<![CDATA[Researchers Receive NIH Funds for Adjuvant Research to Boost Coronavirus Vaccines]]> 27195 Researchers have received funding from the National Institute of Allergy and Infectious Diseases, part of the National Institutes of Health, to screen and evaluate certain molecules known as adjuvants that may improve the ability of coronavirus vaccines to stimulate the immune system and generate appropriate responses necessary to protect the general population against the virus.

“The adjuvants that we are studying, known as pathogen-associated molecular patterns (PAMPs), are molecules often found in viruses and bacteria, and can efficiently stimulate our immune system,” explained Krishnendu Roy, a professor and Robert A. Milton Chair in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University. “Most viruses have several of these molecules in them, and we are trying to mimic that multi-adjuvant structure.”

Adjuvants are used with some vaccines to help them create stronger protective immune responses in persons receiving the vaccine. The research team will screen a library of various adjuvant combinations to quickly identify those that may be most useful to enhance the effects of both protein- and RNA-based coronavirus vaccines under development.

“We are trying to understand how adjuvant combinations affect the vaccine response,” Roy said. “We will look at how the immune system shifts and changes with the adjuvant combinations. The ultimate goal is to determine how to generate the most effective, strongest, and most durable immune response against the virus. There are more than a hundred vaccine candidates being developed for the SARS-CoV-2 virus, which causes COVID-19, and it is likely that many will generate initial antibody responses. It remains to be seen how long those responses will last and whether they can generate appropriate immunological memory that protects against subsequent virus exposures in the long-term.” 

The parent grant to Georgia Tech is part of a program called “Molecular Mechanisms of Combination Adjuvants (MMCA).” For the past four years, the agency has been supporting Roy and his research team to pursue studies to understand how adjuvants work, and this additional funding will allow them to apply their research to potential coronavirus vaccines.

For more coverage of Georgia Tech’s response to the coronavirus pandemic, please visit our Responding to COVID-19 page.

“It has been difficult to develop safe and durable vaccines against respiratory viruses,” explained Roy, who also directs the Center for ImmunoEngineering.  “Over the past several years, we have been looking mostly at the basic science and understanding how the immune system integrates signals from multiple adjuvants to create a unified immune response in mammals. This new funding will allow us to pursue more translational aspects related to COVID-19 and provide the scientific community with potentially new tools to fight this devastating pandemic.”

The team has developed a technique that uses micron- and nanometer-scale polymer particles to present both the vaccine antigen and adjuvant compounds to the mammalian immune system. The medical polymer that is the basis for the particles is used for other purposes in the body. 

The synthetic particles, which Roy’s team calls pathogen-like particles (PLPs), are designed to mimic real pathogens in terms of how they elicit immune responses – without causing infection. “They have an antigen and multiple synergistic adjuvants on a particle-structure that is very similar to how native pathogens present these molecules to our immune system,” he said. 

The PLPs combined with adjuvants encourage the immune system to develop antibodies and T cell responses that can battle the real pathogen if it attacks. Having existing antibodies and the appropriate virus-fighting T cells to the novel coronavirus will enable the body’s immune system to respond quickly to the threat of infection and potentially destroy the virus quickly.

The researchers will first evaluate how the adjuvants affect the interaction of specific immune cells, called dendritic cells and macrophages, with T cells – a key component of generating immune system response – and then follow up with animal studies using the promising combinations. Whether or not a vaccine can be created that will provide long-term protective immunity against the coronavirus is still an open question in the research community, and Roy said the research into adjuvants will help provide new tools to answer that question.

“Part of the knowledge gap right now is that we don’t know how the immune system is influenced by various adjuvants,” he said. “We need to look at how the vaccine formulations, our particles and the adjuvants affect T cell proliferation and T cell response, and how we can optimize that response to generate durable immunity.”

The adjuvant Alum has been used since the 1930s to boost the action of the immune system as it responds to antigens in vaccines that elicit protection against many pathogens. However, for those pathogens that require alternative adjuvants, only a few other adjuvants are currently used in commercial vaccines. Research on modern adjuvants aims to understand the way they specifically activate our immune systems and can be designed to protect against infections. Another approach is to find out if combinations of adjuvants are safe and more effective than a single adjuvant providing highly effective and long-lasting protective immunity.

Roy and his team will be evaluating existing adjuvants in combination, along with potential protein and RNA-based antigens currently under evaluation. The goal is to develop novel combinations of current adjuvants, including adjuvants approved for use and others that are still in development. “In this work, the strategy is to take existing platforms and see how we can pivot them to understand how to make the COVID vaccines better, and do it rapidly.”

As with other research into potential coronavirus vaccines, the work is being accelerated with the goal of creating a safe and effective vaccine against the pandemic virus as soon as possible.

“There are multiple efforts that the NIH and others are funding to really accelerate the pace of the work to see how many different approaches we can come up with and to evaluate the differences,” Roy said. “The goal is to determine what data we can generate very quickly to move toward a successful vaccine that is safe, durable, affordable, scalable, and effective. Evaluating different approaches will help increase the likelihood that we’ll find one or more that meet these criteria.”

This research is supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under supplemental funding to award number U01AI124270. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.


Research News
Georgia Institute of Technology
177 North Avenue
Atlanta, Georgia  30332-0181  USA

Media Relations Contact: John Toon (404-894-6986) (jtoon@gatech.edu)

Writer: John Toon

]]> Colly Mitchell 1 1590079509 2020-05-21 16:45:09 1653584976 2022-05-26 17:09:36 0 0 news Researchers have received funding from the National Institute of Allergy and Infectious Diseases, part of the National Institutes of Health, to screen and evaluate certain molecules known as adjuvants that may improve the ability of coronavirus vaccines to stimulate the immune system and generate appropriate responses necessary to protect the general population against the virus.

]]>
2020-05-20T00:00:00-04:00 2020-05-20T00:00:00-04:00 2020-05-20 00:00:00 John Toon - Research News

]]>
635549 635550 635549 image <![CDATA[Vaccine Vials]]> image/jpeg 1590021017 2020-05-21 00:30:17 1590021017 2020-05-21 00:30:17 635550 image <![CDATA[Krishnendu Roy Vaccine Adjuvants]]> image/jpeg 1590021254 2020-05-21 00:34:14 1590021254 2020-05-21 00:34:14
<![CDATA[Petit Institute Seed Grants Awarded to Three Teams]]> 28153 Three pairs of interdisciplinary researchers have been awarded 2019 Petit Institute Seed Grants.

The program annually pairs two researchers from the Petit Institute as co-principal investigators, providing early stage funding opportunities that serve as a catalyst for bio-related breakthroughs.

The teams and their projects are:

Vinny Agarwal (assistant professor, School of Chemistry and Biochemistry) and Shu Takayama (professor, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory) are working on a project called, “Marine bromine bleach bomb net to fight pseudomonas aeruginosa,” which is an aggressive pathogen that has high antibiotic resistance. Infections caused by P. aeruginosa usually occur in people in the hospital or weakened immune systems, and they can be deadly. Agarwal and Takayma are joining forces to develop new interventions to treat this and other antibiotic resistant pathogens.

Greg Sawicki (associate professor, Woodruff School of Mechanical Engineering) and Tim Cope (Coulter Department of Biomedical Engineering) submitted a project called, “Modifying musculotendon neuromechanics to improve proprioception in aging.” Through their research into understanding the interaction between biological and engineering systems, the Sawicki-Cope team plans to develop a roadmap for designing better exoskeleton controllers that may improve mobility in aging by restoring proprioception.

Gabe Kwong (assistant professor, Coulter Department of Biomedical Engineering) and M.G. Finn (professor, School of Chemistry and Biochemistry) have a project called, “Activity biosensors that implement Boolean logic as precision diagnostics for immunotherapy.” The researchers reason that disease detection and evaluation of treatment responses in vivo depend on the ability to extract clinically useful information from complex biological systems. Noting that previous work in biological computing led to the use of genetic and cell-based tools, they propose that developing programmable biomaterials to perform basic computations, such as Boolean logic, may provide a new framework to increase detection precision and resistance to biological noise.

The Petit Institute Seed Grants provide year-one funding of $50,000 with equivalent year-two funding contingent on submission of an NIH R21/R01 or similar collaborative grant proposal within 12 to 24 months of the year-one start date (July 1, 2019).

]]> Jerry Grillo 1 1562091609 2019-07-02 18:20:09 1562091633 2019-07-02 18:20:33 0 0 news Annual funding program supports diverse range of interdisciplinary research projects

]]>
2019-07-02T00:00:00-04:00 2019-07-02T00:00:00-04:00 2019-07-02 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
622950 622948 622949 622950 image <![CDATA[Vinny Agarwal and Shu Takayama]]> image/jpeg 1562090984 2019-07-02 18:09:44 1562090984 2019-07-02 18:09:44 622948 image <![CDATA[Tim Cope and Greg Sawicki]]> image/jpeg 1562090880 2019-07-02 18:08:00 1562090880 2019-07-02 18:08:00 622949 image <![CDATA[M.G. Finn and Gabe Kwong]]> image/jpeg 1562090929 2019-07-02 18:08:49 1562090929 2019-07-02 18:08:49
<![CDATA[Cracking the Code]]> 28153 Nearly one out of three people in the United States will have cancer during their lifetimes, according to the American Cancer Society.

While a cure remains at large, innovative treatments like immunotherapies, stem cell replacement and gene therapy are advancing quickly.  Screening tests are also playing a role in catching cancer early, so doctors can apply aggressive treatment to send cancer into remission.    

Among those working to change the story on cancer are researchers from the Petit Institute of Bioengineering and Bioscience at Georgia Tech, three of whom are featured in a story from the College of Engineering.

Read about the work of Fatih Sarioglu, Susan Thomas, and Julie Champion right here.

]]> Jerry Grillo 1 1537543625 2018-09-21 15:27:05 1538169431 2018-09-28 21:17:11 0 0 news Petit Institute researchers developing new technologies to battle cancer

]]>
2018-09-21T00:00:00-04:00 2018-09-21T00:00:00-04:00 2018-09-21 00:00:00 405151 405151 image <![CDATA[Fatih Sarioglu in lab]]> image/jpeg 1449254135 2015-12-04 18:35:35 1475895127 2016-10-08 02:52:07
<![CDATA[Rubbing Shoulders with the Giants]]> 28153 Dennis Zhou, a fifth-year BioEngineering Ph.D. student at the Georgia Institute of Technology, has been invited to attend the 68th Lindau Nobel Laureate Meeting, June 24-29, in Lindau, Germany.

