First standard operation procedures should be adhered to in every laboratory to eliminate the risks that are associated with nanoparticles (Schulte, et al., 2016). These procedures should be contained in a Standard Operating Procedures (SOP) that contains relevant contact information, experiment overview, risk assessments, and controls in the lab.

The researchers should wear personal protective equipment (PPE) and follow other relevant rules to bring the risk as low as possible.

Moreover, a chart that stipulates a hierarchy of controls ought to be used to reduce the risks involved when using nanomaterials.

These controls should be placed at the source where the hazard originates from, along the path where the hazard travels to, and on the worker's PPE. Safety can also be maintained using a special flooring sticky mat, door signs, labeling and storage, and through correct disposal.

Continue reading: Why Nanotoxicology Should be the First Step Towards a Nanotechnology Future.

Allan, J., Belz, S., Hoeveler, A., & Hugas, M. (2021). Regulatory landscape of nanotechnology and nanoplastics from a global perspective. Regulatory Toxicology and Pharmacology. Available at:www.sciencedirect.com/science/article/pii/S0273230021000258

Asmatulu, R. (2011). Toxicity of Nanomaterials and Recent Developments in Lung Disease. In I. Martn-Loeches, Bronchitis.Available at: http://www.intechopen.com/chapters/17355

Boraschi, D., Costantino, L., & Italiani, P. (2012). Interaction of nanoparticles with immunocompetent cells: nanosafety considerations. Nanomedicine (Lond). Available at:www.futuremedicine.com/doi/10.2217/nnm.11.169

Boverhof, D. R., Bramante, C., Butala, J., Clancy, S., & Gordon, S. C. (2015). Comparative assessment of nanomaterial definitions and safety evaluation considerations. Regulatory Toxicology and Pharmacology. Available at:www.sciencedirect.com/science/article/pii/S0273230015001488

Chhantyal, P. (2020). How has Nanotechnology Developed Over Time? [Online] AZoNano. Available at: http://www.azonano.com/article.aspx?ArticleID=5610

Karakoti, A., Hench, L., & Seal , S. (2006). The potential toxicity of nanomaterialsThe role of surfaces. JOM. Available at:link.springer.com/article/10.1007/s11837-006-0147-0

Schulte, P., Roth, G., Hodson, L., Murashov, V., Hoover, M., & Zumwalde, R. (2016). Taking stock of the occupational safety and health challenges of nanotechnology. J Nanopart Res. Available at:pubmed.ncbi.nlm.nih.gov/27594804/

Yao, Y., Zhou, Y., Liu, L., Xu, Y., Chen, Q., Wang, Y., . . . Shao, A. (2020). Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Nanobiotechnology. Available at:www.frontiersin.org/articles/10.3389/fmolb.2020.00193/full

Yazdi, A. S., Guarda, G., Riteau, N., Drexler, S. K., Tardivel, A., Couillin, I., & Tschopp, J. (2010). Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1 and IL-1. Proc Natl Acad Sci U S A. Available at:www.pnas.org/content/early/2010/10/20/1008155107

Yuvaraj, M., Yuvaraj, V., Arunkumar, V., Pandiyan, M., & Subramanian, K. S. (2020). Nanosafety. In M. Ince, Biochemical Toxicology - Heavy Metals and Nanomaterials. Available at:www.intechopen.com/chapters/71950

Disclaimer: The views expressed here are those of the author expressed in their private capacity and do not necessarily represent the views of AZoM.com Limited T/A AZoNetwork the owner and operator of this website. This disclaimer forms part of the Terms and conditions of use of this website.

Read more from the original source:
What is Nanosafety and Why is it Important? - AZoNano

Anti-viral Drugs Can Be Final Solution As WHO Warns Against Lowering Our Guard To COVID-19 – Scoop.co.nz

Wednesday, 26 January 2022, 7:07 amPress Release: World Nano Foundation

Suggestions that COVID-19 is on the wane have beenstrongly contradicted by the World Health Organizationssenior pandemics scientist, Dr Maria Van Kerkhove.

Andher criticism of virus complacency has fuelled calls forresearch and development of anti-viral drugs to stop allcoronaviruses at source, in addition to ongoing vaccines andtesting for COVID-19 variants.

DrVan Kerkhove, a highly regarded infectious diseaseepidemiologist and World Health Organization (WHO) Head ofthe Emerging Diseases and Zoonoses Unit, delivered herwake-up call in a BBC TV interview where she insisted thatCOVID-19 was still evolving and the world must evolve withit:

It will not end with this latest wave (Omicron)and it will not be the last variant you will hear us (WHO)speaking about unfortunately, she told BBCinterviewer Sophie Raworth.

Countries with highimmunity and vaccination levels were starting to think thepandemic is over, she added, but despite 10 billion vaccinedoses delivered globally, more than three billion peoplewere yet to receive one dose, leaving the world highlysusceptible to further COVID mutations - a global problemfor which a global solution was needed.

She alsochallenged assumptions that the COVID Omicron variant wasmild: It is still putting people in hospitaland itwill not be the last (variant). There is no guarantee thatthe next one will be less severe. We must keep the pressureup we cannot give it a free ride.

This drew aresponse from the World Nano Foundation (WNF), anot-for-profit organization that promotes many of theinnovations including nanomedicines, AI andcomputational drug development platforms, testing andvaccine development that have played vital roles infighting the COVID pandemic.

WNF Chairman PaulStannard said: We welcome Dr Van Kerkhoves timelyintervention. Too many people think we can sit back withCOVID now, forgetting lessons learned the hardway.

Such as theres always another variant justaround the corner, and testing and vaccines are not thecomplete answer.

Even if Omicron seems milder thanits predecessors though this may be due to vaccinationsand growing herd immunity who can say that a more fatalCOVID mutation will not follow, or an all-new virus iswaiting to strike.

Many other pathogens haveentered humans in last 15 years including SARs Ebola, Zikavirus and Indian Flu variants, so permanent pandemicprotection investment is vital to restoring confidence inour way of life and the global markets.

An evenolder lesson is Spanish Flu (1918-20): the death toll wasrelatively contained initially, lulling people alreadyfatigued by WW1 devastation into thinking the worst wasover.

But that virus then mutated into its mostdeadly strain, killing 50 million people when Earthspopulation numbered less than two billion. All of whichsuggests we must maintain or redouble our efforts againstCOVID-19 and other potential threats.

We havealready benefitted from greater healthcare investment andresearch due to the pandemic: experts say the first sixmonths of the emergency delivered sector progress equivalentto the previous 10 years.

This helped unusuallyrapid deployment of new and better testing and vaccines thathave driven down infection, hospitalization and deaths, butwe hope that the WHO view will now foster a new andpotentially more effective development against COVID andother threats anti-viral drugs.

Instead ofattacking the virus like a vaccine, anti-viral drugs aim tostop it functioning in the human body. Merck and Pfizer saythey have re-purposed existing drugs to do justthat.

But a better option is gathering momentumusing nanomedicine, AI and advanced computational technologyto develop all-new drugs more quickly and effectively,potentially delivering breakthroughs against many seriouskillers, including viruses, cancers and heartdisease.

WNF believes these can disrupt thetraditional pharmaceutical industry as Tesla has done in theauto industry, or SpaceX and Blue Origin have done inspace.

California-based Verseonhas developed an AI and computational drug developmentplatform and has six drug candidates, including ananti-viral drug to potentially block all coronaviruses andsome flu variants, potentially transforming pandemicprotection.

This could be on the market within 18months after securing a final $60 million investment, asmall amount compared to the $1 billion pharma industry normfor a single new drug (source: Biospace),and weighed against 5.6 million COVID deaths globally and anestimated $3 trillion in economic output (source: Statista)lost since the start of the pandemic.

Verseon Head ofDiscovery Biology Anirban Datta said: Vaccines and thecurrent anti-viral drugs are retrospective solutions thatdont treat newly emergent strains. We need a differentstrategy to avoid always being one step behind viralmutations.

So, we switched target from the virus tothe human host. If we stop SARS-CoV-2 (COVID-19) enteringour cells which, unlike viruses, dont mutate then we havea long-term solution.

Even better, the strategyshould work against other coronaviruses and influenzastrains that use the same mechanism as SARS-CoV-2 to infectcells a key point, since it surely wont be the lastpandemic to affecthumanity.

Scoop Media

Become a member Get our free pnui

Excerpt from:
Anti-viral Drugs Can Be Final Solution As WHO Warns Against Lowering Our Guard To COVID-19 - Scoop.co.nz

Growth Opportunities in mRNA Therapeutics, Genomics Platforms, Microbiome Testing, Would Healing Solutions, and AI-Based Diagnostics -…

DUBLIN--(BUSINESS WIRE)--The "Growth Opportunities in mRNA Therapeutics, Genomics Platforms, Microbiome Testing, Would Healing Solutions, and AI-Based Diagnostics" report has been added to ResearchAndMarkets.com's offering.

The increase in clinical utility of liquid biopsies and other diagnostic assays, which is driven by AI has been discussed. The TOE issue also highlights developments in enabling technologies for microbiome analysis, and personalized health solutions based on microbiome testing. A few emerging technologies around DNA synthesis, next generation mRNA therapeutics and diagnostics have been highlighted. Few innovations highlight developments in wound healing solutions, which include smart dressings and plant-based solutions.

The Life Science, Health & Wellness TOE will feature disruptive technology advances in the global life sciences industry. The technologies and innovations profiled will encompass developments across genetic engineering, drug discovery and development, biomarkers, tissue engineering, synthetic biology, microbiome, disease management, as well as health and wellness among several other platforms.

