Taiwan’s AI thermometer draws interest in SE Asia – Taiwan News

Taiwan develops AI infrared thermometer. (NHRI image) Taiwan develops AI infrared thermometer. (NHRI image)

TAIPEI (Taiwan News) Many Southeast Asian nations have expressed interest in acquiring a next-generation infrared thermometer developed by Taiwan that is being touted as smart and able to reduce erroneous readings.

Designed by the National Health Research Institutes (NHRI), a government-sponsored and mission-oriented medical research center, the thermometer incorporates artificial intelligence (AI) technologies.

The AI feature allows the device to detect people's faces before reading their temperatures, which reduces errors due to interference by radiation emitted from objects held by the individual being measured for example, a cup of hot coffee or cold drink. The feature is particularly commercially attractive at a time when countries around the world are deploying thermometers to screen suspected patients of the coronavirus.

Compared to other types of contactless thermometer, the system delivered satisfactory results in an experiment of 200 people in open spaces, said Liao Lun-de (), assistant investigator of NHRIs Institute of Biomedical Engineering and Nanomedicine. The masks, hats, and glasses the individuals wore were not found to have affected the temperature readings.

The device will sound an alarm and automatically take photos of individuals whose temperatures exceed a certain level. The alerts will be dispatched to disease control staff so they can respond in an efficient and timely fashion.

The mechanism, which integrates real-time thermal sensor software and AI neuroscience, among other technologies, is also cost-competitive, as some thermometers are priced at over NT$200,000 (US$6,653) a unit, said Liao.

Continued here:
Taiwan's AI thermometer draws interest in SE Asia - Taiwan News

Nanomedicine Market: Industry Analysis and forecast 2026 – Research Columnist

Nanomedicine Marketwas valued US$ XX Bn in 2018 and is expected to reach US$ XX Bn by 2026, at CAGR of XX% during forecast period of 2019 to 2026.

The report study has analyzed revenue impact of covid-19 pandemic on the sales revenue of market leaders, market followers and disrupters in the report and same is reflected in our analysis.

Nanomedicine Market Drivers and Restrains:Nanomedicine is an application of nanotechnology, which are used in diagnosis, treatment, monitoring, and control of biological systems. Nanomedicine usages nanoscale manipulation of materials to improve medicine delivery. Therefore, nanomedicine has facilitated the treatment against various diseases. The nanomedicine market includes products that are nanoformulations of the existing drugs and new drugs or are nanobiomaterials. The research and development of new devices as well as the diagnostics will become, more effective, enabling faster response and the ability to treat new diseases are likely to boost the market growth.

REQUEST FOR FREE SAMPLE REPORT:https://www.maximizemarketresearch.com/request-sample/39223/

The nanomedicine markets are driven by factors such as developing new technologies for drug delivery, increase acceptance of nanomedicine across varied applications, rise in government support and funding, the growing need for therapies that have fewer side effects and cost-effective. However, long approval process and risks associated with nanomedicine (environmental impacts) are hampering the market growth at the global level. An increase in the out-licensing of nanodrugs and growth of healthcare facilities in emerging economies are likely to create lucrative opportunities in the nanomedicine market.

Nanomedicine Market Segmentation Analysis:Based on the application, the nanomedicine market has been segmented into cardiovascular, neurology, anti-infective, anti-inflammatory, and oncology. The oncology segment held the dominant market share in 2018 and is projected to maintain its leading position throughout the forecast period owing to the rising availability of patient information and technological advancements. However, the cardiovascular and neurology segment is projected to grow at the highest CAGR of XX% during the forecast period due to presence of opportunities such as demand for specific therapeutic nanovectors, nanostructured stents, and implants for tissue regeneration.

Nanomedicine Market Regional Analysis:Geographically, the Nanomedicine market has been segmented into North America, the Europe, Asia Pacific, Latin America, and Middle East & Africa. North America held the largest share of the Nanomedicine market in 2018 due to the rising presence of patented nanomedicine products, the availability of advanced healthcare infrastructure and the rapid acceptance of nanomedicine. The market in Asia Pacific is expected to expand at a high CAGR of XX% during the forecast period thanks to rise in number of research grants and increase in demand for prophylaxis of life-threatening diseases. Moreover, the rising investments in research and development activities for the introduction of advanced therapies and drugs are predicted to accelerate the growth of this region in the near future.

DO INQUIRY BEFORE PURCHASING REPORT HERE:https://www.maximizemarketresearch.com/inquiry-before-buying/39223/

Nanomedicine Market Competitive landscapeMajor Key players operating in this market are Abbott Laboratories, CombiMatrix Corporation, General Electric Company, Sigma-Tau Pharmaceuticals, Inc, and Johnson & Johnson. Manufacturers in the nanomedicine are focusing on competitive pricing as the strategy to capture significant market share. Moreover, strategic mergers and acquisitions and technological innovations are also the key focus areas of the manufacturers.

The objective of the report is to present a comprehensive analysis of Nanomedicine Market including all the stakeholders of the industry. The past and current status of the industry with forecasted market size and trends are presented in the report with the analysis of complicated data in simple language. The report covers all aspects of the industry with a dedicated study of key players that includes market leaders, followers and new entrants by region. PORTER, SVOR, PESTEL analysis with the potential impact of micro-economic factors by region on the market are presented in the report. External as well as internal factors that are supposed to affect the business positively or negatively have been analyzed, which will give a clear futuristic view of the industry to the decision-makers. The report also helps in understanding Nanomedicine Market dynamics, structure by analyzing the market segments and project the Nanomedicine Market size. Clear representation of competitive analysis of key players By Type, Price, Financial position, Product portfolio, Growth strategies, and regional presence in the Nanomedicine Market make the report investors guide.Scope of the Nanomedicine Market:

Nanomedicine Market by Modality:

Diagnostics TreatmentsNanomedicine Market by Diseases:

Oncological Diseases Infectious Diseases Cardiovascular Diseases Orthopedic Disorders Neurological Diseases Urological Diseases Ophthalmological Diseases Immunological DiseasesNanomedicine Market by Application:

Neurology Cardiovascular Anti-Inflammatory Anti-Infectives OncologyNanomedicine Market by Region:

Asia Pacific North America Europe Latin America Middle East AfricaNanomedicine Market Major Players:

Abbott Laboratories CombiMatrix Corporation General Electric Company Sigma-Tau Pharmaceuticals, Inc Johnson & Johnson Mallinckrodt plc. Merck & Company, Inc. Nanosphere, Inc. Pfizer, Inc. Teva Pharmaceutical Industries Ltd. Celgene Corporation UCB (Union Chimique Belge) S.A. AMAG Pharmaceuticals Nanospectra Biosciences, Inc. Arrowhead Pharmaceuticals, Inc. Leadiant Biosciences, Inc. Epeius Biotechnologies Corporation Cytimmune Sciences, Inc.

MAJOR TOC OF THE REPORT

Chapter One: Nanomedicine Market Overview

Chapter Two: Manufacturers Profiles

Chapter Three: Global Nanomedicine Market Competition, by Players

Chapter Four: Global Nanomedicine Market Size by Regions

Chapter Five: North America Nanomedicine Revenue by Countries

Chapter Six: Europe Nanomedicine Revenue by Countries

Chapter Seven: Asia-Pacific Nanomedicine Revenue by Countries

Chapter Eight: South America Nanomedicine Revenue by Countries

Chapter Nine: Middle East and Africa Revenue Nanomedicine by Countries

Chapter Ten: Global Nanomedicine Market Segment by Type

Chapter Eleven: Global Nanomedicine Market Segment by Application

Chapter Twelve: Global Nanomedicine Market Size Forecast (2019-2026)

Browse Full Report with Facts and Figures of Nanomedicine Market Report at:https://www.maximizemarketresearch.com/market-report/nanomedicine-market/39223/

About Us:

Maximize Market Research provides B2B and B2C market research on 20,000 high growth emerging technologies & opportunities in Chemical, Healthcare, Pharmaceuticals, Electronics & Communications, Internet of Things, Food and Beverages, Aerospace and Defense and other manufacturing sectors.

Contact info:

Name: Lumawant Godage

Organization: MAXIMIZE MARKET RESEARCH PVT. LTD.

Email: sales@maximizemarketresearch.com

Contact: +919607065656/ +919607195908

Website: http://www.maximizemarketresearch.com

Originally posted here:
Nanomedicine Market: Industry Analysis and forecast 2026 - Research Columnist

A new, inhaled siRNA therapeutic option for asthma – Advanced Science News

Share

Share

Email

After more than 20 years of research, we are now witnessing a breakthrough of small interfering RNA (siRNA)-based therapies. In 2018, the first-ever siRNA drug, Onpattro, reached the market, followed by the approval of Givlaari in 2019, and many other clinical trials are in progress.

Holding the potential to treat a wide range of diseases from cancer to immunological disorders, siRNA therapeutics have received plenty of attention. With the support of a suitable delivery system, they can be directed to downregulate a specific target gene. Both approved siRNA drugs Onpattro and Givlaari are only able to reach the liver, however. Other organs that can be treated by loco-regional administration, such as the lung, are, in principle, good targets for siRNA therapies as well.

In this view, siRNA-baseddrugs could not only act as an ally in the battle against the current COVID-19pandemic but also against other severe lung diseases such as asthma. Despitethe great advances in asthma treatment, this disease still represents an unmetmedical need in about 510% of patients.Moreover, most of the available drugs work symptomatically rather than causally.

In a recent article published in WIREs Nanomedicine and Nanobiotechnology, Domizia Baldassi and Tobias Keil, graduate students in Prof. Olivia Merkels research group at the University of Munich, discuss the groups advances towards developing a nanocarrier that can deliver siRNA into T cells in the lung.

The aim of T-cell delivery is downregulation of GATA-3, the transcription factor of T helper 2 (TH2) cells overexpressed in asthmatic patients, which is recognized as a key factor in the asthmatic inflammatory cascade. Based on their observation that transferrin receptor is overexpressed in activated T cells, the researchers sought to find a virus-like tool to target activated TH2 cells specifically and efficiently in a receptor-mediated manner.

They accomplishedthis goal by creating a conjugate formed by transferrin and low-molecular-weightpolyethylenimine (Tf-PEI). On the one hand, they used a well-known cationic polymerto electrostatically interact with the negatively charged siRNA and protect itfrom degradation during the journey through the airways. And on the other hand,transferrin served as a targeting moiety to mediate a specific, targeteddelivery of siRNA only to activated T cells.

Since theendosomal escape is considered the rate-limiting step in cytoplasmaticdelivery of nanoparticle-based therapies, improving this aspect of theformulation was the focus, and Tf-PEI was blended with a second conjugatecomposed of melittin and PEI (Mel-PEI). Melittin is a well-known membranolyticagent from bee venom that was chemically modified to react in a pH-dependentmanner.

The researchersexploited the intrinsic lytic characteristic of the peptide to improve therelease of siRNA into the cytosol, reaching knockdown levels as high as 70% exvivo. But further steps such as the validation of these results in vivo on anasthma mouse model are needed, as well as possible alternative polymericmaterials.

In the process of developing a new pharmaceutical product, it is crucial to keep the administration route in mind. Spray drying is the most straightforward technique to produce inhalable particles for pulmonary delivery, according to the researchers. In a proof-of-concept study, they obtained nano-in-microparticles by spray drying PEI-pDNA polyplexes together with a cryoprotectant agent. After seeing promising results, their studies to obtain a dry powder formulation of siRNA-based polyplexes are ongoing.

Ultimately, both research fields will be combined and hopefully result in a new therapy for the treatment of severe, uncontrolled asthma and many other lung diseases, concluded Baldassi, Keil, and Merkel.

Reference: Tobias W. M. Keil et al. T-cell targeted pulmonary siRNA delivery for the treatment of asthma. WIREs Nanomedicine and Nanobiotechnology (2020). DOI: 10.1002/10.1002/wnan.1634

Link:
A new, inhaled siRNA therapeutic option for asthma - Advanced Science News

University of Waterloo researchers developing non-invasive COVID-19 vaccine – Toronto Sun

A new, non-invasive COVID-19 vaccine is being developed by researchers at the University of Waterloo.

The DNA-based vaccine will be delivered as a nasal spray and have both preventative and therapeutic effect.

Dr. Roderick Slavcev, a professor in the School of Pharmacy, explained Wednesday that the vaccine uses bacteriophage the process allows the vaccine to replicate within bacteria already in the body and is being designed to target tissues in the nasal cavity and lower respiratory tract.

Once complete, our DNA-based vaccine delivers nanomedicine engineered to immunize and decrease COVID-19 infections, said Slavcev, who specializes in designing vaccines, pharmaceuticals and gene-therapy treatments.

The genetic cargo to be delivered attaches to receptors in the respiratory tract, where COVID-19 would attach so its competing for the same spots.

Dr. Roderick Slavcev, a professor in the UW School of Pharmacy.

The DNA vaccine, he adds, will both stimulate an immune response, firing up the bodys capacity to fight off COVID-19, and also prevent further infection.

The vaccine enters cells in targeted tissues and causes them to produce a virus-like particle (VLP) that will stimulate an immune response.

The VLP will look similar to the structure of SARS-CoV-2 the virus which causes COVID-19 but is harmless. However, the similarity activates the bodys natural immune response to protect against viral infections comparable to the VLP, including SARS-CoV-2. In binding to the same receptors that SARS-CoV-2 would bind to, it limits possible sites for transmission.

In this way, the vaccine can build immunity against COVID-19 and decrease the severity of infections in progress, working as a therapeutic and a vaccine.

Said Slavcev, Its a cool way to mimic a virus for maximal protection.

Slavcev is teaming up with Emmanuel Ho, another professor at the School of Pharmacy, and Marc Aucoin, professor of chemical engineering.

Hos team is designing the nanomedication that will be delivered by the nasal spray, which is currently being tested. Aucoins lab is constructing and purifying the VLP and boosting immunity following the initial administration of the therapeutic vaccine.

Asked for a time line, Slavcev said, We are expediting quickly. We anticipate going to animal trials within six months and hope to have completed them by May of 2021.

lbraun@postmedia.com

Continue reading here:
University of Waterloo researchers developing non-invasive COVID-19 vaccine - Toronto Sun

NANOBIOTIX Announces Fast Track Designation Granted By U.S. FDA For Investigation of First-in-class NBTXR3 In Head and Neck Cancer – Business Wire

PARIS & CAMBRIDGE, Mass.--(BUSINESS WIRE)--Regulatory News:

NANOBIOTIX (Paris:NANO) (Euronext: NANO - ISIN: FR0011341205 the Company), a clinical-stage nanomedicine company pioneering new approaches to the treatment of cancer, today announced that the U.S. Food and Drug Administration (FDA) has granted Fast Track designation for the investigation of NBTXR3 activated by radiation therapy, with or without cetuximab, for the treatment of patients with locally advanced head and neck squamous cell cancer who are not eligible for platinum-based chemotherapy.

Fast Track is a process designed to facilitate the development and accelerate the review of drugs for serious conditions and that have the potential to address unmet medical needs. The purpose is to expedite the availability of new treatment options for patients.

A product that receives Fast Track designation is eligible for1:

About NBTXR3

NBTXR3 is a first-in-class product designed to destroy tumors through physical cell death when activated by radiotherapy. NBTXR3 has a high degree of biocompatibility, requires one single administration before the first radiotherapy treatment session, and has the ability to fit into current worldwide radiotherapy radiation therapy standards of care. The physical mode of action of NBTXR3 makes it applicable across solid tumors such as lung, prostate, liver, glioblastoma, and breast cancers.

NBTXR3 is actively being evaluated locally advanced head and neck squamous cell carcinoma (HNSCC) of the oral cavity or oropharynx in elderly and frail patients unable to receive chemotherapy or cetuximab with limited therapeutic options. Promising results have been observed in the phase I trial regarding local control. In the United States, the company has started the regulatory process for the clinical authorization of a phase II/III trial in locally advanced head and neck cancers.

Nanobiotix is also running an Immuno-Oncology development program. The Company received FDA approval to launch a clinical trial of NBTXR3 activated by radiotherapy in combination with anti-PD-1 antibodies in locoregional recurrent (LRR) or recurrent and metastatic (R/M) HNSCC amenable to re-irradiation of the HN and lung or liver metastases (mets)from any primary cancer eligible for anti-PD-1.

The other ongoing NBTXR3 trials are treating patients with hepatocellular carcinoma (HCC) or liver metastases, locally advanced or unresectable rectal cancer in combination with chemotherapy, head and neck cancer in combination with concurrent chemotherapy, and prostate adenocarcinoma. Furthermore, the company has a large-scale, comprehensive clinical research collaboration with The University of Texas MD Anderson Cancer Center (9 new phase I/II clinical trials in the United States) to evaluate NBTXR3 across head and neck, pancreatic, thoracic, lung, gastrointestinal and genitourinary cancers.

About NANOBIOTIX: http://www.nanobiotix.com

Incorporated in 2003, Nanobiotix is a leading, clinical-stage nanomedicine company pioneering new approaches to significantly change patient outcomes by bringing nanophysics to the heart of the cell.

The Nanobiotix philosophy is rooted in designing pioneering, physical-based approaches to bring highly effective and generalized solutions to address unmet medical needs and challenges.

Nanobiotixs first-in-class, proprietary lead technology, NBTXR3, aims to expand radiotherapy benefits for millions of cancer patients. Nanobiotixs Immuno-Oncology program has the potential to bring a new dimension to cancer immunotherapies.

Nanobiotix is listed on the regulated market of Euronext in Paris (Euronext: NANO / ISIN: FR0011341205; Bloomberg: NANO: FP). The Companys headquarters are in Paris, France, with a US affiliate in Cambridge, MA, and European affiliates in France, Spain and Germany

1https://www.fda.gov/patients/fast-track-breakthrough-therapy-accelerated-approval-priority-review/fast-track

Disclaimer

This press release contains certain forward-looking statements concerning Nanobiotix and its business, including its prospects and product candidate development. Such forward-looking statements are based on assumptions that Nanobiotix considers to be reasonable. However, there can be no assurance that the estimates contained in such forward-looking statements will be verified, which estimates are subject to numerous risks including the risks set forth in the reference document of Nanobiotix registered with the French Financial Markets Authority (Autorit des Marchs Financiers) under number R.19-018 on April 30, 2019 (a copy of which is available on http://www.nanobiotix.com) and to the development of economic conditions, financial markets and the markets in which Nanobiotix operates. The forward-looking statements contained in this press release are also subject to risks not yet known to Nanobiotix or not currently considered material by Nanobiotix. The occurrence of all or part of such risks could cause actual results, financial conditions, performance or achievements of Nanobiotix to be materially different from such forward-looking statements.