Zhou will be among the 600 young scientists (undergraduates, graduate students, and post-doctoral researchers) from across the world sharing the unique atmosphere of the annual event, which brings together more than 40 Nobel Laureates to meet and inspire this next generation of researchers.

“Basically, it’s a chance for us to share our excitement in science,” says Zhou, whose home school is the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University. “I’m pretty overwhelmed and beyond excited about this. I mean, I’ve never met a Nobel Laureate before, so it truly is the opportunity of a lifetime.”

In the lab of Petit Institute researcher Andrés García, Zhou’s research focuses on cell adhesion. Specifically, his studies explore how cells generate forces as they adhere to their environment, “and how these forces are transfused in the signaling pathways within the cell,” he says.

“It’s very basic cell biology, but since adhesion is such an essential process, we hope our results may be applicable in the clinic one day,” Zhou adds. “For example, adhesion is implicated in the disease processes of diseases like cancer and atherosclerosis.”

While the notion of rubbing shoulders with past winners of the Nobel Prize is overwhelming to Zhou, he hasn’t really had time to catch his breath – he’s been busy with research.

“It’ll really sink in over the next few months,” he says. “But Georgia Tech has such a strong history of sending people to this meeting, so I’m going to talk to some of the previous attendees from Tech and learn from their experience.”

 

]]> Jerry Grillo 1 1520719273 2018-03-10 22:01:13 1520882463 2018-03-12 19:21:03 0 0 news BioEngineering/BME grad student Dennis Zhou invited to attend annual meeting of Nobel Laureates

]]>
2018-03-10T00:00:00-05:00 2018-03-10T00:00:00-05:00 2018-03-10 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
603607 603607 image <![CDATA[Dennis Zhou]]> image/jpeg 1520718826 2018-03-10 21:53:46 1520718962 2018-03-10 21:56:02
<![CDATA[Thomas Wins Biomaterials Honor]]> 28153 Susan Thomas, a researcher in the Petit Institute for Bioengineering and Bioscience at the Georgia Institute for Technology, has been selected to receive the 2018 Young Investigator Award from the Society for Biomaterials.

The award is specifically given to recognize an individual who has demonstrated outstanding achievements in the field of biomaterials research within 10 years following his or her terminal degree or formal training.

Thomas, assistant professor in the George W. Woodruff School of Mechanical Engineering, was nominated by fellow Petit Institute researcher Andrés J. García, professor in the Woodruff School.

She’ll be recognized at the 2018 Society for Biomaterials Annual Meeting, held in Atlanta this year (April 11-14), and her research is being considered for publication in the Journal of Biomedical Materials Research or Applied Biomaterials.

The Thomas lab studies the role of fluid transport phenomena in regulating the dynamics and kinetics of cellular and molecular transport processes with the goal of providing novel design principles for targeted drug delivery strategies in disease therapy.

]]> Jerry Grillo 1 1515786042 2018-01-12 19:40:42 1515786076 2018-01-12 19:41:16 0 0 news Petit Institute researcher selected for 2018 Young Investigator Award

]]>
2018-01-12T00:00:00-05:00 2018-01-12T00:00:00-05:00 2018-01-12 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
600755 600755 image <![CDATA[Susan Thomas]]> image/jpeg 1515785825 2018-01-12 19:37:05 1515785825 2018-01-12 19:37:05
<![CDATA[Riding the Next Wave]]> 28153 Krishnendu Roy was very familiar with the news coming out of Philadelphia, about the progress of clinical studies assessing an experimental treatment for leukemia, developed with Novartis Pharmaceuticals by University of Pennsylvania researchers. He knew all about Emily Whitehead, the young girl who was the first patient to test the first engineered cell therapy in history.

Emily’s leukemia is still in remission five years after undergoing a 2012 clinical trial at Children’s Hospital of Philadelphia of the groundbreaking drug by Novartis, Kymriah (tisagenlecleuce). In a global trial, 83 percent of terminally-ill patients went into complete remission. In August of this year, Emily and her family celebrated the U.S. Food and Drug Administration’s (FDA) approval of the revolutionary T-cell therapy for acute lymphocytic leukemia, the world’s first genetically engineered immune therapy.

It’s the kind of epic story that reminds Roy, a researcher at the Petit Institute for Bioengineering and Bioscience at the Georgia Institution of Technology, why he does what he does.

“These patients, like Emily, are incredibly brave,” says Roy, who holds the Robert Milton Chair in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University. “They face the unknown – these potentially risky, potentially promising therapies. At the end of the day, it is for them, for the patients. I think everybody who works in the cellular therapies space, works with that motivation in mind.”

Over the past few years, Roy has taken the lead role in expanding that space at Georgia Tech, where he is director of the Center for ImmunoEngineering, the Marcus Center for Cell-Therapy Characterization and Manufacturing (MC3M), and the recently-established National Science Foundation Engineering Research Center for Cell Manufacturing Technologies (CMaT).

Roy is running point in a widespread endeavor at Georgia Tech to develop cutting-edge cell therapies, and CMaT, which launched this fall, is the latest highlight in a surge of cell therapy-related activity that goes back to the early 1990s, “when we went after a Whitaker Foundation development award and decided the focus should be on tissue engineering – the engineering of replacement tissues using living cells,” notes Bob Nerem, founding director of the Petit Institute.

“We got that award and began our initiative with three initial focus areas,” Nerem adds. “The cardiovascular area, diabetes, and orthopaedic tissue engineering.  Later, we added the neural area, which in many ways has to be considered the holy grail, because there aren’t any viable treatments for many neural issues.”

And that’s the point of cell therapies: they offer a powerful alternative to patients with a dwindling supply of hope.

“We believe cell-based therapies can provide new treatment options," Nerem says. "We feel that’s the future, and we hope to make a major contribution for patients who are running out of options."

 

Collaborative Heft

In September 2017, the National Science Foundation (NSF) announced it was awarding $20 million to a Georgia Tech-led consortium of universities to establish CMaT, which will serve as the catalyst for a new epoch in the evolution of cell therapies. CMaT researchers will collaborate with industry and clinical partners to develop tools and technologies for the consistent, scalable, and affordable production of living therapeutic cells, which could be used to battle cancer, heart disease, autoimmune diseases, and other disorders.

Almost 20 years earlier, NSF sparked cell therapy research with a $12.5 award to establish GTEC – the Georgia Tech/Emory Center for the Engineering of Living Tissues.

“Georgia Tech has been showing great leadership in cell therapy research for a number of years,” says Bob Guldberg, executive director of the Petit Institute and professor in the Woodruff School of Mechanical Engineering. “GTEC really helped build a critical mass of people here doing regenerative medicine research and established Georgia Tech as a national leader in tissue engineering and regenerative medicine.”

GTEC has since evolved into the Regenerative Engineering and Medicine (REM) research center, a collaboration of Tech, Emory University, and the University of Georgia (UGA).

With CMaT, the number of collaborators has grown. In addition to Georgia Tech, major partners include UGA, the University of Wisconsin-Madison, and the University of Puerto Rico (Mayaguez campus), as well as affiliate partners Emory, the Gladstone Institutes, Michigan Technological University, and the University of Pennsylvania.

A collaboration across many disciplines is going to be necessary to tackle the complex challenge of properly manufacturing cell therapies. Georgia Tech seems well-poised to lead such an effort, because of its own multidisciplinary capacity.

“Now we need a broad group of stakeholders to come into play, not only the clinicians and the biomedical and chemical engineers that have traditionally dominated the field,” says Roy, who led the National Cell Manufacturing Consortium – a collaboration of more than 25 companies and 15 academic institutions, along with government agencies and private foundations, that produced a national roadmap for large-scale cell therapy manufacturing.

“We’re bringing in electrical engineers, mechanical engineers, industrial engineers, basic scientists, as well as automation and robotics personnel and experts, data scientists, computational scientists,” Roy adds. “Bring all of that together with policy experts and our natural science programs, and it makes an ideal coalition. That crosstalk between so many disciplines at Georgia Tech makes it an ideal place for an effort like CMaT.”

 

Tech Marks the Spot

In the spring of 2014, the National Institute of Standards and Technology (NIST) awarded a $500,000 advanced technology planning grant to Georgia Tech, funds specifically allotted for creating a national roadmap and consortium targeting cell manufacturing. The NCMC emerged from that, under the direction of Georgia Tech and the Georgia Research Alliance (GRA).

In June 2016 at the White House Organ Summit, the consortium presented its 10-year plan, Technology Roadmap 2025, that basically details the critical stages in the manufacturing pipeline, including cell processing, cell preservation, distribution and handling, quality control, standardization, and workforce development.

Six months before the roadmap’s public unveiling, Georgia Tech was already on the right path. In January 2016, The Marcus Foundation awarded Tech $15.7 million to build a new research center for the development of processes and techniques to ensure the consistent, low-cost, large-scale manufacture of high-quality cells to be used in cell therapies. With additional funding from the GRA and Tech, the $23 million MC3M – the first research facility of its kind – was launched.

“The Marcus Center is already serving as a cell characterization hub for a network of clinical trials around the country,” says Guldberg. “This is a tremendously exciting role, because Georgia Tech will have a lot of the data that will be used to correlate what the important attributes of a cell are that determine whether it’s going to work clinically.”

Initial funding for the MC3M was slated for five years, after which time the center would be expected to support itself with corporate, government, and nonprofit funding. So, through MC3M, Georgia Tech (in partnership with institutions around the state and country) applied for federal funding from NSF to further augment its research and development in cell manufacturing.

With a strategic roadmapping plan in place and funding for a unique cell manufacturing and characterization center secured, Georgia Tech was well positioned to apply for one of the highly competitive NSF Engineering Research Centers. From an initial group of more than 170 proposals nationwide, CMaT was selected as one of just four newly funded centers for 2017.  CMaT is headquartered in the Petit Institute for Bioengineering and Bioscience and will focus on developing enabling technologies as well as the workforce needed by the emerging cell manufacturing industry.   

