Tag Archives: Warren Chan

Shooting drugs to an infection site with a slingshot

It seems as if I’ve been writing up nanomedicine research a lot lately, so I would have avoided this piece. However, since I do try to cover Canadian nanotechnology regardless of the topic and this work features researchers from l’Université de Montréal (Québec, Canada), here’s one of the latest innovations in the field of nanomedicine. (I have some additional comments about the nano scene in Canada and one major issue concerning nanomedicine at the end of this posting.) From a May 8, 2017 news item on ScienceDaily,

An international team of researchers from the University of Rome Tor Vergata and the University of Montreal has reported, in a paper published this week in Nature Communications, the design and synthesis of a nanoscale molecular slingshot made of DNA that is 20,000 times smaller than a human hair. This molecular slingshot could “shoot” and deliver drugs at precise locations in the human body once triggered by specific disease markers.

A May 8, 2017 University of Montreal news release (also on EurekAlert), which originated the news item, delves further into the research (Note: A link has been removed),

The molecular slingshot is only a few nanometres long and is composed of a synthetic DNA strand that can load a drug and then effectively act as the rubber band of the slingshot. The two ends of this DNA “rubber band” contain two anchoring moieties that can specifically stick to a target antibody, a Y-shaped protein expressed by the body in response to different pathogens such as bacteria and viruses. When the anchoring moieties of the slingshot recognize and bind to the arms of the target antibody the DNA “rubber band” is stretched and the loaded drug is released.

“One impressive feature about this molecular slingshot,” says Francesco Ricci, Associate Professor of Chemistry at the University of Rome Tor Vergata, “is that it can only be triggered by the specific antibody recognizing the anchoring tags of the DNA ‘rubber band’. By simply changing these tags, one can thus program the slingshot to release a drug in response to a variety of specific antibodies. Since different antibodies are markers of different diseases, this could become a very specific weapon in the clinician’s hands.”

“Another great property of our slingshot,” adds Alexis Vallée-Bélisle, Assistant Professor in the Department of Chemistry at the University of Montreal, “is its high versatility. For example, until now we have demonstrated the working principle of the slingshot using three different trigger antibodies, including an HIV antibody, and employing nucleic acids as model drugs. But thanks to the high programmability of DNA chemistry, one can now design the DNA slingshot to ‘shoot’ a wide range of threrapeutic molecules.”

“Designing this molecular slingshot was a great challenge,” says Simona Ranallo, a postdoctoral researcher in Ricci’s team and principal author of the new study. “It required a long series of experiments to find the optimal design, which keeps the drug loaded in ‘rubber band’ in the absence of the antibody, without affecting too much its shooting efficiency once the antibody triggers the slingshot.”

The group of researchers is now eager to adapt the slingshot for the delivery of clinically relevant drugs, and to demonstrate its clinical efficiency. [emphasis mine] “We envision that similar molecular slingshots may be used in the near future to deliver drugs to specific locations in the body. This would drastically improve the efficiency of drugs as well as decrease their toxic secondary effects,” concludes Ricci.

Here’s a link to and a citation for the paper,

Antibody-powered nucleic acid release using a DNA-based nanomachine by Simona Ranallo, Carl Prévost-Tremblay, Andrea Idili, Alexis Vallée-Bélisle, & Francesco Ricci. Nature Communications 8, Article number: 15150 (2017) doi:10.1038/ncomms15150 Published online: 08 May 2017

This is an open access paper.

A couple of comments

The Canadian nanotechnology scene is pretty much centered in Alberta and Québec. The two provinces have invested a fair amount of money in their efforts. Despite the fact that the province of Alberta also hosts the federal government’s National Institute of Nanotechnology, it seems that the province of Québec is the one making the most progress in its various ‘nano’ fields of endeavour. Another province that should be mentioned with regard to its ‘nano’ efforts is Ontario. As far as I can tell, nanotechnology there doesn’t enjoy the same level of provincial funding support as the other two but there is some important work coming out of Ontario.