The Health & Wellness cluster tracks developments in a myriad of areas including genetic engineering, regenerative medicine, drug discovery and development, nanomedicine, nutrition, cosmetic procedures, pain and disease management and therapies, drug delivery, personalized medicine, and smart healthcare.

Innovations in Life Sciences, Health & Wellness

Companies Mentioned

For more information about this report visit https://www.researchandmarkets.com/r/hzvh03

Excerpt from:
Growth Opportunities in mRNA Therapeutics, Genomics Platforms, Microbiome Testing, Would Healing Solutions, and AI-Based Diagnostics -...

Complementary Protection May Be at Hand With a COVID-19-Preventing Nasal Spray – Newsweek

Vaccinated citizens can still transmit the COVID-19 virus and its variants to other people. Salvacion USA Inc. is therefore excited to introduce the development of a complementary product, designed for adults 18 and older, it hopes could accompany COVID-19 vaccines to offer additional protection: a nasal spray to shield the nasal passages and prevent further viral transmission. (However, CDC guidelines should still be followed, and those who are eligible should receive the COVID-19 vaccination.)

COVID-19 vaccination numbers in the U.S. have now reached nearly 50 percent, and Americans are eager to resume life post-pandemic. However, as flu season approaches and the COVID-19 vaccines' efficacy and longevity have come under question, communities are increasingly concerned about the virus and its Delta and unknown variantsespecially as children return to school this fall. Experts have also speculated this pandemic could become endemic, cycling from season to season. For these reasons, we must continue to stay ahead of the rapidly updating situation and arrive at innovative strategies.

Salvacion's new technology is gaining momentum among the scientific community. The National Cancer Instituteestablished Nanotechnology Characterization Laboratory (NCL) recently selected Salvacion USA Inc. as one of its Assay Cascade awardees for Salvacion's nasal spray, trade name: COVIXYL-V. The announcement appeared in NCL's June 2021 quarterly newsletter, in which Salvacion noted, "COVIXYL-V is intended to reduce SARS-CoV-2 in nasal passages, a main point of entry for the virus in humans. Our unique virus-blocking product, optimized in collaboration with NCL, contains agents which block the virus from attaching to tissue and reducing the viral load in the tissue milieu."

According to NCL's announcement, "Nanomedicines accepted into the program will undergo a rigorous evaluation that may include sterility and endotoxin testing, physicochemical characterization, in vitro hemato- and immunotoxicity, and in vivo studies to evaluate safety, efficacy and pharmacokinetics. The studies are tailored to each individual nanomedicine and are designed to promote the clinical translation of these novel therapies."

Among Nanotechnology Characterization Laboratory's's five awardees, Salvacion is the only one working on a product intended to avert the spread of COVID-19. As an Assay Cascade awardee, NCL commits to funding Salvacion studies free of charge.

Ryan Hwang, a Salvacion spokesman, said, "Our product is preventative and protects nasal passages, halting transmission. Vaccines are developed to protect against COVID-19, but they are not designed to stop transmission. Our strategy is complementary to the effectiveness of vaccines by deterring COVID-19 infection by blocking the transmission."

Salvacion's clinical human trials are currently underway. In vitro and in vivo testing performed thus far suggested that the nasal spray inhibited 99.99 percent of COVID-19. This spray effectively blocked COVID-19 activity in the nasal passages of hamsters, the prime entry points for the virus. One study, performed by an independent BSL-3 laboratory (which, according to Public Health Emergency, is a lab "used to study infectious agents or toxins that may be transmitted through the air and cause potentially lethal infections"), showed that COVID-19 was 99.99 percent inactivated post-spray, with no clinical symptoms experienced by Syrian hamsters from the treatment. No adverse reactions were reported in the hamsters following administration. The data developed in this study showed that the nasal spray product was effective in neutralizing the virus within low concentrations. An additional barrier effect animal study undertaken at an independent laboratory also assessed the COVID-19 blocking effects of the nasal spray. It concluded that the spray created a physical barrier to block the viral particles from taking hold on the surface of the nasal passages. The testing was to prove the mechanism of the nasal spray is capable of blocking the transmission of COVID-19 by creating a physical barrier. The next step is the conducting of a human clinical trial, which is now underway. Salvacion is currently seeking an Emergency Use Authorization (EAU) for its COVIXYL-V nasal spray from the FDA.

This nasal spray is made of ingredients listed as GRAS, or "Generally Recognized As Safe," by the FDA. Unlike other products based on isopropyl alcohol currently being tested, it appears Salvacion's nasal spray could offer a unique blocking system with enhanced effectiveness at a very low concentration. A worldwide patent has been filed for this technology.

"Our nasal spray product may well be the key to moving back to a world that some have thought lost forever to the 'new normalcy,'" said Abdul Gaffar, a Salvacion chemist and recipient of the American Chemistry Society's Heroes of Chemistry Award, who invented this nasal spray.

The contents of this article are for informational purposes only and do not constitute medical advice. It's important to consult with your medical providers and the CDC before making any medical decisions or changes to your health plan, particularly with regard to COVID-19 and its variants.

Read more from the original source:
Complementary Protection May Be at Hand With a COVID-19-Preventing Nasal Spray - Newsweek

The Role of Nanomedicine in Transdermal Vaccine Alterantives – AZoNano

The World Health Organization has estimated the SARS-CoV-2 (COVID-19) pandemic to have caused more than 5.3 million deaths worldwide to date, and with the emergence of new variants, the urgency of effective immunization has never been more critical.

Study:The Importance of Nanocarrier Design and Composition for an Efficient Nanoparticle-Mediated Transdermal Vaccination. Image Credit:BaLL LunLa/Shutterstock.com

Alternative means of effective delivery have been explored in a recently published article from the journal vaccines, highlighting how nanomedicine can aid the effectiveness oftransdermal antigen delivery.

The COVID-19 infection caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2) has been catastrophic for the global population, with high rates of morbidity and mortality.

Schematic representation of the routes of skin penetration of active compounds. On the left, a transpedicular route consists of a. entry through hair follicle, b. entry through sweat glands, c. entry through sebaceous glands. On the right, transepidermal route. d. Transcellular pathway, e. Intercellular pathway.Image Credit:Valdivia-Olivares, R.,et al

The World Health Organization has estimated approximately 19.4 million infants globally have not been provided with vaccines. As the severity of the pandemic continues, the health of these children is at increased risk for deterioration.

This has led to the need to find alternative routes for immunization, which are effective and safe.

Transdermal routes of antigen delivery consist of a topical application to the skin where the active ingredients are absorbed systemically.

This type of method can be beneficial as it reduces first-pass metabolism and reduces the level of adverse effects. Transdermal administration provides an easy, simple, non-traumatic alternative that allows for self-administration, making this delivery route more attractive than the traditional injection method that involves needles.

However, a challenge for this delivery route for vaccine development consists of the protective barrier of the skin and the associated difficulties with overcoming the stratum corneum to enable antigen absorption.

Schematic representation of the mechanisms involved in immunization based on nanoparticles, either using combined techniques or design of nanoparticles by passive diffusion. Once the stratum corneum has been crossed, the antigens can interact with cells of the immune system already described. Image Credit:Valdivia-Olivares, R., et al

The skin is the largest organ in the body and consists of up to 20 million cell types, comprising but not limited tokeratinocytes, Langerhans cells, dendritic cells, T cells, and mast cells. These cells hold critical functions in the skin, contributing to immunocompetence and preventing pathogens from entering.

However, this also would prevent antigen delivery in the circumstance of vaccine development. Overcoming the stratum corneum barrier for transdermal permeability has been a focal point for researchers who have developed innovative technologies, such as iontophoresis, sonophoresis, and magnetophoresis electroporation, and laser microporation.

These methods can be effective but have their own limitations. For example, they are economically inefficient, and due to this, the most ubiquitous method for the administration of micro- and macromolecules via the skin has been the use of microneedles.

While microneedles can be effective, it is not considered to be completely needle-free and so with the emergence of the innovative field of nanotechnology, optimized nanosystems have been developed to cross the skin barrier without the dependence on invasive techniques.

Nanosystems which have been used for antigen delivery have been researched within literature and include nanoparticles and liposomes. Liposomes can be utilized effectively to transport antigens within parenteral administration;however, their use for needle-free transdermal immunization can be less effective.

This is due to their rigid structure which makes them inefficient for crossing the skin barrier, though incorporating nanotechnology within this nano-sized delivery particle, can allow this obstacle to be overcome.

Transferosomes are elastic liposomes that consist of phospholipidsthat form deformable vesicles. This mode of antigen delivery can be promising for use within the transdermal route as these particles can increase transdermal permeability in the stratum corneum in the presence of a hydration gradient.

Intriguingly, the biological makeup of transferosomes includes being highly flexible, effective for encapsulating hydrophobic and hydrophilic compounds as well peptides, and most importantly, they can pass through the pores of the skin. These key characteristics make transferosomes a perfect candidate for needle-free antigen delivery.

While there are significant benefits utilizing nanosized particles for nanovaccine development in innovative alternatives for parenteral antigen delivery within vaccinations, this area still requires further research.

Nanomedicine has innovated several fields of medicine, and this has benefitted research into the quality of patient care; however, the current state of medicine has proven immunizations of entire populations to be the most cost-effective method in disease prevention. This route can only be challenged with a collaborative effort with production laboratories to reduce production costs.

The advancement of needle-free administration would also require further research into the size and stability of nanosystems and the optimization of strategiesto tackle designs in a short space of time with limited resources, such as through utilizing computational techniques. This would be beneficial within a pandemic where resources are being shared between countries and urgent demand for solutions.