“I think we’re at a critical juncture in cell manufacturing,” says Johnna Temenoff, Petit Institute researcher, professor in the Coulter Department, and deputy director of CMaT.

“If we do this right, there’s a huge potential,” adds Temenoff, co-director of REM. “The long term goal is to have a large pool of high-quality cells that we can get to people around the world in developed and developing countries.”

This idea of creating affordable new-age medicine for a global population is a major goal for cell therapy and manufacturing researchers like Roy, who notes the high cost of cell therapies. The pioneering Novartis T-cell therapy that cured Emily Whitehead’s cancer is listed at $475,000.

“These treatments can be very expensive, and inaccessible to a majority of the people in the world,” Roy says. “So, the burning question is, how do we bring this to scale and make these therapies cost-effective and available for a broad population across the world, regardless of socioeconomic status? As long as we can develop reproducible product at a much lower cost and achieve manufacturing that is tightly controlled and delivers consistent high-quality cells, we’re going to get the answer.”

 

Homegrown Meds

Cell therapies can come from a couple of different sources. The two most common types of stem cell transplants for cell therapies are autologous and allogeneic. With an autologous transplant, the patient’s own cells are removed, expanded, modified for a therapeutic purpose – finding and attacking cancer, for example. In an allogeneic transplant, the patient receives cells – say, bone marrow or peripheral blood stem cells – from a matching donor, typically a sibling.

“Cells can do many things that a single molecule can’t do,” Roy says. “A cell is a complex entity – a living and breathing entity. They can multiply inside the body, attack and kill certain other cells, like cancer, change the behavior of other cells, like immune cells. These can be extremely powerful drugs. If not harnessed properly, they can be deleterious to a patient as well.”

The side effects with the Novartis drug almost killed Emily Whitehead. These include high fevers, low blood pressure, seizures, liver abnormalities, and heart irregularities. The company and clinicians have developed strategies to manage and minimize the risks.

In the end, for the great majority of patients in the study, the reward was well worth the risks. “Our daughter was going to die, and now she leads a normal life,” Emily’s father, Tom Whitehead, told the FDA panel that endorsed the therapy.

Wilbur Lam is a physician – a hemotologist/oncologist – as well as a researcher in the Petit Institute. He’s seen what happens when standard therapies fail and much prefers having an alternative.

“Cell therapies are the next generation of therapeutics. They offer hope,” says Lam, associate professor in the Coulter Department, and a pediatrician with Children’s Healthcare of Atlanta and the Emory School of Medicine.

His lab has developed a technology in which the patient’s own platelets – the cells that control blood clotting – can be used as a delivery system for drugs. “When it gets to a bleed, it can release its cargo, because the platelet is fine tuned to react to the environment,” Lam says.

It’s a treatment that can be used for patients with hemophilia, or patients who have experienced trauma and are bleeding. “We can also fine tune this system to go in the opposite direction, use it to deliver anti-clotting medications for patients who have heart attacks or strokes,” Lam says. “All of which is enabled by the patient’s own platelets, which act as the brain and muscle, releasing the drug only where it needs to be.”

Petit Institute researcher Melissa Kemp has some personal reasons for her interest and work in cell therapy research, which is based in computational systems biology. Her lab is interested in how intracellular and extracellular environments control the transmission of cellular information, studying living systems using engineering and computational tools, basically looking at complex protein networks the way an electrical engineer might look at a power grid.

“We want to understand how these things are connected together – if you have a failure at one spot, how does that propagate and cause a blackout in another location,” says Kemp, associate professor in the Coulter Department, whose family medical history includes conditions that cell therapies would address.

“Some of the target applications that Georgia Tech researchers have in mind include cardiovascular disease, osteoarthritis, cancer – all of which run in my family,” says Kemp, whose father is a cancer survivor. “So, I’m really excited about the potential of these end applications. The exciting aspect about cellular manufacturing is the ability to really revolutionize medicine in this century.”

 

Watch the Video

]]> Jerry Grillo 1 1512749967 2017-12-08 16:19:27 1513015645 2017-12-11 18:07:25 0 0 news Georgia Tech leading the effort to develop manufacturing expertise and expand cell therapies 

]]>
2017-12-08T00:00:00-05:00 2017-12-08T00:00:00-05:00 2017-12-08 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
599701 599702 599704 596321 592996 391861 446811 599701 image <![CDATA[Cell Therapies - manipulation]]> image/jpeg 1512748229 2017-12-08 15:50:29 1512748229 2017-12-08 15:50:29 599702 image <![CDATA[Krishnendu Roy]]> image/jpeg 1512748339 2017-12-08 15:52:19 1512748339 2017-12-08 15:52:19 599704 image <![CDATA[Bob Guldberg]]> image/jpeg 1512749196 2017-12-08 16:06:36 1512749196 2017-12-08 16:06:36 596321 image <![CDATA[Melissa Kemp, associate professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory]]> image/jpeg 1506027899 2017-09-21 21:04:59 1506027899 2017-09-21 21:04:59 592996 image <![CDATA[Bob Nerem]]> image/jpeg 1498511453 2017-06-26 21:10:53 1498511453 2017-06-26 21:10:53 391861 image <![CDATA[Johnna Temenoff]]> image/jpeg 1449246332 2015-12-04 16:25:32 1475894406 2016-10-08 02:40:06 446811 image <![CDATA[Wilbur Lam and patient]]> image/jpeg 1449256246 2015-12-04 19:10:46 1512765459 2017-12-08 20:37:39
<![CDATA[Boost for Breast Cancer Research]]> 28153 Susan Thomas, a researcher with the Petit Institute for Bioengineering and Bioscience at the Georgia Institute of Technology, is one of is one of three breast cancer researchers from three different Georgia universities to be awarded $50,000 in funding from It’s the Journey and The Georgia Center for Oncology Research and Education (CORE).

Thomas, assistant professor in the Woodruff School of Mechanical Engineering, is researching in collaboration with M.G. Finn, professor and chair of Georgia Tech’s School of Chemistry, a proposed ‘two-stage delivery and release’ drug delivery system with the goal of ultimately eliminating HER2 positive breast tumors. HER2 is a breast cancer that tests positive for a protein called human epidermal growth factor receptor 2 (HER2), which promotes the growth of cancer cells. 

She is a winner of the Rita Schaffer Young Investigator Award from the Biomedical Engineering Society (2013) and the Young Investigator Award from the Society for Biomaterials (2018), and her interdisciplinary research program has been supported by the National Cancer Institute, the Department of Defense, the National Science Foundation, and the Susan G. Komen Foundation, among others.

It’s the Journey and Georgia CORE teamed up to provide $175,000 to recognize creative ideas that may advance progress toward detecting, treating or curing breast cancer. 

In addition to Thomas, the two other $50,000 grant awardees are Mandi Murph, associate professor in the Department of Pharmaceutical and Biomedical Sciences at University of Georgia, and Aneja Ritu, adjunct professor for the Center for Inflammation, Immunity and Infection in the Department of Biology at Georgia State University. Dora Il’yasova, associate professor of epidemiology in Georgia State’s School of Public Health was granted a $25,000 award.

The awards were announced at the end of the Georgia 2-Day Walk for Breast Cancer, Nov. 12. The event, produced annually by It’s the Journey, Inc., founded 15 years ago by breast cancer survivor Randi Passoff. Georgia CORE is an independent non-profit organization (comprised of clinicians, scientists, educations, researchers, and people affected by cancer) that supports clinical research.

 

]]> Jerry Grillo 1 1512147576 2017-12-01 16:59:36 1512147576 2017-12-01 16:59:36 0 0 news Petit Institute researcher Susan Thomas awarded funding from It’s the Journey and Georgia CORE

]]>
2017-12-01T00:00:00-05:00 2017-12-01T00:00:00-05:00 2017-12-01 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
599374 599375 599374 image <![CDATA[Susan Thomas in lab]]> image/jpeg 1512147112 2017-12-01 16:51:52 1512147112 2017-12-01 16:51:52 599375 image <![CDATA[2 Day Walk]]> image/jpeg 1512147209 2017-12-01 16:53:29 1512147209 2017-12-01 16:53:29
<![CDATA[Technology Developed at Petit Institute Gets Test Run]]> 28153 An estimated 120 million people worldwide are infected with lymphatic filariasis, a parasitic, mosquito-borne disease that can cause major swelling and deformity of the legs known as elephantiasis. Health-care workers rely on leg measurements to assess the severity of the condition. However, measuring legs that are severely swollen often proves cumbersome and impractical.

But now, scientists at Washington University School of Medicine in St. Louis, working with collaborators in Sri Lanka, have shown that a portable scanning device, developed in the Petit Institute for Bioengineering and Bioscience at the Georgia Institute of Technology, can measure limb enlargement and disfigurement more quickly and easily in patients with elephantiasis. The research tool makes it easy to obtain accurate measurements and determine whether treatments to reduce swelling are effective.

The study is published this month in the American Journal of Tropical Medicine and Hygiene.

“This is important because it will allow doctors and researchers to take very accurate limb measurements in developing nations, where there are often limited tools to monitor swollen limbs,” said senior author Philip J. Budge, M.D., Ph.D., an assistant professor of medicine in the Division of Infectious Diseases.

In patients with elephantiasis, the parasitic worms that cause the disease make their way into the lymphatic system and prevent the lymph vessels from working properly, which leads to swollen legs. This condition also is referred to as lymphedema.

“Unfortunately, the medication does not usually reverse lymphedema in those already affected,” Budge said. “The ability to get these measurements rapidly will make it much easier to treat patients, including those in clinical trials exploring better treatment therapies.”

The device, created by Atlanta-based LymphaTech, is essentially an infrared sensor, mounted on an iPad, that produces a highly accurate, virtual 3-D reconstruction of the legs using scanning technology similar to that found in Microsoft’s Xbox Kinect video game system.

“The technology was developed in our lab as part of a study funded by the Georgia Research Alliance,” said Petit Institute researcher and LymphaTech co-founder Brandon Dixon, associate professor in the Wallace H. Coulter Department of Biomedical Engineering and the Woodruff School of Mechanical Engineering at Georgia Tech, and LymphaTech’s chief scientific advisor. LymphaTech CEO Mike Weiler, earned his Ph.D. in Bioengineering as a member of Dixon’s lab. Both Dixon and Weiler are authors in this new study, spearheaded by Washington University.