My other comment has to do with nanomedicine. While it is an exciting field, there is a tendency toward a certain hyperbole. For anyone who got excited about the ‘slingshot’, don’t forget this hasn’t been tested on any conditions close to the conditions found in a human body nor have they even used, “... clinically relevant drugs,  … .”  It’s also useful to know that less than 1% of the drugs used in nanoparticle-delivery systems make their way to the affected site (from an April 27, 2016 posting about research investigating the effectiveness of nanoparticle-based drug delivery systems). By the way, it was a researcher at the University of Toronto (Ontario, Canada) who first noted this phenomenon after a meta-analysis of the research,

More generally, the authors argue that, in order to increase nanoparticle delivery efficiency, a systematic and coordinated long-term strategy is necessary. To build a strong foundation for the field of cancer nanomedicine, researchers will need to understand a lot more about the interactions between nanoparticles and the body’s various organs than they do today. …

It’s not clear from the news release, the paper, or the May 8, 2017 article by Sherry Noik for the Canadian Broadcasting Corporation’s News Online website, how this proposed solution would be administered but presumably the same factors which affect other nano-based drug deliveries could affect this new one,

Scientists have for many years been working on improving therapies like chemo and radiation on that score, but most efforts have focused on modifying the chemistry rather than altering the delivery of the drug.

“It’s all about tuning the concentration of the drug optimally in the body: high concentration where you want it to be active, and low concentration where you don’t want to affect other healthy parts,” says Prof. Alexis Vallée-Bélisle of the University of Montreal, co-author of the report published this week in Nature Communications.

“If you can increase the concentration of that drug at the specific location, that drug will be more efficient,” he told CBC News in an interview.

‘Like a weapon’

Restricting the movement of the drug also reduces potentially harmful secondary effects on other parts of the body — for instance, the hair loss that can result from toxic cancer treatments, or the loss of so-called good bacteria due to antibiotic use.

The idea of the slingshot is to home in on the target cells at a molecular level.

The two ends of the strand anchor themselves to the antibody, stretching the strand taut and catapulting the drug to its target.

“Imagine our slingshot like a weapon, and this weapon is being used by our own antibody,” said Vallée-Bélisle, who heads the Laboratory of Biosensors & Nanomachines at U of M. “We design a specific weapon targeting, for example, HIV. We provide the weapon in the body with the bullet — the drug. If the right solider is there, the soldier can use the weapon and shoot the problem.”

Equally important: if the wrong soldier is present, the weapon won’t be deployed.

So rather than delay treatment for an unidentified infection that could be either viral or bacterial, a patient could receive the medication for both and their body would only use the one it needed.

Getting back to my commentary, how does the drug get to its target? Through the bloodstream?  Does it get passed through various organs? How do we increase the amount of medication (in nano-based drug delivery systems) reaching affected areas from less than 1%?

The researchers deserve to be congratulated for this work and given much encouragement and thanks as they grapple with the questions I’ve posed and with all of the questions I don’t know how to ask.

Nominations open for Kabiller Prizes in Nanoscience and Nanomedicine ($250,000 for visionary researcher and $10,000 for young investigator)

For a change I can publish something that doesn’t have a deadline in three days or less! Without more ado (from a Feb. 20, 2017 Northwestern University news release by Megan Fellman [h/t Nanowerk’s Feb. 20, 2017 news item]),

Northwestern University’s International Institute for Nanotechnology (IIN) is now accepting nominations for two prestigious international prizes: the $250,000 Kabiller Prize in Nanoscience and Nanomedicine and the $10,000 Kabiller Young Investigator Award in Nanoscience and Nanomedicine.

The deadline for nominations is May 15, 2017. Details are available on the IIN website.

“Our goal is to recognize the outstanding accomplishments in nanoscience and nanomedicine that have the potential to benefit all humankind,” said David G. Kabiller, a Northwestern trustee and alumnus. He is a co-founder of AQR Capital Management, a global investment management firm in Greenwich, Connecticut.

The two prizes, awarded every other year, were established in 2015 through a generous gift from Kabiller. Current Northwestern-affiliated researchers are not eligible for nomination until 2018 for the 2019 prizes.

The Kabiller Prize — the largest monetary award in the world for outstanding achievement in the field of nanomedicine — celebrates researchers who have made the most significant contributions to the field of nanotechnology and its application to medicine and biology.

The Kabiller Young Investigator Award recognizes young emerging researchers who have made recent groundbreaking discoveries with the potential to make a lasting impact in nanoscience and nanomedicine.

“The IIN at Northwestern University is a hub of excellence in the field of nanotechnology,” said Kabiller, chair of the IIN executive council and a graduate of Northwestern’s Weinberg College of Arts and Sciences and Kellogg School of Management. “As such, it is the ideal organization from which to launch these awards recognizing outstanding achievements that have the potential to substantially benefit society.”

Nanoparticles for medical use are typically no larger than 100 nanometers — comparable in size to the molecules in the body. At this scale, the essential properties (e.g., color, melting point, conductivity, etc.) of structures behave uniquely. Researchers are capitalizing on these unique properties in their quest to realize life-changing advances in the diagnosis, treatment and prevention of disease.