Read the original:
NANOBIOTIX Announces Fast Track Designation Granted By U.S. FDA For Investigation of First-in-class NBTXR3 In Head and Neck Cancer - Business Wire

Multifunctional nanomedicine: Developing smarter therapeutics – Advanced Science News

Share

Share

Email

Althoughthe medical and pharmaceutical fields have come a long way in diagnosing diseasestates and producing highly potent drugs, the lack of effective delivery ofsuch therapeutics to the target organ with desired pharmacokinetics remains oneof the major challenges in this process.

The advent of nanotechnology, along with advances in protein engineering and materials science, have brought new hope to patients. The impact of nanotechnology on medicine nanomedicine is recognized by the development of novel nanoscale therapeutics and diagnostic and imaging modalities.

Ina recent review published in WIREs Nanomedicine and Nanobiotechnology, Professor Joerg Lahann and his team from the University of Michigan discuss state-of-the-art nanoparticle drug delivery platforms, their advantages and shortcomings, and future directions towards clinical translation.

The ability to impart multiple functions to a single delivery system, engineering both bulk and surface properties, provides a means to answer some of the greatest remaining challenges in the field of drug delivery, said Jason Gregory, a PhD student in the Lahann Lab.

In fact, approaches to address this conundruminclude the development of multifunctional particles, cell-mediated transportmechanisms, and the use of biologically derived materials. Multifunctionalparticles can possess two or more dissimilar properties through surface or bulkanisotropy.

For example, the electrohydrodynamic co-jetting process, which was pioneered in the Lahann lab, permits the creation of multicompartmental particles. Independently engineering individual compartments of the nanoparticle leads to an ability to incorporate materials with orthogonal properties that may offer a solution to simultaneously address multiple biological barriers.

Multicompartmental particles provide a set ofunique features for nanoparticle targeting and controlled release ofcombination drugs, said Dr. Joerg Lahann, the Wolfgang Pauli CollegiateProfessor of Chemical Engineering and Director of the Biointerfaces Instituteat the University of Michigan.

While traditional nanoparticles fail to efficiently deliver the drug to target sites, our bodys circulatory cells as natural carriers of many substances have evolved properties to optimally perform delivery functions. Imparting these properties into the design of the drug delivery platforms by combining nanoparticles with circulatory cells enhances the overall outcome of the system, added Nahal Habibi, a PhD student in the Lahann Lab working on cell-mediated drug delivery strategies.

Leukocytes are particularly good candidates because they can naturally migrate to disease-relevant regions that are often inaccessible by traditional nanoparticles, and have been used to carry therapeutic nanoparticles to cross the bloodbrain barrier in a Parkinsons disease model.

Synthetic protein nanoparticles are another emerging trend in nanomedicine.

Advances in designing novel multicompartmental polymer/protein nanoparticles utilizing the intersection of polymer chemistry and protein biochemistry offers promise in engineering the next generation of nanoparticle formulations, said Daniel Quevedo, another PhD candidate in Prof. Lahanns group.

Reference: Nahal Habibi et al. Emerging methods in therapeutics using multifunctional nanoparticles. WIREs Nanomedicine and Nanobiotechnology (2020). DOI: 10.1002/wnan.1625

Excerpt from:
Multifunctional nanomedicine: Developing smarter therapeutics - Advanced Science News

Biomedical Applications of Zeolitic Nanoparticles, with an Emphasis on | IJN – Dove Medical Press

Hossein Derakhshankhah, 1, 2,* Samira Jafari, 1, 2,* Sajad Sarvari, 3 Ebrahim Barzegari, 4 Faezeh Moakedi, 5 Milad Ghorbani, 6 Behrang Shiri Varnamkhasti, 1 Mehdi Jaymand, 7 Zhila Izadi, 1, 8 Lobat Tayebi 9

1Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; 2Zistmavad Pharmed Co., Tehran, Iran; 3Department of Pharmaceutical and Pharmacological Science, School of Medicine, West Virginia University, Morgantown, WV, USA; 4Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; 5Department of Biochemistry and Molecular Biology, School of Medicine, West Virginia University, Morgantown, WV, USA; 6Department of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran; 7Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; 8Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; 9Marquette University School of Dentistry, Milwaukee, WI 53201, USA

*These authors contributed equally to this work

Correspondence: Zhila Izadi; Lobat Tayebi Email izadi_zh@razi.tums.ac.ir; lobat.tayebi@marquette.edu

Abstract: The advent of porous materials, in particular zeolitic nanoparticles, has opened up unprecedented putative research avenues in nanomedicine. Zeolites with intracrystal mesopores are low framework density aluminosilicates possessing a regular porous structure along with intricate channels. Their unique physiochemical as well as physiological parameters necessitate a comprehensive overview on their classifications, fabrication platforms, cellular/macromolecular interactions, and eventually their prospective biomedical applications through illustrating the challenges and opportunities in different integrative medical and pharmaceutical fields. More particularly, an update on recent advances in zeolite-accommodated drug delivery and the prevalent challenges regarding these molecular sieves is to be presented. In conclusion, strategies to accelerate the translation of these porous materials from bench to bedside along with common overlooked physiological and pharmacological factors of zeolite nanoparticles are discussed and debated. Furthermore, for zeolite nanoparticles, it is a matter of crucial importance, in terms of biosafety and nanotoxicology, to appreciate the zeolite-bio interface once the zeolite nanoparticles are exposed to the bio-macromolecules in biological media. We specifically shed light on interactions of zeolite nanoparticles with fibrinogen and amyloid beta which had been comprehensively investigated in our recent reports. Given the significance of zeolite nanoparticles interactions with serum or interstitial proteins conferring them new biological identity, the preliminary approaches for deeper understanding of administration, distribution, metabolism and excretion of zeolite nanoparticles are elucidated.

Keywords: zeolite, mesoporous, nanostructure, biosafety, biomedical applications

This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License.By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.

See the article here:
Biomedical Applications of Zeolitic Nanoparticles, with an Emphasis on | IJN - Dove Medical Press

Healthcare Nanotechnology (Nanomedicine) Market Share, Size, Future Demand, Global Research, Top Leading Player, Emerging Trends and Forecast to 2015 …

In 2018, the market size of Casing Heads Market is million US$ and it will reach million US$ in 2025, growing at a CAGR of from 2018; while in China, the market size is valued at xx million US$ and will increase to xx million US$ in 2025, with a CAGR of xx% during forecast period.

In this report, 2018 has been considered as the base year and 2018 to 2025 as the forecast period to estimate the market size for Casing Heads .

This report studies the global market size of Casing Heads , especially focuses on the key regions like United States, European Union, China, and other regions (Japan, Korea, India and Southeast Asia).

Request Sample Report @ https://www.researchmoz.com/enquiry.php?type=S&repid=2566962&source=atm

This study presents the Casing Heads Market production, revenue, market share and growth rate for each key company, and also covers the breakdown data (production, consumption, revenue and market share) by regions, type and applications. Casing Heads history breakdown data from 2014 to 2018, and forecast to 2025.

For top companies in United States, European Union and China, this report investigates and analyzes the production, value, price, market share and growth rate for the top manufacturers, key data from 2014 to 2018.

In global Casing Heads market, the following companies are covered:

TechnipFMCJMP Petroleum TechnologiesDelta CorporationGE Oil & GasIntegrated EquipmentWellhead Systems Incorporated (WSI)TIGER VALVE COMPANYHorizon WellheadWorldwide Oilfield Machine (WOM)Larkin ProductsIAL ENGINEERING SERVICES

Segment by RegionsNorth AmericaEuropeChinaJapan

Segment by TypeMonolithic TypeCombined Type

Segment by ApplicationOil/Gas WellsOther

Make An EnquiryAbout This Report @ https://www.researchmoz.com/enquiry.php?type=E&repid=2566962&source=atm

The content of the study subjects, includes a total of 15 chapters:

Chapter 1, to describe Casing Heads product scope, market overview, market opportunities, market driving force and market risks.

Chapter 2, to profile the top manufacturers of Casing Heads , with price, sales, revenue and global market share of Casing Heads in 2017 and 2018.

Chapter 3, the Casing Heads competitive situation, sales, revenue and global market share of top manufacturers are analyzed emphatically by landscape contrast.

Chapter 4, the Casing Heads breakdown data are shown at the regional level, to show the sales, revenue and growth by regions, from 2014 to 2018.

Chapter 5, 6, 7, 8 and 9, to break the sales data at the country level, with sales, revenue and market share for key countries in the world, from 2014 to 2018.

You can Buy This Report from Here @ https://www.researchmoz.com/checkout?rep_id=2566962&licType=S&source=atm

Chapter 10 and 11, to segment the sales by type and application, with sales market share and growth rate by type, application, from 2014 to 2018.

Chapter 12, Casing Heads market forecast, by regions, type and application, with sales and revenue, from 2018 to 2024.

Chapter 13, 14 and 15, to describe Casing Heads sales channel, distributors, customers, research findings and conclusion, appendix and data source.

Link:
Healthcare Nanotechnology (Nanomedicine) Market Share, Size, Future Demand, Global Research, Top Leading Player, Emerging Trends and Forecast to 2015 ...

Students of the Month: November | Local – Fredericksburg.com

Hannah Williams, the daughter of Donald Williams and Sandra Williams, is a senior at Brooke Point High School. Hannah has ben a member of National Honor Society, Spanish Honor Society, Inspire BP, HOSA, Model UN and Spirit Club. Hannah has been a member of the varsity gymnastics team and has received awards for Most Improved. She has been named to the honor roll every year and received academic honors and academic excellence awards. She has also been recognized by the Virginia House of Delegates. Hannah has been a junior volunteer at Stafford Hospital, and she is a certified EMT volunteer at Brooke Fire Station. She also volunteers through her church and through her membership in NHS. Hannah plans to attend a four-year university to study either nursing or biology.

Alanda Guan, the daughter of Min Gao and Xue Guan, is a senior at Colonial Forge High School. Alanda has served as co-president of Environmental Club, president of Science Honor Society and vice president of Spanish Honor Society. She has been a member of National Honor Society, Math Honor Society, Fossil Free Fredericksburg and Chamber Orchestra. She tutors a middle school student, privately. Alanda has been named an AP Scholar with Distinction and has been placed on the AB honor roll. Alanda has served her community at the Stafford Junction Brainbuilders Program, working as tutor during the school year and as a group leader during summer camp. She has also been a Mary Washington Hospital junior volunteer and helped with Friends of the Rappahannock River cleanups. Alanda plans to attend the University of Virginia and major in biology.

Carter Sprinkle is a senior at Courtland High School. Carter has served as treasurer of National Honor Society, vice president of Spanish Honor Society, and has been a member of Environmental club, which completed bulb and tree planting projects, and donated to help the school purchase filtered water fountains. She is also a senior leader with the Youth In Philanthropy program of The Community Foundation. Carter is a member of Symphonic Orchestra and received Superior rating for Assessments. She placed second for the 2-mile during Track District Championship. Carter has also served as captain of the varsity field hockey team. She was named first team All-District and All-Region, and she has received the Coachs Award for field hockey, demonstrating leadership on and off the field. She also holds the saves record at Courtland High School with 28 saves during one game.

Natalie Polanosky is a senior at Fredericksburg Christian High School. Natalie has been involved in FCS Leadership Team since her freshman year. Through this, she has had the opportunity to attend ACSIs National Leadership Conference two years in a row. She has also been the chaplin for Student Council, served as junior class vice president and senior class president. Natalie has choreographed the schools spring musical, Freaky Friday. She has been a member of the varsity swim team for four years, qualifying for VISAA states every year and holding two records for FCS. She has also been a varsity cheerleader. Natalie has coached gymnastics at Stafford Gymnastics for three years, taught acrobatics at Polaris Dance Institute and InSuRGe Dance company, and choreographed/assistant choreographed multiple shows with Christian Youth Theater. Natalie will attend college to pursue a degree in linguistics, she has taken both French and Spanish throughout the past four years and grown very passionate about language.

Emma Elizabeth Kruus, the daughter of Mikael and Marijayne Kruus, is a senior at James Monroe High School. Emma has been a member of National Honor Society and the JUMP Mentorship Program. She has served as executive treasurer of SCA. Emma is a member of DECA; she has served as secretary and vice-president. She was awarded second place for Community Service Project at DECA International Career Development Conference and second place at DECA State Leadership Conference. Emma is a Monroe Scholar. She has received the School Board Merit Award, and she has been named Outstanding Student in IB English and AP Virginia/U.S. History. Emma has played varsity lacrosse and in the JM Winter Lacrosse League. She has been named All Region, All District second team, and All Area honorable mention. Emma was 2019 Fredericksburg Area Teen Gala co-chair for St. Jude Childrens Research Hospital which raised $62,212 and 2020 Fredericksburg Area Teen Gala co-chair for St. Jude Childrens Research Hospital. She has served as teen volunteer coordinator at the Josh Hardy Superhero Celebration and volunteered at the Ceili Leahy Day of Service. Emma plans to attend a four-year university and major in pre-law.

Alex Catahan, the son of Rodolfo and Susan Catahan, is a senior at Massaponax High School. Throughout high school, Alex has been awarded AP Calculus BC award at the Commonwealth Governors School program, and he has received an award for above 4.0 GPA all four years of high school. He has also received the all-around CGS award, awarded to one student who is unanimously chosen by the teachers in the program. Alex was also nominated and selected to attend Boys State. The class he enjoyed most in high school was calculus. Alex wants to attend the University of Virginia and major in biomedical engineering. In the future, he hopes to be on the forefront of biomedical research and research nanomedicine in cancer therapeutics.

William Schmitt, son of Corinne and Daniel Schmitt of Stafford County, is a senior at Mountain View High School. William is a full IB student who tutors other students for math and chemistry. He is president of Latin Club, part of the Lacrosse team, and has been awarded the title Face of IB within the full IB program. He plans to continue his education at the college level, majoring in computer science.

Dulanda Saintcyr, the daughter of Gertrude Bonheur and Jean Saintcyr, is a senior at North Stafford High School. Dulanda is known as a compassionate and strong person, so it is no surprise that her goal is becoming a civil rights attorney. Dulanda has pushed for equality and recognition of others in a variety of ways for the North Stafford community. She is the founder in North Staffords Black History Month, president of the Stafford NAACP Youth Council for three years, treasurer of the National Honor Society, a member of the superintendents Equity, Diversity and Opportunity Committee, and cofounder and communications director of the Mobilizing Youth Project (MYP). MYP was specifically designed to increase youth involvement in local politics. Additionally, Dulandas four-year-long independent research project is on Racial Inequality on Youth Development. This process has afforded her the opportunity to publish her information in her first novel, For the Souls with Different Tones, in 2018. Dulanda has made a positive and empowering impact on North Stafford High School, where she encourages her teachers, friends and family in so many ways.

Sarah Statler, the daughter of Kristi and John Statler, is a senior at Riverbend High School. Sarah is a member of National Honor Society, Math Honor Society, Science Honor Society, Marketing Honor Society, Spanish Honor Society and Spanish Club. She is a member of SCA, serving as Bear Squad leader and as a senate council member. She has been named to distinguished honor rolls and received academic letters. She has been named Student of the Year in AP Calculus and honors math analysis. She has been named an AAU Academic All-American. As a member of DECA, Sarah has served as district president and vice president of state regions three and four. She has earned a DECA letter, is an international competition finalist, a state competition finalist, and customer service and school-based enterprise certified. She has received a merit award and been named Marketing Student of the Year. She has also served DECA as community service coordinator. Sarah has played varsity volleyball, serving as captain, and she has been libero for Fredericksburg Juniors Volleyball Club. She has been a member of the varsity swim team, receiving the award for Best Work Ethic. She is a summer swim coach and has received the summer swim coaches award. She has also been a member of the varsity tennis team. Sarah is an Ambassador Girl Scout. She serves her community as a tutor for elementary and high school students. She has also volunteered at Mary Washington Hospital, the SPCA and at Harvest Hayride. Sarah will continue her education at a four-year college or university and plans to major in kinesiology, biology or chemistry.

Melani Corral, the daughter of Dina Corral and Juan Corral, is a senior at Spotsylvania High School. Melani is president of Spanish Club, and she is a member of French Club, Future Business Leaders of America and Newspaper Club. Melani has been a member of the varsity girls soccer team and Phoenix travel soccer team, and she has served both as captain, playing defender and midfielder. She is an Ambassador Girl Scout. Melani has received three academic letters, and was recognized for three years with the highest achievement in French. Melani is a volunteer translator and intern at LUCHA Ministries Inc. She also volunteers as coach of a Latinas Soccer team of mothers. Melani is a Taekwondo Black Belt and has volunteered as an instructor assistant. Melani plans to pursue a bachelors degree in pre-law and business, and a minor in Spanish. She would like to attend law school and become an immigration lawyer.

Michaline Johnston, the daughter of Michael and Jacqueline Johnston, is a senior at Stafford High School. Michaline has been a member of National Honor Society, French Honor Society, Math Honor Society, Science Honor Society, French Club and Science Olympiad. She has also been a member of the track and cross country teams. Michaline has also served the JROTC Drill Team as commander. Michaline hopes to attend West Point or Penn State to major in either computer science or physics.

Amy Paz Cuervo, the daughter of Gustavo Paz Cuervo and Maribel Lamas, was selected as Student of the Month of October at Brooke Point High School. Amy is president of Cafe Espanol, vice president of career development in DECA and co-founder of IB Ambassadors. She has also been a member of National Honor Society, Spanish Honor Society, FBLA and Inspire BP. Amy has received academic excellence awards, and the IB Passion awards for physics and Spanish. She was awarded second place in DECA competition for restaurant and food service management. Amy tutors in Spanish, and she helps set up and clean up at school events like IB info night and Flight School. Amy plans to major in aerospace engineering at a four-year university and to pursue a masters degree.

Read more:
Students of the Month: November | Local - Fredericksburg.com

Growing Demand for Eco-friendly Products to Bolster the Growth of the Healthcare Nanotechnology (Nanomedicine) Market during 2015 2021 – Primo…

The comprehensive report published by Persistence Market Research offers an in-depth intelligence related to the various factors that are likely to impact the demand, revenue generation, and sales of the Healthcare Nanotechnology (Nanomedicine) Market. In addition, the report singles out the different parameters that are expected to influence the overall dynamics of the Healthcare Nanotechnology (Nanomedicine) Market during the forecast period 2015 2021.