“The study was extremely beneficial to Georgia Tech and LymphaTech’s efforts to develop technology for commercialization in clinical lymphedema monitoring,” Dixon said. “It provided third-party validation of the accuracy of the scanning approach by placing the scanner in the hands its intended clinical users.”

After learning about the technology, Washington University researchers Budge and Ramakrishna Rao, Ph.D., teamed up with international partners to test the device on 52 patients with varying stages of lymphedema at a clinic in Galle, Sri Lanka. Working with physicians at the clinic, the team compared scanner results with results from two other techniques frequently used to ascertain the severity of elephantiasis: use of a tape measure, and water displacement.

Tape measures allow researchers to measure limb circumference near the knees, feet and ankles. However, Budge said, the method can be difficult to standardize and unreliable in assessing leg volume because of bumpy, uneven skin surfaces caused by the swelling.

The water displacement procedure entails patients submerging a leg in a water tank and then measuring how much water is displaced. Each leg is done separately. “This is the gold standard for measuring limb volume, but it is cumbersome and impractical to use in field studies,” Budge said. “Some patients have lymphedema so severe, they have difficulty getting a leg into the water tank or standing still long enough for all the water to drain out. Or they may have open wounds that complicate the process.”

The study showed that the infrared scanner provided measurements of leg volume and of limb circumference at multiple points that were just as accurate and precise as those obtained by tape measure and water displacement. 

“But the most encouraging news is that the scanner produced highly accurate results in only a fraction of the time of the other tests,” Budge said.

Researchers found that the average time required for scanner measurements of both legs was 2.2 minutes. In comparison, the tape measure and water displacement methods took an average of 7.5 minutes and 17.4 minutes, respectively. 

“The scanning tool also offers convenience,” Budge said. “Many patients with swollen limbs often have great difficulty traveling from their homes to the clinic to have their measurements taken. The scanner should make it possible to take extremely accurate limb measurements in the patients’ homes or villages, without cumbersome equipment or inconveniencing patients.”

“To our knowledge, this is the first time that infrared 3-D scanning technology has been used in patients with filarial lymphedema,” Budge said. “It worked so well that it has been added as a measurement tool in future clinical trials in which we are collaborating.”

That study is a two-year, multisite, international clinical trial to determine whether the antibiotic, doxycycline, can reduce the severity of swelling and disfigurement in patients with lymphatic filariasis. Enrollment for Washington University’s partner site in Sri Lanka is scheduled to start this fall.

• • •

Yahathugoda C, Weiler MJ, Rao R, De Silva L, Dixon JB, Weerasooriya MV, Weil GJ, Budge PJ. Use of a Novel Portable Three-Dimensional Scanner to Measure Limb Volume and Circumference in Patients with Filarial Lymphedema. Published online October 9, 2017. DOI: 10.4269/ajtmh.17-0504.

This research was funded by Washington University School of Medicine in St. Louis and the U.S. Agency for International Development.

Disclosures: Co-author Michael J. Weiler is employed by LymphaTech. Weiler and J. Brandon Dixon, a professor of mechanical and biomedical engineering at Georgia Institute of Technology, have an equity stake in the company. 

 

]]> Jerry Grillo 1 1508174246 2017-10-16 17:17:26 1508188789 2017-10-16 21:19:49 0 0 news Portable 3-D scanner developed in lab of Brandon Dixon assesses patients with elephantiasis

]]>
2017-10-16T00:00:00-04:00 2017-10-16T00:00:00-04:00 2017-10-16 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
394511 394511 image <![CDATA[Mike Weiler and Brandon Dixon]]> image/jpeg 1449246346 2015-12-04 16:25:46 1475895110 2016-10-08 02:51:50
<![CDATA[Digging Deeper into RSV]]> 28153 Respiratory syncytial virus (RSV) can affect almost anyone of any age, showing itself like a bad cold in adults and older children. But in younger children, particularly infants, it can become something much worse.

RSV is the most common cause of acute lower respiratory infections in infants and young children globally, often leading to bronchiolitis or pneumonia, sending about 3 million children to the hospital each year. In spite of its prevalence, there is no effective vaccine yet. But researchers at the Georgia Institute of Technology are on the case.

“Treatments and vaccines are currently being investigated, and there might be a vaccine soon, but we really don’t know a lot about the cellular events that occur during RSV,” says Phil Santangelo, associate professor in the Wallace H. Coulter Department of Biomedical Engineering and a researcher with the Petit Institute for Bioengineering and Bioscience.

Santangelo’s lab decided to look closer at RSV, to dig a little deeper,

“We’re imaging the genome of the virus, the guts of it, looking at what happens inside the cell in the hopes of developing new drug targets,” says Santangelo, whose lab’s research was published recently in the journal Nature Communications. “Live cell imaging was the key to this research.”

The research, entitled RSV glycoprotein and genomic RNA dynamics reveal filament assembly prior to the plasma membrane, could lead to the development of antivirals against RSV and other viruses that use the secretory membrane system during assembly.

 

Finding the Unexpected

RSV is a cell membrane-wrapped, single-stranded RNA virus (which is closely related to other RNA viruses, such as measles and mumps) that assembles into viral filaments that can be seen on the outside of the cell.

The researchers utilized live cell imaging (with a major assist from research scientist Aaron Lifland, technical director of the microscopy core facility), as well as protein probes developed in Santangelo’s lab, and bioconjugation techniques –  Lead author Daryll Vanover (a grad student in Santangelo’s lab) used fluorescently-labeled soybean agglutinin to selectively label the RSV G protein (which plays an important role in the assembly of filamentous virions) in living cells. And the results were remarkable, something Santangelo calls, “a mind-blowing event.”

It turns out, most of the viral components needed for filament formation in RSV assemble within the cytosol, not at the plasma membrane.

“The long filamentous structures we see on the outside of the cell, are made inside of the cell,” Santangelo says. “This is not what we expected at all. The dynamics we saw inside the cell are amazing. We’d never seen these structures inside the cell.”

There’s a lot of protein traffic inside of a cell. Future research from the Santangelo team will explore, in a deeper way, what components of the secretory membrane system are critical for specific protein trafficking into the assembly pathway, and applications of this research may lead to the development of new, effective drugs, “small molecules that would inhibit the trafficking and assembly process – assembly inhibitors,” Santangelo says.

“We haven’t seen that class of drugs – that actually inhibit assembly,” he adds. “It would be fantastic if you trap the virus inside the cell. If the virus stays there, it’s going to be degraded.”

In addition to Vanover, Santangelo, and Lifland, authors of the research include biomedical engineering (BME) grad students Emmeline Blanchard and Jonathan Kirschman, BME undergrad Daisy Smith, as well as Eric Alonas (a Santangelo lab alum who earned his Ph.D. last year), and Coulter Department research scientist Chiara Zurla.

 

CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]> Jerry Grillo 1 1507583891 2017-10-09 21:18:11 1507730490 2017-10-11 14:01:30 0 0 news Santangelo lab makes startling discovery in research of common, widespread virus.

]]>
2017-10-09T00:00:00-04:00 2017-10-09T00:00:00-04:00 2017-10-09 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
597142 597142 image <![CDATA[Santangelo and Vanover]]> image/jpeg 1507582187 2017-10-09 20:49:47 1507833219 2017-10-12 18:33:39
<![CDATA[Researchers join the Cancer Systems Biology Consortium with $3.2 Million NCI Grant]]> 27513 The National Cancer Institute (NCI) has awarded Melissa Kemp, associate professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, and a multidisciplinary team of researchers a five year, $3.2 million grant.

 

The researchers aim to identify metabolic features in head and neck cancers that are predictive of tumor response to a new chemotherapeutic drug, ß-lapachone, currently in clinical trial at the University of Texas-Southwestern (UTSW). Fellow leaders of the project are David Boothman, Ph.D., from the UTSW Medical Center and Cristina Furdui, Ph.D., from the Wake Forest School of Medicine.

 

Joshua Lewis, an Emory M.D./BME Bioinformatics Ph.D. student in Kemp’s lab, developed a genome-wide model of metabolism in head and neck cancer that explained why the cytotoxicity to ß-lapachone differed between radiation-sensitive and radiation-resistant cancer cells.

 

The research team identified new molecular targets for enhancing cell death with the drug—validating the results with a 332 gene RNAi screen. The modeling analysis suggests that the radiation-resistant cells rerouted metabolism and altered the enzymatic cycling of ß-lapachone, rendering them more susceptible to the chemotherapy.

 

“I’ve learned through this project how devastating head and neck cancer (HNC) is for patients, and the incidence of HNC is particularly high here in the Southeast compared to the rest of the US,” said Kemp, a researcher with the Petit Institute for Bioengineering and Bioscience at Georgia Tech.

 

“There are very few FDA-approved drugs for HNC and the survival rate for the late-stage cancer patients we are examining has been relatively stagnant for the past three decades," Kemp added. “Our goals are to develop computational models that factor in patient-to-patient variability in HNC metabolism and use these tools to predict who will respond well to the new ß-lapachone therapies.”

 

Head and neck cancers include cancers of the larynx (voice box), throat, lips, mouth, nose, and salivary glands.

 

As part of the award, the researchers will join and participate in the NCI Cancer Systems Biology Consortium. The multidisciplinary Cancer Systems Biology Consortium, funded by the National Cancer Institute, aims to tackle the most perplexing issues in cancer to increase our understanding of tumor biology, treatment options, and patient outcomes.

 

Media Contact:

Walter Rich
Communications Manager
Wallace H. Coulter Department of Biomedical Engineering

]]> Walter Rich 1 1506028078 2017-09-21 21:07:58 1507724142 2017-10-11 12:15:42 0 0 news 2017-09-21T00:00:00-04:00 2017-09-21T00:00:00-04:00 2017-09-21 00:00:00 Walter Rich

]]>
596321 596321 image <![CDATA[Melissa Kemp, associate professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory]]> image/jpeg 1506027899 2017-09-21 21:04:59 1506027899 2017-09-21 21:04:59
<![CDATA[Engineering Research Center Will Help Expand Use of Therapies Based on Living Cells]]> 27561 The National Science Foundation (NSF) has awarded nearly $20 million to a consortium of universities to support a new engineering research center (ERC) that will work closely with industry and clinical partners to develop transformative tools and technologies for the consistent, scalable and low-cost production of high-quality living therapeutic cells. Such cells could be used in a broad range of life-saving medical therapies now emerging from research laboratories.