“Nanotechnology is one of the key areas of distinction at Northwestern,” said Chad A. Mirkin, IIN director and George B. Rathmann Professor of Chemistry in Weinberg. “We are very grateful for David’s ongoing support and are honored to be stewards of these prestigious awards.”

An international committee of experts in the field will select the winners of the 2017 Kabiller Prize and the 2017 Kabiller Young Investigator Award and announce them in September.

The recipients will be honored at an awards banquet Sept. 27 in Chicago. They also will be recognized at the 2017 IIN Symposium, which will include talks from prestigious speakers, including 2016 Nobel Laureate in Chemistry Ben Feringa, from the University of Groningen, the Netherlands.

2015 recipient of the Kabiller Prize

The winner of the inaugural Kabiller Prize, in 2015, was Joseph DeSimone the Chancellor’s Eminent Professor of Chemistry at the University of North Carolina at Chapel Hill and the William R. Kenan Jr. Distinguished Professor of Chemical Engineering at North Carolina State University and of Chemistry at UNC-Chapel Hill.

DeSimone was honored for his invention of particle replication in non-wetting templates (PRINT) technology that enables the fabrication of precisely defined, shape-specific nanoparticles for advances in disease treatment and prevention. Nanoparticles made with PRINT technology are being used to develop new cancer treatments, inhalable therapeutics for treating pulmonary diseases, such as cystic fibrosis and asthma, and next-generation vaccines for malaria, pneumonia and dengue.

2015 recipient of the Kabiller Young Investigator Award

Warren Chan, professor at the Institute of Biomaterials and Biomedical Engineering at the University of Toronto, was the recipient of the inaugural Kabiller Young Investigator Award, also in 2015. Chan and his research group have developed an infectious disease diagnostic device for a point-of-care use that can differentiate symptoms.

BTW, Warren Chan, winner of the ‘Young Investigator Award’, and/or his work have been featured here a few times, most recently in a Nov. 1, 2016 posting, which is mostly about another award he won but also includes links to some his work including my April 27, 2016 post about the discovery that fewer than 1% of nanoparticle-based drugs reach their destination.

Warren Chan and a distinguished career in nanobioengineering

I’m always happy to find out more about Canada’s nanotechnology scene and this Nov. 1, 2016 University of Toronto (UofT) news release by Carolyn Farrell provides an informative overview with its description of Warren Chan’s current achievements and recent career acknowledgement,

Institute of Biomaterials and Biomedical Engineering (IBBME) Professor Warren Chan has been named the University of Toronto Distinguished Professor of Nanobioengineering. The Distinguished Professor Award recognizes individuals with highly distinguished accomplishments and those who display exceptional promise. Chan will hold the professorship for a five-year term starting November 1, 2016. He is one of nine Distinguished Professors in the Faculty.

Chan leads a world-renowned research program in biomedical nanotechnology that has garnered international recognition for its exceptional innovation, breadth, and impact. His group has created a rapid, point-of-care nanotechnology-based diagnostic system that can detect multiple diseases from a single drop of blood.  The device is based on a combination of quantum dot barcoding technology — which picks out genetic markers for diseases — and techniques that allow the signals to be imaged and identified by a smartphone. The device costs less than $100 and can detect sequences from viruses like HIV or hepatitis B in less than one hour at 90 per cent accuracy.

Another focus of Chan’s research has been the development of technology for delivering chemotherapy drugs directly into tumours, avoiding the side-effects of traditional chemotherapy treatments. Chan and his research group have designed a targeted molecular delivery system that uses modular nanoparticles whose shape, size and chemistry can be altered by the presence of specific DNA sequences. This work has been published in the Proceedings of the National Academy of Sciences and the journal Science.

Chan’s most recent work, featured on the cover of ACS Nano, has provided unique insights into the fate and distribution of nanoparticles injected into the body. Chan’s lab developed techniques to visualize interactions between nanoparticles and the body’s various organs using 3D optical microscopy, revealing for the first time the distribution of these structures within tumour tissue. They have also set up an open online database that will enable the collection and analysis of data on nanoparticle delivery efficiency from any published study.

Professor Chan has received several Canadian and international awards for his research, including a NSERC Steacie Fellowship, the BF Goodrich Young Inventors Award, the Lord Rank Prize Fund Award in Optoelectronics, and the Dennis Gabor Award. He was recently the inaugural winner of the Kabiller Young Investigator Award from Northwestern University’s International Institute for Nanotechnology.