As per the findings of the presented study, the Healthcare Nanotechnology (Nanomedicine) Market is poised to surpass the value of ~US$ XX by the end of 2029 growing at a CAGR of ~XX% over the assessment period. The report includes a thorough analysis of the upstream raw materials, supply-demand ratio of the Healthcare Nanotechnology (Nanomedicine) in different regions, import-export trends and more to provide readers a fair understanding of the global market scenario.

ThisPress Release will help you to understand the Volume, growth with Impacting Trends. Click HERE To get SAMPLE PDF (Including Full TOC, Table & Figures) athttps://www.persistencemarketresearch.co/samples/6370

The report segregates the Healthcare Nanotechnology (Nanomedicine) Market into different segments to provide a detailed understanding of the various aspects of the market. The competitive analysis of the Healthcare Nanotechnology (Nanomedicine) Market includes valuable insights based on which, market players can formulate impactful growth strategies to enhance their presence in the Healthcare Nanotechnology (Nanomedicine) Market.

Key findings of the report:

The report aims to eliminate the following doubts related to the Healthcare Nanotechnology (Nanomedicine) Market:

Get Access To TOC Covering 200+ Topics athttps://www.persistencemarketresearch.co/toc/6370

Key players in the global nanomedicine market include: Abbott Laboratories, CombiMatrix Corporation, GE Healthcare, Sigma-Tau Pharmaceuticals, Inc., Johnson & Johnson, Mallinckrodt plc, Merck & Company, Inc., Nanosphere, Inc., Pfizer, Inc., Celgene Corporation, Teva Pharmaceutical Industries Ltd., and UCB (Union chimique belge) S.A.

Key geographies evaluated in this report are:

Key features of this report

In order to get a strategic overview of the market,Access Research Methodology Prepared By Experts athttps://www.persistencemarketresearch.co/methodology/6370

Reasons to buy from PMR

About us:

Persistence Market Research (PMR) is a third-platform research firm. Our research model is a unique collaboration of data analytics and market research methodology to help businesses achieve optimal performance.

To support companies in overcoming complex business challenges, we follow a multi-disciplinary approach. At PMR, we unite various data streams from multi-dimensional sources. By deploying real-time data collection, big data, and customer experience analytics, we deliver business intelligence for organizations of all sizes.

Contact us:

Persistence Market Research

305 Broadway, 7th Floor

New York City, NY 10007

United States

Ph.no. +1-646-568-7751

The rest is here:
Growing Demand for Eco-friendly Products to Bolster the Growth of the Healthcare Nanotechnology (Nanomedicine) Market during 2015 2021 - Primo...

Healthcare IT Market trends research and projections – GroundAlerts.com

The consumerization in healthcare information technology has reached a tipping point, the impact of which has been felt across healthcare IT market. The need to ensure comfort and security for patients has brought about a major transformation in the medical sphere, leading to a path-breaking intersection of IT and healthcare. The deployment of IT has equipped the healthcare industry with nanomedicine, virtual healthcare, 3D printing, robot-assisted surgery, and more. These advancements, aided by the investments by the governments worldwide, have brought about a massive change in the healthcare IT industry outlook.

The global healthcare IT market has also gained traction on account of innumerable parameters, prominent among them being, the increasing geriatric populace and the changing consumer lifestyles. Indeed, these have led to a spate of diseases worldwide, consequently surging the demand for a highly sophisticated healthcare IT network in order to lower errors in administration processes and ensure efficient medical data and patient record management.

Request sample copy of this report @ http://decresearch.com/request-sample/detail/232

Consumer expectations have also changed in the last decade or so, leading to healthcare providers focusing on prioritizing efficient management of healthcare data. This has subsequently led to the implementation of innovative technologies in the medical ecosphere, augmenting the revenue graph of the global healthcare IT market. Indeed, estimates claim that healthcare IT industry is expected to exceed $441.8 billion by the year 2025.

While numerous IT solutions are deployed in the healthcare space, one of the most significant ones is that of electronic health records. Undeniably, healthcare IT industry has gained much via rapid adoption of the EHR technology by healthcare specialists in the U.S. and other economies. According to the National Electronic Health Records Survey, 2017, approximately 9 out of 10 office-based physicians had adopted any type of EHR, while certified EHRs were adopted by 4 out of 5 office-based physicians. Since 2008, the rate of EHR adoption has more than doubled from 42 percent to nearly 86 percent in 2017. The accelerated adoption of EHR will thus drive the growth graph of healthcare IT industry from electronic health records. As a matter of fact, estimates claim that EHR-based healthcare IT market size will cross $97.8 billion by 2025.

Request for discount @ http://decresearch.com/request-discount/232

Regionally speaking, it has been forecast that the United States will crop up as a prime growth avenue for the global healthcare IT industry, primarily driven by numerous investments in medical care infrastructure and government mandates. More than half a decade ago, The U.S. Department of Health and Human Services (HHS) had mandated the adoption of information technology by healthcare providers. A substantial growth has also been recorded in terms of investments by the U.S. government in healthcare IT since 2008.

A few years ago, the U.S. government had made an investment of about $20 billion through the HITECH (Health Information Technology for Economic and Clinical Health) Act, for setting up electronic health records. Aided by numerous government initiatives and the escalating need for an efficient healthcare management system, the U.S. healthcare IT industry is estimated to record substantial revenues by 2025.

Speaking of government initiatives, yet another regional ground touted to garner extensive proceeds in healthcare IT market is the United Kingdom. In December 2018, a new collaboration had been announced between the government and the life sciences industry, backed by a government fund worth 79 million, in order to study 5 million people and develop AI-centric diagnostic tests. Back in 2014, the UK government has also announced an investment of $5.4 billion in healthcare IT for a five-year period. It comes as no surprise therefore, that the UK healthcare IT market, powered by government initiatives, will reach $24.7 billion by the year 2025.

Get In-depth table of contents @ http://decresearch.com/toc/detail/healthcare-it-market

With the increasing prevalence of diseases and the subsequently rising demand for a sophisticated medical infrastructure, healthcare IT market contenders have been working to bring forth a slew of advancements in their service portfolio. For instance, McKesson Corporation has recently collaborated with technology leader Navigating Cancer to provide an enhanced Patient Relationship Management (PRM) platform for oncologists.

IT aided healthcare has come a long way since its inception. With massive changes in the technological landscape, many more innovations have been touted to disrupt the healthcare space. Powered by huge investments and the incorporation of advanced technologies in medical care management, healthcare IT market is expected to chart out a lucrative growth map in the forthcoming years.

Table Of Content

Chapter 1. Methodology

1.1. Methodology

1.2. Market definition

1.3. Forecast parameters

1.4. Data sources

1.4.1. Secondary

1.4.1.1. Paid sources

1.4.1.2. Unpaid sources

1.4.2. Primary

Chapter 2. Executive Summary

2.1. Healthcare information technology industry 3600 synopsis, 2014 - 2025

2.1.1. Business trends

2.1.2. Solution trends

2.1.3. End-use trends

2.1.4. Regional trends

Chapter 3. Healthcare Information Technology Industry Insights

3.1. Industry segmentation

3.2. Industry landscape, 2014 - 2025

3.3. Industry impact forces

3.3.1. Growth drivers

3.3.1.1. Growing investment towards healthcare infrastructure development in Asia Pacific region

3.3.1.2. Growing adoption of artificial intelligence

3.3.1.3. High adoption of electronic health records in developed countries such as the U.S.

3.3.1.4. Favorable government initiatives

3.3.1.5. Increasing demand for cost-saving in healthcare delivery

3.3.2. Industry pitfalls & challenges

3.3.2.1. High cost associated with implementation and maintenance

3.3.2.2. Security and privacy concerns

3.4. Growth potential analysis

3.4.1. By solution

3.4.2. By end-use

3.5. Regulatory landscape

3.5.1. U.S.

3.5.2. Europe

3.5.3. China

3.6. Technology landscape

3.7. Porter's analysis

3.8. Competitive landscape, 2017

3.8.1. Strategy dashboard

3.9. PESTEL analysis

Read the original:
Healthcare IT Market trends research and projections - GroundAlerts.com

New Class of Drugs Harnesses Gold Nanocrystals to Heal and Protect the Brain – BioSpace

Clene Chief Executive Officer Rob Etherington. Photo courtesy of Clene.

Clene Nanomedicine is trying to set a new gold standard in neurodegenerative diseases through the development of a new class of drugs called bioenergetic nanotherapeutics that harnesses the properties of gold nanocrystals.

The gold nanocrystals are used to amplify bioenergetic reactions in patients in order to drive intracellular biological reactions. Bioenergetic nanotherapeutics are a clean break from pharmaceutical drug development that uses classical synthetic chemistry, Clene Chief Executive Officer Rob Etherington told BioSpace in an interview. Clenes lead asset is CNM-Au8, a bioenergetic nanocatalyst under development as an add-on treatment for neurodegenerative diseases like Parkinsons disease, multiple sclerosis and Amyotrophic Lateral Sclerosis (ALS). CNM-Au8 is designed to catalyze bio cellular reactions, and so far the company has seen the asset live up to its promise in clinical studies. The companys gold nanocrystals are grown in water and patients drink the asset down. Research has so far indicated that Clene and its golden asset could become a pioneer in therapeutic neurorepair and neuroprotection.

To date, CNM-Au8 has demonstrated safety in Phase I studies, remyelination and neuroprotective effects in preclinical models and is currently being assessed in a Phase II study for the treatment of chronic optic neuropathy in patients with multiple sclerosis. Additionally, CNM-Au8 is being studies in Phase II and Phase III studies for disease progression in patients with ALS. In September, Clene presented interim results from the REPAIR-MS and REPAIR-PD Phase II studies demonstrating the effects of its lead nanocatalytic therapeutic, CNM-Au8. The preliminary data demonstrate CNM-Au8-mediated modulation of key brain bioenergetic metabolites in relapsing multiple sclerosis (MS) and Parkinson's disease (PD) patients. Data from the studies indicate catalytic bioenergetic improvements across important CNS bioenergetic metabolites, including total nicotinamide adenine dinucleotide (NAD) levels, NAD+/NADH ratio, and adenosine triphosphate (ATP) levels, indicating a homeostatic effect of CNM-Au8 on brain bioenergetics, the company said.

Etherington said the data from the REPAIR-MS and REPAIR-PD studies indicate that CNM-Au8 is working mechanistically to address a foundational challenge common to many neurodegenerative diseases, which is that stressed or failing neurons need additional energy to survive and repair.

We now have insights that CNM-Au8 is driving bioenergetics within the brain, which is a foundational insight for the further development of Clene's neurorepair clinical programs, Etherington said. He added that should the data from the interim analysis pan out, it indicated that CNM-Au8 could effectively benefit millions of people across the globe suffering from multiple sclerosis and other neurodegenerative diseases.

There are multiple drugs already on the market for these neurodegenerative diseases. CNM-Au8 is not meant to replace those drugs, but to work alongside them. Etherington explained that CNM-Au8 is not designed to target a specific protein, nor it is designed to block or antagonize something, like most drugs. Rather, Clenes compound is designed to enhance the intracellular biological actions necessary to repair and reverse neuronal damage, Etherington said.

We are purposely seeking to reverse neurodegernation. We want to let the cell take care of its own housekeeping and enhance whats naturally occurring in the central nervous system, he said.

Etherington acknowledged the concept of drinking bits of gold nanocrystals may sound like something out of a Star Trek episode, but insisted the idea is sound. Gold-salt injections were historically used to treat rheumatoid arthritis decades ago, but were dropped due to health concerns. Clene had the idea to build a stable, oral nanotherapeutic, so they could see less toxicity and drive bioenergetics targets for a suite of neurodegenerative diseases, he said.

Its so out of the box that it can be a bit mind boggling. Were breaking with the traditional path and shifting the paradigm to how we think neurodegenerative disease should be treated, he said.

Not only is Clene moving forward in its clinical assessment of CNM-Au8, the company is planning to go public with a special purpose acquisition companies (SPAC) merger before the end of 2020. 2020 has been the busiest year for this kind of stock entry, with a 250% surge. As BioSpace recently reported, there have been nearly two dozen SPAC mergers in the biotech sector this year, targeting more than $3.5 billion in proceeds. When the company goes public, Etherington said Clenes management team will remain the same and the funding raised from this reverse stock merger will provide the finances that can support the companys ongoing research.

Featured Jobs on BioSpace

Excerpt from:
New Class of Drugs Harnesses Gold Nanocrystals to Heal and Protect the Brain - BioSpace

Nanomedicine Market Forecast to 2026 : How it is Going to Impact on Global Industry to Grow in Near Future – Gazette Quest

Global Nanomedicine Market is forecast to bring about a fairly desirable remuneration portfolio by the end of the forecast period 2019-2025. Certainly, the report not only includes a modest growth rate over the forecast time frame but also contains a reliable overview of this business. The study involves overall growth opportunities and valuation currently this market is holding. Additionally, the report involves classified segmentation of Nanomedicine market.

The global Nanomedicine Market report comprises a thorough outline and upcoming view. Get sample copy of Nanomedicine Market Report at https://www.stratagemmarketinsights.com/sample/9943

Some of key competitors or manufacturers included in the study are:

Regional analysis covers:

Market Scenario:

The report further highlights the development trends in the global Nanomedicine market. Factors that are driving the market growth and fueling its segments are also analyzed in the report. The report also highlights on its applications, types, deployments, components, developments of this market.

Additionally, the report quotes worldwide certainties and countenance of Nanomedicine industry along with downstream and upstream analysis of leading players. Numerous research findings and conclusions stated in the report will help decision-makers to take imperative decisions in the near future.

Ask more details or request a custom report sample to our experts at https://www.stratagemmarketinsights.com/quiry/9943

The Nanomedicine Market report is a compilation of first-hand information, qualitative and quantitative assessment by industry analysts, inputs from industry experts and industry participants across the value chain. The report provides an in-depth analysis of parent market trends, macro-economic indicators, and governing factors along with market attractiveness as per segments. The report also maps the qualitative impact of various market factors on market segments and geographies.

Nanomedicine Market report Segmented By Product Type:

Nanomedicine Market report Applications:

Chapters involved in Nanomedicine market report:

Chapter 1: Market Overview, Drivers, Restraints and Opportunities, Segmentation overviewChapter 2: Market Competition by ManufacturersChapter 3: Production by RegionsChapter 4: Consumption by RegionsChapter 5: Production, By Types, Revenue and Market share by TypesChapter 6: Consumption, By Applications, Market share (%) and Growth Rate by ApplicationsChapter 7: Complete profiling and analysis of ManufacturersChapter 8: Manufacturing cost analysis, Raw materials analysis, Region-wise manufacturing expensesChapter 9: Industrial Chain, Sourcing Strategy and Downstream BuyersChapter 10: Marketing Strategy Analysis, Distributors/TradersChapter 11: Market Effect Factors AnalysisChapter 12: Market ForecastChapter 13: Nanomedicine Research Findings and Conclusion, Appendix, methodology and data source

Hurry Up limited period offer to get discount on the report: https://www.stratagemmarketinsights.com/cart/9943

About Us:

Stratagem Market Insights is a management consulting organization providing market intelligence and consulting services worldwide. We bring the expertise of consultants with an cumulative industry experience of more than 70 years. The firm has been providing quantified B2B research and currently offers services to over 350+ customers worldwide. Our reports cover various end-use industries such as Aerospace and Defense, Agriculture, Food and Beverages, Automotive, Chemicals and Materials, Consumer Goods and Retail, Electronics, Energy, Mining, and Utilities, Pharmaceuticals, Manufacturing and Construction, Services, and Healthcare, and ICT.

Get in touch @ https://healthcareinsights786.blogspot.com/

Read more:
Nanomedicine Market Forecast to 2026 : How it is Going to Impact on Global Industry to Grow in Near Future - Gazette Quest

NanoViricides to Participate in Panel Discussion at the B. Riley FBR Virtual Infectious Disease Summit Today, July 21, 2020 – BioSpace

SHELTON, CT / ACCESSWIRE / July 21, 2020 / NanoViricides, Inc. (NYSE American:NNVC) (the "Company") a global leader in the development of highly effective antiviral therapies based on a novel nanomedicines platform, today announced that Anil R. Diwan, PhD, President and Executive Chairman of the Company, will participate in the "B. Riley FBR Virtual Infectious Disease Summit - Therapeutics Day" on Tuesday, July 21, 2020. The Conference is organized by B. Riley FBR, Inc. (https://brileyfbr.com/).

Dr. Diwan is invited to participate in Panel #3 at 2020 at 2:10 p.m. ET, entitled "Taming the Severe Disease Presentations". He will briefly discuss the Company's novel nanomedicines platform and the Company's progress in the lead IND-ready candidate for the treatment of shingles rash, NV-HHV-101, as well as in developing a drug candidate against SARS-CoV-2, the cause of COVID-19 global pandemic.

The Company believes that it is close to selecting a clinical candidate worthy of advancing into human clinical trials for the treatment of SARS-CoV-2 infection, based on (i) cell culture effectiveness studies against multiple circulating coronaviruses that employ different cell surface receptors, (ii) a lethal lung infection animal model effectiveness study using hCoV-NL63 infection (a coronavirus that uses the same receptor, ACE2, as SARS-CoV-2, and produced similar disease in the animal model), and (iii) preliminary safety studies in animal model at maximum feasible dosage levels. The Company has disclosed its findings from these studies in previous press releases.

Prior to filing for human clinical trials, NanoViricides plans on conducting studies, towards clinical candidate selection, to further determine the effectiveness against SARS-CoV-2, perform additional drug development studies as may be necessary, and request a pre-IND Meeting with the US FDA for regulatory guidance.

The Company is also working with its regulatory consultants on completing an IND with the US FDA to advance its lead drug candidate NV-HHV-101 into human clinical trials for topical dermal treatment of Shingles rash as the initial indication. In particular, the Company is working on finalizing the clinical trials plan for the anticipated human clinical trials for shingles rash treatment. The Company is also in the process of finalizing clinical trial sites. This process has been adversely affected by the current global COVID-19 pandemic, and in particular, its effects across the USA.

Importantly, nanoviricides are designed to act by a novel mechanism of action, trapping the virus particle like the "Venus-fly-trap" flower does for insects. Antibodies, in contrast, only label the virus for other components of the immune system to take care of. It is well known that the immune system is not functioning properly at least in severe COVID-19 patients.

Additionally, it is well known that viruses escape antibody-drugs via mutations. The Company's "nanoviricide" drug candidates, in contrast, are designed to be broad-spectrum, and therefore virus escape by mutations is expected to be unlikely.