Led by the Georgia Institute of Technology, the NSF Engineering Research Center for Cell Manufacturing Technologies (CMaT) could help revolutionize the treatment of cancer, heart disease, autoimmune diseases and other disorders by enabling broad use of potentially curative therapies that utilize living cells – such as immune cells and stem cells – as “drugs.” Examples of these highly promising therapies include T cell-based immunotherapies for blood cancers, such as the one developed at the University of Pennsylvania and approved in August by the U.S. Food & Drug Administration, and a gene-modified stem cell therapy recently approved in Europe for a form of the so-called “bubble boy” syndrome.

To facilitate the widespread application of these cutting-edge emerging treatments, CMaT will develop robust and scalable technologies, innovative analytical tools, and engineering systems that will enable industry and clinical facilities to reproducibly manufacture efficient, safe and affordable cell-therapy products. The center, one of four ERCs announced September 12 by the NSF, will also develop improved models for a robust supply chain, storage and distribution system for these therapeutic cell products.

“For over 30 years, NSF Engineering Research Centers have promoted innovation, helped to maintain our competitive edge, and added billions of dollars to the U.S. economy,” said NSF Director France Córdova. “They bring together talented innovators and entrepreneurs with resources from academia, industry and government to produce engineers and engineering systems that solve real-world problems.  I am confident that these new ERCs will strengthen U.S. competitiveness for the next generation and continue our legacy of improving the quality of life for all Americans.”

In addition to the consistent manufacture of  cell-based therapies, the public-private CMaT initiative will also help develop a skilled, diverse and inclusive bio-manufacturing workforce through extensive education and training activities at the K-12, technical college, undergraduate, graduate and postdoctoral levels.

Living cells become “drugs”

“Unlike pharmaceuticals and other products now used in medical treatments, cells are living entities whose properties can significantly change depending on nuances in the way they are grown, stored or otherwise manipulated,” said Krishnendu Roy, director of CMaT and the Robert A. Milton chair professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University. “The center will develop new engineering tools and scalable methods to better characterize, expand, differentiate, separate, transport and store high-quality cells so they provide consistent therapeutic effects, allowing them to be used in standardized therapies by clinicians to serve large numbers of patients worldwide.”

Beyond Georgia Tech, the center will include major partners – the University of Georgia, the University of Wisconsin-Madison and the University of Puerto Rico, Mayaguez Campus – as well as affiliate partners such as the University of Pennsylvania, Emory University, the Gladstone Institutes and Michigan Technological University. Additional international academic partners, as well as industry and the U.S. national laboratories, will also be critical collaborators in the effort.

Moving discoveries into application

“Georgia Tech has a long history of building collaborative partnerships with industry, the national labs and other research universities. With the support of the NSF and this new ERC, we will be able to capitalize on expertise in multiple areas, taking transformative research from the laboratory to practice much more quickly,” said Georgia Tech President G. P. “Bud” Peterson. “The Center for Cell Manufacturing Technologies will also help us educate, train and prepare the workforce in a new industry, thereby continuing to strengthen the U.S. economy.”

Clinical trials have already established the effectiveness of several cell-based therapies and many other trials are underway. But for these exciting therapies to advance into broad healthcare use, the cells will have to be produced in much larger quantities and with more consistent quality than is now available. There are also very few, if any, established industry standards for analytics and processes in cell manufacturing, which hinders consistent production of safe and efficacious cells. Another key limitation identified by industry is the need for a highly-trained workforce.

CMaT would address these barriers through transformative innovations that build upon a series of earlier efforts, including the National Cell Manufacturing Consortium (NCMC) roadmap, infrastructure established at Georgia Tech with support from the Marcus Foundation, quality and other standards programs from the National Institute of Standards and Technology (NIST) and independent industry-led bodies, and translational activities by industry, entrepreneurs and other partners.

The NSF’s multidisciplinary engineering research centers address unique, complex engineering challenges by stimulating knowledge and tech transfer between different sectors, from electronics to energy to infrastructure. Each center takes on a specific engineering research challenge.

“The overall goal of the NSF Engineering Research Centers program is nothing less than to revolutionize engineering research and education in the United States,” said Dawn Tilbury, NSF assistant director for engineering. “We look forward to the exciting advances and outcomes in these important areas.”

Accelerating clinical trials

Beyond established cell-based therapies, the work of CMaT should accelerate the development of new therapies and the testing needed to bring them into the clinic, said Steven Stice, director of the University of Georgia’s Regenerative Bioscience Center (RBC). Regenerative medicine applications could offer new ways of treating diseases for which there are now essentially no treatments, including Parkinson's, Alzheimer’s, heart disease and stroke.

“There are a significant number of cell therapy clinical trials and investments in the field,” Stice said. “But there is little or no investment in a set of consistent standardization methods to optimize how these therapies should work. For instance, we know that cell therapies will improve human health, but right now it’s difficult to guarantee that each dose produced will be as potent as the next. The work done by CMaT researchers will help solve some of these problems.”

The University of Pennsylvania develops cellular therapies and has conducted more than 40 clinical trials of cell-based therapies, including those for engineered T cell therapies and chimeric antigen receptor (CAR) T cells. An example is recently-approved treatment for relapsed and refractory acute lymphoblastic leukemia in pediatric and young adult patients.

“The cell and gene therapy fields are on the cusp of multiple regulatory approvals in the near term,” said Bruce Levine, Barbara and Edward Netter Professor in Cancer Gene Therapy in the Perelman School of Medicine at the University of Pennsylvania. “The challenges ahead lie in developing manufacturing and testing processes incorporating automation that can bring costs down and allow access to more patients.”

Developing broad-based innovations

Critical innovations often occur at the boundaries of disciplines, and CMaT will bring together relevant specialties for both research and workforce development, noted Madeline Torres-Lugo, a professor in the Department of Chemical Engineering at the University of Puerto Rico, Mayaguez Campus.

“Due to the complexity of cells as living organisms, a team with a strong background in biology, chemistry, physics, materials science, and engineering is required for this initiative,” Torres-Lugo said. “Our participation and contribution to CMaT will ensure that Puerto Rico not only remains at the forefront of pharma manufacturing, but also supports cell manufacturing technologies here and around the world by educating highly talented engineering students.”

CMaT testbeds have been selected to address several cell types that are in early stages of clinical adoption or moving toward clinical applications, but it isn't yet clear what cell types will have the greatest therapeutic impacts, noted Sean Palecek, the Milton J. and A. Maude Shoemaker Professor in chemical and biological engineering at the University of Wisconsin-Madison. Therefore, one of the center’s challenges will be to ensure that fundamental discoveries, and tool and technology development efforts, will apply to multiple cell types.

“Our work will provide safer and more potent cell products that will allow clinical studies to establish the effectiveness of these cells as therapeutics,” Palecek said. “In addition, our work on scaling cell production will enable manufacturing of sufficient numbers of cells to replace damaged organs, such as the loss of heart muscle after a heart attack, at a cost that makes these therapies accessible to broad segments of society. We will also train the future leaders of the emerging therapeutic cell manufacturing industry. These students and their work establishing this industry will be the most significant impact of CMaT.”

New centers among 19 ERCs

Since the program’s inception in 1985, NSF has funded a total of 74 ERCs and will support 19 in this fiscal year, including four new centers. Each center receives NSF funding for up to 10 years. During this time, centers build partnerships with industry, universities and other government agencies that will sustain them for years to come.

In May, the National Academies published a report, “A new vision for center-based engineering research,” which was the result of an NSF-funded study to examine the future of the NSF ERC program.

The report identifies and recommends strategies to enable NSF multidisciplinary engineering research centers to continue addressing key research, education and innovation needs of the United States in a changing global context.

“ERCs are widely known as outstanding examples of successful partnerships between universities, private industry and government that have made significant contributions to address national challenges,” said Don Millard, acting division director for the NSF Division of Engineering Education and Centers. “We are continually working with the scientific and engineering communities, as well as private industry and government partners, to ensure NSF-funded centers and grantees are best-equipped to match societal needs with research abilities.”

 

Research News

Georgia Institute of Technology

177 North Avenue

Atlanta, Georgia  30332-0181  USA

 

Media Relations Contacts: John Toon (404-894-6986) (jtoon@gatech.edu) or Ben Brumfield (404-660-1408) (ben.brumfield@comm.gatech.edu).

Writer: John Toon

]]> Angela Ayers 1 1505238773 2017-09-12 17:52:53 1507554309 2017-10-09 13:05:09 0 0 news The National Science Foundation (NSF) has awarded nearly $20 million to a consortium of universities to support a new engineering research center (ERC) that will work closely with industry and clinical partners to develop transformative tools and technologies for the consistent, scalable and low-cost production of high-quality living therapeutic cells. 

]]>
2017-09-12T00:00:00-04:00 2017-09-12T00:00:00-04:00 2017-09-12 00:00:00 John Toon

Research News

(404) 894-6986

]]>
595805 595806 595807 595808 595809 595805 image <![CDATA[Cell manufacturing lab]]> image/jpeg 1505149092 2017-09-11 16:58:12 1505149092 2017-09-11 16:58:12 595806 image <![CDATA[Cell manufacturing lab2]]> image/jpeg 1505149268 2017-09-11 17:01:08 1505149268 2017-09-11 17:01:08 595807 image <![CDATA[Krishnendu Roy, director of CMaT]]> image/jpeg 1505149393 2017-09-11 17:03:13 1505149393 2017-09-11 17:03:13 595808 image <![CDATA[Human fibroblast cells]]> image/jpeg 1505149532 2017-09-11 17:05:32 1505149532 2017-09-11 17:05:32 595809 image <![CDATA[Cell bioreactor]]> image/jpeg 1505149639 2017-09-11 17:07:19 1505149639 2017-09-11 17:07:19
<![CDATA[Training the Next Generation]]> 28153 A new five-year grant from the National Institutes of Health (NIH) will help the Georgia Institute of Technology train the next generation of leaders in ImmunoEngineering – a new wave of researchers applying the tools and principles of engineering to study the immune system in health and disease in the quest for breakthrough solutions to improve the lives of patients.