“I am profoundly grateful that UofT has recognized Warren Chan’s groundbreaking research applying nano-engineered materials to the diagnosis and treatment of disease,” said Dean Cristina Amon. “His research, which has the potential to revolutionize healthcare, has contributed tremendously to U of T’s growing reputation as a leading centre for biomedical engineering.”

Warren Chan has been mentioned here before with regard to his groundbreaking work, most recently in a Sept. 9, 2016 post about how the liver prevents nanoparticles from reaching cancer cells and in an April 27, 2016 post about the discovery that fewer than 1% of nanoparticle-based drugs reach their destination.

Congratulations Professor Chan!

Discovering how the liver prevents nanoparticles from reaching cancer cells

There’s a lot of excitement about nanoparticles as enabling a precise drug delivery system but to date results have been disappointing as a team of researchers at the University of Toronto (Canada) noted recently (see my April 27, 2016 posting). According to those researchers, one of the main problems with the proposed nanoparticle drug delivery system is that we don’t understand how the body delivers materials to cells and disappointingly few nanoparticles (less than 1%) make their way to tumours. That situation may be changing.

An Aug. 19, 2016 news item on Nanowerk announces the latest research from the University of Toronto,

The emerging field of nanomedicine holds great promise in the battle against cancer. Particles the size of protein molecules can be customized to carry tumour-targeting drugs and destroy cancer cells without harming healthy tissue.

But here’s the problem: when nanoparticles are administered into the body, more than 99 per cent of them become trapped in non-targeted organs, such as the liver and spleen. These nanoparticles are not delivered to the site of action to carry out their intended function.

To solve this problem, researchers at the University of Toronto and the University Health Network have figured out how the liver and spleen trap intact nanoparticles as they move through the organ. “If you want to unlock the promise of nanoparticles, you have to understand and solve the problem of the liver,” says Dr. Ian McGilvray, a transplant surgeon at the Toronto General Hospital and scientist at the Toronto General Research Institute (TGRI).

An Aug. 15, 2016 University of Toronto news release by Luke Ng, which originated the news item, expands on the theme,

In a recent paper in the journal Nature Materials, the researchers say that as nanoparticles move through the liver sinusoid, the flow rate slows down 1,000 times, which increases the interaction of the nanoparticles all of types of liver cells. This was a surprising finding because the current thought is that Kupffer cells, responsible for toxin breakdown in the liver, are the ones that gobbles [sic] up the particles.  This study found that liver B-cells and liver sinusoidal endothelial cells are also involved and that the cell phenotype also matters.

“We know that the liver is the principle organ controlling what gets absorbed by our bodies and what gets filtered out—it governs our everyday biological functions,” says Dr. Kim Tsoi (… [and] research partner Sonya MacParland), a U of T orthopaedic surgery resident, and a first author of the paper, who completed her PhD in biomedical engineering with Warren Chan (IBBME). “But nanoparticle drug delivery is a newer approach and we haven’t had a clear picture of how they interact with the liver—until now.”

Tsoi and MacParland first examined both the speed and location of their engineered nanoparticles as they moved through the liver.

“This gives us a target to focus on,” says MacParland, an immunology post-doctoral fellow at U of T and TGRI. “Knowing the specific cells to modify will allow us to eventually deliver more of the nanoparticles to their intended target, attacking only the pathogens or tumours, while bypassing healthy cells.”

“Many prior studies that have tried to reduce nanomaterial clearance in the liver have focused on the particle design itself,” says Chan. “But our work now gives greater insight into the biological mechanisms underpinning our experimental observations — now we hope to use our fundamental findings to help design nanoparticles that work with the body, rather than against it.”

Here’s a link to and a citation for the paper,

Mechanism of hard-nanomaterial clearance by the liver by Kim M. Tsoi, Sonya A. MacParland, Xue-Zhong Ma, Vinzent N. Spetzler, Juan Echeverri, Ben Ouyang, Saleh M. Fadel, Edward A. Sykes, Nicolas Goldaracena, Johann M. Kaths, John B. Conneely, Benjamin A. Alman, Markus Selzner, Mario A. Ostrowski, Oyedele A. Adeyi, Anton Zilman, Ian D. McGilvray, & Warren C. W. Chan. Nature Materials (2016) doi:10.1038/nmat4718 Published online 15 August 2016

This paper is behind a paywall.