The market size for the treatment of shingles is estimated at approximately one billion dollars by various estimates. These estimates take into account the Shingrix vaccine as well as existing vaccines. About 500,000 to 1 million cases of shingles occur in the USA alone every year.

The market size for our immediate target drugs in the HerpeCide program is variously estimated at billions to tens of billions of dollars. The Company believes that its dermal topical cream for the treatment of shingles rash will be its first drug heading into clinical trials. The Company believes that additional topical treatment candidates in the HerpeCide program, namely, HSV-1 "cold sores" treatment, and HSV-2 "genital ulcers" treatment are expected to follow the shingles candidate into IND-enabling development and then into human clinical trials. These additional candidates are based on NV-HHV-101, thereby maximizing return on investments and shareholder value.

The Company develops its class of drugs, that we call nanoviricides, using a platform technology. This approach enables rapid development of new drugs against a number of different viruses. A nanoviricide is a "biomimetic" - it is designed to "look like" the cell surface to the virus. The nanoviricide technology enables direct attacks at multiple points on a virus particle. It is believed that such attacks would lead to the virus particle becoming ineffective at infecting cells. Antibodies in contrast attack a virus particle at only a maximum of two attachment points per antibody.

In addition, the nanoviricide technology also simultaneously enables attacking the rapid intracellular reproduction of the virus by incorporating one or more active pharmaceutical ingredients (APIs) within the core of the nanoviricide. The nanoviricide technology is the only technology in the world, to the best of our knowledge, that is capable of simultaneously (a) attacking extracellular virus to break the reinfection cycle, and (b) disrupting intracellular production of the virus, thus enabling complete control of a virus infection.

About NanoViricidesNanoViricides, Inc. (www.nanoviricides.com) is a development stage company that is creating special purpose nanomaterials for antiviral therapy. The Company's novel nanoviricide class of drug candidates are designed to specifically attack enveloped virus particles and to dismantle them. Our lead drug candidate is NV-HHV-101 with its first indication as dermal topical cream for the treatment of shingles rash. The Company is in the process of completing an IND application to the US FDA for this drug candidate. The Company cannot project an exact date for filing an IND because of its dependence on a number of external collaborators and consultants, and the effects of recent COVID-19 restrictions.

The Company is also developing drugs against a number of viral diseases including oral and genital Herpes, viral diseases of the eye including EKC and herpes keratitis, H1N1 swine flu, H5N1 bird flu, seasonal Influenza, HIV, Hepatitis C, Rabies, Dengue fever, and Ebola virus, among others. NanoViricides' platform technology and programs are based on the TheraCour nanomedicine technology of TheraCour, which TheraCour licenses from AllExcel. NanoViricides holds a worldwide exclusive perpetual license to this technology for several drugs with specific targeting mechanisms in perpetuity for the treatment of the following human viral diseases: Human Immunodeficiency Virus (HIV/AIDS), Hepatitis B Virus (HBV), Hepatitis C Virus (HCV), Rabies, Herpes Simplex Virus (HSV-1 and HSV-2), Varicella-Zoster Virus (VZV), Influenza and Asian Bird Flu Virus, Dengue viruses, Japanese Encephalitis virus, West Nile Virus and Ebola/Marburg viruses. The Company has executed a Memorandum of Understanding with TheraCour that provides a limited license for research and development for drugs against human coronaviruses. The Company intends to obtain a full license and has begun the process for the same. The Company's technology is based on broad, exclusive, sub-licensable, field licenses to drugs developed in these areas from TheraCour Pharma, Inc. The Company's business model is based on licensing technology from TheraCour Pharma Inc. for specific application verticals of specific viruses, as established at its foundation in 2005.

This press release contains forward-looking statements that reflect the Company's current expectation regarding future events. Actual events could differ materially and substantially from those projected herein and depend on a number of factors. Certain statements in this release, and other written or oral statements made by NanoViricides, Inc. are "forward-looking statements" within the meaning of Section 27A of the Securities Act of 1933 and Section 21E of the Securities Exchange Act of 1934. You should not place undue reliance on forward-looking statements since they involve known and unknown risks, uncertainties and other factors which are, in some cases, beyond the Company's control and which could, and likely will, materially affect actual results, levels of activity, performance or achievements. The Company assumes no obligation to publicly update or revise these forward-looking statements for any reason, or to update the reasons actual results could differ materially from those anticipated in these forward-looking statements, even if new information becomes available in the future. Important factors that could cause actual results to differ materially from the company's expectations include, but are not limited to, those factors that are disclosed under the heading "Risk Factors" and elsewhere in documents filed by the company from time to time with the United States Securities and Exchange Commission and other regulatory authorities. Although it is not possible to predict or identify all such factors, they may include the following: demonstration and proof of principle in preclinical trials that a nanoviricide is safe and effective; successful development of our product candidates; our ability to seek and obtain regulatory approvals, including with respect to the indications we are seeking; the successful commercialization of our product candidates; and market acceptance of our products. FDA refers to US Food and Drug Administration. IND application refers to "Investigational New Drug" application. CMC refers to "Chemistry, Manufacture, and Controls". ]

Contact:NanoViricides, Inc.info@nanoviricides.com

Public Relations Contact:MJ ClyburnTraDigital IRclyburn@tradigitalir.com

SOURCE: NanoViricides, Inc.

View source version on accesswire.com:https://www.accesswire.com/598261/NanoViricides-to-Participate-in-Panel-Discussion-at-the-B-Riley-FBR-Virtual-Infectious-Disease-Summit-Today-July-21-2020

See the original post:
NanoViricides to Participate in Panel Discussion at the B. Riley FBR Virtual Infectious Disease Summit Today, July 21, 2020 - BioSpace

Aviceda Therapeutics Announces Formation of Scientific Advisory Board – BioSpace

Oct. 27, 2020 12:00 UTC

CAMBRIDGE, Mass.--(BUSINESS WIRE)-- Aviceda Therapeutics, a late-stage, pre-clinical biotech company focused on developing the next generation of immuno-modulators by harnessing the power of glycobiology to manipulate the innate immune system and chronic, non-resolving inflammation, is announcing the members of its Scientific Advisory Board who will help shape ongoing development efforts.

The Aviceda Scientific Advisory Board includes Pamela Stanley, PhD; Ajit Varki, MD; Christopher Scott, PhD; Geert-Jan Boons, PhD; Salem Chouaib, PhD; and Peng Wu, PhD.

Aviceda has assembled an extraordinary multi-disciplinary team of world-class scientists and renowned researchers to join our efforts in developing the next generation of glyco-immune therapeutics for the treatment of immune-dysfunction conditions, said Mohamed A. Genead, MD, Founder, CEO & President of Aviceda Therapeutics. Each individual offers a fresh perspective and unique strategic acumen that complements and strengthens the insights of our in-house leadership development team.

Prof. Scott, Aviceda Scientific Co-Founder, is Director of the Patrick G Johnston Centre for Cancer Research and Cell Biology at Queens University Belfast. He is internationally renowned for his work in development of novel approaches in the field of antibody and nanomedicine-based therapies for the treatment of cancer and other conditions. Prof. Scott has a background in both the pharmaceutical industry and academia and was a founding scientist of Fusion Antibodies Plc. Research in his laboratory is funded by agencies such as Medical Research Council, UK charities and various industrial sources. He also held a Royal Society Industrial Fellowship with GSK from 2012 to 2015 and won the Vice Chancellors Prize for Innovation in 2015 with his groups work on developing a novel Siglec targeting nanomedicine for the treatment of sepsis and other inflammatory conditions.

The novelty of Avicedas platform technology is its potential to affect immune responses associated with a wide range of disease states, many of which are currently unmet or underserved needs. I look forward to the continued development of Avicedas core technology and moving forward to clinical trials that will pave the way for truly disruptive therapeutic strategies to enter the clinic that will significantly impact and improve patients lives in the not-too-distant future, said Prof. Scott.

Avicedas Scientific advisory chairwoman, Prof. Stanley, is the Horace W. Goldsmith Foundation Chair; Professor, Department of Cell Biology; and Associate Director for Laboratory Research of the Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York. She obtained a doctorate degree from the University of Melbourne, Australia, for studies of influenza virus, and was subsequently a postdoctoral fellow of the Medical Research Council of Canada in the laboratory of Louis Siminovitch, University of Toronto, where she studied somatic cell genetics. Prof. Stanleys laboratory is focused on identifying roles for mammalian glycans in development, cancer and Notch signaling. Among her many varied contributions, Prof. Stanleys laboratory has isolated a large panel of Chinese hamster ovary (CHO) glycosylation mutants; characterized them at the biochemical, structural and genetic levels; and used them to identify new aspects of glycan synthesis and functions. She serves on the editorial boards of Scientific Reports, Glycobiology and FASEB Bio Advances; she is an editor of the textbook Essentials of Glycobiology; and her laboratory is the recipient of grants from the National Institutes of Health. Prof. Stanley has received numerous awards, including a MERIT award from the National Institutes of Health, an American Cancer Society Faculty Research Award, the Karl Meyer Award from the Society for Glycobiology (2003) and the International Glycoconjugate Organization (IGO) Award (2003).

Working with Aviceda represents a unique opportunity to contribute to science at the cutting edge. Its pipeline contains a broad range of candidates that represents numerous first-in-class opportunities, said Prof. Stanley.

Prof. Varki is currently a distinguished professor of medicine and cellular and molecular medicine, Co-director of the Glycobiology Research and Training Center and Executive Co-director for the UCSD/Salk Center for Academic Research and Training in Anthropogeny at the University of California, San Diego; and an Adjunct Professor at the Salk Institute for Biological Studies. Dr. Varki is also the executive editor of the textbook Essentials of Glycobiology. He received basic training in physiology, medicine, biology and biochemistry at the Christian Medical College, Vellore, The University of Nebraska, and Washington University in St. Louis, as well as formal training and certification in internal medicine, hematology and oncology. Dr. Varki is the recipient of numerous awards and recognitions, including election to the American Academy of Arts and Sciences and the US National Academy of Medicine, a MERIT award from the National Institutes of Health, an American Cancer Society Faculty Research Award, the Karl Meyer Award from the Society for Glycobiology and the International Glycoconjugate Organization (IGO) Award (2007).

The Aviceda team is already building on the foundational work in the emerging field of glycobiology to develop potential therapeutics and interventional strategies. Their work could be critically important for growing the understanding of how glycobiology and glycochemistry are applicable to immunology, and more broadly, to the field of drug and therapeutic development, said Prof. Varki.

Prof. Boons is a Distinguished Professor in Biochemical Sciences at the Department of Chemistry and the Complex Carbohydrate Research Center (CCRC) of the University of Georgia (USA) and Professor and Chair of the Department of Medicinal and Biological Chemistry of Utrecht University (The Netherlands). Prof. Boons directs a research program focused on the synthesis and biological functions of carbohydrates and glycoconjugates. The diversity of topics to which his group has significantly contributed includes the development of new and better methods for synthesizing exceptionally complex carbohydrates and glycoconjugates. Highlights of his research include contributions to the understanding of immunological properties of complex oligosaccharides and glycoconjugates at the molecular level, which is being used in the development of three-component vaccine candidates for many types of epithelial cancer; development of convergent strategies for complex oligosaccharide assembly, which make it possible to synthesize large collections of compounds with a minimal effort for structure activity relationship studies; and creation of a next generation glycan microarray that can probe the importance of glycan complexity for biological recognition, which in turn led to identification of glycan ligands for various glycan binding proteins that are being further developed as glycomimetics for drug development for various diseases. Among others, Prof. Boons has received the Creativity in Carbohydrate Science Award by the European Carbohydrate Association (2003), the Horace Isbell Award by the American Chemical Society (ACS) (2004), the Roy L. Whistler International Award in Carbohydrate

Chemistry by the International Carbohydrate Organization (2014), the Hudson Award (2015) and the Cope Mid-Career Scholar Award from ACS (2016).

Aviceda is leading the field of glycoimmunology in exciting new directions. I look forward to working with the company as it pursues multiple lines of development efforts that will someday transform the way immune-inflammatory conditions are treated in the clinic, said Prof. Boons.

Prof. Chouaib is the Director of Research, Institute Gustave Roussy, Paris, where he is active in research in tumor biology. Previously, Prof. Chouaib worked at the French National Institute of Health and Biomedical Research (INSERM) where he led a research unit focused on the investigation of the functional cross talk between cytotoxic cells and tumor targets in the context of tumor microenvironment complexity and plasticity. His research was directed at the transfer of fundamental concepts in clinical application in the field of cancer vaccines and cancer immunotherapy. Prof. Chouaib is a member of the American Association of Immunologists, New York Academy of Sciences, French Society of Immunologists, International Cytokine Society, American Association for Cancer Research, International Society for Biological Therapy of Cancer and American Association of Biological Chemistry. He was awarded the cancer research prize of the French ligue against cancer in 1992 and in 2004 the presidential prize in biotechnology. He was awarded for translational research and scientific excellency by INSERM. His research has resulted in more than 310 scientific articles and several reviews in the field of human immunology, tumor biology and cancer immunotherapy; he has also been an editor for several textbooks.

Dr. Wu is an Associate Professor in the Department of Molecular Medicine at Scripps Research. The current research in the Wu laboratory integrates synthetic chemistry with glycobiology to explore the relevance of protein glycosylation in human disease and cancer immunotherapy. In 2018, Dr. Wu developed a platform to construct antibody-cell conjugates for cancer immunotherapy, which does not require genetic engineering. Previously, while working as a postdoctoral fellow in the group of Professor Carolyn R. Bertozzi at the University of California, Berkeley, Dr. Wu developed an aldehyde-tag (SMARTag) based technology for site-specific labeling of monoclonal antibodies, which served as the foundation for Redwood Biosciences Inc., a biotech company co-founded by Bertozzi. In 2014, Redwood Bioscience Inc. and the SMARTag Antibody-Drug Conjugate technology platform was acquired by Catalent Pharma Solutions.

About Aviceda Therapeutics

Founded in 2018 and based in Cambridge, Massachusetts, Aviceda Therapeutics is a late-stage, pre-clinical biotechnology company with a mission to develop the next generation of glyco-immune therapeutics (GITs) utilizing a proprietary technology platform to modulate the innate immune system and chronic, non-resolving inflammation. Aviceda has assembled a world-class, cross-disciplinary team of recognized scientists, clinicians and drug developers to tackle devastating ocular and systemic degenerative, fibrotic, oncologic and immuno-inflammatory diseases. At Aviceda, we exploit a unique family of receptors found expressed on all innate immune cells and their associated glycobiological interactions to develop transformative medicines. Combining the power of our biology with our innovative cell-based high-throughput screening platform and proprietary nanoparticle technology, we can modulate the innate immune response specifically and profoundly. Aviceda is developing a pipeline of GITs that are delivered via biodegradable nanoparticles and which safely and effectively target numerous immune-inflammatory conditions. Avicedas lead ophthalmic optimized nanoparticle, as an intravitreal formulation, AVD-104, is being developed to target various immune system responses that contribute to pathology associated with age-related macular degeneration (AMD).

View source version on businesswire.com: https://www.businesswire.com/news/home/20201027005101/en/

Read the original:
Aviceda Therapeutics Announces Formation of Scientific Advisory Board - BioSpace

Nanomedicine Market Growth Is Boosted By Increasing Healthcare Applications And Medical Device Sales – News Succeed

A report on the Nanomedicine Market added by Reports and Data, features the current and future growth trends of this industry in addition to significant details regarding the geographies that comprise the regional spread of the Nanomedicine market. Also, the report simplifies complex information about the supply-demand analysis, market share, growth statistics, and participation of prominent players in the Nanomedicine market.

An extensive analysis of the Nanomedicine market has been provided through this research report, which also includes a detailed evaluation of the business space. Moreover, the Nanomedicine market segmentation given in this report covers the market extensively, in addition to a general overview of this Nanomedicine market in the context of its present scenario.

Request a Sample Report of the Nanomedicine Market @ https://www.reportsanddata.com/sample-enquiry-form/1048

The influential players of the Nanomedicine market that are included in the report are:

Arrowhead Pharmaceuticals, Inc., Nanospectra Biosciences, Inc., AMAG Pharmaceuticals, Bio-Gate AG, Celgene and Johnson & Johnson

Market segmentation of the Nanomedicine market by Application (Value and Volume from 2020 to 2027):

Oncology

Infectious diseases

Cardiology

Orthopedics

Others

Market segmentation of the Nanomedicine market by Type (Value and Volume from 2020 to 2027):

Therapeutics

Regenerative Medicine

In-vitro diagnostics

In-vivo diagnostic

Vaccines

The study assesses the important and crucial data concerning the geographical extent of this market and as well as the companies that have successfully established their positions in the Nanomedicine market.

How far does the scope of the Nanomedicine market traverse?

Get a Discount on the Nanomedicine Market Report @ https://www.reportsanddata.com/discount-enquiry-form/1048

An overview of the competitive landscape:

A detailed outline of the regional spread:

A summary of the segmentation:

Purchase this Report @ 4590$, Visit:https://www.reportsanddata.com/checkout-form/1048

Some of the Major Highlights of TOC covers:

Chapter 1: Methodology & Scope

Chapter 2: Executive Summary

Chapter 3: Nanomedicine Industry Insights

Chapter 4: Nanomedicine Market, By Region

Chapter 5: Company Profiles

Read Full Report details @ https://www.reportsanddata.com/report-detail/nanomedicine-market

Thank you for reading this article. You can also get chapter-wise sections or region-wise report coverage for North America, Europe, Asia Pacific, Latin America, and Middle East & Africa.

Larry Norris is a journalism graduate with keen interest in covering news specifically top trending. He has as a keen eye for technologies and has predicted quite a few successful startups over the last couple of years. Larry goal with this website is to report accurately on all kinds of stock news, and have a great deal of passion for technical and active reporting. Larry is diligent and proactive when it comes to news reporting.

Email: [emailprotected]

Address: 1083 Republic Avenue East, Monroe, IA 50170, USA

Contact number: 641-741-7672

See the article here:
Nanomedicine Market Growth Is Boosted By Increasing Healthcare Applications And Medical Device Sales - News Succeed

Nanomedicine Market Size Analysis, Top Manufacturers, Shares, Growth Opportunities and Forecast to 2026 – Science In Me

New Jersey, United States: Market Research Intellect has added a new research report titled, Nanomedicine Market Professional Survey Report 2020 to its vast collection of research reports. The Nanomedicine market is expected to grow positively for the next five years 2020-2026.