Benefitting and improving patient care should be the highest priority of medicine and with advanced nanocarrier design and innovative researchers, transdermal vaccinations could potentially become the future of disease control.

Continue reading: Antiviral Activity of Intermetallic Nanoparticles Incorporated into Polymeric Fibers.

Valdivia-Olivares, R., Rodriguez-Fernandez, M., lvarez-Figueroa, M., Kalergis, A. and Gonzlez-Aramundiz, J., (2021)The Importance of Nanocarrier Design and Composition for an Efficient Nanoparticle-Mediated Transdermal Vaccination.Vaccines, 9(12), p.1420. Available at: https://www.mdpi.com/2076-393X/9/12/1420

Gheibi Hayat, S. and Darroudi, M., (2019)Nanovaccine: A novel approach in immunization.Journal of Cellular Physiology, 234(8), pp.12530-12536. Available at: https://doi.org/10.1002/jcp.28120

Disclaimer: The views expressed here are those of the author expressed in their private capacity and do not necessarily represent the views of AZoM.com Limited T/A AZoNetwork the owner and operator of this website. This disclaimer forms part of the Terms and conditions of use of this website.

Here is the original post:
The Role of Nanomedicine in Transdermal Vaccine Alterantives - AZoNano

PhD within Development of Amine-Based nanofluids with Improved CO2 Capture Performance job with NORWEGIAN UNIVERSITY OF SCIENCE & TECHNOLOGY -…

About the position

We have a vacancy for a PhD candidate position within theParticle Engineering Centreat the Department of Chemical Engineering.

For a position as a PhD Candidate, the goal is a completed doctoral education up to an obtained doctoral degree.

TheParticle Engineering Centreat the Department of Chemical Engineering at NTNU was established in early 2021. The basis for the research in the Centre is an extensive experience at the department within particle synthesis and characterization, which quite recently was put into practical application as part of the COVID-19 test method developed at NTNU. The objective of the Centre is to advance the scientific knowledge within fundamental and applied particle engineering. Ongoing research activities focus on synthesis, characterization and tailoring of particles for use in nanomedicine and environmental applications.

The proposed PhD project aims to improve CO2 absorption efficiency of conventional amine solvents by developing their corresponding nanofluids. A comprehensive study is planned to prepare stable nanofluids with optimal thermophysical properties and to examine their effects on mass transfer kinetics and overall CO2 absorption performance under conditions resembling real CO2 capture plants.

You will report to your supervisor.

Duties of the position

The PhD research work will involve:

The candidate is further expected to:

Required selection criteria

The appointment is to be made in accordance with the regulations in force concerning State Employees and Civil Servants andRegulations concerning the degrees of Philosophiae Doctor (PhD) and Philosodophiae Doctor (PhD) in artistic research national guidelines for appointment as PhD, post doctor and research assistant

Preferred selection criteria

Personal characteristics

We offer

Salary and conditions

PhD candidates are remunerated in code 1017, and are normally remunerated at gross from NOK 491 200 per annum before tax, depending on qualifications and seniority. From the salary, 2% is deducted as a contribution to the Norwegian Public Service Pension Fund.

The period of employment is 3years.

Appointment to a PhD position requires that you are admitted to the PhD programme inChemical Engineeringwithin three months of employment, and that you participate in an organized PhD programme during the employment period.

The engagement is to be made in accordance with the regulations in force concerning State Employees and Civil Servants, and the acts relating to Control of the Export of Strategic Goods, Services and Technology. Candidates who by assessment of the application and attachment are seen to conflict with the criteria in the latter law will be prohibited from recruitment to NTNU. After the appointment you must assume that there may be changes in the area of work.

The position is subject to external funding.

It is a prerequisite you can be present at and accessible to the institution daily.

About the application

The application and supporting documentation to be used as the basis for the assessment must be in English.

Publications and other scientific work must follow the application. Please note that applications are only evaluated based on the information available on the application deadline. You should ensure that your application shows clearly how your skills and experience meet the criteria which are set out above.

The application must include:

If all, or parts, of your education has been taken abroad, we also ask you to attach documentation of the scope and quality of your entire education, both bachelor's and master's education, in addition to other higher education. Description of the documentation required can be foundhere. If you already have a statement from NOKUT, please attach this as well

Joint works will be considered. If it is difficult to identify your contribution to joint works, you must attach a brief description of your participationand personal and interpersonal qualities. Motivation, ambitions, and potential will also count in the assessment of the candidates.

NTNU is committed to following evaluation criteria for research quality according toThe San Francisco Declaration on Research Assessment - DORA.

General information

Working at NTNU

A good work environment is characterized by diversity. We encourage qualified candidates to apply, regardless of their gender, functional capacity or cultural background.

The city of Trondheimis a modern European city with a rich cultural scene. Trondheim is the innovation capital of Norway with a population of 200,000. The Norwegian welfare state, including healthcare, schools, kindergartens and overall equality, is probably the best of its kind in the world. Professional subsidized day-care for children is easily available. Furthermore, Trondheim offers great opportunities for education (including international schools) and possibilities to enjoy nature, culture and family life and has low crime rates and clean air quality.

As an employeeatNTNU, you must at all times adhere to the changes that the development in the subject entails and the organizational changes that are adopted.

Information Act (Offentleglova), your name, age, position and municipality may be made public even if you have requested not to have your name entered on the list of applicants.

If you have any questions about the position, please contact Professor Jana Poplsteinova Jakobsen, emailjana.p.jakobsen@ntnu.no. If you have any questions about the recruitment process, please contact HR-Consultant Unni M. Myhre, e-mail:unni.m.myhre@ntnu.no

Please submit your application electronically via jobbnorge.no with your CV, diplomas and certificates. Applications submitted elsewhere will not be considered. Diploma Supplement is required to attach for European Master Diplomas outside Norway. Chinese applicants are required to provide confirmation of Master Diploma fromChina Credentials Verification (CHSI).

If you are invited for interview you must include certified copies of transcripts and reference letters. Please refer to the application number 140/2021 when applying.

Application deadline: 10.01.2022

NTNU - knowledge for a better world

The Norwegian University of Science and Technology (NTNU) creates knowledge for a better world and solutions that can change everyday life.

Department of Chemical Engineering

We take chemistry from laboratory scale to industrial production. This demands a wide range of knowledge, from molecular processes and nanotechnology to building and operation of large processing plants. We educate graduates for some of Norway's most important industries. The Department of Chemical Engineering is one of eight departments in the Faculty of Natural Sciences.

Deadline10th January 2022EmployerNTNU - Norwegian University of Science and TechnologyMunicipalityTrondheimScopeFulltimeDurationTemporaryPlace of serviceGlshaugen

Originally posted here:
PhD within Development of Amine-Based nanofluids with Improved CO2 Capture Performance job with NORWEGIAN UNIVERSITY OF SCIENCE & TECHNOLOGY -...

Well, well, well! Notes from Session 3 of TEDMonterey – TED Blog

Engineer Kathryn A. Whitehead takes us down to the nano level to break down how lipid nanoparticles could revolutionize the way vaccines our delivered to our bodys cells. She speaks at TEDMonterey: The Case for Optimism on August 2, 2021. (Photo: Bret Hartman / TED)

The central topic of Session 3 is, surely, on many of our minds: health. From the technology powering the Pfizer-BioNTech COVID-19 vaccine to new Alzheimers treatment possibilities, these amazing speakers give a glimpse at exciting new frontiers of medicine. Plus, an artist drawing live onstage!

The event: TEDMonterey: Session 3, hosted by TEDs Chris Anderson on Monday, August 2, 2021

Speakers: Uur ahin, zlem Treci, Kathryn Whitehead, Jarrett J. Krosoczka, Ian Kerner, Li-Huei Tsai, Nabiha Saklayen

The talks in brief:

Uur ahin and zlem Treci, cofounders of BioNTech

Big idea: When COVID-19 first reared its many-crowned head in January of 2020, immunologists zlem Treci and Uur ahin saw an opportunity to develop a vaccine at light speed, using revolutionary mRNA technology to turn the bodys own immune system into a COVID-fighting factory.

How? Treci and ahins development of the first approved COVID-19 vaccine impacted the lives of millions if not billions of people worldwide, who were previously defenseless against the deadliest virus to hit humanity in a century. The husband-and-wife team had started their firm BioNTech as a way to bypass the glacial channels of traditional research, and thus were uniquely suited to bring a vaccine to market quickly. Their revolutionary approach used messenger RNA (mRNA) to address the bodys immune system at the cellular level, compiling chemical source code to teach cells how to produce antibodies personalized to each patient. As Treci puts it: mRNA strands are the generals which call all the different special forces and train them on the wanted poster of the attackers. And far from being effective only against COVID-19, mRNA therapy offers the promise of individually adaptable treatments for a variety of cellular disorders, including cancer.

Kathryn A. Whitehead, engineer, teacher, innovator

Big idea: Messenger RNA (or mRNA) is about to change the world forever.

How? After years of research, Kathryn A. Whitehead and her team have finally created the perfect vehicle for delivering life-saving yet delicate mRNA information through vaccines: lipid nanoparticles. She takes us down to the nano level to break down this fatty packaging into its four key ingredients: phospholipids, cholesterol, ionizable lipids and a polymer called PEG. These components make up the ideal shipping materials for delivering life-saving vaccines to our bodys cells. Amazingly, her work is paving the way for mRNA therapies that can treat or cure diseases that have unrelentingly plagued us, including cancer, Type 1 diabetes, muscular dystrophy and cystic fibrosis as well as the flu, malaria, Ebola, Zika and HIV. As she puts it: mRNA therapies are going to usher in a new era of medicine in human health forever, and its all thanks to these fatty little balls that deliver this miracle medicine to exactly where it needs to go.