How many nanoparticle-based drugs does it take to kill a cancer tumour? More than 1%

According to an April 27, 2016 news item on Nanowerk researchers at the University of Toronto (Canada) along with their collaborators in the US (Harvard Medical School) and Japan (University of Tokyo) have determined that less than 1% of nanoparticle-based drugs reach their intended destination (Note: A link has been removed),

Targeting cancer cells for destruction while leaving healthy cells alone — that has been the promise of the emerging field of cancer nanomedicine. But a new meta-analysis from U of T’s [University of Toronto] Institute of Biomaterials & Biomedical Engineering (IBBME) indicates that progress so far has been limited and new strategies are needed if the promise is to become reality.

“The amount of research into using engineered nanoparticles to deliver cancer drugs directly to tumours has been growing steadily over the last decade, but there are very few formulations used in patients. The question is why?” says Professor Warren Chan (IBBME, ChemE, MSE), senior author on the review paper published in Nature Reviews Materials (“Analysis of nanoparticle delivery to tumours”). “We felt it was time to look at the field more closely.”

An April 25, 2016 U of T news release, which originated the news item, details the research,

Chan and his co-authors analysed 117 published papers that recorded the delivery efficiency of various nanoparticles to tumours — that is, the percentage of injected nanoparticles that actually reach their intended target. To their surprise, they found that the median value was about 0.7 per cent of injected nanoparticles reaching their targets, and that this number has not changed for the last ten years. “If the nanoparticles do not get delivered to the tumour, they cannot work as designed for many nanomedicines,” says Chan.

Even more surprising was that altering nanoparticles themselves made little difference in the net delivery efficiency. “Researchers have tried different materials and nanoparticle sizes, different surface coatings, different shapes, but all these variations lead to no difference, or only small differences,” says Stefan Wilhelm, a post-doctoral researcher in Chan’s lab and lead author of the paper. “These results suggest that we have to think more about the biology and the mechanisms that are involved in the delivery process rather than just changing characteristics of nanoparticles themselves.”

Wilhelm points out that nanoparticles do have some advantages. Unlike chemotherapy drugs which go everywhere in the body, drugs delivered by nanoparticles accumulate more in some organs and less in others. This can be beneficial: for example, one current treatment uses nanoparticles called liposomes to encapsulate the cancer drug doxorubicin.

This encapsulation reduces the accumulation of doxorubicin in the heart, thereby reducing cardiotoxicity compared with administering the drug on its own.

Unfortunately, the majority of injected nanoparticles, including liposomes, end up in the liver, spleen and kidneys, which is logical since the job of these organs is to clear foreign substances and poisons from the blood. This suggests that in order to prevent nanoparticles from being filtered out of the blood before they reach the target tumour, researchers may have to control the interactions of those organs with nanoparticles.

It may be that there is an optimal particle surface chemistry, size, or shape required to access each type of organ or tissue.  One strategy the authors are pursuing involves engineering nanoparticles that can dynamically respond to conditions in the body by altering their surfaces or other properties, much like proteins do in nature. This may help them to avoid being filtered out by organs such as the liver, but at the same time to have the optimal properties needed to enter tumors.

More generally, the authors argue that, in order to increase nanoparticle delivery efficiency, a systematic and coordinated long-term strategy is necessary. To build a strong foundation for the field of cancer nanomedicine, researchers will need to understand a lot more about the interactions between nanoparticles and the body’s various organs than they do today. To this end, Chan’s lab has developed techniques  to visualize these interactions across whole organs using 3D optical microscopy, a study published in ACS Nano this week.

In addition to this, the team has set up an open online database, called the Cancer Nanomedicine Repository that will enable the collection and analysis of data on nanoparticle delivery efficiency from any study, no matter where it is published. The team has already uploaded the data gathered for the latest paper, but when the database goes live in June, researchers from all over the world will be able to add their data and conduct real-time analysis for their particular area of interest.

“It is a big challenge to collect and find ways to summarize data from a decade of research but this article will be immensely useful to researchers in the field,” says Professor Julie Audet (IBBME), a collaborator on the study.

Wilhelm says there is a long way to go in order to improve the clinical translation of cancer nanomedicines, but he’s optimistic about the results. “From the first publication on liposomes in 1965 to when they were first approved for use in treating cancer, it took 30 years,” he says. “In 2016, we already have a lot of data, so there’s a chance that the translation of new cancer nanomedicines for clinical use could go much faster this time. Our meta-analysis provides a ‘reality’ check of the current state of cancer nanomedicine and identifies the specific areas of research that need to be investigated to ensure that there will be a rapid clinical translation of nanomedicine developments.”