The Nanomedicine market report studies past factors that helped the market to grow as well as, the ones hampering the market potential. This report also presents facts on historical data from 2011 to 2019 and forecasts until 2026, which makes it a valuable source of information for all the individuals and industries around the world. This report gives relevant market information in readily accessible documents with clearly presented graphs and statistics. This report also includes views of various industry executives, analysts, consultants, and marketing, sales, and product managers.

Market Segment as follows:

The global Nanomedicine Market report highly focuses on key industry players to identify the potential growth opportunities, along with the increased marketing activities is projected to accelerate market growth throughout the forecast period. Additionally, the market is expected to grow immensely throughout the forecast period owing to some primary factors fuelling the growth of this global market. Finally, the report provides detailed profile and data information analysis of leading Nanomedicine company.

Nanomedicine Market by Regional Segments:

The chapter on regional segmentation describes the regional aspects of the Nanomedicine market. This chapter explains the regulatory framework that is expected to affect the entire market. It illuminates the political scenario of the market and anticipates its impact on the market for Nanomedicine .

The Nanomedicine Market research presents a study by combining primary as well as secondary research. The report gives insights on the key factors concerned with generating and limiting Nanomedicine market growth. Additionally, the report also studies competitive developments, such as mergers and acquisitions, new partnerships, new contracts, and new product developments in the global Nanomedicine market. The past trends and future prospects included in this report makes it highly comprehensible for the analysis of the market. Moreover, The latest trends, product portfolio, demographics, geographical segmentation, and regulatory framework of the Nanomedicine market have also been included in the study.

Ask For Discount (Special Offer: Get 25% discount on this report) @ https://www.marketresearchintellect.com/ask-for-discount/?rid=201321&utm_source=SI&utm_medium=888

Table of Content

1 Introduction of Nanomedicine Market1.1 Overview of the Market1.2 Scope of Report1.3 Assumptions

2 Executive Summary

3 Research Methodology3.1 Data Mining3.2 Validation3.3 Primary Interviews3.4 List of Data Sources

4 Nanomedicine Market Outlook4.1 Overview4.2 Market Dynamics4.2.1 Drivers4.2.2 Restraints4.2.3 Opportunities4.3 Porters Five Force Model4.4 Value Chain Analysis

5 Nanomedicine Market, By Deployment Model5.1 Overview

6 Nanomedicine Market, By Solution6.1 Overview

7 Nanomedicine Market, By Vertical7.1 Overview

8 Nanomedicine Market, By Geography8.1 Overview8.2 North America8.2.1 U.S.8.2.2 Canada8.2.3 Mexico8.3 Europe8.3.1 Germany8.3.2 U.K.8.3.3 France8.3.4 Rest of Europe8.4 Asia Pacific8.4.1 China8.4.2 Japan8.4.3 India8.4.4 Rest of Asia Pacific8.5 Rest of the World8.5.1 Latin America8.5.2 Middle East

9 Nanomedicine Market Competitive Landscape9.1 Overview9.2 Company Market Ranking9.3 Key Development Strategies

10 Company Profiles10.1.1 Overview10.1.2 Financial Performance10.1.3 Product Outlook10.1.4 Key Developments

11 Appendix11.1 Related Research

Complete Report is Available @ https://www.marketresearchintellect.com/product/nanomedicine-market-size-and-forecast/?utm_source=SI&utm_medium=888

We also offer customization on reports based on specific client requirement:

1-Freecountry level analysis forany 5 countriesof your choice.

2-FreeCompetitive analysis of any market players.

3-Free 40 analyst hoursto cover any other data points

About Us:

Market Research Intellect provides syndicated and customized research reports to clients from various industries and organizations with the aim of delivering functional expertise. We provide reports for all industries including Energy, Technology, Manufacturing and Construction, Chemicals and Materials, Food and Beverage and more. These reports deliver an in-depth study of the market with industry analysis, market value for regions and countries and trends that are pertinent to the industry.

Contact Us:

Mr. Steven FernandesMarket Research IntellectNew Jersey ( USA )Tel: +1-650-781-4080

Email: [emailprotected]

Get Our Trending Report

https://www.marketresearchblogs.com/

https://www.marktforschungsblogs.com/

Tags: Nanomedicine Market Size, Nanomedicine Market Growth, Nanomedicine Market Forecast, Nanomedicine Market Analysis, Nanomedicine Market Trends, Nanomedicine Market

Read this article:
Nanomedicine Market Size Analysis, Top Manufacturers, Shares, Growth Opportunities and Forecast to 2026 - Science In Me

Global Nanomedicine Market Executive Summary and Analysis by Top Players 2020 – 2025 : GE Healthcare, Johnson & Johnson – Stock Market Herald

A market study dependent on the Nanomedicine Market over the globe, as of late added to the storehouse of Market Research, is titled Worldwide Nanomedicine Market 2019. The exploration report examinations the chronicled just as present execution of the overall Nanomedicine industry and makes expectations on the future status of Nanomedicine advertise based on this investigation.

Get Free Sample Copy of Report Here:https://www.marketresearchstore.com/report/global-nanomedicine-market-status-trend-report-2018-2023-260464#RequestSample

Top Companies Include (from a broad pool of working players over the globe):GE Healthcare, Johnson & Johnson, Mallinckrodt plc, Merck & Co. Inc., Nanosphere Inc., Pfizer Inc., Sigma-Tau Pharmaceuticals Inc., Smith & Nephew PLC, Stryker Corp, Teva Pharmaceutical Industries Ltd., UCB (Union chimique belge) S.A

The report reads the business for Nanomedicine over the globe taking the current business chain, the import and fare measurements in Nanomedicine advertise and elements of interest and supply of Nanomedicine into thought. The Nanomedicine examine study covers every single part of the Nanomedicine showcase comprehensively, which begins from the meaning of the Nanomedicine business and creates towards Nanomedicine advertise divisions. Further, every fragment of the Nanomedicine advertise is grouped and broke down based on item types, applications, and the end-use businesses of the Nanomedicine showcase. The land division of the Nanomedicine business has likewise been canvassed finally in this report.

Market Size Segmentation by Type (Customizable):Regenerative Medicine, In-vitro & In-vivo Diagnostics, Vaccines, Drug Delivery

Market Size Segmentation by Application (Customizable):Clinical Cardiology, Urology, Genetics, Orthopedics, Ophthalmology

The focused scene of the overall market for Nanomedicine is controlled by assessing the different business members, creation limit, Nanomedicine markets creation chain, and the income produced by every producer in the Nanomedicine advertise around the world.

Enquire Here:https://www.marketresearchstore.com/report/global-nanomedicine-market-status-trend-report-2018-2023-260464#InquiryForBuying

The worldwide Nanomedicine showcase 2020is additionally examined based on item evaluating, Nanomedicine creation volume, information with respect to request and Nanomedicine supply, and the income accumulated by the item. Different precise instruments, for example, speculation returns, plausibility, and market engaging quality investigation has been utilized in the exploration to introduce a far-reaching investigation of the business for Nanomedicine over the globe.

About Us:

MarketResearchStore.comis a single destination for all the industry, company and country reports. We feature large repository of latest industry reports, leading and niche company profiles, and market statistics released by reputed private publishers and public organizations.

Contact US:

Joel JohnSuite #8138, 3422 SW 15 Street,Deerfield Beach, Florida 33442United StatesToll Free: +1-855-465-4651 (USA-CANADA)Tel: +1-386-310-3803Web:http://www.marketresearchstore.comEmail:sales@marketresearchstore.com

The rest is here:
Global Nanomedicine Market Executive Summary and Analysis by Top Players 2020 - 2025 : GE Healthcare, Johnson & Johnson - Stock Market Herald

Nanoparticle interactions with immune cells dominate tumor retention and induce T cellmediated tumor suppression in models of breast cancer – Science…

INTRODUCTION

Nanoparticles provide unique opportunities and challenges for cancer therapy and diagnosis. They have the potential to interact with the immune system and solid tumor microenvironment (TME) in unexpected ways to ultimately and critically affect performance and tumor response (13). The premise that nanoscale materials can be engineered to selectively detect and destroy cancer cells in solid tumors is undergoing a critical reevaluation (411). Yet, relatively little analysis of nanoparticle fate and intratumor accumulation across biological models and immune cell or tumor compartments has been completed, particularly with histology or flow cytometry (6).

As with many cancer drug development scenarios, nanotechnology-based formulations are often tested and optimized using a specific mouse model of human cancer. These xenograft tumor studies rely on immunodeficient animal models, which provide a permissive environment for cross-species tissue grafting. Therefore, how well these models predict the potential and mechanisms for nano-targeting becomes a relevant question when the construct itself demonstrates strong interactions with the recipients immune system (13, 6).

Polysaccharide (starch or dextran)coated iron oxide nanoparticles have been used for decades in biomedicine as agents for parenteral anemia therapy, magnetic resonance contrast, cancer hyperthermia, drug delivery, cell sorting, and most recently for inducing ferroptosis in cancer cells (4, 5, 1219). Thus, they present an interesting and important class of nanoparticles for applications in medicine.

Here, we show that host immune status and the immune components of the TME are key factors influencing retention of 100-nm hydroxyethyl starchcoated iron oxide nanoparticles in orthotopic mammary tumors. When labeled with an antibody, the nanoparticles were retained by tumors regardless of the presence of the target antigen, whereas retention of the unlabeled counterpart was not substantial. Additional experiments demonstrated that systemic exposure of tumor-bearing immune competent mice to the nanoparticles induced immune-mediated tumor growth inhibition with evidence of later infiltration by CD8+ T cells. Both plain and antibody-labeled nanoparticles initiated similar immune responses with similar tumor growth inhibition and T cell infiltration into tumors, despite different tumor retention. This suggests that complex interdependencies exist between host and tumor immune responses to nanoparticle exposure. Together, these results offer intriguing possibilities to explore nanoparticle targeting of the tumor immune microenvironment, and they demonstrate an exciting potential to develop nanoparticles as cancer immune therapy platforms.

We used amine-functionalized starch-coated bionized nanoferrite (BNF) nanoparticles with trastuzumab (BH), a humanized antihuman epidermal growth factor receptor 2 (HER2/neu) monoclonal antibody approved for clinical use in the management of HER2+ breast cancer (Fig. 1A). The ability of trastuzumab to target HER2+ cancer cells in tumors has been validated and well documented, as has its use for nanoparticle-targeting studies (20, 21). The precursor BNF-Plain (BP) nanoparticles comprise a magnetic iron oxide core that is coated with hydroxyethyl starch (core shell) to provide biocompatibility and colloid stability in biological media (1519).

(A) Schematic of particle chemistry showing amine functionalization of BP nanoparticles using maleimide precursors for conjugation with thiol moieties of the antibody (trastuzumab). (B) Western blot analysis showing HER2 protein expression by human breast cancer cell lines used in the study. (C) Immunofluorescence results showing HER2 protein surface expression in six human breast cancer cell lines. MDA-MB-231 is a triple-negative ER/PR/HER2- cell line. MCF7/neo and MCF7/HER2 are an isogenic pair with HER2-expressing (MCF7/HER2) variant having a single copy of HER2 gene and HER2- (MCF7/neo), which received a scrambled gene. Other cell lines are wild type and express varying amounts of HER2 protein. (D) In vitro iron content analysis (ferene-s assay) after exposure of cells to BP and BH nanoparticles shows a positive correlation with HER2 protein level and iron uptake in the breast cancer cells. For the assay, cells were incubated at 37C for 3 hours with BP or BH nanoparticles (0.5 mg/ml) and evaluated for total iron content after washing unbound particles. Untreated cells, Herceptin alone, and BNF-IgG were used as controls. The average of three independent experiments is shown. Statistical differences among BP, BH, and BNF-IgG were obtained by two-tailed Students t test (*P < 0.05 and **P < 0.01). (E) Schematic of the overall study design using mouse models of human breast cancers. See text for details.

The choice of 100-nm BNF nanoparticles was motivated from our previous study, which demonstrated higher accumulation by the 100-nm nanoparticles to tumors than with 30-nm nanoparticles, despite the longer blood circulation time of the latter construct (1519). When measured by dynamic light scattering, BP nanoparticles had a mean measured (z average) hydrodynamic diameter of 99 nm (3) with a mean polydispersity index of 0.07 (0.02) (table S1). Zeta potential, a measure of surface charge density, was slightly negative (2.2 0.2 mV) at pH 7.4. Overall, addition of trastuzumab to the BP nanoparticles had only a modest effect on the measured physical properties of the nanoparticles. Several of antibody-labeled nanoparticles were prepared and assayed using a modified in vitro test to confirm selective binding of the BH construct. In all cases, successful binding of antibody was confirmed by a modified bicinchoninic acid assay (BCA) and immunofluorescence (tables S2 to S5 and fig. S1, A and B). A BNFimmunoglobulin G (IgG) construct was synthesized with a nonspecific human polyclonal antibody, as an additional control. The measured physical properties of the BNF-IgG nanoparticles were similar to those of BH nanoparticles (tables S2 and S4).

We limited our selection of cancer models to those for which a stable transmembrane protein/marker is well documented and for which multiple cell lines and mouse models are readily available. In general, HER2+ breast cancer biology has been extensively studied, providing numerous human and mouse cell lines to yield xenograft, syngeneic, and spontaneous models (22, 23). For in vitro and xenograft studies, we selected six human breast cancer cell lines (Fig. 1, B and C, and table S3). HER2 protein expression was verified by Western blotting (Fig. 1B). We used an isogenic pair derived from a HER2- MCF7 parental line, MCF7/HER2 (+), and MCF7/neo () (Fig. 1, B and C). The variable total protein and surface expression of HER2 were evident in three HER2+ lines: HCC1954, BT474, and SKBR3 from both Western blotting and immunofluorescence, whereas MDA-MB-231 cells showed no HER2 expression.

Residual iron concentration was measured in cells using a modified ferene-s assay (24) and correlated with HER2/neu expression following exposure to BH nanoparticles. Both iron concentration and HER2/neu expression followed the same progression: MCF7/HER2 < HCC1954 < BT474 < SKBR3 (Spearman correlation coefficient, = 0.89, P = 0.03; Fig. 1D, inset, and fig. S1C), confirming that in vitro targeting occurred via the expected antibody-antigen binding.

We used two immunodeficient strains of mice [athymic nude and nonobese diabetic/severe combined immunodeficiency (NOD/scid) gamma (NSG)] engrafted with five human breast cancer cell lines: two HER2- cell lines (MDA-MB-231 and MCF7/neo) and three HER2+ lines (MCF7/HER, HCC1954, and BT474). The xenograft study design is illustrated in Fig. 1E, and details are provided in Materials and Methods and table S6. Visibly evident 24 hours after injection by discoloration of tumors, BH nanoparticles were retained by tumors to a greater extent than were BP nanoparticles (Fig. 2A).

(A) Gross morphology of tumors following intravenous injection with BP or BH nanoparticles shows different tissue color. Darker (black) color indicates greater particle uptake. Tumors from NOD/scid (NSG) mice show more BH than BP. Photo credit: Preethi Korangath, Johns Hopkins University. (B) Representative images of HER2 immunohistochemistry (IHC) from breast xenografts showing that expression correlates with in vitro expression. (C and D) Inductively coupled plasma mass spectrometry (ICP-MS) of Fe recovered from tumors excised from mice injected with BH nanoparticles demonstrates consistently higher Fe content than tumors from mice injected with BP nanoparticles regardless of HER2 status of the tumor. Recovered iron was higher in tumors excised from NSG mice (D) than that from athymic nude mice (C) (*P < 0.05, **P < 0.01, and ***P < 0.001). (E and F) Prussian bluestained tissue slides recovered from the same tumors as in (C) and (D) and digitally analyzed for percent positive area that showed a similar trend as observed with ICP-MS. (G and H) ICP-MS analysis of Fe from the livers showed higher iron content in mice injected with BP nanoparticles than mice injected with BH nanoparticles, mirroring the results of Fe recovered from tumors (**P 0.01 and ***P < 0.0001).

Volumetric analysis of iron by inductively coupled plasma mass spectrometry (ICP-MS) recovered from tumors grown in nude mice corroborated our observations of gross tumor presentation. HER2 status/expression of tumors was confirmed by immunohistochemistry (IHC) (Fig. 2B). Higher iron concentrations were present in tumors of mice injected with BH relative to phosphate-buffered saline (PBS) or BP-injected mice (P < 0.001) irrespective of HER2 status (Fig. 2C). In contrast, intratumor iron concentrations measured from mice receiving BP was only slightly higher than PBS-injected controls (MCF7/HER, HCC1954, and BT474; P > 0.05; see Fig. 2C). Iron recovered from nude mice bearing MCF7/neo tumors injected with BH was comparable to those recovered from MCF7/HER tumors. Comparable iron recovery in these two isogenic (HER2+/) tumor models following injection with BH, which was higher than either PBS- or BP-injected controls, suggests that biological factors other than antibody-antigen binding were responsible for nanoparticle retention. In other words, the BH nanoparticle targeting observed in vitro was not evident in vivo.

This pattern of retention was also measured in tumors recovered from NSG mice (Fig. 2D and fig. S2); however, HER2 expression by the tumor slightly correlated more with BH retention in NSG mice than in nude mice (Fig. 3, A and B). In contrast to results obtained from nude mice, iron recovered from HCC1954 and BT474 tumors in NSG mice was slightly higher than in MCF7/HER, consistent with higher HER2/neu protein expression in these cell lines (Fig. 1, B and C).

(A) Analysis of Prussian bluepositive (nanoparticle-rich) areas of tumors from nude mice injected with BH nanoparticles reveals only weak correlation with HER2 expression. (B) Conversely, this correlation is stronger in tumors from NSG mice. (C and D) Weak or no correlation was observed between BH nanoparticle presence and CD31+ (vascular endothelium) regions. (E) Representative histology images of sequential sections showing IBA-1+ cells associated with Prussian bluepositive areas in HCC1954 (HER2+) tumors grown in NSG mice and treated with BH (a) hematoxylin and eosin (H&E), (b) Prussian blue, (c) HER2 IHC, (d) IBA-1 IHC, (e) CD-31 IHC, (f) H&E of another area from same tumor, (g) sequential section stained for Prussian blue shows positive staining for iron nanoparticles, and (h) immunofluorescence (IF) staining for IBA-1 shows positivity in the nanoparticle accumulated region. (F and G) Iron recovery from HER2+ (HCC1954) or HER2 (MDA-MB-231) tumors is similar whether BNF nanoparticles have trastuzumab (anti-HER2) or human IgG (polyclonal), suggesting that antibody-antigen binding does not drive intratumor nanoparticle accumulation. ns, not statistically significant.