Jarrett J. Krosoczka delights the crowd with live drawing presentation paired with a poignant autobiographical journey. He speaks at TEDMonterey: The Case for Optimism on August 2, 2021 (Photo: Bret Hartman / TED)

Jarrett J. Krosoczka, author, illustrator

Big idea: Stories keep people alive and help us remember, share and process the human experience.

How? Equipped with sheets of paper and a mug full of markers, Jarrett J. Krosoczka takes us on an autobiographical journey during a live drawing presentation. He begins by drawing his younger self surrounded by his grandfather and the ancestors he was introduced to through family stories. Krosoczka used his sketchbook as a means of escape from the chaos of his upbringing and as a way to connect with his incarcerated mother, who was also an artist. As a teenager, he volunteered at a camp for children with cancer, befriending a four-year-old boy named Eric who had recently been diagnosed with Leukemia. Krosoczkas drawing of Eric is vibrant, depicting the boy with a Power Rangers sword in hand and a huge grin. Krosoczka shares the difficulties of recounting his experience at the camp in his graphic memoir, Sunshine, and how its creation forced him to come face-to-face with unspoken losses. While this can be painful, he explains, stories are an opportunity to understand the human experience, deal with absence and bring loved ones back to life on the page.

Ian Kerner, psychotherapist

Big idea: If youre experiencing a lack of sexual desire, increasing psychological stimulation can help boost feelings of arousal.

How? Failure to launch, or the inability to build and maintain sexual momentum, is a common problem plaguing the couples who work with sex therapist Ian Kerner. His solution? An arousal runway of psychological stimulation. In other words, he suggests getting the mind in on the action before anything physical begins. For couples dealing with shame around sex, he recommends starting with side-to-side experiences like listening to a sexy podcast together or reading literary erotica aloud. Other couples might try face-to-face experiences, such as sharing sexual fantasies to bring some psychological stimuli back into the relationship. After a year spent in a pandemic (and in our pajamas), Kerner reminds us that if were not feeling super sexy, theres nothing wrong with our libidos we just need to try some new strategies to get the sparks flying.

Li-Huei Tsai, professor, neuroscientist

Big idea: A promising approach to Alzheimers and dementia treatment lies in a mind-blowing (or rather, mind-healing) application of gamma wave stimulation.

How? Of the many signals that fire our synapses, the brain relies on gamma frequencies, or waves, to coordinate cellular activity and keep everything in sync. So when these specific brain waves become weaker, it is often an early indication of dementia or Alzheimers. In seeking to understand and treat this degenerative disease, Li-Huei Tsai and her team asked an intriguing question and found promising answers: What if we artificially boosted the brains gamma waves? They started with mice and, through light and sound stimulation carrying the gamma frequency, discovered profound benefits, such as improved memory and less brain decay. But as Tsai says, mice are nice but people are the point. Now theyre testing on humans with an at-home device that emits the same gamma output and the results, so far, are exciting: reduced brain atrophy, improved mental function and increased synchrony. While its early days and theres still work to be done, shes already seeing a lot of evidence that this approach seems safe and that humans tolerate gamma wave stimulation well meaning this non-invasive treatment could prove accessible for those who need it and usher in a better world and brighter future for everyone.

Nabiha Saklayen, biotech entrepreneur

Big idea: The future of regenerative medicine is personalized.

How? What if diseases could be treated with a patients own cells precisely and on demand? This may sound like science fiction, but through personalized, stem-cell-derived therapies, Nabiha Saklayen says this future is closer than we think. How could this work? The answer lies in automation through machine learning. Currently, stem cells are painstakingly difficult (and expensive) to engineer, requiring scientists to manually remove unwanted cells from stem cell cultures. Saklayen describes how we could leverage physics, biology and algorithms to scale up an alternative, affordable approach gathering the perfect culture of your own personal stem cells by utilizing the precision of a computer. Imagining a revolution in personalized pharmaceuticals, forecasts a world where every person could have a personalized bank of these cells to be used as needed.

Read more:
Well, well, well! Notes from Session 3 of TEDMonterey - TED Blog

Global Nanobiotechnologies Markets Report 2021: Comprehensive and Thorough Review of the Current Status of Nanobiotechnology, Research Work in…

DUBLIN, May 21, 2021 /PRNewswire/ -- The "Nanobiotechnologies - Applications, Markets & Companies" report from Jain PharmaBiotech has been added to ResearchAndMarkets.com's offering.

Nanobiotechnology, an integration of physical sciences, molecular engineering, biology, chemistry and biotechnology holds considerable promise of advances in pharmaceuticals and healthcare.

The report starts with an introduction to various techniques and materials that are relevant to nanobiotechnology. It includes some of the physical forms of energy such as nanolasers. Some of the technologies are scaling down such as microfluidics to nanofluidic biochips and others are constructions from bottom up. Application in life sciences research, particularly at the cell level sets the stage for the role of nanobiotechnology in healthcare in subsequent chapters.

Some of the earliest applications are in molecular diagnostics. Nanoparticles, particularly quantum dots, are playing important roles. In-vitro diagnostics, does not have any of the safety concerns associated with the fate of nanoparticles introduced into the human body. Numerous nanodevices and nanosystems for sequencing single molecules of DNA are feasible. Various nanodiagnostics that have been reviewed will improve the sensitivity and extend the present limits of molecular diagnostics.

An increasing use of nanobiotechnology by the pharmaceutical and biotechnology industries is anticipated. Nanotechnology will be applied at all stages of drug development - from formulations for optimal delivery to diagnostic applications in clinical trials. Many of the assays based on nanobiotechnology will enable high-throughput screening. Some of nanostructures such as fullerenes are themselves, drug candidates, as they allow precise grafting of active chemical groups in three-dimensional orientations.

The most important pharmaceutical applications are in drug delivery. Apart from offering a solution to solubility problems, nanobiotechnology provides and intracellular delivery possibilities. Skin penetration is improved in transdermal drug delivery. A particularly effective application is as nonviral gene therapy vectors. Nanotechnology has the potential to provide controlled release devices with autonomous operation guided by the needs.

Nanomedicine is now within the realm of reality starting with nanodiagnostics and drug delivery facilitated by nanobiotechnology. Miniature devices such as nanorobots could carry out integrated diagnosis and therapy by refined and minimally invasive procedures, nanosurgery, as an alternative to crude surgery. Applications of nanobiotechnology are described according to various therapeutic systems. Nanotechnology will markedly improve the implants and tissue engineering approaches as well.

Of the over 1,000 clinical trials of nanomedicines, approximately 100 are selected and tabulated in major therapeutic areas. Other applications such as for management of biological warfare injuries and poisoning are included. Contribution of nanobiotechnology to nutrition and public health such as the supply of purified water are also included.

There is some concern about the safety of nanoparticles introduced in the human body and released into the environment. Research is underway to address these issues. As yet there are no FDA directives to regulate nanobiotechnology but as products are ready to enter the market, these are expected to be in place.

Future nanobiotechnology markets are calculated on the basis of the background markets in the areas of application and the share of this market by new technologies and state of development at any given year in the future. This is based on a comprehensive and thorough review of the current status of nanobiotechnology, research work in progress and anticipated progress.

There is a definite indication of large growth of the market but it will be uneven and cannot be plotted as a steady growth curve. Marketing estimates are given according to areas of application, technologies and geographical distribution starting with 2020. The largest expansion is expected between the years 2024 and 2030.

Profiles of 252 companies, out of over 500 involved in this area, are included in the last chapter along with their 185 collaborations. The report is supplemented with 51 Tables, 32 figures and 800 references to the literature.

The report contains information on the following:

Key Topics Covered:

Part One: Applications & Markets

Executive Summary

1. Introduction

2. Nanotechnologies

3. Nanotechnologies for Basic Research Relevant to Medicine

4. Nanomolecular Diagnostics

5. Nanopharmaceuticals

6. Role of Nanotechnology in Biological Therapies

7. Nanodevices & Techniques for Clinical Applications

8. Nanooncology

9. Nanoneurology

10. Nanocardiology

11. Nanopulmonology

12. Nanoorthopedics

13. Nanoophthalmology

14. Nanomicrobiology

15. Miscellaneous Healthcare Applications of Nanobiotechnology

16. Nanobiotechnology and Personalized Medicine

17. Nanotoxicology

18. Ethical and Regulatory Aspects of Nanomedicine

19. Research and Future of Nanomedicine

20. Nanobiotechnology Markets

21. References

Part Two: Companies

22. Nanobiotech Companies

For more information about this report visit https://www.researchandmarkets.com/r/wk4qzo

Media Contact:

Research and Markets Laura Wood, Senior Manager [emailprotected]

For E.S.T Office Hours Call +1-917-300-0470 For U.S./CAN Toll Free Call +1-800-526-8630 For GMT Office Hours Call +353-1-416-8900

U.S. Fax: 646-607-1907 Fax (outside U.S.): +353-1-481-1716

SOURCE Research and Markets

http://www.researchandmarkets.com

More:
Global Nanobiotechnologies Markets Report 2021: Comprehensive and Thorough Review of the Current Status of Nanobiotechnology, Research Work in...