The NIH T32 grant, entitled “Research Training Program in ImmunoEngineering,” starts this month and will support five biomedical engineering and bioengineering doctoral students this academic year, and new cohorts of students in subsequent years.

“There’s a lot of enthusiasm for this program,” says Julia Babensee, director of the training program, associate professor in the Wallace H. Coulter Department of Biomedical Engineering and a researcher in the Petit Institute for Bioengineering and Bioscience.

And it may be the first of its kind.

“NIH didn’t have a training grant program in this space before,” says Babensee, who applied for the grant and is managing the program. “It’s the first ImmunoEngineering training grant that we know of. NIH recognizes that this is an important, emerging discipline.”

The five trainees selected for 2017-2018 are Nicholas Beskid (from Babensee’s lab), David Francis (from the lab of Susan Thomas), Midori Maeda (from the lab of Shuichi Takayama), Katily Ramirez (from the lab of Todd Sulcheck), Cory Sago (from the lab of James Dahlman). Another trainee, Jeff Noble (from the lab of M.G. Finn) deferred to 2018-2019.

The program’s co-directors, Susan Thomas (Petit Institute researcher, assistant professor in the Woodruff School of Mechanical Engineering at Georgia Tech) and Rafi Ahmed (director of the Emory Vaccine Center) will leverage the $191,090 award to prepare engineering students for advanced careers in immunoengineering.

Competition for the inaugural slots was stiff, according to Babensee, who is on the executive committee of the Center for ImmunoEngineering at Georgia Tech. That research center, based in the Petit Institute, brings together multi-disciplinary researchers, engineers and scientists, to develop solutions for patients battling cancer, infectious diseases (HIV, hepatitis, etc.), autoimmune and inflammatory disorders (diabetes, multiple sclerosis, arthritis, etc.), as well as those undergoing regenerative therapies (organ transplantation, spinal cord injury, etc.).

“These trainees will have the ability to tackle those complex problems, having both an engineering approach and a deep understanding of immunology,” Babensee says. “Our intention is to train people who will be able to take on leadership positions in either academia or industry. This is a prestigious designation from NIH – it says a lot about Georgia Tech and Emory, about our faculty and students.”

 

CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]> Jerry Grillo 1 1507054325 2017-10-03 18:12:05 1507550413 2017-10-09 12:00:13 0 0 news Georgia Tech wins NIH grant to develop new wave of ImmunoEngineers

]]>
2017-10-03T00:00:00-04:00 2017-10-03T00:00:00-04:00 2017-10-03 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
596894 596894 image <![CDATA[ImmunoEngineering Grant]]> image/jpeg 1507054105 2017-10-03 18:08:25 1507054105 2017-10-03 18:08:25
<![CDATA[First Possible Drug Treatment for Lymphedema]]> 28153 Tracey Campbell has lived for seven years with lymphedema, a chronic condition that causes unsightly swelling in her left leg.

The disease, which stems from a damaged lymphatic system, can lead to infections, disfigurement, debilitating pain and disability. There is no cure. The only available treatment is to wear compression garments or use massage to suppress the swelling, which can occur throughout the body in some cases. Campbell — who had two quarts of excess water in her left leg by the time she was diagnosed — has for years worn restrictive garments 24 hours a day and has spent an hour each night massaging the lymph fluid out of her leg. 

Lymphedema is uncomfortable, exhausting and dangerous if left uncontrolled. As many as 10 million Americans and hundreds of millions of people worldwide suffer from the condition, many from the after-effects of cancer therapy treatments. 

“There’s this extra layer of emotional burden,” said Campbell, who added that she has to be constantly vigilant to protect against infection. “All you want to be is normal.”

Now there’s new hope for a possible pharmaceutical treatment for patients like Campbell. A study led by scientists at the Stanford University School of Medicine has uncovered for the first time the molecular mechanism responsible for triggering lymphedema, as well as a drug with the potential for inhibiting that process. Contributing to the study was the lab of Brandon Dixon, researcher with the Petit Institute for Bioengineering and Bioscience at Georgia Tech.

“Our main role was to provide the functional imaging of the lymphatics that showed that the therapeutic directly resulted in improved lymphatic function,” said Dixon, associate professor in the Woodruff School of Mechanical Engineering, and one of the study’s co-authors.

The study was published May 10 in Science Translational Medicine.

“We figured out that the biology behind what has been historically deemed the irreversible process of lymphedema is, in fact, reversible if you can turn the molecular machinery around,” said Stanley Rockson, MD, professor of cardiovascular medicine and the Allan and Tina Neill Professor of Lymphatic Research and Medicine at Stanford. Rockson shares senior authorship of the study with Mark Nicolls, MD, professor of pulmonary and critical care medicine. Stanford research scientists Wen “Amy” Tian, PhD, and Xinguo Jiang, MD, PhD, share lead authorship of the study and are also affiliated with the Veterans Affairs Palo Alto Health Care System.

 

‘Fundamental new discovery’

“This is a fundamental new discovery,” said Nicolls, who is also a researcher at the VA Palo Alto.

The researchers found that the buildup of lymph fluid is actually an inflammatory response within the tissue of the skin, not merely a “plumbing” problem within the lymphatic system, as previously thought.

Working in the lab, scientists discovered that a naturally occurring inflammatory substance known as leukotriene B4, or LTB4, is elevated in both animal models of lymphedema and in humans with the disease, and that at elevated levels it causes tissue inflammation and impaired lymphatic function.

Further research in mice showed that by using pharmacological agents to target LTB4, scientists were able to induce lymphatic repair and reversal of the disease processes.

“There is currently no drug treatment for lymphedema,” Tian said. Based on results of the study, the drug bestatin, which is not approved for use in the United States but which has been used for decades in Japan to treat cancer, was found to work well as an LTB4 inhibitor, with no side effects, she said.

Based on the research, bestatin (also known as ubenimex), is being tested in a clinical trial that started in May 2016 — known as ULTRA — as a treatment for secondary lymphedema, which occurs because of damage to the lymphatic system from surgery, radiation therapy, trauma or infection. Primary lymphedema, on the other hand, is hereditary. The results of the research pertain to both types.

Rockson is principal investigator for this multisite phase-2 clinical trial.

“The cool thing about this story — which you almost never see — is that a clinical trial testing the therapy has already started before the basic research was even published,” Nicolls said. “This is the first pharmaceutical company-sponsored trial for a medical treatment of lymphedema, a condition that affects millions.” 

Nicolls and Tian are co-founders of Eiccose LLC. Eiccose is now part of Eiger BioPharmaceuticals, which gets the drug from Nippon Kayaku in Japan. Eiger is sponsoring the clinical trial. Nicolls and Rockson are both scientific advisers to the company.

 

Two labs, two diseases

The study, which got underway about four years ago, began somewhat uniquely as a collaboration between two labs that were studying two completely different diseases. At the time, the Nicolls lab, where Tian works, was studying pulmonary hypertension. The Rockson lab was conducting lymphedema research. 

The two teams met through SPARK, a Stanford program designed to help scientists translate biomedical research into treatments for patients. 

“I was in a privileged position of seeing two faculty conducting important research and recognizing the possible link in causality,” said Kevin Grimes, MD, associate professor of chemical and systems biology and co-founder of SPARK. “It occurred to me that both diseases affected vascular tissues and had strong inflammatory components.” 

“He blind-dated us,” Nicolls said. “When Amy Tian and I looked at the data from Stan’s research, Amy said, ‘It looks like it could be the same molecular process.’”

“It was an arranged marriage between us and Stan which worked out great,” Tian said. 

At the time, Rockson had begun to suspect that lymphedema was an inflammatory disease. This led to his team’s discovery that the anti-inflammatory drug ketoprofen successfully helped to relieve lymphedema symptoms, although it wasn’t a perfect drug; side effects were a concern, and it remained unclear how the drug worked at the molecular level.

Meanwhile, the Nicolls lab had discovered that LTB4 was part of the cycle of inflammation and injury that keeps pulmonary hypertension progressing. When researchers blocked LTB4 in rats with the disease, their symptoms lessened and blood vessels became less clogged, lowering blood pressure in the lungs. 

“When we became aware of Mark’s work, we began to realize that we were both possibly dealing with the activation of steps downstream of the 5-LO [5-lipoxygenase] pathway,” Rockson said. “This became intriguing and formed the basis of our relationship.”

 

Joining forces

The two teams joined forces to figure out the mechanism that triggered lymphedema, hopefully revealing a target for drug treatment in humans. After determining that ketoprofen was primarily working on the 5-LO pathway, the researchers began blocking the various endpoint pathways after 5-LO activation in mouse models of lymphedema, Rockson said.

“It turned out that, in fact, we were both dealing with the same branch, which is LTB4,” Rockson said.

“So now it became clear we really were dealing with a very similar biological process in two different diseases,” he said. “Because of Mark’s work in pulmonary hypertension, we knew that we had an ideal form of therapy that we could try in lymphedema as well.”

The Nicolls lab had used the drug bestatin, which blocks the enzyme that generates LTB4, to reverse pulmonary hypertension disease processes. When researchers tested bestatin in the mouse lymphedema model, it worked to reverse symptoms of that disease.

“I’m still in awe,” Rockson said. “There are few situations where you take a problem at the bedside, and go into the lab, and then take discoveries back to the bedside. It’s amazingly gratifying.”

Campbell, who is now participating in the double-blinded, placebo-controlled bestatin trial at Stanford, remains hopeful.

“When all of the sudden one of your limbs begins to swell, you want to understand what the heck is going on,” she said. “It’s a tough condition that few people seem to care about, even though millions and millions suffer with it. We’re hoping for something that gives some relief.”

In addition to Stanford and Georgia Tech, researchers from Virginia Commonwealth University, the University of Michigan Health Systems and the University of Illinois at Chicago are also co-authors.