We analyzed tumor tissue sections stained with Perls reagent (also known as Prussian blue) to visualize the nanoparticle-rich regions across all models (Fig. 2, E and F, and fig. S3). The trends observed with gross presentation and ICP-MS were consistent with tumor histopathology (Fig. 2, C to F, and fig. S2) and also revealed notable spatial heterogeneity of iron localization. Nevertheless, all tumor models studied showed significantly more nanoparticle retention when mice were injected with BH, but localization to cancer cells was not evident.

As previously observed, a substantial amount of systemically injected nanoparticles will accumulate in the liver (611, 2527). It is widely held that resident macrophages (liver) and circulating macrophages along with other phagocytic immune cells will sequester nanoparticles of about 100 nm in diameter, clearing them from blood circulation and depositing them into the liver and other organs. Our ICP-MS analysis of iron recovered from the livers showed that all mice injected with nanoparticles exhibited higher iron concentration in the liver than PBS-injected controls. However, the livers of mice injected with BP had higher iron content than the livers of mice injected with BH (Fig. 2, G and H). We conclude that BH retention in tumors (and perhaps other tissues not assayed) contributed to the reduced liver content when compared with BP-injected mice.

Higher tumor retention of Herceptin (Her/trastuzumab)labeled nanoparticles having varied composition and sizes (15 to 500 nm) following systemic delivery into nude female mice bearing MCF7 tumors has been noted (2831). It is worth emphasizing, however, that MCF7 cancer cells express no HER2 antigen on their membranes, begging the question of the mechanisms of targeting observed in these previous studies. Together, results reported here and elsewhere indicate that retention of nanoparticles in (xenograft human-mouse) tumors may depend on complex biological responses that are intertwined with the host immune system. We note in our results that immune status of the mouse seemed to play a role in nanoparticle retention in tumors, whereas antigen expression by cancer/tumor cells seemed to have very little influence, especially in nude mice (Fig. 2 and fig. S2). Further study was needed to determine nanoparticle association with cell type.

We analyzed comparable regions of stained serial tissue sections in detail by scoring to determine whether intratumor nanoparticle localization correlated with tumor-specific factors. Digitally scored Prussian bluestained sections were compared with manual scoring of the corresponding HER2-stained tumor sections using Spearmans rank correlation coefficient from mice injected with BH nanoparticles (Materials and Methods). A positive but weak correlation was found between BH localization and HER2/neu protein expression in nude mice ( = 0.3827; Fig. 3A). We measured a stronger, positive correlation between BH localization with HER2+ sections in tumors from NSG mice ( = 0.8462; Fig. 3B). These results were consistent with both ICP-MS and digital scoring of Prussian bluestained slides among all tumor models (Fig. 2, C to F) further supporting our observations that immune status of the host animal was an important factor determining BH retention in tumors but not for BP (fig. S4A).

To test whether BH nanoparticle retention in tumors correlated with the tumor microvascular network, we compared Prussian bluestained areas with corresponding sections stained with CD31 for visualizing the vascular endothelium (32). No correlation was found between BH score and CD31+ score in sections obtained from nude mice ( = 0.018; Fig. 3C), but a weak positive correlation was measured in sections obtained from NSG mice ( = 0.3241; Fig. 3D). By contrast, slight positive correlations were found with CD31+ regions in both nude and NSG mice injected with BP (fig. S4A).

Both athymic nude and NSG mice lack mature T cells, but NSG mice, in addition, also lack functional components of their innate immune system (table S6) (33). We speculated that subpopulations of innate immune cells in the TME contributed to BH retention. We compared Prussian bluestained sections with corresponding sections stained for ionized calcium-binding adapter molecule 1 (IBA-1), a pan-(murine) macrophage marker that also labels other myeloid cells including subpopulations of dendritic cells, monocytes, activated neutrophils, and some types of endothelial cells (Fig. 3E) (34). Comparing IBA-1+ tissue sections with Prussian bluepositive regions revealed that antibody-labeled nanoparticles were found in similar locations as IBA-1+ regions within the TME in both nude and NSG mice (HCC1954 tumor grown in NSG mice, Fig. 3E; BT474 tumor grown in NSG mice, fig. S4B). However, we found no significant differences in the content (number) of IBA-1+ cells among any of the tumor models or treatment (fig. S4C).

Next, we tested the notion that antibody-antigen binding to cancer cells does not determine tumor localization of BH to tumors by using BNF nanoparticles labeled with a nonspecific human polyclonal IgG. BNF-IgG nanoparticles were intravenously injected into cohorts of both nude and NSG mice bearing HER2+ (HCC1954) and HER2 (MDA-MB-231) tumors. ICP-MS analysis of tissue iron content of tumors extracted from mice injected with BNF-IgG was similar to that measured from mice injected with BH in both tumor models and immune backgrounds of mice (Fig. 3, F and G, and fig. S5, A and B). These results support that retention of antibody-labeled nanoparticles (i.e., BH or BNF-IgG) was independent of antibody-antigen binding.

From the results obtained across the five human tumor xenograft models in two immunodeficient mouse strains and with two antibody nanoparticle types (trastuzumab and nonspecific IgG), we hypothesized that BNF nanoparticle retention by tumors was determined by active biological processes influenced (or directed) by cells of the innate immune system, residing within the TME and reacting to the presence of an antibody on the nanoparticle surface. Our analysis of xenograft tumors of the IBA-1stained tissue sections provided no evidence of measurable (aggregate innate) immune cell infiltration into or depletion from the tumors following nanoparticle exposure. To the contrary, the area of IBA-1+ regions among PBS- and nanoparticle-injected cohorts was comparable (fig. S4C), indicating that tumor-associated immune cell subpopulations internalized antibody-labeled nanoparticles (trastuzumab or IgG; see Fig 3, F and G). To test whether macrophages were responsible for these observations, we depleted macrophages by treatment with clodronate liposomes in athymic nude mice growing HCC1954 tumors and injected with BH (35). Unexpectedly, macrophage depletion alone failed to decrease the amount of BH nanoparticles retained in tumors (fig. S5C), suggesting involvement by other immune cells.

BNF nanoparticle localization in tumors across multiple xenograft mouse models suggested that immune status contributed to, and perhaps dominated, nanoparticle retention. To test this concept further, we used a syngeneic tumor model derived from the transgenic huHER2 mouse (Fig. 4A) and transplanted to NSG, nude, and immune competent FVB/N mice. HER2 protein expression in the tumors was confirmed by IHC (Fig. 4B).

(A) Schema of transgenic huHER2 tumor allograft development and IHC confirmation of HER2 protein expression on cancer cells in tumors. (B) IHC analysis demonstrates that HER2 protein expression in syngeneic huHER2 allografts is comparable among the range of immune strains of mice tested: FVB/N, athymic nude, and NSG mice. (C) Gross appearance of huHER2 allograft tumors grown to 150 to 200 mm3 in FVB/N, athymic nude, or NSG mice 24 hours after they were injected via tail vein with BP or BH nanoparticles shows that BH accumulation is greatest in tumors growing in immune competent host(s). Photo credit: Preethi Korangath, Johns Hopkins University. (D) ICP-MS results showing absolute iron recovery from tumors grown in all mice reveals highest accumulation of BH nanoparticles in FVB/N mice (*P < 0.05, **P < 0.005, and ***P 0.0001). (E) Histology analysis revealed that Prussian bluepositive area was seen in stromal area and colocalized more with IBA-1+ cells than HER2+ tumor cells.

The intensity of coloration, 24 hours after injection by BH nanoparticles into FVB/N mice, was visibly greater than that displayed by tumors in either NSG or nude mice (Fig. 4C). Iron content analysis by ICP-MS and analysis of Prussian bluestained slides demonstrated a notable uptake of BH by huHER2 allograft tumors grown in FVB/N mice (Fig. 4D and fig. S6, A and B). Similar to results obtained from xenograft models (Fig 2), FVB/N mice showed retention of less BH in the liver than BP, and higher iron content was detected in the lymph nodes and spleens of both BP- and BH-injected mice (fig. S6, C to E). Prussian bluepositive areas appeared more prominently in stromal regions associated with IBA-1+ areas than in the HER2+ regions (Fig. 4E and fig. S7). These results provided strong evidence that immune status of mouse models is a critical biological variable for nanoparticle targeting studies; however, the nature of this interaction was unclear.

Across all models studied, the presence of immune cells within tumors was detected. Colocalization of nanoparticles and IBA-1+ cells occurred at the tumor periphery (Fig. 3E and fig. S3) in xenograft tumors and in tumor-stromal interfaces in the immune competent huHER2 allograft model (Fig. 4E and fig. S7). It has been well documented that the cancer tissue boundary of tumors often exhibits proinflammatory features (36). We hypothesized that tumor-associated immune cells exhibiting an inflammatory phenotype preferentially sequestered and retained antibody-labeled nanoparticles.

To test this hypothesis and to further elucidate the mechanism of nanoparticle retention in the TME, we performed tests in vitro with murine macrophages and neutrophils. Macrophages were activated with lipopolysaccharide (LPS) and interferon- (IFN-) to mimic a T helper 1 (TH1)type induction (M1) or with interleukin-4 (IL-4) to mimic a TH2-type induction (M2). When exposed to either BP or BH, macrophages always sequestered more BH than BP; however, M1 macrophages sequestered significantly more nanoparticles, especially BH (Fig. 5A). Uninduced neutrophils showed no preference for either construct; however, when activated with LPS (TH1-type induction), neutrophils demonstrated significantly greater preference for BH (Fig. 5B).

(A) Undifferentiated RAW 264.7 (M0) or differentiated M1 or M2 (LPS + IFN- or IL-4, respectively) macrophages were incubated for 24 hours with BP or BH, and ferene-s assay was conducted to measure the total amount of iron uptake per cell. As a control, BP and Her, added together, were also used. As shown in the figure, BH nanoparticles were taken up more significantly than BP by macrophages irrespective of their phenotype. The uptake was significantly higher in M1 macrophages than either M0 or M2, which indicates that proinflammatory macrophages take up more BP and BH nanoparticles with preference toward BH. (B) Likewise, LPS-activated neutrophils (induced) preferentially sequestered BH over BP, whereas no difference in uptake was observed with nave bone marrow neutrophils (uninduced). (C) Total cell count obtained from magnetically separated BP- or BH-injected tumors shows significant difference. Immune competent FVB/N mice (n = 3 per group, two tumors each) bearing huHER2 tumors were intravenously injected with BP or BH. After 24 hours, tumors were harvested and digested to isolate single cells and were magnetically separated to collect nanoparticle-associated cells to determine the total cell count. (D) Analysis of magnetically sorted cells obtained from in vivo tumors showed that nanoparticles were associated with immune cells, not tumor cells. Immune competent FVB/N mice (n = 5 to 8 per group) bearing huHER2 tumors were intravenously injected with PBS, BP, or BH. After 24 hours, tumors were harvested and digested to isolate single cells and were magnetically separated to collect nanoparticle-associated cells for analysis by flow cytometry. Gating strategy is provided in fig. S8. Cell numbers measured from BP- and BH-injected mice are shown as change in ratio relative to PBS-injected mice (PBS ratio = 1). (a) Populations of cancer cells were not changed in nanoparticle-associated cancer cells. Ratios of NK cells (b), monocytes (c), TAMs (d), neutrophils (e), and dendritic cells (f) are increased in nanoparticle fractions, suggesting uptake of nanoparticles by immune cells rather than tumor cells. (*P 0.05, **P 0.01, and ***P < 0.001).

Magnetic nanoparticles provide a unique tool to query biological responses because they enable magnetic sorting to isolate specific cell populations containing the nanoparticles. To further elucidate the in vivo tumor immune response to BNF nanoparticle exposure, tumor digests were placed on a permanent magnet. Cells containing nanoparticles were sedimented, whereas cells devoid of nanoparticles remained suspended. Sedimented (nanoparticle-associated) cells were collected and analyzed for total number (Fig. 5C). Consistent with in vitro results, the total number of cells containing iron was higher in tumors of mice injected with BH than in those injected with BP. To distinguish among tumor-associated cell populations that sequestered nanoparticles, both sedimented (nanoparticle associated) and suspended (supernatant, no nanoparticle) cells were collected and analyzed by polychromatic flow cytometry. Figures S8 and S9 provide graphical gating strategy and complete results of analysis, respectively. Results of magnetic sorting of equal (initial) numbers of tumor-derived cell populations are displayed in Fig. 5D (a to f) as ratios of cell number by type and fraction relative to cell numbers obtained from PBS-injected mice. PBS ratios are expressed as unity and all others as <1 or >1 depending on the number of cells detected in each fraction. Among cancer cells, it is notable that for either BP or BH, numbers were lower than from PBS-injected controls, indicating little nanoparticle association with the HER2+ cancer cells (Fig. 5D, a). This is consistent with histopathology (Fig. 3E). Following intravenous delivery, evidence indicates that nanoparticle association with cancer cells was minimal regardless of HER2+ expression, further confirming the different performance of antibody-labeled nanoparticles in vivo versus in vitro.

On the basis of the evidence, nanoparticle retention in the studied models was likely determined by tumor-associated leukocytes, but what effect did systemic exposure to nanoparticles have on the tumor immune microenvironment? We used polychromatic flow cytometry to identify changes of individual tumor immune cell populations in huHER2 allograft tumors growing in FVB/N mice following injection with nanoparticle or free antibody (Fig. 6A and fig. S8, A and B). Twenty-four hours after intravenous injection, we measured a slight decrease of live cell populations in tumors derived from mice receiving either BP or BH relative to PBS-injected controls. No measurable differences were detected in cancer cell populations among the four cohorts, but a significant decrease in CD45+ population was noted (fig. S9B, a to c).

Immune competent FVB/N mice (n = 5 to 8 per group) bearing huHER2 tumors were intravenously injected with PBS, BP, BH, or Herceptin (Her). After 24 hours, tumors were harvested and digested to isolate single cells and evaluated by polychromatic fluorescence-activated cell sorter (FACS). Gating strategy is provided in fig. S8. (A) Relative decreases in T cell (a) and B cell (b) populations were observed following injection of nanoparticles. By contrast, relative increases were measured in many innate immune cell populations within the TME: NK cells (c), neutrophils (d), TAMs (e), and monocytes (f) 24 hours after nanoparticle exposure. Except for TAMs, no significant increase was seen in any other immune cell population after Her injection. (*P 0.05 and **P 0.01). (B) Graphic representation of distributions of nanoparticle-associated CD45+ immune cells among the cohorts.

Nanoparticle exposure induced many changes across a number of tumor immune cell lineages, with a notable decrease in T cells and an increase in the relative fraction (i.e., ratio) of innate immune cells initiating a restructuring of the immune compartment of the TME (Fig. 6A, a). B cell populations also decreased in BH- and Her-treated groups (Fig. 6A, b). Relative to PBS controls, natural killer (NK) cell and monocyte fractions increased following BH injection but not in mice receiving BP or Her (Fig. 6A, c and f). Populations of other phagocytic innate immune cells, specifically neutrophils, and tumor-associated macrophages (TAMs) increased with either BP or with BH injection relative to controls (Fig. 6A, d and e, and fig. S9B), but dendritic cell populations remained relatively unchanged 24 hours after injection (fig. S9B, d) as did the fraction of T cells (GD T cells) (fig. S9B, e). However, we found no evidence in histology data indicating that depletion or infiltration of innate immune cells carrying nanoparticles to or from tumors occurred after nanoparticle injection, suggesting capture of nanoparticles by the resident population(s) of innate immune cells in the TME (Fig. 3 and fig. S4C) (37). Nevertheless, for conclusive quantification of this process, further study is needed. Exposure to free trastuzumab (Her) elevated TAMs, reflecting a specific interaction (Fig. 6A, e).

Trastuzumab is a humanized monoclonal antibody with a human IgG1 (hIgG1) that can elicit a response in murine macrophages (38). Furthermore, it is recognized that Fc receptors on murine macrophages can recognize hIgG1 (38), and the response observed in our flow cytometry with free trastuzumab (Her) is consistent with this observation (Fig. 6A, e). Note that, however, macrophages were the only tumor immune population that elevated within 24 hours following injection with free trastuzumab, whereas multiple immune cell subpopulations responded to BP and BH exposure (Fig. 6, A and B, and fig. S9B). The tumor immune response to BH was more complex than that to free trastuzumab (Her)including T cells, NK cells, monocytes, neutrophils, dendritic cells, and macrophagesand it was similar to that of BP. Thus, while the potential exists for specific interactions between murine macrophages and hIgG1-containing nanoparticles, our evidence demonstrates that labeling the surface of a nanoparticle with a hIgG1 monoclonal antibody alters the immune response to recognize the nanoparticle-antibody construct as an entity distinguishable from free antibody.

The data indicate that, in addition to macrophages (TAMs), many other lineages of phagocytic innate immune cellsNK cells, monocytes, neutrophils, and dendritic cellsreside in the TME sequestered nanoparticles (Fig. 5D, b to f, and fig. S9A, b to i). It seemed that an intact immune system is a critical component in determining the retention of nanoparticles in solid tumors. To challenge this notion, we pretreated tumor-bearing mice with a pan-leukocyte inhibitor, azathioprine (39, 40), before injecting with BH. Iron recovered from tumors in azathioprine-treated mice was significantly reduced and similar to BP-injected mice (fig. S10, A and B), confirming the role of a wider immune involvement in nanoparticle retention.

These results support a model that tumor-associated phagocytic immune cells significantly influence the degree of retention of systemically delivered nanoparticles within the TME. Furthermore, our results demonstrate that an intact host immune system can manifest decidedly different tumor retention when compared with comparable immunodeficient models, raising an important question about clinical relevance of studies performed in the latter. Depending on environmental chemical cues, tumor-associated leukocytes may display a greater sensitivity to the chemical signatures of nanoparticles than their counterparts residing in other tissues. This offers potential for tumor targeting with nanomedicines.

In a complex manner, while the restructuring of the immune compartment of the TME, likely mirroring a systemic immune response to nanoparticle exposure, was similar for both BH and BP nanoparticles, it is only the BH nanoparticles that were significantly retained within the TME. These complex and seemingly contradictory immune responses may indicate potential for anticancer effects.