Nano silver-induced toxicity and associated mechanisms | IJN – Dove Medical Press

Introduction

Nano silver refers to silver particles that have at least one dimension <100 nm on a three-dimensional scale.1 It presents unique physical and chemical properties such as its nano scale, high specific surface area, strong surface reactivity and strong interaction between particles,2 which makes nano silver widely used in various fields, such as imaging, diagnostics, and medicine,3 as well as in paints for the production and preservation of artistic work,4 in cosmetics to improve product safety and stability, in the processing industry as a packaging material to improve food freshness and prolonged release of biologically active ingredients,5 in agrifoods sector to fight against agricultural pest and pathogen, and support food production, in poultry industry sector to product vaccine, control animal skin infections, stimulate immune responses and diagnose.68 Compared to ordinary silver, nano silver has unique biological properties, such as stronger antibacterial activity. Nano silver can be added to toothpaste to achieve an oral sterilization activity, and can be prepared in gel form to treat cervicitis.9 Nano silver has quietly become more common in daily life, and people are increasingly exposed to products that contain nano silver. Nevertheless, individuals are not fully aware of the toxic effects of nano silver, the mechanisms involved in its toxic effects, and potential approaches to modify its toxicity profile are limited. Therefore, this article summarizes recent data to elaborate on these issues to provide a better understanding of the properties of nano silver and to provide insight into its real-life applications.

The methodology adopted to search and summarize this literature review was as follows: (1) an initial search based on key words, including AgNPs, nano silver, silver nanoparticles, metal nanoparticles, toxicity, safety issues and hazard effects, in PubMed, Science Direct, Crossref and other databases; (2) preliminary screening of literature according to the title, keywords, and guideline; (3) addition of new references like a snow ball from original references; and (4) summary and organization of literatures.

Due to the widespread use of nano silver in the environment and everyday products, individuals encounter these nanoparticles in a variety of ways. Nano silver mainly enters the human body via ingestion, inhalation, skin contact, and may directly enter the systemic circulation through intraperitoneal or intravenous injection.10 Silver nanomaterials are used in industrial production processes, resulting in a great amount of silver in the form of nanoparticles being discharged into groundwater with the release of industrial wastewater. Urban and industrial effluents enter the aquatic ecosystem and accumulate along trophic chains, which results in unconscious intake of nano silver.

There are several ways for nano silver to enter the human body and exert its activity (Figure 1). After oral intake, nano silver is absorbed and distributed to organs.11 Studies have shown that after silver nanoparticles enter the body through the respiratory tract, they mainly accumulate in the lungs. After passing through the lung epithelial mucosal system, because of their small particle diameter, the nanoparticles are transported from the lungs to other tissues and diffuse throughout the body.12 The skin is the first barrier between the internal environment of the human body and the external environment as it is directly exposed to the air.13 Nanoparticles are able to penetrate both damaged and healthy skin. Nano silver penetrates the epidermis, diffuses to the dermis, and even the underlying structure of the skin such as the subcutaneous tissue.14 Therefore, there is a strong possibility that nano silver present in cosmetic wound dressings and antibacterial textiles would diffuse through the skin in large amounts. Nano silver injected through the abdominal cavity or intravenously enters the systemic circulation directly. After entering the systemic circulation, they are distributed to the heart, liver, kidney, brain, testes, and ovary and cause organ-specific pathophysiological effects.15

Figure 1 Various routes of exposure to nano silver in human body.

Nano silver enters the biological system through various ways. Routes of exposure and time, size and state of aggregation, and doses of silver nanoparticles link to their bioavailability, biodistribution, and pathological symptoms. To explore the toxicity of nano silver to organs, different animal models are established and employed (Table 1). 1619

Table 1 Toxicity of Nano Silver in Different Organs

Compared to ordinary materials, nano-silver materials have better barrier function, antibacterial ability, and higher mechanical strength, and are widely used in various daily necessities and packaging materials.20 After oral intake, silver nanoparticles reach the stomach rapidly, where they dissolve under acidic conditions. After passing through the intestine, the properties of nano silver are affected. Once absorbed by the intestinal mucosa, nano silver reaches the liver.21

Studies have shown that after a 24-hour intravenous injection of nano silver in rats, higher levels of silver can be detected in the liver, feces, and colon.22 Approximately 30 to 99% of the nano-silver dose will accumulate and sequester in the liver after being administered to the body. This leads to a decrease in delivery to the target diseased tissues and potentially an increase in toxicity at the hepatocyte level.23

Research by Jia et al found that nano silver increased the level of protein phosphorylation of normal human colonic epithelial cells NCM460 and human colorectal cancer HCT116 and promoted the expression of the p53 and Bcl-2-associated X protein (Bax). When the exposure to nano silver was higher than 15 gmL1, the survival rate of both cell types began to decrease. The study also showed that nano silver can promote the downregulation of B cell lymphoma/leukemia-2 (Bcl-2), leading to an increase in the Bax/Bcl-2 ratio and activation of p21, further accelerating cell death.24 DArcy et al showed that silver nanoparticles can induce focal hepatocyte necrosis and apoptosis.25 The apoptosis induced in the liver of mice treated with 10-nm silver nanoparticles indicates that nano silver may induce intercellular stress leading to cell death. Silver nanoparticles may also lead to the destruction of the endoplasmic reticulum (ER) and partial degranulation, causing severe liver tissue and ultrastructural changes that affect the metabolism and function of the liver and other important organs.16

Animal and human studies have shown that inhaled nanoparticles are less efficiently eliminated by macrophage removal mechanisms than other large particles. Nano silver is retained in the lungs and causes damage, or is transported through the circulation, nervous system, and to distal tissues and organs.26 The lung and liver are the main target tissues after exposure to silver nanoparticles via inhalation for 90 days, and the resulting toxicity is dose-dependence.27

The chemical characterization of silver nanoparticles endows them with redox ability. The reaction involves the elements Ag and H2O2 to generate hydroxyl and oxidize silver ions.28 This mechanism allows silver nanoparticles to induce oxidative stress, and this interaction with cellular matter interacts to produce oxidants.29 Surface oxidation of silver nanoparticles may contribute to the release of silver ions, thus amplifying toxicity. Mitochondrial function is impaired when lung epithelial cells are exposed to nano silver. In the process, NADPH oxidase (NOX) activity increases, leading to damage to oxidative stress. Tight junction proteins in the lung epithelium are a known target of oxidative stress damage, which alters epithelial transport processes and damages the homeostasis and integrity of the lung epithelial barrier.30

Lin et al evaluated the physiological toxicity of nano silver for the heart and concluded that nano silver acts quickly and inhibits the activity of rectifying the inward potassium current (IK1) and inward sodium current (INa) channels of cardiomyocytes, leading to rapid collapse of cardiac cell transmembrane potential (TMP) with subsequent loss of excitability. Toxic effects of nano silver on similar channels of the cardiac conduction system and autonomic nerves can also be expected, but the exact mechanism of action needs further study.31

Recombinant myosin heavy chain 6 (MYH6) is a cardiomyocyte marker gene that encodes the alpha heavy chain subunit of cardiac myosin.32 The treatment of silver nanoparticles triggers abnormal changes in ISL1, MYH6, and alpha heavy chain subunits, which seriously damage the process of embryogenesis, germ layer, and heart development. The steps of nano silver to sabotage cardiomyocytes are as follows: (1) silver ions are slowly released from silver nanoparticles; (2) protein crowns are formed by the combination of silver nanoparticles with different serum proteins; and (3) changes occur in the total surface charge of silver nanoparticles, which will disrupt the ion balance in the body and affect the electrophysiology of cardiomyocytes.33

The rapid development of the nanotechnology industry has brought many potential risks that are of serious concern. In order to safely use nanomaterials in consumer products and pharmaceuticals, regulatory health risk assessment of such particles should be mandatory, including the potential impact on reproduction and fertility.34

Silver nanoparticles are able to cross the blood-testis barrier and locate directly in the testes after intraperitoneal or intravenous injection.35 The human testicular embryonic carcinoma cell line (NT2) Ntera2 and primary testicular cells from C57BL6 mice were used as cell models to simulate the repair state and oxidative damage of human testicular cells exposed to silver nanoparticles of 20 and 200 nm in size. Nano silver exhibited strong cytotoxicity and cytostatic properties, causing apoptosis, necrosis, and reduction of proliferation in a concentration- and time-dependent manner. Silver nanoparticles with a size of 200 nm even caused DNA strand breaks in NT2 cells.36

At the cellular level, nano silver generates a large amount of reactive oxygen species (ROS) by activating the inhibitory kappa B kinase/transcription factor nuclear factor-kappa B (IKK/NF-B) signaling pathway, destroying the cytoskeleton and DNA, damaging DNA repair enzymes, and upregulating autophagy to activate p53-dependent or mitochondrial-dependent apoptosis pathways to induce cell apoptosis and exert its cytotoxic effects.37 At the genetic level, a lower dose of silver nanoparticles will lead to changes in human skin fibroblast energy metabolism, oxidative stress, changes in the cell cycle, and in other related genes. Even very low doses of nano silver are capable of causing structural or functional damage to target cells.38 As shown in Table 2, the following mainly describes the cytotoxicity of silver nanoparticles based on the progressive effect induced on cell layers.24,3032,36 Figure 2 shows the potential mechanisms of nano silver-induced cytotoxicity in the cell.

Table 2 Toxicity of Nano Silver in Different Cells

Figure 2 Mechanisms of entry of silver nanoparticles into the organism and potential mechanisms of nano silver-induced cytotoxicity in the cell.