I made time to read this paper,

Analysis of nanoparticle delivery to tumours by Stefan Wilhelm, Anthony J. Tavares, Qin Dai, Seiichi Ohta, Julie Audet, Harold F. Dvorak, & Warren C. W. Chan. Nature Reviews Materials 1, Article number: 16014 (2016  doi:10.1038/natrevmats.2016.14 Published online: 26 April 2016

It appears to be open access.

The paper is pretty accessible but it does require that you have some tolerance for your own ignorance (assuming you’re not an expert in this area) and time. If you have both, you will find a good description of the various pathways scientists believe nanoparticles take to enter a tumour. In short, they’re not quite sure how nanoparticles gain entry. As well, there are discussions of other problems associated with the field such as producing enough nanoparticles for general usage.

More than an analysis, there’s also a proposed plan for future action (from Analysis of nanoparticle delivery to tumours ),

UofT_30yrCancerMedicine

Current research in using nanoparticles in vivo has focused on innovative design and demonstration of utility of these nanosystems for imaging and treating cancer. The poor clinical translation has encouraged the researchers in the field to investigate the effect of nanoparticle design (for example, size, shape and surface chemistry) on its function and behaviour in the body in the past 10 years. From a cancer-targeting perspective, we do not believe that nanoparticles will be successfully translated to human use if the current ‘research paradigm’ of nanoparticle targeting continues because the delivery efficiency is too low. We propose a long-term strategy to increase the delivery efficiency and enable nanoparticles to be translated to patient care in a cost-effective manner from the research stage. A foundation for the field will be built by obtaining a detailed view of nanoparticle–organ interaction during nanoparticle transport to the tumour, using computational strategies to organize and simulate the results and the development of new tools to assess nanoparticle delivery. In addition, we propose that these results should be collected in a central database to allow progress in the field to be monitored and correlations to be established. A 30-year strategy was proposed and seemed to be a reasonable time frame because the first liposome system was reported in 1965 (Ref. 122) and the first liposome formulation (Doxil) was approved by the US Food and Drug Administration (FDA) in 1995 (Refs 91,92). This 30-year time frame may be shortened as a research foundation has already been established but only if the community can parse the immense amount of currently published data. NP, nanoparticle.

Another paper was mentioned in the news release,

Three-Dimensional Optical Mapping of Nanoparticle Distribution in Intact Tissues by Shrey Sindhwani, Abdullah Muhammad Syed, Stefan Wilhelm, Dylan R Glancy, Yih Yang Chen, Michael Dobosz, and Warren C.W. Chan.ACS Nano, Just Accepted Manuscript Publication Date (Web): April 21, 2016 DOI: 10.1021/acsnano.6b01879

Copyright © 2016 American Chemical Society

This paper is behind a paywall.

Finally, Melanie Ward in an April 26, 2016 article for Science News Hub has another approach to describing the research. Oddly, she states this,

However, the study warns about the lack of efficiency despite major economic investments (more than one billion dollars in the US in the past decade).

She’s right; the US has spent more than $1B in the last decade. In fact, they’ve allocated over $1B every year to the National Nanotechnology Initiative (NNI) for almost two decades for a total of more than $20B. You might want to apply some caution when reading. BTW, I think that’s a wise approach for everything you read including the blog postings here.

Shape-shifting nanoparticles for better chemotherapy from the University of Toronto (Canada)

A research team from the University of Toronto and its shape-shifting nanoparticles are being touted in a Feb. 19, 2016 news item on Nanowerk,

Chemotherapy isn’t supposed to make your hair fall out — it’s supposed to kill cancer cells. A new molecular delivery system created at U of T [University of Toronto] Engineering could help ensure that chemotherapy drugs get to their target while minimizing collateral damage.

Many cancer drugs target fast-growing cells. Injected into a patient, they swirl around in the bloodstream acting on fast-growing cells wherever they find them. That includes tumours, but unfortunately also hair follicles, the lining of your digestive system, and your skin.

U of T Engineering Professor Warren Chan has spent the last decade figuring out how to deliver chemotherapy drugs into tumours — and nowhere else. Now his lab has designed a set of nanoparticles attached to strands of DNA that can change shape to gain access to diseased tissue.

A Feb. 18, 2016 University of Toronto news release (also on EurekAlert), which originated the news item, expands on the theme,

“Your body is basically a series of compartments,” says Chan. “Think of it as a giant house with rooms inside. We’re trying to figure out how to get something that’s outside, into one specific room. One has to develop a map and a system that can move through the house where each path to the final room may have different restrictions such as height and width.”