To explore the potential clinical relevance of our findings, we used the huHER2 allograft tumor model to ascertain effects of nanoparticle exposure on tumor growth in FVB/N and athymic nude mice. Five days after implantation of huHER2 tumors, FVB/N or athymic nude mice received a single intravenous injection of PBS, BP, BH, or Her as previously described. Exposure to either BP or BH significantly delayed tumor growth in FVB/N mice but not in athymic nude mice (Fig. 7, A to C, and fig. S11, A to C). As expected, trastuzumab alone was effective to significantly inhibit tumor growth in both FVB/N and athymic nude mice, however, its mode of action involves direct binding via HER2 antigen to cancer cell membranes. Our evidence shows that neither BP nor BH nanoparticles associated appreciably with cancer cells in vivo; thus, the therapeutic effect seen only in FVB/N mice due to nanoparticle exposure must involve an alternate mechanism that we hypothesized was mediated by the adaptive immune system. To gain further insight, we repeated the experiment in FVB/N mice and conducted flow cytometry analysis of immune populations in tumors 3, 7, and 14 days after injection. Beginning at 7 days after injection, significant increases in activated T cells (CD3+/CD4+/CD8+) were measured in tumors, reversing the depletion observed at 24 hours and 3 days and supporting a model of immune-mediated tumor suppression induced by systemic exposure to nanoparticles (Fig. 7, D and E, and figs. S11D to S14). Immune cells known to be involved in adaptive immune signaling, i.e., dendritic and T cells, displayed a complex time-dependent patternincreasing to day 3 and decreasing thereafterconsistent with adaptive immune signaling response (Fig. 6 and figs. S9 and S13) (41, 42). On the other hand, phagocytic effector immune cells, i.e., macrophages and monocytes, initially displayed relatively elevated numbers at day 1 but displayed no such increases afterward relative to PBS controls (Fig. 6 and fig. S9 and S14). These complex and time-dependent immune cell responses observed in the TME resemble systemic responses observed in mice following acute and nonlethal infection by some pathogens, i.e., Listeria monocytogenes, which can also lead to anticancer immune stimulation (41, 42). Note that both BH and BP nanoparticles induced similar effects on tumor immune cell populations and on tumor growth, despite the fact that BP nanoparticles were not significantly retained within the tumor. This suggests that exposure to nanoparticles has the potential to induce both systemic and local (tumor) effects, which bear further study and offer potential for developing another paradigm in cancer nanomedicine (fig. S15).

(A) Female FVB/N mice bearing huHER2 allograft tumors (n = 7 to 18 per group) were intravenously injected with either PBS, BP, BH (5 mg per mouse), or Herceptin (175 g per mouse) 5 days after tumor implantation (day 0). Growth of the tumors was monitored by caliper measurements twice per week for 4 weeks (means SEM). Final tumor weight is given in inset (**P < 0.005 and &P 0.0001). (B) On day 28, all mice were euthanized, and representative images of tumors are shown. Photo credit: Preethi Korangath, Johns Hopkins University. [C (a and b)] Female athymic nude mice bearing huHER2 allograft tumors (n = 6 to 7 per group) were similarly treated as above, and 3 weeks of tumor growth and tumor weight is reported (means SEM, *P < 0.05). [D (a and b) and E (a and b)] Flow analysis of tumors: As in (A), mice (n = 5 per group) were intravenously injected with either PBS, BP, BH (5 mg per mouse), or Herceptin (175 g per mouse) on the 10th day after tumor implantation. Seven days after injection, mice were euthanized; tumors were harvested, and single cells were isolated and evaluated by FACS. Infiltration of CD3+ T cells with increases in CD8+ T cells was measured following nanoparticle exposure, likely leading to growth inhibition observed in (A) (*P < 0.05). FITC, fluorescein isothiocyanate.

In summary, targeting nanoparticles has been a topic of considerable debate, even controversy, in the cancer nanomedicine community (17, 16, 2531). In most previous studies, the biology of tumor and/or host was not studied in detail with analysis of tissue histology and flow cytometry, thus motivating our efforts to understand the role of host biology in nanoparticle-tumor interactions (610). Across all models studied, we found strong evidence pointing to immune status of the host as a key factor determining the retention of antibody-labeled nanoparticles in tumors. Using an immune intact model, we discovered that the retention of nanoparticles in tumors was dominated by multiple lineages of tumor-associated immune cells when the nanoparticles included an antibody and found no in vivo evidence supporting a mechanism of antibody-antigen binding (i.e., the mechanism operating in vitro) to cancer cells in the tumor. Yet, the amount of nanoparticle retained by the tumor within 24 hours was most pronounced in an immune intact model, further emphasizing the significance of an intact immune system in studies of nanoparticle delivery to solid tumors. Our results demonstrate that the host immune system can be a substantial factor in studies of cancer nanomedicine and that macrophages are only one among many immune cell lineages that determine nanoparticle fate. It was only when we pharmacologically inhibited the entire host immune system that we measured a reduced retention of the BH nanoparticles. While these findings reveal new insights, they also raise many questions regarding complexities of nanoparticleimmune cell interactions in vivo across the many biological models used in cancer research and how immune cell receptors distinguish among nanoparticle coatings.

Related to this, but in a different manner, we observed that the immune response to nanoparticle exposure measured in tumors was equally profound and seemed insensitive to nanoparticle composition (BP or BH). As measured by population changes of immune cells in the TME, the immune response included an initial T cell depletion and later T cell infiltration into the tumor with significant tumor growth inhibition.

The presence of immune cells within an established solid tumor implies that immune cells are performing surveillance and homeostasis functions to support the growth and maintenance of the tumor. Our results show that exposure to nanoparticles can disrupt this delicate balance, potentially enabling a transient immune recognition of the tumor. In an immune-intact model of cancer, the systemic delivery of a nanoparticle construct can initiate a complex immune response, which can affect tumor growth regardless of retention. These results highlight the notion that the biology of the host and cancer tumor forms an interconnected and inextricably linked biological network that interacts in complex ways to determine the biological fate and retention of nanoparticles. Host immune status and, consequently, composition of the immune compartment(s) within the TME are critical variables in developing and testing the performance of cancer nanomedicines. Results presented here motivate more questions of mechanism of host and tumor immune cell interactions with nanoparticles. They also point to new possibilities for nanoparticle anticancer immunotherapy technologies.

MDA-MB-231 [ER/PR/HER2 () negative], MCF7 [ER/PR (+) positive/HER2 () negative], and BT474 [ER/PR/HER2 (+) positive] were purchased from the American Type Culture Collection (ATCC; Manassas, VA) and maintained according to the suppliers recommendations. They were grown in Dulbeccos modified Eagles medium (DMEM) containing 10% fetal bovine serum (FBS). HCC1954 [ER/PR () negative/HER2 (+) positive] was grown in RPMI containing 10% FBS. MCF/neo and MCF7/HER were provided by K. Osborne (University of Texas Health Science Center). All cell lines were authenticated using short tandem repeat analysis (data provided upon request) and matched against ATCC and Deutsche Sammlung von Mikroorganismen und Zellkulturen databases to ensure the genetic origins.

The nanoparticles used for this study are commercially available aqueous suspensions of hydroxyethyl starchcoated magnetite (Fe3O4) core-shell particles (BNF; Micromod Partikeltechnologie GmbH, Rostock, Germany). The synthesis and physical characterization of the BNF particles have been extensively documented (1519). Briefly, BNF particles were produced by precipitating ferric and ferrous sulfate salts from solution at high pH in a high-pressure homogenization reaction vessel, which controls both crystal formation and aggregation. According to the manufacturer, they have a mean hydrodynamic diameter of ~100 nm and an iron content of >50% (w/w) [or iron oxide of >70% (w/w)].

The mean hydrodynamic diameter of the magnetic iron oxide nanoparticles (BNF) and their zeta potential were measured in 1 mM PBS buffer (pH 7.4) with a Zetasizer Nano ZS90 (Malvern Instruments Limited, UK) at an iron concentration of 0.1 mg/ml. The mean particle diameter Z(Ave) is given as a result of the cumulative analysis of the autocorrelation function. The polydispersity index is a measure of the quality of the size distribution. Monodisperse suspensions have a polydispersity index of <0.25.

The monoclonal anti-HER2/neu antibody (Her), or trastuzumab (trade name) (Genentech, South San Francisco, CA), was purchased from Johns Hopkins Pharmacy and was shipped to micromod for conjugation with BNF particles to form BH. The Her was formulated according to the prescribing information. The lyophilized powder that contained 440 mg of Her was dissolved in 20 ml of bacteriostatic water for injection (provided). The Her solution was purified by washing with PBS buffer (pH 4) using a desalting column (PD-10, GE Healthcare, UK) to remove the stabilizing agents. The obtained Her solution was thiolated with Trauts reagent (2-iminothiolane) as follows: The antibody solution (390 l, 1.7 mg/ml in PBS buffer) was mixed with 160 l of 1.4 mM 2-iminothiolane in 450 l of PBS-EDTA buffer. After shaking for 1 hour at room temperature, the excess of 2-iminothiolane was removed by washing with PBS-EDTA buffer (PBS buffer, 1 mM EDTA) in a desalting column (G-25, GE Healthcare, UK). In parallel, an aqueous suspension of 80-nm BNF-starch nanoparticles with amino groups on the surface (2.25 ml, [Fe] = 8.0 mg/ml; product code: 10-01-801, micromod Partikeltechnologie GmbH) was mixed with 250 l of 10 PBS-EDTA buffer. Sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (sulfo-SMCC) (3.6 mg) was dissolved in 100 l of dimethyl sulfoxide and added to the BNF-starch suspension. After 1 hour of shaking at room temperature, the excess of sulfo-SMCC was removed by washing with PBS-EDTA buffer in a PD-10 desalting column. The maleimide-functionalized nanoparticles were mixed with the thiolated antibody solution and shaken for 3 hours at room temperature. Then, 200 l of 20 mM cysteine solution was added to quench the remaining maleimide groups on the nanoparticle surface. Last, the nanoparticles were washed by magnetic separation in a high-gradient magnetic field column (QuadroMACS with LD columns, Miltenyi Biotec GmbH, Bergisch-Gladbach, Germany) with 5 ml of PBS-Tween buffer (pH 7.4, 0.05% Tween 20) and 5 ml of PBS buffer (pH 7.4) per column filling. The magnetic column was removed from the magnet, and the nanoparticles were eluted with 2 ml of water per column filling. The high gradient magnetic field (HGMF) wash was repeated until the suspension was completely washed. The suspension was filtered using 0.22-m polyethersulfone filter (Carl Roth GmbH, Karlsruhe, Germany).

After conjugation, BH nanoparticles were rigorously characterized for their physical and biological properties in vitro to ensure nanoparticle stability, and BNF-Her binding was successful and retained sufficient protein. Antibody immunoreactivity of the BH construct was separately tested using a cell culturebased assay (see below).

The iron content of the antibody-conjugated nanoparticles (BH) was determined after the digestion of a 20 l of sample with 80 l of concentrated HCl. After addition of 4.9 ml of a citrate phosphate buffer (pH 3.6), the iron concentration was calorimetrically determined with the Spectroquant Kit (Merck, Germany) against a Titrisol Iron Standard (Merck, Germany).

The amount of the conjugated antibody in the sample was measured by a modified BCA method. The BCA reagents were obtained from Thermo Fisher Scientific (Germany). The calibration curve was obtained by adding increasing amounts of an albumin standard solution to aminated BNF-starch particles (without antibody on the surface) at a constant iron concentration of 0.25 mg/ml. The antibody-conjugated nanoparticles were adjusted to the same iron concentration of 0.25 mg/ml and developed with the BCA reagent together with the calibration curve for 2 hours at 37C.

Polyclonal normal hIgG was purchased from R&D Systems (Minneapolis, MN) for conjugation with BNF nanoparticles for BNF-IgG nanoparticles. Methods to conjugate the IgG antibody to BNF nanoparticles were same as for trastuzumab, except that proportions of reagents were altered to accommodate differences between the antibodies. The lyophilized hIgG (2 mg) was dissolved in 1 ml of PBS buffer (pH 7.4) and purified by washing with PBS buffer (pH 4) using a desalting column (G-25, GE Healthcare, UK). The antibody solution used was 510 l (1.3 mg/ml) in PBS buffer and was mixed with 160 l of 1.4 mM 2-iminothiolane in 330 l of PBS-EDTA buffer. After shaking for 1 hour at room temperature, the excess 2-iminothiolane was removed by washing with PBS-EDTA buffer in a desalting column. In parallel, BP nanoparticles with amino groups on the surface were prepared as described above. The maleimide-functionalized nanoparticles were mixed with the thiolated antibody solution, reacted, washed, and purified as above.

The detailed protocol for conducting the modified ferene-s measurement of iron associated with cells after exposure to BNF nanoparticles has been previously described (24). Briefly, cells were trypsinized and washed with PBS thoroughly and were incubated at 37C with BP (0.5 mg/ml), BH, or trastuzumab (Her 2 g/ml) alone for 3 hours in growth media (DMEM + 10% FBS) with occasional shaking/tapping of tubes to maximize distribution and prevent settling of cells. After incubation, cells were pelleted by centrifugation and washed with PBS to remove unbounded particles and again pelleted by centrifugation. This washing with PBS was repeated three more times. The final cell pellet was resuspended in PBS and counted using a Cellometer (Nexcelom Bioscience, Lawrence, MA) to obtain the total number of cells. The cells in the tubes were then centrifuged, and the supernatant was removed to add working solution (acetate buffer with ascorbic acid). Cell pellets were digested in working solution by incubating at room temperature for at least 20 hours before reading in a colorimeter. A known quantity of ferene-s was used along with other external standard reference materials to quantify the iron concentration of the test samples according to previously described procedures (24). For the entire study, we used only those batches of BH showing more than fourfold retention by SKBR3 cells, as measured by iron concentration with the ferene-s assay when compared to BP (table S5). In addition, we used MDA-MB-231 (HER2-, control) to confirm that nonspecific binding of BH particles by those cells was minimal (<1 pg of Fe per cell).

Cells were trypsinized and washed in PBS and incubated in DMEM + 10% FBS at 37C with trastuzumab (2 g/ml) for 3 hours with occasional shaking/tapping of tubes to maximize distribution and prevent settling of cells. After incubation, cells were washed four times with PBS and plated on poly-lysinecoated coverslips in six-well plates. After overnight incubation, they were washed with PBS, fixed with methanol for 10 min, and blocked with 1% bovine serum albumin for 30 min at 37C. Dye-labeled secondary antibody (anti-human Alexa Fluor 488, Life Technologies, Eugene, OR) was added and incubated for 1 hour in the dark at room temperature, followed by washing three times in PBS and mounting with mounting media containing DAPI (4,6-diamidino-2-phenylindole). They were then visualized and photographed using a fluorescent microscope (Zeiss Axioimager Z1, Carl Zeiss Microscopy GmbH, Jena, Germany). To visualize BNF-HER nanoparticles alone, 30 l of BNF-HER or BP nanoparticles was separated on a magnet for 2 hours at 4C. The particles suspended in 1 ml of PBS volume and the concentration of BH nanoparticle suspensions were incubated with anti-human Alexa Fluor 488 secondary antibody (1:1000) for 1 hour at room temperature. The particles were then separated on a magnet for 1 hour, washed with PBS, and dropped on a clean slide to mount and visualize with a fluorescent microscope.

Cells were lysed with radioimmunoprecipitation assay buffer (Sigma-Aldrich, St. Louis, MO) containing protease and phosphatase inhibitors on ice for 30 min. The lysates were centrifuged at 13,000 rpm for 15 min. The supernatant was collected and quantified by BCA (Thermo Fisher Scientific, Waltham, MA) assay. Thirty to 50 g of total protein were used for SDSpolyacrylamide gel electrophoresis gel after being heated with sample buffer. The proteins were then transferred to nitrocellulose membranes. After blocking with 5% milk solution in PBS-T (1% Tween 20) for 30 min, the membranes were blotted with primary antibody (anti-human HER2 antibody, 1:1000; Cell Signaling Technology, 29D8) overnight and with secondary horseradish peroxidase (HRP)conjugated antibody (GE Healthcare, UK) for 1 hour. The membranes were developed using chemiluminescence reagent (Amersham Biosciences, Marlborough, MA).

RAW264.7 cells were purchased from the ATCC (Manassas, VA) and maintained in DMEM with 10% heat-inactivated FBS. Low-passage cells were used for the study (P3 to P5). For M1 macrophage activation, cells were treated with LPS (100 ng/ml; Sigma-Aldrich, St. Louis, MO) and IFN- (50 ng/ml; Miltenyi Biotech, Germany) for 24 hours. To differentiate cells into M2, phenotype cells were treated with IL-4 (10 ng/ml; Miltenyi Biotech, Germany) for 24 hours (43). Induced and uninduced cells (1 million) were collected and treated with either BP or BH nanoparticles (0.5 mg/ml) or cotreated with BP and Her (16.3 g/ml; equivalent to protein content of BH) for 24 hours. After incubation, cells were washed thoroughly with PBS four times and processed for iron content analysis with the ferene-s assay as described above. Experiments were repeated three times.

Neutrophils were activated in vivo with LPS by the method described by Rnnefarth et al. (44). Briefly, 50 g of LPS was intraperitoneally injected into FVB/N mice (n = 3). After 18 hours, activated peritoneal neutrophils were collected by injecting 5 ml of PBS to peritoneum, cells were harvested, and red blood cells (RBCs) were lysed with ammonium-chloride-potassium (ACK) lysis buffer and thoroughly washed. Nave neutrophils were prepared using methods described by Mcsai et al. (45). For this, bone marrow cells were collected to Hanks balanced salt solution (HBSS) from femur and tibia of FVB/N mice (n = 3). RBCs were lysed from bone marrow cells with ACK lysis buffer, and cells were passed through a 70-m strainer. These cells were then centrifuged after layering on 62.5% freshly prepared Percoll in HBSS for 30 min at 1000g without brake. The cloudy pellet of neutrophils was collected. Uninduced bone marrowderived neutrophils and activated peritoneal-derived neutrophils were incubated with BP or BH nanoparticles (0.5 mg/ml) for 24 hours, and ferene-s assay was conducted to measure the amount of iron uptake per cell as described above.