 

CONTACT

Tracie White, Stanford University

traciew@stanford.edu

 

]]> Jerry Grillo 1 1494438800 2017-05-10 17:53:20 1498248782 2017-06-23 20:13:02 0 0 news Petit Institute researcher Brandon Dixon contributes to groundbreaking Stanford-led study

]]>
2017-05-10T00:00:00-04:00 2017-05-10T00:00:00-04:00 2017-05-10 00:00:00 176201 176201 image <![CDATA[Brandon Dixon]]> image/jpeg 1449179022 2015-12-03 21:43:42 1494438878 2017-05-10 17:54:38
<![CDATA[A Joint Effort]]> 28153 Johnna Temenoff is only jesting a little when she describes her lab’s recent collaboration with two other labs at the Petit Institute for Bioengineering and Bioscience.

“This was very much, pardon the pun, a joint effort,” Temenoff says about the research, which demonstrates for the first time the degenerative effects of tendon overuse (tendinopathy) on surrounding tissues in the shoulder joint.

The Temenoff team worked with the labs of Manu Platt and Robert Guldberg, resulting in a research article recently published in the Journal of Orthopaedic Research, entitled “Supraspinatus Tendon Overuse Results in Degenerative Changes to Tendon Insertion Region and Adjacent Humeral Cartilage in a Rat Model.”

It’s a partnership that could lead, down the road, to new therapeutics and preventive medicine for people with shoulder injuries – “athletes, or quite honestly, anyone, particularly people who do a lot of overhead reaching,” says Temenoff, professor in the Wallace H. Coulter Department of Biomedical Engineering and co-director of the center for Regenerative Engineering and Medicine (a partnership with Emory University and the University of Georgia).

“We wanted to understand how tissues degenerate, particularly the supraspinatus, one of the major rotator cuff tendons,” Temenoff adds. “So we paired with Dr. Platt’s lab to better understand and characterize the enzymes that were present at various stages.”

In previous work, Temenoff and her research partners analyzed torn rotator cuff (supraspinatus) tendon tissue that had been damaged from overuse for the presence of proteases (an enzyme that breaks down proteins and peptides), and also examined structural damage changes in rats, where the tendon meets the bone. They saw more degeneration in the area close to the bone and cartilage, rather than where the tendon enters into muscle tissue.

“Our work has really been trying to demonstrate how members of this class of enzymes are involved in more tissue destructive diseases than are being investigated,” says Platt, associate professor in the Coulter Department. 

“There have also been major pushes by pharmaceutical companies to develop inhibitors to block these enzyme’s activities,” he adds. “They keep failing in clinical trials due to side effects, not efficacy, indicating their importance in the disease progression, but also in many regulatory functions that still need to be understood.”

In the most recent study, the researchers wanted to focus just on the area of the humeral head – where the tendon inserts into the bone and the articular cartilage that covers the head. What they found confirmed some suspicions, showing degeneration in multiple tissues adjacent to the humeral head – in both tendon and cartilage – as a result of an overuse protocol.

“Indeed, we found damage in both places,” Temenoff says. “Now we have a better idea of the enzyme activity in the tendon over time. Going forward, we have a better of understanding of what enzymes to target and what tissues might need to be targeted for some effective therapies.”

This is the first confirmation showing that overuse injury in the shoulder tendon could damage the adjacent cartilage. 

The Guldberg lab employed its expertise in micro computed tomography (microCT) to assess the damage to the articular cartilage, “and it showed that tendon overuse resulted in significant changes in the joint surface, consistent with the early stages of osteoarthritis,” says Guldberg, executive director of the Petit Institute and professor in the Woodruff School of Mechanical Engineering.

The research, funded by the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) of the National Institutes of Health (NIH), offers a new, broader view of rotator cuff disease.

“We and others are starting to think of it as a disease of the entire joint rather than just the tendons,” Temenoff says. “The aim is to prevent further damage. Of course, over the longer term we’d also love to be able to regenerate what’s been lost.”

The findings suggest a necessity to treat both the tendon and nearby cartilage to slow or reverse tissue damage during overuse injuries. 

“It’s important to let clinicans know that they should monitor this because they may have patients that might be putting themselves at risk for a total shoulder replacement,” Temenoff says.

Lead author of the paper is Akia Parks, a biomedical engineering graduate student who is based in the Platt lab. In addition to Guldberg, Platt, and Temenoff, her co-authors include Jennifer McFaline-Figueroa (research technician in the Temenoff lab), and BME undergrads Anne Coogan and Emma Poe-Yamagata. 

Parks, whose studies are supported by the NIH’s Cellular and Tissue Engineering (CTEng) grant, served as a critical human link, straddling different research areas and exemplifying the multidisciplinary approach that is emblematic of the Petit Institute.

“Akia has been a great bridge between the Platt and Temenoff labs by interfacing with the enzymology/biochemistry from our lab with the tendon structure, remodeling, and mechanical engineering insights of the Temenoff lab,” says Platt. “She is a great example of the education and preparation these scholars receive to communicate across a number of disciplines.” 

 

LINKS:

"Supraspinatus tendon overuse results in degenerative changes to tendon insertion region and ajacent humeral cartilage in a rat model"

 

CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]> Jerry Grillo 1 1486556791 2017-02-08 12:26:31 1494869261 2017-05-15 17:27:41 0 0 news Trio of Petit Institute labs link tendon overuse injury to degenerative changes in shoulder cartilage

]]>
2017-02-08T00:00:00-05:00 2017-02-08T00:00:00-05:00 2017-02-08 00:00:00  

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
587114 319751 587114 image <![CDATA[Shoulder injury]]> image/jpeg 1486555594 2017-02-08 12:06:34 1486555594 2017-02-08 12:06:34 319751 image <![CDATA[Johnna Temenoff, PhD - Director of GTBioMAT program, associate professor in the Wallace H. Coulter Department of Biomedical Engineering]]> image/jpeg 1449244997 2015-12-04 16:03:17 1475895029 2016-10-08 02:50:29
<![CDATA[Self-Repaired Eyesight]]> 28153 Researchers with the Regenerative Engineering and Medicine research center (REM) have developed a new way to identify and sort stem cells that may one day allow clinicians to restore vision to people with damaged corneas using the patient’s own eye tissue. They published their findings in Biophysical Journal.

The cornea is a transparent layer of tissue covering the front of the eye, and its health is maintained by a group of cells called limbal stem cells. But when these cells are damaged by trauma or disease, the cornea loses its ability to self-repair.

“Damage to the limbus, which is where the clear part of the eye meets the white part of the eye, can cause the cornea to break down very rapidly,” said James Lauderdale, an associate professor of cellular biology in the University of Georgia's (UGA) Franklin College of Arts and Sciences and paper co-author. “The only way to repair the cornea right now is do a limbal cell transplant from donated tissue.”

In their study, researchers used a new type of highly sensitive atomic force microscopy, or AFM, to analyze eye cell cultures. Created by Todd Sulchek, a researcher at the Petit Institute for Bioengineering and Bioscience and an associate professor in the George W. Woodruff School of Mechanical Engineering at Georgia Tech, the technique allowed researchers to probe and exert force on individual cells to learn more about the cell’s overall health and its ability to turn into different types of mature cells. 

Read the complete story here.

]]> Jerry Grillo 1 1489756309 2017-03-17 13:11:49 1494869105 2017-05-15 17:25:05 0 0 news Stem cell treatment may restore vision to patients with damaged corneas

]]>
2017-03-17T00:00:00-04:00 2017-03-17T00:00:00-04:00 2017-03-17 00:00:00 588598 588598 image <![CDATA[Todd Sulchek and microfluidic device]]> image/jpeg 1489179074 2017-03-10 20:51:14 1489179074 2017-03-10 20:51:14
<![CDATA[The Perfect Patient]]> 28153 Robert Mannino was curious about his disease, beta thalassemia. He wanted to study it. And that’s exactly what he’s been doing for the past six years at the Georgia Institute of Technology and Emory University.

Now a grad student, Mannino conducts his work in the lab of Petit Institute researcher Wilbur Lam, assistant professor in the Wallace H. Coulter Department of Biomedical Engineering.

And he’s making great progress.

Often serving as his own test subject, Mannino is the perfect combination of patient and researcher as he continues developing innovative solutions, including an app that can monitor hemoglobin levels.

Read the whole story here.

]]> Jerry Grillo 1 1492540455 2017-04-18 18:34:15 1494869070 2017-05-15 17:24:30 0 0 news Rob Mannino is poised to advance the field of mobile health technology

]]>
2017-04-18T00:00:00-04:00 2017-04-18T00:00:00-04:00 2017-04-18 00:00:00 590583 590583 image <![CDATA[Lam and Mannino]]> image/jpeg 1492540244 2017-04-18 18:30:44 1492540244 2017-04-18 18:30:44
<![CDATA[Exosomes Have a Sense of Urgency]]> 28153 Exosomes, tiny vesicles smaller than red blood cells, were once thought of as molecular trash bins. And it’s true, these nanoparticles do carry off a cell’s discarded material.

But that disposable payload, which can include mRNAs and proteins, can be picked up by other cells, which means that exosomes play an important role as messengers, helping to carry out the critical cell-to-cell communication that multicellular organisms depend on for survival.

Not only can exosomes communicate and provide transport over long distances – they also happen to be in the ideal size range for lymphatic transport, a concept that has long captivated Brandon Dixon and Fred Vannberg, researchers in the Petit Institute for Bioengineering and Bioscience, and others interested in the future of the lymphatic targeted drug delivery systems.

Last year, Dixon and Vannberg collaborated on a groundbreaking research paper in the Nature journal, Scientific Reports, entitled, “Lymphatic transport of exosomes as a rapid route of information dissemination to the lymph node.” Their results suggested that exosomes facilitate the rapid exchange of infection-specific information from peripheral tissue to the lymph node, essentially priming the node for an effective innate immune response.

Their latest paper, “TLR-exosomes exhibit distinct kinetics and effector function,” published recently in the same journal (Scientific Reports, March 2017), digs deeper, making a striking new discovery along the way: Exosomes move with what looks like an increased sense of urgency depending on their payload.

“Not only do we find out that these exosomes can inform the node of what kind of specific immune response to initiate – is it viral, or a bacterial infection? It’s that specific – but we found out the uptake of exosomes from viral-infected cells was different from the control exosomes,” says Vannberg, an assistant professor in the School of Biological Sciences.