One thing we know about cancer: no two tumours are identical. Early-stage breast cancer, for example, may react differently to a given treatment than pancreatic cancer, or even breast cancer at a more advanced stage. Which particles can get inside which tumours depends on multiple factors such as the particle’s size, shape and surface chemistry.

Chan and his research group have studied how these factors dictate the delivery of small molecules and nanotechnologies to tumours, and have now designed a targeted molecular delivery system that uses modular nanoparticles whose shape, size and chemistry can be altered by the presence of specific DNA sequences.

“We’re making shape-changing nanoparticles,” says Chan. “They’re a series of building blocks, kind of like a LEGO set.” The component pieces can be built into many shapes, with binding sites exposed or hidden. They are designed to respond to biological molecules by changing shape, like a key fitting into a lock.

These shape-shifters are made of minuscule chunks of metal with strands of DNA attached to them. Chan envisions that the nanoparticles will float around harmlessly in the blood stream, until a DNA strand binds to a sequence of DNA known to be a marker for cancer. When this happens, the particle changes shape, then carries out its function: it can target the cancer cells, expose a drug molecule to the cancerous cell, tag the cancerous cells with a signal molecule, or whatever task Chan’s team has designed the nanoparticle to carry out.

“We were inspired by the ability of proteins to alter their conformation — they somehow figure out how to alleviate all these delivery issues inside the body,” says Chan. “Using this idea, we thought, ‘Can we engineer a nanoparticle to function like a protein, but one that can be programmed outside the body with medical capabilities?’”

Applying nanotechnology and materials science to medicine, and particularly to targeted drug delivery, is still a relatively new concept, but one Chan sees as full of promise. The real problem is how to deliver enough of the nanoparticles directly to the cancer to produce an effective treatment.

“Here’s how we look at these problems: it’s like you’re going to Vancouver from Toronto, but no one tells you how to get there, no one gives you a map, or a plane ticket, or a car — that’s where we are in this field,” he says. “The idea of targeting drugs to tumours is like figuring out how to go to Vancouver. It’s a simple concept, but to get there isn’t simple if not enough information is provided.”

“We’ve only scratched the surface of how nanotechnology ‘delivery’ works in the body, so now we’re continuing to explore different details of why and how tumours and other organs allow or block certain things from getting in,” adds Chan.

He and his group plan to apply the delivery system they’ve designed toward personalized nanomedicine — further tailoring their particles to deliver drugs to your precise type of tumour, and nowhere else.

Here are links to and citations for the team’s two published papers,

DNA-controlled dynamic colloidal nanoparticle systems for mediating cellular interaction by Seiichi Ohta, Dylan Glancy, Warren C. W. Chan. Science  19 Feb 2016: Vol. 351, Issue 6275, pp. 841-845 DOI: 10.1126/science.aad4925

Tailoring nanoparticle designs to target cancer based on tumor pathophysiology by Edward A. Sykes, Qin Dai, Christopher D. Sarsons, Juan Chen, Jonathan V. Rocheleau, David M. Hwang, Gang Zheng, David T. Cramb, Kristina D. Rinker, and Warren C. W. Chan. PNAS     doi: 10.1073/pnas.1521265113 published online Feb. 16, 2016.

Both papers are behind paywalls.

Home pregnancy tests inspire simple diagnostics containing gold nanoparticles

PhD student Kyryl Zagorovsky and Professor Warren Chan of the University of Toronto’s Institute of Biomaterials and Biomedical Engineering (IBBME) have created a rapid diagnostic biosensor according to a Feb. 28, 2013 news item on phys.org,

A diagnostic “cocktail” containing a single drop of blood, a dribble of water, and a dose of DNA powder with gold particles could mean rapid diagnosis and treatment of the world’s leading diseases in the near future. …

The recent winner of the NSERC E.W.R. Steacie Memorial Fellowship, Professor Chan and his lab study nanoparticles: in particular, the use of gold particles in sizes so small that they are measured in the nanoscale. Chan and his group are working on custom-designing nanoparticles to target and illuminate cancer cells and tumours, with the potential of one day being able to deliver drugs to cancer cells.

But it’s a study recently published in Angewandte Chemie that’s raising some interesting questions about the future of this relatively new frontier of science.

Zagorovsky’s rapid diagnostic biosensor will allow technicians to test for multiple diseases at one time with one small sample, and with high accuracy and sensitivity. The biosensor relies upon gold particles in much the same vein as your average pregnancy test. With a pregnancy test, gold particles turn the test window red because the particles are linked with an antigen that detects a certain hormone in the urine of a pregnant woman.