All animal studies were approved by the Institutional Animal Care and Use Committee at Johns Hopkins University and were conducted using female mice. All mice were fed normal diet and water ad libitum. They were maintained in the normal 12-hour light/12-hour dark cycle. All animals were closely monitored for any distress or pain throughout the study period. The weight range of animals during the study was 20 to 30 g. Strains of mice used in all studies were athymic nude (Charles River Laboratories, Frederick, MD), NSG (Sydney Kimmel Comprehensive Cancer Center colony, Johns Hopkins University School of Medicine, Baltimore, MD), and FVB/N (Jackson laboratory, Bar Harbor, ME); all mice were aged 6 to 8 weeks. The characteristics of cell lines and mice used are provided above and in tables S3 and S6. A schematic of the xenograft tumor study design is provided in Fig. 1E. An overview of the numbers of mice divided by strain and group used for the studies is provided in table S10. Depending on cohort, 3 106 MDA-MB-231 or HCC1954 or 5 106 MCF-7(HER/neo) or BT474 cells were suspended in 50 l of PBS and Matrigel (1:1) and injected into the fourth mammary gland on either side of female mice under anesthesia. For MCF-7(HER/neo) and for BT474 xenograft studies, mice received estrogen by implanting a 60-day release estrogen pellet (0.72 mg per pellet; Innovative Research of America, Sarasota, FL) 5 days before cell line injection on the dorsal neck region through a small subcutaneous insertion made using sterile scissors while mice were under ketamine/xylazine anesthesia[ketamine (10 mg/ml) Vedco Inc., St. Joseph, MO] and xylazine (2 mg/ml; Lloyd Inc., Shenandoah, IA) mixed in sterile PBS and intraperitoneally injected at 0.01 ml/g body weight. Tumor volume was monitored by caliper measurements twice weekly when tumors became palpable. When the measured tumor volume was 125 to 200 mm3, mice were randomly assigned into cohorts containing five animals in each group. Group I received intravenous (tail vein) injections of PBS and served as (negative) control. Group II received intravenous injections to tail vein of BP (5 mg of Fe per animal), and group III received intravenous tail vein injections of BH (5 mg of Fe per animal). Group IV received injections of BNF-IgG (intravenous tail vein injections; 5 mg of Fe per animal) only for mice bearing either MDA-MB-231 or HCC1954 xenografts. The total volume of injection was 150 l in all cases. Twenty-four hours after injection, all mice were euthanized to collect tumors and liver for analysis.

Athymic nude mice growing HCC1954 tumors (n = 3 with two tumors each) were treated with two consecutive doses of clodronate liposome (CL) (Liposoma, Netherlands) via intraperitoneal (300 l per animal) injection. After the second dose of CL, BH nanoparticles were injected (5 mg of Fe per mouse intravenously) and euthanized 24 hours later to harvest tumors for ICP-MS.

The second half of each tumor and whole livers were weighed, lyophilized, and stored at 20C until analysis by ICP-MS using methods previously described (46). Briefly, each tissue sample was transferred to a 7-ml Teflon microwave digestion vessel (Savillex Corporation, Eden Prairie, MN) to which 1 ml of optima-grade HNO3 (Fisher Scientific, Columbia, MD) was added. The vessel was sealed and placed into a 55-ml Teflon microwave digestion vessel (CEM Corporation, Matthews, NC) to which 10 ml of ultrapure H2O (Millipore Corporation, Billerica, MA), and samples were digested in a MARS5 Xpress microwave (CEM Corporation, Matthews, NC) using a single-stage ramp-to-temperature of 15-min ramp to 130C with a hold of 30 min. After cooling, each sample was diluted: 35 l of sample digest and 300 l of HNO3 were added to 14.665 ml of ultrapure H2O to achieve a final HNO3 concentration of 2% (w/v). External reference standards scandium (CPI Incorporated, Santa Rosa, CA) and Seronorm Trace Elements Whole Blood (SERO AS, Billingstad, Norway) were added to normalize instrument counts and sample iron content, respectively. In addition, four reagent blanks were digested and analyzed in each run to correct for background iron content.

An Agilent 7500ce ICP-MS (Agilent Technologies, Santa Clara, CA) was used to measure iron content of each sample. Measurements were blank-corrected using the average iron value of the reagent blanks and corrected using external standard reference materials. An eight-point calibration curve (0, 1, 5, 10, 50, 100, 500, and 1000 g/liter) was obtained from Standard Reference Material (SRM) measurements. The analytical limit of detection (LOD) was calculated by multiplying the SD of the lowest detectable calibration standard (1 g/liter) by three. For samples with values below the analytical LOD, one-half of the LOD was substituted (46).

Fresh tumors were fixed in 10% formalin and sectioned on positively charged slides. For HER2 staining, a VECTASTAIN ABC kit (Vector Laboratories, Burlingame, CA) was used to perform IHC. After hydration with serial dilutions of ethanol, antigen retrieval was performed using 10 mM citrate buffer. The sections were then treated with 3% hydrogen peroxide for 10 min and incubated with normal serum to block nonspecific binding. The sections were later incubated overnight with anti-human HER2 antibody (1:400; Cell Signaling Technology, 29D8). Secondary antibody (provided in the kit) was added the next day after washing, followed by incubation with ABC reagent and developed with 3,3-Diaminobenzidine (DAB) (DAB peroxidase substrate kit, Vector Laboratories, Burlingame, CA) reagent and counterstained with hematoxylin (Dako North America Inc., Carpinteria, CA.) as specified by the manufacturer. For CD31 (Dianova, DIA 310), and IBA-1 (Wako, 019-19741), after deparaffinization and hydration, the slides were steamed in HTTR or EDTA buffer for 45 min in a steamer followed by washing in PBS containing Tween. They were then blocked in peroxidase solution and incubated with CD31 (1:40) or IBA-1 (1:2500) antibody for 45 min at room temperature. After washing, sections were incubated with secondary antibody (PowerVision Poly-HRP anti-Rabbit IHC Detection Systems Novocastra, Leica Biosystems, Buffalo Grove, IL) for 30 min at room temperature followed by washing. The slides were then washed and developed with DAB fast (Sigma-Aldrich, St. Louis, MO) for 20 min at room temperature and counter stained with hematoxylin.

One-half of each tumor was fixed with 10% formalin and submitted for paraffin embedding and sectioning for hematoxylin and eosin (H&E) staining, Prussian blue (also known as Perls reagent) staining to visualize nanoparticle (iron oxide) distributions, and IHC (HER2, CD31, and IBA-1). All stained slides were evaluated by a pathologist (B.W.S.) and quantitated in a blinded study. For manual analysis, HER2 immunostains were semiquantiatively scored to determine the percentage of tumor cells with positive, membranous staining. For automated image analysis, whole slides were digitized using the Aperio ScanScope At or CS system (Aperio, Vista CA) at 40 magnification. Analysis was performed using Aperio ImageScope software (version 12.3.0.5056) with the included Positive Pixel Count algorithm. Images were manually annotated to select a region of interest representing a full cross section of each graft and a 50-m border of surrounding subcutaneous tissue. Artifacts and necrotic regions of the tumor were excluded from analysis. Default hue values (brown, positive; blue, negative) were used for immunostains (DAB Chromogen) and were adjusted for Prussian blue (blue, positive; pink, negative). Digital analysis settings that were used are provided in tables S7 and S8. One slide per condition per tumor was analyzed, and results represent as percent positive pixels over negative pixels in region of interest.

Transgenic (huHER2) mice (FVB/N background) that develop mouse mammary tumor virusdriven mammary-specific human HER2overexpressing tumors were provided under a material transfer agreement (Genentech, South San Francisco, CA). These mice are well characterized for their tumor development and response to trastuzumab as described elsewhere (22, 23). The primary tumor from a donor mouse was collected in normal media and finely minced. Approximately 3 to 4 mm3 of the mash were implanted into the fourth mammary gland on either side of FVB/N females (Jackson laboratory, Bar Harbor, ME) at 6 to 8 weeks of age under anesthesia. Tumor growth was monitored twice weekly by caliper measurements. When the measured tumor volume was ~1000 mm3, tumors were collected and minced to repeat the transplantation into other FVB/N recipient mice for expansion by serial transplantation for up to six generations. At each generation, a section of tumor was fixed in formalin and was analyzed for tumor morphology by H&E and (human) HER2/neu expression by IHC. Nanoparticle uptake studies commenced when a sufficient number of tumors was established to ensure completion of the huHER2 study design. To establish tumors for the nanoparticle studies, huHER2 tumors were collected from five to eight FVB/N donor mice and minced. Portions of the mashes (3 to 4 mm3) were implanted into the fourth mammary gland on either side of female recipient mice comprising immune strains FVB/N, athymic nude, or NSG (18 to 24 animals in each group) under anesthesia. When the measured tumor volume reached 150 to 200 mm3, animals were randomly assigned into cohorts comprising five to nine animals in each group and treated according to their cohort as described for the xenograft studies (see above). For tumor growth delay, huHER2 allografts were implanted in either FVB/N or athymic nude mice and intravenously treated with PBS, BP, BH (5 mg per animal, or Her 175 g/ml, equivalent dose of Her on BH particles) 5 days after implantation (day 0). Tumors were measured and recorded twice weekly up to 28 days. On day 28, all animals were euthanized, tumors were collected, and weight was recorded.

FVB/N female mice (five to eight animals per group) growing single huHER2 allograft tumors of 150 to 200 mm3 received intravenous (tail vein) injections of PBS, BP, or BH (5 mg of Fe per mouse) or trastuzumab (Her; 175 g per mouse). Mice were euthanized 24 hours after injection. For the later time point (14 day after injection) flow analysis, mice bearing huHER2 allograft tumors were injected with PBS, BP, BH, or Her (same concentrations as above) 10 days after tumor implantation. Tumors were minced with a sterile blade in a petri dish and transferred to a 50-ml conical tube containing digestion media [DMEM + 10% FBS (heat inactivated) and 0.1% collagenase + 0.005% hyaluronidase]. The tubes were rotated at 37C for 30 min, and the dissociated tissue was filtered through a 100-m filter. After centrifugation at 1400 rpm for 10 min, the pellets were washed with 30 ml of DMEM with 10% serum and centrifuged again for 10 min. Supernatant was discarded, and RBCs were lysed with ACK lysis buffer at room temperature for 3 to 5 min. An additional 30 ml of media was added, and the mixture was centrifuged at 1400 rpm for 10 min. The supernatant was discarded, and 500 l of media was added to resuspend the pellet. An aliquot of this whole tumor was removed and labeled with the panel of flow cytometry antibodies (see table S9 for list). The remaining single cells were incubated on a rare-earth permanent magnet at 37C for 30 min. After incubation, the supernatant was carefully separated to a fresh tube for flow cytometry. Three milliliters of media was added to the adhered (remaining) fraction cells, which contained magnetic (nanoparticle) material. Media was added to all sample tubes to make up a final volume of 3 ml, and they were then centrifuged at 1400 rpm for 10 min. The supernatant was discarded, and pellets were suspended in PBS and counted to measure the total number of cells. Cells (1 106 to 2 106) were collected and treated with Fc blocker (2 l of 100 l of PBS; anti-CD16/32, BioLegend, San Diego, CA) and incubated on ice for 10 min. Samples were then centrifuged at 1400 rpm for 3 min, after which the cells were incubated with LIVE/DEAD solution (1 l of 100 l of PBS; Zombie Aqua, BioLegend, San Diego, CA) at room temperature in the dark for 30 min and then centrifuged. After centrifugation, cells were washed with 100 l of PBS with 5% heat-inactivated FBS and again centrifuged. Cells were then stained with 50 l of the solution of panel of antibodies (table S9) in appropriate dilution and incubated at room temperature in the dark for 30 min, centrifuged at 1400 rpm for 3 min, washed with 100 l of PBS and 5% heat-inactivated FBS, and again centrifuged. Cells were then fixed with 50 l of fix/perm solution and incubated at room temperature in the dark for 30 min, centrifuged, and washed. Two hundred microliters of PBS with 5% heat-inactivated FBS was added, and the suspensions were stored at 4C until they were measured by polychromatic flow cytometry (LSR-II, BD Biosciences, San Jose, CA). The gating and selection of quadrants were based on fluorescence minus one controls. Analysis of data was done using FlowJo (version 10) software. Gating strategy is provided in figs. S8 and S11.

FVB/N female mice (three animals per group with two tumors each) growing huHER2 allograft tumors at ~100 mm3 received intravenous (tail vein) injections of BP or BH (5 mg of Fe per mouse). Mice were euthanized 24 hours after injection. Tumors were processed as above for flow cytometry and placed on permanent magnet for 30 min. After discarding supernatant, total numbers of magnetically attached cells were counted in a cell counter (Nexcelom, MA).

huHER2 allograft tumors were grown in FVB/N female mice (n = 4). When the tumor volumes reached ~150 mm3, mice were treated with subcutatneous injections of azathioprine (Sigma-Aldrich, St. Louis, MO), a pan leukocyte inhibitor at a dose of 200 mg/kg body weight for three consecutive days (39, 40). On the third day and 4 hours after azathioprine injection, mice were intravenously injected with BH (5 mg of Fe per animal). Mice were euthanized 24 hours after BH injection to collect tumor for ICP-MS analysis.

Results of all statistical analyses are provided in tables S11 to S33. ICP-MS and Prussian blue Aperio scored data were modeled as following log-normal distributions on the basis of proper exploratory analyses. Generalized mixed-effect models were used, with treatment, strain, and cell line as the fixed effects and mice as the random effect (intercept), such that the ratios between any two levels of fixed effects may be reported directly from the mixed-effect models. Models with fixed effect only and with two-way and three-way interactions were considered. Multiple comparison adjustments were made using the Bonferroni method to strongly control the overall family-wise type I error at 0.05.

Flow cytometry and tumor growth analysis. For flow cytometry and tumor growth data, it seemed unreasonable to assume commonly used parametric distributions. Therefore, pure nonparametric comparisons were made across all comparisons with Dwass-Steel-Critchlow-Fligner procedure for the pairwise comparisons to properly adjust for the potential inflation of family-wise type I errors.

In vitro cell count and ferene-s assay. All in vitro cell count and ferene-s assay data were analyzed by Students t test.

Correlation analysis. Rank-based, nonparametric Spearman correlation was performed using SAS 9.4, R, and Graphpad software.

Supplementary material for this article is available at http://advances.sciencemag.org/cgi/content/full/6/13/eaay1601/DC1

Supplementary Materials and Methods

Table S1. Summary of analytical data of (80 nm) BP nanoparticles.

Table S2. Summary of analytical data of all BNF-HER nanoparticles prepared.

Table S3. Characteristics of breast cancer cell lines used in the study.

Table S4. Summary of analytical data of BNF-IgG nanoparticles.

Table S5. Summary of analytical data of BNF-HER nanoparticles that passed in vitro qualification testing.

Table S6. Summary of immune modifications in mouse strains used for study.

Table S7. Summary of Aperio imaging settings used for digital analysis of tissue sections.

Table S8. Definitions of parameters used for Aperio imaging settings.

Table S9. Antibodies used for flow cytometry and their dilutions.

Table S10. Summary of numbers and strains of mice used in the study.

Table S11. Summary of one-factor model statistical analysis of iron measurements in xenograft models.

Table S12. Summary of two-factor model statistical analysis of iron measurements in xenograft models.

Table S13. Summary of three-factor model statistical analysis of iron measurements in xenograft models.

Table S14. Summary of one-factor model statistical analysis of Prussian blue histopathology analyses in xenograft models.

Table S15. Summary of two-factor model statistical analysis of Prussian blue histopathology analyses in xenograft models.

Table S16. Summary of three-factor model statistical analysis of Prussian blue histopathology analyses in xenograft models.

Table S17. Summary of statistical analysis of whole tumor digests flow cytometry in huHER2 allograft model.

Table S18. Summary of statistical analysis of nanoparticle-associated fractions (magnetic-sorted sediment) from flow cytometry in huHER2 allograft model.

Table S19. Summary of statistical analysis of nanoparticle-depleted fractions (magnetic-sorted supernatant) from flow cytometry in huHER2 allograft model.

Table S20. Summary of statistical analysis of iron measurements (ICP-MS) obtained from the livers of xenograft models.

Table S21. Ratio of Fe level between groups (treatment).

Table S22. Ratio of Fe level between groups (strains).

Table S23. Statistical analysis of ICP-MS huHER2-FVB/N lymph node data.

Table S24. Statistical analysis of ICP-MS huHER2-FVB/N spleen data.

Table S25. Statistical analysis of ICP-MS huHER2-FVB/N liver data.

Table S26. Ratio of percent positive between groups.

Table S27. Statistical analysis of tumor weight in huHER2-FVB/N.

Table S28. Statistical analysis of tumor growth in huHER2-FVB/N.

Table S29. Statistical analysis of whole tumor flow data third day.

Visit link:
Nanoparticle interactions with immune cells dominate tumor retention and induce T cellmediated tumor suppression in models of breast cancer - Science...

Children’s cancer researcher named Woman of the Year – UNSW Newsroom

Professor Maria Kavallaris, a leading childhood cancer researcher and a pioneer of nanomedicine in Australia, is the 2020 NSW Premier's Woman of the Year.

Professor Kavallaris is Head of Translational Cancer Nanomedicine at Childrens Cancer Institute and Founding Director of the Australian Centre for NanoMedicine at UNSW Sydney.

The prestigious award, announced a ceremony in Sydney this morning,recognises NSW women who have excelled in their chosen career, field or passion; are exceptional achievers who have made a significant contribution to NSW; and whose accomplishments make them a strong role model for other women.

I am truly honoured to have received this award and I hope it inspires young women to do what they love, grow and learn, and to lead with generosity and respect, Professor Kavallaris said.

Professor Kavallaris is internationally renowned for her research in cancer biology and therapeutics. She has been widely recognised for the innovation and impact of her research, her leadership as well as her mentoring of talented young scientists. She is passionate about training the next generation of research leaders.

Her personal journey with cancer began at the age of 21 and has driven her research to develop effective and less toxic cancer treatments.

As one of the original three scientists appointed at the Childrens Cancer Institute when its laboratories first opened in 1984, she has made important discoveries in relation to the mechanisms of clinical drug resistance and tumour aggressiveness in childhood cancer.

Her studies have not only identied how some tumours can grow and spread;she has also applied this knowledge to develop eective, less toxic cancer therapies using nanotechnology.

To be able to make a difference to the lives of children with cancer and their families by developing better treatments and improving survival rates is very humbling. Even if you can save one childs life, thats an incredible feat, Professor Kavallaris said.

As a conjoint professor in the UNSW Faculty of Medicine, Professor Kavallaris relishes her role of mentor and has supervised many Honours and PhD students, several of whom have become research leaders.

Professor Kavallariss extensive research and leadership contributions have been recognised withnumerous awards including the NSW Premiers Prize for Science and Engineering (Leadership in Innovation in NSW) in 2017, the Australian Society for Biochemistry and Molecular Biologys Lemberg Medal in 2019 and she was made aMember of the Order of Australia (AM) for significant service to medicine, and to medical research, in the field of childhood and adult cancerson Australia Day 2019.

The rest is here:
Children's cancer researcher named Woman of the Year - UNSW Newsroom