Silver nanoparticles can interact with membrane proteins and activate signaling pathways, thereby inhibiting cell proliferation. They directly interact with the macromolecular structure of living cells and affect cellular metabolism.39 Nano silver interferes with Na and K ion channels on the cell membrane, causing an imbalance in the cell membrane potential, or reacts with sulfhydryl (-SH) protein on the cell membrane destroying the barrier function and the material exchange function of the cell membrane, resulting in direct cell necrosis.40 Gunawan et al used attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy to detect the toxic mechanism of silver nanoparticles in bacteria. The results showed that nanoparticles caused major structural changes in the cell membrane components and interfered with the peptides and lipid chains (phospholipids) as well as sugar and phosphate groups leading to the breakdown of the cell structure.41

Anuj et al explored a scheme to improve the bactericidal effect of linezolid on gram-negative bacteria with nano silver. The change in the zeta-potential caused by the interaction between nano silver and bacterial membrane protein enhanced the permeability of the bacterial cell membrane and the alteration of integrity, which allowed linezolid to penetrate into the cell, thereby increasing the cytoplasmic concentration of linezolid to an effective level. This study demonstrated that silver nanoparticles can change the permeability of the cell membrane, causing the leakage of intracellular material or the entry of extracellular material to cause cell death.42

The cell membrane only allows for free diffusion of oxygen, carbon dioxide, water, small hydrophobic or non-polar molecules, and 1030 nm particles. Various particles enter the cell through different cell internalization pathways. These internalization pathways are classified as endocytosis. The endocytosis mechanism includes phagocytosis and pinocytosis.43 Depending on the size of the vesicles and the proteins involved in the formation of the vesicle, pinocytosis can be further divided into four mechanisms, which include (1) macropinocytosis; (2) clathrin-mediated endocytosis; (3) caveolae-mediated endocytosis; and (4) non-clathrin- and non-caveolin-mediated endocytosis.44

Once the nano silver is internalized, it will migrate to the mitochondria and nucleus and induce changes in cell morphology, oxidative stress, DNA damage, inflammation, genotoxicity, mitochondrial dysfunction, and subsequent apoptosis or necrosis.45

Free nano silver in the extracellular fluid causes only a limited release of ROS in the cell.46 Silver nanoparticles that enter the cell through endocytosis were then transferred to the lysosome. Under the action of the acidic environment of the lysosome, the oxidative dissolution releases silver ions, and the cell itself degrades and releases nano silver, causing a higher degree of ROS release, thereby destroying the lysosome. In the cell membrane, particles escape from the lysosomal sequestration into the cytosol, and then target other subcellular compartments, resulting in a higher degree of cytotoxicity.47 Bouallegui et al used the uptake inhibitor amantadine to evaluate the effects of blocking clathrin-mediated endocytosis on nano silver protein-induced toxicity in mussel gills and digestive glands. Blocking clathrin-mediated endocytosis may protect cells from nano silver toxicity, which indicates that this uptake of clathrin-mediated endocytosis is a key mechanism for silver nanoparticles to exert their toxic effects.48

In a recent study, using 15, 50, and 100 nm silver nanoparticles, Chen et al showed that the smallest 15 nm silver nanoparticles exerted the strongest cytotoxicity. The 100-nm silver nanoparticles aggregate and cannot pass through the plasma membrane, and thus cannot be captured by endocytosis or cause toxicity to the cell.49

Autophagy is a mechanism in which cellular materials are delivered to lysosomes for degradation, leading to the basic turnover of cellular components, and providing energy and macromolecular precursors.50 Autophagy is activated at the basic level under normal physiological conditions, selectively removing stress-mediated protein aggregates, and removing damaged organelles. Autophagy also actively participates in the elimination of cell invaders and maintaining intracellular balance. Studies have shown that exposure of cells to silver nanoparticles activates the cellular defense mechanism defined as autophagy. However, silver nanoparticle-activated autophagy results in defective autophagosome-lysosome fusion, which leads to autophagy defects and increases cell toxicity.51

Ubiquitination confers autophagy selectivity and regulates the stabilization, activation, and transport of proteins involved in the autophagy pathway.52 Silver nanoparticles have been shown to increase the level of enzymes involved in ubiquitination processes or weaken ubiquitination.53 The reactivity of silver nanoparticles can interfere with the formation of ubiquitin. The interference of silver nanoparticles on ubiquitination may be the cause of autophagy defects and cytotoxicity caused by silver nanoparticles.54,55 As a multi-domain adaptor protein, p62 binds microtubule-associated protein 1 light chain 3 (LC3) and ubiquitin. The accumulation of the p62 subunit caused by defective autophagy may also be a potential cause of silver nanoparticle cytotoxicity.56

Lee et al showed for the first time in vitro that nano silver led to the formation of numerous cytoplasmic acid vesicle organelles (AVOs) (autophagosomes and autolysates). In addition, exposure to nano silver resulted in a dose-dependent increase in the conversion of LC3-I to LC3-II and a dose-dependent accumulation of p62 protein, indicating that although nano silver activates autophagy, it may eventually lead to the interruption of autophagy flow.50

Previous investigations have shown that exposure of cells to silver nanoparticles can cause mitochondrial damage. Silver nanoparticles are capable of inducing mitochondrial swelling, increasing intracellular ROS levels, and disrupting mitochondrial membrane potentials, whose breakdown leads to mitochondrial pathway-induced apoptosis.57,58 Silver nanoparticles induce changes in the morphology and structure of mitochondria. The expression of nuclear fission-related protein 1 (p-Drp1) (Ser616) was significantly up-regulated, and the expression of mitochondrial biogenesis protein (PGC-1) in cells treated with nano silver decreased, indicating that silver nanoparticles induce cytotoxicity by targeting mitochondria, leading to the destruction of mitochondrial function and the damage to the mitochondrial structure and morphology that interferes with mitochondrial dynamics and biogenesis.59

The mitochondrial respiratory chain is the main source of ROS in cells. Under normal circumstances, ROS are balanced by the mitochondrial antioxidant system. In the process of cellular stress, mitochondria may malfunction, with increased ROS production, leading to cell damage and cell death.60

Holmila et al studied the effects of silver nanoparticles and ionizing radiation on the mitochondrial redox state and function in lung cell lines (A549, BEAS-2B, Calu-1, and NCI-H358). In Calu-2 cells, exposure to nano silver reduced cell proliferation by inducing cell cycle arrest. Nano silver increased mitochondrial reactive oxygen and protein oxidation in sensitive cell lines in a time- and dose-dependent manner, but did not significantly change mitochondrial respiration mechanisms.61

To demonstrate that nano silver would induce cell death through both the apoptotic p53 pathway and the independent p53 pathway, a model system containing two osteosarcoma cell lines was used and the cell response after nano silver administration was tested.62 Loss of mitochondrial membrane potential, increased leakage of cytochrome C protein into the cytoplasm, and increased ROS levels were detected in both U2OS cells harboring sufficient levels of p53 and in Saos-2 cells lacking functional p53, indicating that nano silver in both cell lines induced mitochondrial stress.63,64 Although nano-silver treatment activates p53 in p53-containing osteosarcoma cells, the main property of nano silver is to induce mitochondrial stress, thus driving cancer cell p53-independent apoptosis.

The ER is a multifunctional subcellular compartment in charge of protein synthesis, assembly and modification, lipid biosynthesis, protein output, calcium ion storage and its regulation and release to the cytoplasm, and redox signals.65 A series of protein-related activities are extremely susceptible to events that interfere with ER homeostasis, leading to accumulation of unfolded and misfolded proteins in the ER. During the process of solving protein folding defects and restoring ER homeostasis, an unfolded protein response is activated, involving three signal branches: RNA-dependent protein kinase-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE 1) and X box binding protein-1 (XBP-1), and activation of transcription factor 6 (ATF6). Many studies have shown that exposure of the body to metal nanoparticles induces the ER stress signaling pathway.

P-glycoprotein (P-gp) is an ATP-binding cassette transporter located on the plasma membrane, which is intrinsically linked to the occurrence of multidrug-resistant cancer.66 Silver nanoparticles of 75 nm in size induce stress in the endoplasmic reticulum in drug-resistant cells, reducing the number of correctly folded P-gp of the plasma membrane. The endoplasmic reticulum cavity is rich in calcium, which is essential for the sustained effect of endoplasmic reticulum protein quality control mechanisms, such as the calnexin/calreticulin cycle. Treatment of drug-resistant cells with 75-nm silver particles will deplete the calcium levels of the endoplasmic reticulum, which may be the cause of the induction of endoplasmic reticulum stress.67

Prolonged exposure of human neuroblastoma cell line (SH-SY5Y) to nano silver has been reported to increase the length of the ER-mitochondria contact site. The expression of phosphatase and tensin homolog deleted on chromosome ten (PTEN) protein in ER and mitochondria-associated membranes (MAMs) is enhanced, and the function of inositol-3-phosphate receptor (IP3R) is altered. Transfer of Ca2+ from the endoplasmic reticulum to the mitochondria increases, and finally the overload of mitochondrial Ca2+ triggers cell death through the mitochondrial apoptosis pathway.68

Lysosomes contain a variety of acid hydrolases, such as cathepsins, which are involved in autophagy and phagocytosis. Autophagy is related to the removal of intracellular (endogenous) debris, and phagocytosis digests exogenous substances.69

The release of silver ions induces only a modest generation of ROS; in contrast, the simultaneous release of silver nanoparticles and silver ions (oxidative dissolution of silver nanoparticles in an acid lysosome environment) induces higher levels of ROS.70 The generation of a large amount of ROS destroys the integrity of the lysosomal membrane and allows the release of silver nanoparticles from the enclosed vesicle into the cytosol. Lysosomal dysfunction due to loss of integrity of the lysosomal membrane or reduced acidity also leads to the release of silver nanoparticles and is closely associated with impaired autophagosome-lysosome fusion.71

Subcytotoxic concentrations of silver nanoparticles (10 gmL1) induce lysosomal dysfunction in liver cancer cells, leading to activation of NOD-like receptor protein 3 (NLRP3) inflammasome-dependent caspase-1. The activation of inflammatory mediators is a biological response induced by silver nanoparticles. NLRP3 inflammatory mediators directly or indirectly interact with nano silver to produce a cellular inflammatory response that leads to cytotoxicity.72

Transcription factor EB (TFEB) plays a key role in the regulation of lysosomal function.73 The activity of TFEB is regulated by its subcellular location. Under certain conditions, such as starvation or lysosomal dysfunction, TFEB transfers to the nucleus and activates the transcription of its target genes. After A549 cells were exposed to nano silver, the gene and protein levels of TFEB binding protein in the cytosol and nucleus decreased, indicating that TFEB expression was transcriptionally inhibited and affected the normal activity of lysosomes.72

The cytotoxicity of nano silver is associated with the available concentration of silver nanoparticles, the duration of activity, the size of the particle, the presence or absence of stabilizers, the type of stabilizer, and the pH of the environment. In addition, the toxic reactions of different types of body cells to nano silver also differ. Below are several approaches that have been proposed to overcome the cytotoxicity induced by nano silver based on the research progress in the recent years.