(Until now, I’d never thought about how a pregnancy test actually works and always assumed it was similar to a litmus paper test of acid.) The University of Toronto’s Feb. 28, 2013 news release, which originated the news item, describes the technology in more detail,

Currently, scientists can target a particular disease by linking gold particles with DNA strands. When a sample containing the disease gene (e.g., Malaria) is present, it clumps the gold particles, turning the sample blue.

Rather than clumping the particles together, Zagorovsky immerses the gold particles in a DNA-based enzyme solution (DNA-zyme) that, when the disease gene is introduced, ‘snip’ the DNA from the gold particles, turning the sample red.

“It’s like a pair of scissors,” said Zagorovsky. “The target gene activates the scissors that cut the DNA links holding gold particles together.”

The advantage is that far less of the gene needs to be present for the solution to show noticeable colour changes, amplifying detection. A single DNA-zyme can clip up to 600 ‘links’ between the target genes.

Just a single drop from a biological sample such as saliva or blood can potentially be tested in parallel, so that multiple diseases can be tested in one sitting.

But the team has also demonstrated that [it] can transform the testing solution into a powder, making it light and far easier to ship than solutions, which degrade over time. Powder can be stored for years at a time, and offers hope that the technology can be developed into efficient, cheap, over-the-counter tests for diseases such as HIV and malaria for developing countries, where access to portable diagnostics is a necessity. [emphases mine]

I think the fact that the testing solution can be made into powder is exciting news. Medical technologies can be wonderful but if they require special handling and training (e.g., a standard vaccine is in a solution which needs to be refrigerated [that’s expensive in some parts of the world] and someone who is specially trained has to administer the injection) then they’re confined to the few who have access and can afford it.

Here’s a citation and a link to the researchers’ paper,

A Plasmonic DNAzyme Strategy for Point-of-Care Genetic Detection of Infectious Pathogens by Kyryl Zagorovsky, and Dr. Warren C. W. Chan. Angewandte Chemie International Edition DOI: 10.1002/anie.201208715 Article first published online: 10 FEB 2013

Copyright © 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

This article is behind a paywall.

ETA Mar. 1, 2013 10:42 am PST: I made a quick change to the title. Hopefully this one makes more sense than the first one did.

Heat and light signifying much from a new nanoparticle at the University of Toronto

Paraphrasing from Shakespeare’s play MacBeth for this piece is a stretch but I can’t resist. The title comes from the speech MacBeth gives on hearing of his wife’s death (from The Tragedy of MacBeth webpage on the MIT website),

… Out, out, brief candle!
Life’s but a walking shadow, a poor player
That struts and frets his hour upon the stage
And then is heard no more: it is a tale
Told by an idiot, full of sound and fury,
Signifying nothing. [emphasis mine]

Enough of the digression. Scientists at the Princess Margaret Hospital and the University of Toronto, have engineered a nanoparticle that uses light and heat to destroy tumours and light and sound to find and image tumours. From the March 20, 2011 news release on the University of Toronto website,

“In the lab, we combined two naturally occurring molecules (chlorophyll and lipid) to create a unique nanoparticle that shows promise for numerous diverse light-based (biophotonic) applications,” Professor [Gang] Zheng said. “The structure of the nanoparticle, which is like a miniature and colourful water balloon, means it can also be filled with drugs to treat the tumor it is targeting.”

It works this way, explains first author Jonathan Lovell, a doctoral student at IBBME [Institute of Biomaterials & Biomedical Engineering] and OCI [Ontario Cancer Institute]: “Photothermal therapy uses light and heat to destroy tumors. With the nanoparticle’s ability to absorb so much light and accumulate in tumors, a laser can rapidly heat the tumor to a temperature of 60 degrees and destroy it. The nanoparticle can also be used for photoacoustic imaging, which combines light and sound to produce a very high-resolution image that can be used to find and target tumors.”

Here’s what makes this such a breakthrough,

This nanomaterial is also non-toxic, explained Professor Warren Chan of IBBME, another author of the paper. “Jon Lovell and Gang Zheng created a material that doesn’t have metals, [which] means no toxins, but with similar tunable properties to its metal nanostructure brother,” he said. This is the first reported organic nanostructure with such a unique feature, he noted, and so provides a significant opportunity to explore unique designs of organic nanostructures for biomedical applications without concerns regarding toxicity.

I recently mentioned Professor Zheng’s work in the context of a recent funding announcement from the Canadian Space Agency and the Canadian Institutes of Health Research in my March 17, 2011 posting.

If I recall rightly and this is a pretty simple explanation, organic chemistry includes the element of carbon while inorganic excludes it.