The toxicity of nano silver is closely related to the size of the particles. Most silver nanoparticles are toxic to the human body, and it is precisely because of their small particle size that they can penetrate human tissues. Zhang et al studied two sizes of nano silver to examine the differences in neurotoxic effects of (20- and 70-nm silver nanoparticles). The results show that 20-nm and 70-nm silver nanoparticles significantly reduce neuronal cell viability, and 20-nm silver nanoparticles exert stronger toxic effects than 70-nm-silver nanoparticles.74

Zhang et al studied the effects of two sizes of silver nanoparticles (10- and 50-nm) on the nitrogen fixation of Azotobacter vinelandii. The marked decrease in the number of bacterial cells associated with the smaller silver nanoparticles indicated nano silver with smaller particle size exerted higher toxicity. Cytometry analysis further confirmed this finding. At the same concentration of 10 mgL1 for 12 h of incubation, the apoptotic rates of cells treated with 10- and 50-nm silver nanoparticles were 20.23% and 3.14%, respectively. Observation under the scanning electron microscope of cells revealed obvious damage to the cell structure, indicating that the toxicity of silver nanoparticles was size dependent.75 Given the above findings and to ensure the desired effects of silver nanoparticles, the influence of the size of silver nanoparticles on their toxicity was briefly summarized in Table 3.1618,74,75

Table 3 The Influence of Size Distribution of Silver Nanoparticles on Their Toxicity

Surface modification of nanoparticles is an effective way to reduce the toxicity of nanoparticles.76 Studies have shown that coated and modified nanoparticles do not lose their original characteristics; however, by modifying the surface of the nanoparticles, the inherent toxicity of the nanoparticles could be reduced, and the biocompatibility of the nanoparticles could be improved at the same time.77,78 The surface functionalization may enable further applications of nano silver in various fields.

Borowik et al synthesized silver nanoparticles using thiobarbituric acid and 11-mercaptoundecanoic acid residues (MUA). Silver nanoparticles coated with MUA were compatible with acridine mutagens. Interaction with ICR-191 could regulate cell viability by influencing mutagens in cells.79

Das et al compared the effect of silver nanoparticles, polyethylene glycol (PEG)-coated silver nanoparticles and bovine serum albumin (BSA) functionalized silver nanoparticles on peripheral blood mononuclear cells in vitro, and found that compared with silver nanoparticles, PEG-coated silver nanoparticles and BSA-functionalized silver nanoparticles produced fewer superoxide anions, nitric oxide, intracellular ROS, reduced glutathione (GSH), oxidized glutathione, and NADPH oxidase. Further surface functionalized silver nanoparticles exhibited less toxicity than unmodified silver nanoparticles.80 Hamilton et al adsorbed silver nanoparticles onto carbon nanotubes and graphene oxide. In vitro cellular experiments showed that silver-carbon nanotube-hydroxyapatite and silver-graphene oxide are less toxic than silver nano particles.81

Nano silver has many excellent properties, but premature release and potential toxicity due to accumulation restrain its further application.82 To make better use of this nanomaterial of great potential, nano silver composite preparations that are in combination with other materials have been proposed. However, most of the studies in this field are focused on the functionality of silver nanoparticle preparations; meanwhile, the human safety of silver nanoparticle composite preparations has not drawn much attention. The formulation of silver nanoparticle composite preparations may also be an approach to overcome the toxicity of silver nanoparticles.83,84

Although nano silver is almost nontoxic at low concentrations, the accumulation of silver nanoparticles in mammalian cells may cause side effects and infections, such as silver burns and silver poisoning, by interacting with different organelles and subcellular components of the body.85 Thus, to overcome this problem, the synthesis of nanocomposite materials has been proposed, which consist of loading silver nanoparticles on a magnetic core. Magnetic core-based nanocomposite materials allow to effectively recover residual particles from the medium. In addition, modification of silver nanoparticles on magnetic particles can also provide stability as a result of their magnetic dispersion. After the silver nanoparticles are deposited on a cobalt core, the cell survival rate is improved, and the toxicity of the nanocomposite particles is even lower than that of the silver nanoparticles.86

Madla-Cruz et al synthesized a nano-silver/carboxymethyl cellulose composite using a green synthesis method and then used MTT reduction assay to evaluate the effects of the silver nanoparticles/carboxymethyl cellulose composite on the viability of normal human gingival fibroblasts (HGF). The viability of HGF was not affected at the experimental concentration that inhibits the growth of microorganisms or reduces the area of the biofilm. When the concentration of the composite is less than 15 gmL1, there were no significant toxic effects on HGF cells.87

To overcome the diffusion of nano silver when injected locally at the target site during positioning and labeling therapy, Lee et al combined silver nanoparticles with porous materials to inhibit the diffusion of the nanoparticles and enhance their biocompatibility to iodine. The mixed complex of cesium-nano silver-pSiMP, and subsequent immunotoxicity experiments showed that no hepatotoxicity was observed in mice treated with nano silver-pSiMP, and the main inflammatory cytokine TNF- level in serum did not change significantly. At 8 and 24 h after injection, the nano silver-pSiMPs treatment group did not present activated lymphocytes or histological changes.88

Yu et al synthesized a composite material of cellulose silver nanoparticles. Even when the concentration of the composite treatment reached 1000 gmL1, the number of viable cells did not decrease significantly. Compared to the control group, the cell viability of normal epithelial cells (FHC) of the human colon incubated with the cellulose nanofibrils (CNF)/AgNP complex (501000 gmL1) did not decrease significantly. These results indicate that the CNF/AgNP complex was not toxic to human cells within 24 h.89

This review introduces the in vivo toxicity of nano silver under different exposure routes, and introduces the mechanism induced by silver nanoparticle cytotoxicity from the outer to inner cell structures. Nano silver is introduced to the human body in by different routes, causing damage to various body systems, including the digestive system, respiratory system, and reproductive system.90 At present, most studies on the toxicity of silver nanoparticles are carried out through in vitro cell tests and animal tests, and there are still some challenges. For example, it is not clear to what extent the intact nano silver itself is absorbed by the human body, or whether the nano silver is altered when exposed to the physiological environment, whether the silver ions released from the nano silver are absorbed, or whether the observed effect is induced by the nano silver itself.91 An inflammatory reaction is caused by ions released by the nano silver or nanoparticle itself. Thus, there is no clear approach to elucidate toxicity mechanisms specific to nano silver.

To effectively evaluate the functional effects of nano silver, a variety of related technologies could be employed to characterize silver nanoparticles and to attempt to overcome the limitations of using a single particle characterization method alone.92 The interaction between nano silver and biological fluids will inevitably change the physical characteristics and uptake or absorption of silver nanoparticles. To determine the potential long-term effects of nano silver in a more realistic situation, the characteristics of silver nanoparticles should be evaluated in an appropriate medium. Multigenerational studies are needed to evaluate intergenerational effects in higher mammalian systems.

Because of the versatility of silver nanoparticle compounds in terms of size, physical properties, and the ability to interact and bind with other compounds, their applicability in different fields is immeasurable. These properties also led to some critical issues such as toxicity to human and animal cells, safe use, long-term exposure, and environmental safety. In future nanotoxicology research, persistent in-depth research will be requested to reveal the ultimate mystery of the toxicity mechanisms induced by nano silver. These findings will help to promote the future applications and development of nano silver-loaded preparations and allow for the use of preventive measures against the toxic risks.

This work was supported by the Jiangxi Provincial Department of Science and Technology (20212ACB206004, 20202ACBL216015 and 20202BABL206157), the National Natural Science Foundation of China (No. 81760639), Young Jinggang Scholar of Jiangxi Province (Jing Zhang) and New Century Talents Project of Jiangxi Province (2017082, Xiang Li and 2020028, Jing Zhang), Jiangxi University of Chinese Medicine 1050 Youth Talent Project (Jing Zhang and Xiang Li), and Jiangxi University of Chinese Medicine Science and Technology Innovation Team Development Program.

The authors report no conflicts of interest in this work.

1. Bakand S, Hayes A. Toxicological considerations, toxicity assessment, and risk management of inhaled nanoparticles. Int J Mol Sci. 2016;17(6):929. doi:10.3390/ijms17060929

2. De Matteis V. Exposure to inorganic nanoparticles: routes of entry, immune response, biodistribution and in vitro/in vivo toxicity evaluation. Toxics. 2017;5(4):29. doi:10.3390/toxics5040029

3. Lee SH, Jun BH. Silver nanoparticles: synthesis and application for nanomedicine. Int J Mol Sci. 2019;20(4):865.

4. Sendra M, Yeste MP, Gatica JM, Moreno-Garrido I, Blasco J. Direct and indirect effects of silver nanoparticles on freshwater and marine microalgae (Chlamydomonas reinhardtii and Phaeodactylum tricornutum). Chemosphere. 2017;179:279289. doi:10.1016/j.chemosphere.2017.03.123

5. Zhang XF, Liu ZG, Shen W, Gurunathan S. Silver nanoparticles: synthesis, characterization, properties, applications, and therapeutic approaches. Int J Mol Sci. 2016;17(9):1534. doi:10.3390/ijms17091534

6. Lang C, Mission EG, Ahmad Fuaad AA-H, Shaalan M. Nanoparticle tools to improve and advance precision practices in the agrifoods sector towards sustainability - A review. J Clean Prod. 2021;293:126063.

7. Bansod SD, Bawaskar MS, Gade AK, Rai MK. Development of shampoo, soap and ointment formulated by green synthesised silver nanoparticles functionalised with antimicrobial plants oils in veterinary dermatology: treatment and prevention strategies. IET Nanobiotechnol. 2015;9(4):165171. doi:10.1049/iet-nbt.2014.0042

8. Wang X, Yuying X, Tang M. Research progress on internal toxicity and the toxic mechanism of silver nanoparticles. Asian J Ecotoxicol. 2018;1:5060.

9. Abd El-Ghany WA, Shaalan M, Salem HM. Nanoparticles applications in poultry production: an updated review. Worlds Poult Sci J. 2021;77(4):10011025. doi:10.1080/00439339.2021.1960235

10. Rezvani E, Rafferty A, McGuinness C, Kennedy J. Adverse effects of nanosilver on human health and the environment. Acta Biomater. 2019;94:145159. doi:10.1016/j.actbio.2019.05.042

11. Ferdous Z, Nemmar A. Health impact of silver nanoparticles: a review of the biodistribution and toxicity following various routes of exposure. Int J Mol Sci. 2020;21(7):2375. doi:10.3390/ijms21072375

12. Gambardella C, Costa E, Piazza V, et al. Effect of silver nanoparticles on marine organisms belonging to different trophic levels. Mar Environ Res. 2015;111:4149. doi:10.1016/j.marenvres.2015.06.001

13. Wong R, Geyer S, Weninger W, Guimberteau JC, Wong JK. The dynamic anatomy and patterning of skin. Exp Dermatol. 2016;25(2):9298. doi:10.1111/exd.12832

14. George R, Merten S, Wang TT, Kennedy P, Maitz P. In vivo analysis of dermal and systemic absorption of silver nanoparticles through healthy human skin. Australas J Dermatol. 2014;55(3):185190. doi:10.1111/ajd.12101

15. Sarhan OM, Hussein RM. Effects of intraperitoneally injected silver nanoparticles on histological structures and blood parameters in the albino rat. Int J Nanomedicine. 2014;9:15051517. doi:10.2147/IJN.S56729

16. Al-Doaiss A, Jarrar Q, Moshawih S. Hepatic histopathological and ultrastructural alterations induced by 10 nm silver nanoparticles. IET Nanobiotechnol. 2020;14(5):405411. doi:10.1049/iet-nbt.2020.0039

17. Chen D, Yang Z. Tissue toxicity following the vaginal administration of nanosilver particles in rabbits. Regen Biomater. 2015;2(4):261265. doi:10.1093/rb/rbv016

18. Pecoraro R, Marino F, Salvaggio A, et al. Evaluation of chronic nanosilver toxicity to Adult Zebrafish. Front Physiol. 2017;8:1011. doi:10.3389/fphys.2017.01011

19. Raj A, Shah P, Agrawal N. Sedentary behavior and altered metabolic activity by AgNPs ingestion in Drosophila melanogaster. Sci Rep. 2017;7(1):15617. doi:10.1038/s41598-017-15645-6

20. Schluesener JK, Schluesener HJ. Nanosilver: application and novel aspects of toxicology. Arch Toxicol. 2013;87(4):569576. doi:10.1007/s00204-012-1007-z

21. Ishizaka T, Nagano K, Tasaki I, et al. Optimization and evaluation of pretreatment method for sp-ICP-MS to reveal the distribution of silver nanoparticles in the body. Nanoscale Res Lett. 2019;14(1):180. doi:10.1186/s11671-019-3016-9

22. Juling S, Bachler G, von Gotz N, et al. In vivo distribution of nanosilver in the rat: the role of ions and de novo-formed secondary particles. Food Chem Toxicol. 2016;97:327335. doi:10.1016/j.fct.2016.08.016

23. Zhang YN, Poon W, Tavares AJ, McGilvray ID, Chan WCW. Nanoparticle-liver interactions: cellular uptake and hepatobiliary elimination. J Control Release. 2016;240:332348. doi:10.1016/j.jconrel.2016.01.020

24. Jia M, Zhang W, He T, et al. Evaluation of the genotoxic and oxidative damage potential of silver nanoparticles in human NCM460 and HCT116 cells. Int J Mol Sci. 2020;21(5):1618. doi:10.3390/ijms21051618

25. DArcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43(6):582592. doi:10.1002/cbin.11137

26. Jo MS, Kim JK, Kim Y, et al. Mode of silver clearance following 28-day inhalation exposure to silver nanoparticles determined from lung burden assessment including post-exposure observation periods. Arch Toxicol. 2020;94(3):773784. doi:10.1007/s00204-020-02660-2

27. Sung JH, Ji JH, Park JD, et al. Subchronic inhalation toxicity of silver nanoparticles. Toxicol Sci. 2009;108(2):452461. doi:10.1093/toxsci/kfn246

28. Liu J, Li S, Fang Y, Zhu Z. Boosting antibacterial activity with mesoporous silica nanoparticles supported silver nanoclusters. J Colloid Interface Sci. 2019;555:470479. doi:10.1016/j.jcis.2019.08.009

29. Abdal Dayem A, Hossain MK, Lee SB, et al. The role of Reactive Oxygen Species (ROS) in the biological activities of metallic nanoparticles. Int J Mol Sci. 2017;18(1):120. doi:10.3390/ijms18010120

30. Garces M, Magnani ND, Pecorelli A, et al. Alterations in oxygen metabolism are associated to lung toxicity triggered by silver nanoparticles exposure. Free Radic Biol Med. 2021;166:324336. doi:10.1016/j.freeradbiomed.2021.02.008

31. Lin CX, Yang SY, Gu JL, Meng J, Xu HY, Cao JM. The acute toxic effects of silver nanoparticles on myocardial transmembrane potential, INa and IK1 channels and heart rhythm in mice. Nanotoxicology. 2017;11(6):827837. doi:10.1080/17435390.2017.1367047

32. Szaraz P, Librach M, Maghen L, et al. In vitro differentiation of first trimester human umbilical cord perivascular cells into contracting cardiomyocyte-like cells. Stem Cells Int. 2016;2016:7513252. doi:10.1155/2016/7513252

33. Hu B, Yin N, Yang R, Liang S, Liang S, Faiola F. Silver nanoparticles (AgNPs) and AgNO3 perturb the specification of human hepatocyte-like cells and cardiomyocytes. Sci Total Environ. 2020;725:138433. doi:10.1016/j.scitotenv.2020.138433

34. Elsharkawy EE, Abd El-Nasser M, Kamaly HF. Silver nanoparticles testicular toxicity in rat. Environ Toxicol Pharmacol. 2019;70:103194. doi:10.1016/j.etap.2019.103194

35. Wang Z, Qu G, Su L, et al. Evaluation of the biological fate and the transport through biological barriers of nanosilver in mice. Curr Pharm Des. 2013;19(37):66916697. doi:10.2174/1381612811319370012

36. Asare N, Instanes C, Sandberg WJ, et al. Cytotoxic and genotoxic effects of silver nanoparticles in testicular cells. Toxicology. 2012;291(13):6572. doi:10.1016/j.tox.2011.10.022

37. Danila OO, Berghian AS, Dionisie V, et al. The effects of silver nanoparticles on behavior, apoptosis and nitro-oxidative stress in offspring Wistar rats. Nanomedicine. 2017;12(12):14551473. doi:10.2217/nnm-2017-0029

38. Shati AA, Elsaid FG. Biosynthesized silver nanoparticles and their genotoxicity. J Biochem Mol Toxicol. 2020;34(1):e22418. doi:10.1002/jbt.22418

39. McShan D, Ray PC, Yu H. Molecular toxicity mechanism of nanosilver. J Food Drug Anal. 2014;22(1):116127. doi:10.1016/j.jfda.2014.01.010

40. Dos Santos CA, Seckler MM, Ingle AP, et al. Silver nanoparticles: therapeutical uses, toxicity, and safety issues. J Pharm Sci. 2014;103(7):19311944. doi:10.1002/jps.24001

41. Gunawan C, Faiz MB, Mann R, et al. Nanosilver targets the bacterial cell envelope: the link with generation of reactive oxygen radicals. ACS Appl Mater Interfaces. 2020;12(5):55575568. doi:10.1021/acsami.9b20193

42. Anuj SA, Gajera HP, Hirpara DG, Golakiya BA. Bacterial membrane destabilization with cationic particles of nano-silver to combat efflux-mediated antibiotic resistance in Gram-negative bacteria. Life Sci. 2019;230:178187. doi:10.1016/j.lfs.2019.05.072

43. Zhang T, Wang L, Chen Q, Chen C. Cytotoxic potential of silver nanoparticles. Yonsei Med J. 2014;55(2):283291. doi:10.3349/ymj.2014.55.2.283

Originally posted here:
Nano silver-induced toxicity and associated mechanisms | IJN - Dove Medical Press