“They move much faster,” notes Dixon, associate professor in the Woodruff School of Mechanical Engineering. “It was really dramatic. Their uptake to the node was a lot quicker when they contained pathogen information. This is a completely novel finding. Something on the surface of the exosomes has to be communicating with the micro-environment to enhance lymphatic transport, but we really don’t know why this happens yet.”

The researchers demonstrated the enhanced (if unexpected) trafficking of pathogenic-stimulated exosomes, which also have an inclination to recruit infection-fighting neutrophils (white blood cells) along the way.

So after encountering, say, a virus on a peripheral tissue, the exosome acts in a couple of ways, sending the information rapidly across long distances to the lymph node, then bringing the molecular cavalry.

The lead author of the latest paper was biology Ph.D. candidate Swetha Srinivasan, who was lead author on the last paper and is co-advised by Dixon and Vannberg. Other authors included grad students James Moore and Shashidhar Ravishankar, and undergrads Michelle Su and Pamelasara Head.

The researchers say having cutting-edge core facilities close at hand, within the Petit Institute, was critical to the work (giving a shout-out to research scientist Shweta Biliya, for her management of the High Throughput DNA Sequencing Core in the acknowledgements section of the paper).

The team’s work, partially funded by an interdisciplinary Petit Institute seed grant, produced important findings on the road to targeted therapy. But the findings lead to the inevitable sequel.

“In the next chapter, we’ll talk about immunotherapy,” Dixon says. “These results suggest we can go beyond targeting and enhance the transportation itself. Whether that is a way to improve, say, vaccine efficacy or drug delivery to the lymph nodes for tumor therapy remains to be seen. But those are the avenues where these results can have important implications.”

 

LINKS:

“TLR-exosomes exhibit distinct kinetics and effector function”

Vannberg lab

Laboratory of Lymphatic Biology and Bioengineering 

 

CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]> Jerry Grillo 1 1493085136 2017-04-25 01:52:16 1494869003 2017-05-15 17:23:23 0 0 news New research from Dixon and Vannberg labs illuminate critical role of courier nanoparticles

]]>
2017-04-24T00:00:00-04:00 2017-04-24T00:00:00-04:00 2017-04-24 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
590873 302161 590869 590873 image <![CDATA[Lymphatics]]> image/jpeg 1493125322 2017-04-25 13:02:02 1493125322 2017-04-25 13:02:02 302161 image <![CDATA[Fred Vannberg]]> image/jpeg 1449244592 2015-12-04 15:56:32 1493147592 2017-04-25 19:13:12 590869 image <![CDATA[Brandon Dixon]]> image/jpeg 1493086001 2017-04-25 02:06:41 1493086001 2017-04-25 02:06:41
<![CDATA[Fighting the Good Fight]]> 28153 The war on cancer is 45 years old. And while there have been some significant advances since passage of the National Cancer Act in 1971, the conflict has spread out along many fronts.

With the realization now that there are more than 200 types and subtypes of cancer, the battle plan has evolved from a one-size-fits-all strategy to a data-driven, more personalized approach, which means the army of researchers and clinicians devoted to fighting cancer also has evolved.

“We’re seeing the emergence of the new cancer biology,” says John McDonald, director of the Integrated Cancer Research Center (ICRC) at the Georgia Institute of Technology. “It’s actually being driven now by technologies and expertise that lie outside the traditional framework of cancer biology. That’s why I think you’re probably going to see major breakthroughs in cancer research coming out of places like Georgia Tech and M.I.T., as opposed to traditional medical schools.”

Advances in genomics and high throughput sequencing have generated massive amounts of data, “and it’s opened up the field to people that were not trained as cancer biologists, but have the necessary skillsets for the analysis of all this new, big data,” says McDonald, a faculty researcher with the Petit Institute for Bioengineering and Bioscience and professor in the School of Biological Sciences, who has definitely seen his share of breakthroughs in his own recent research focused on ovarian cancer.

The cancer biology that McDonald knew when he was a college student has moved from an era of specialization into an era of multidisciplinary research, in which researchers from a wide range of areas now work together on common projects.

“Twenty five years ago, these people probably wouldn’t have spoken to each other because they didn’t have any common interests,” says McDonald. “I was like a kid in a candy store when we first came to Georgia Tech, and it still feels like that – the idea of being in a place where all of this expertise and creativity exist. Cancer research is not a one-person endeavor. It’s all about collaboration.”

And McDonald has plenty of collaborators within and beyond the ICRC, which occupies a busy space where molecular biology, computational science, engineering and nanotechnology converge. Together, these scientists and engineers are developing next generation cancer diagnostics and therapeutics.

 

Family Affair

Fatih Sarioglu trained as an electrical engineer in his native Turkey and later at Stanford University, developing particular expertise in microsystems and nanosystems, developing sensitive, small-scale devices to look at atoms. After earning his Ph.D., he says, “I wondered how I could use these skills to benefit humanity.”

Sarioglu, assistant professor in the School of Electrical and Computer Engineering and a Petit Institute faculty researcher, he spent three years as a post-doc at Massachusetts General Hospital and Harvard Medical School, learning about cancer. He found his opportunity, “to give biologists and biomedical scientists and clinicians capabilities they don’t have.”

There was a personal reason for Sarioglu’s interest in cancer, as well. The disease took the life of two grandparents. But he was particularly motivated when his mother-in-law was diagnosed, back in Turkey, with late-stage brain cancer.

“It was devastating. I knew life expectancy was about four or five months,” says Sarioglu. “But their diagnosis was based purely on the pathology, a biopsy slice.”

He asked a colleague at Mass General, David Lewis, one of the world’s top pathologists, for another opinion. Lewis’ conclusions were vastly different. The cancer was benign, operable, and Sagioglu’s mother-in-law is alive and well.

“It showed me that we still have to improve how we diagnose cancer,” says Sarioglu, whose lab develops microfluidic chips that can isolate tumor cells out of billions of other cells. At Mass General, he worked on a device that captures clumps of tumor cells before metastasis, preventing the spread of cancer.

He’s continued that work since arriving at Georgia Tech in 2014, developing microchip technology that analyzes cells accurately and at very high speeds. Essentially, it is a better way to find the needle in the haystack, a minimally invasive way to diagnose cancer, liquid biopsy.

“The possibilities are endless, really,” says Sarioglu, who counts McDonald and Fred Vannberg (an expert in DNA sequencing who specializes in the molecular analysis of cancer) among his research collaborators. “The technology is applicable to all types of cancer.”

 

Doing Better

The primary tumor is rarely the killer in cancer. Nine times out of 10, cancer kills because it spreads to other parts of the body. So when a patient gets a cancer diagnosis, one of his first questions is, “has it metastasized?”

“You can obviously appreciate the anxiety. The physician and patient wonder the same exact thing. That’s the first question,” says Stanislav Emelianov, professor in the Georgia Tech/Emory Wallace H. Coulter Department of Biomedical Engineering (BME), a Georgia Research Alliance Eminent Scholar and the Joseph M. Pettit Chair in School of Electrical and Computer Engineering.

“Then there are more questions. What is the prognosis, the treatment, how do I deal with this – a lot of questions that can be better answered if we know the answer to the first question,” says Emelianov, whose team designs ultrasound imaging devices and algorithms, and has embarked on a project supported by a grant from the Breast Cancer Research Foundation to use light and sound and a non-radioactive molecularly targeted contrast agent, to answer that anxious first question.

The traditional approach has been to inject radioactive material and tracking that, then biopsy, which involves incision of the skin to expose the lymph node and taking pieces out to look for cancer.

“It is accurate, but it is also invasive, complicated and uses radioactive material,” Emelianov says. “We can do better.”

Emelianov speculates that in the future, we may be able to “weaponize” these contrast agents to actually kill cancer cells. Meanwhile, his team also is using its advanced imaging technology in collaboration with colleagues at Emory University’s Winship Cancer Center, to diagnose thyroid cancer and differentiate between malignant and benign tumors.

 

Tech’s Cancer Army

There are more than 40 faculty researchers at Georgia Tech who are members of the ICRC. They come from 12 different departments or schools. And there are an additional 16 researchers from academic and medical institutions that are affiliate members. It’s a diverse intellectual force that is giving Georgia Tech its own identity in cancer research.

“We can be a major player in cancer,” says McDonald. “How many medical schools have this breadth of expertise?”

He’s talking about young researchers like Susan Thomas, awarded Georgia Tech’s first grant from Susan G. Komen (breast cancer research foundation), supporting her work in immunotherapy for breast cancer; and Manu Platt, whose lab developed a new technique to give patients and oncologists more personalized information for choosing breast cancer treatment options.

And he’s referring to computer scientists like Constantine Dovrolis, who has spent the last few years investigating a phenomenon called “the hourglass effect” that is present in both technological and natural systems. He’s adapting what he learned studying embryogenesis with Georgia Tech biologist (and Petit Institute researcher) Soojin Yi to his collaboration with McDonald in cancer research.

He’s also thinking of BME-based researchers James Dahlman and William Lam.

Dahlman, an assistant professor who came to Georgia Tech earlier this year, works on cancer in two ways. Focusing extensively on primary lung tumors as well as lung metastasis, his team works on delivering genetic drugs to tumors.

“We have changed their gene expression, and either slowed tumor growth or caused established tumors to recede,” says Dahlman, an expert in gene editing. “In some cases, we have delivered multiple therapeutic RNAs to tumors, so that tumor cells are hit with a genetic ‘one-two’ punch that affects multiple cancer causing genes.”

His lab also creates tools to understand how cancer genes cause tumor resistance, studying how combinations of genes influence tumor growth, “because cancer is such a complicated disease and the genetics of cancer are notoriously difficult to understand,” Dahlman says. “It’s driven by many genes working together at once.”

For Lam, the war on cancer is waged in a lab and on the front lines, in a clinical setting. In addition to being a biomedical engineer, he’s also a pediatric hematologist-oncologist who treats patients at Children’s Healthcare of Atlanta.

His Ph.D. was actually focused on the biophysics of childhood leukemia, and his research in this area has focused on a small percentage of patients who develop leukostasis (stroke-like symptoms and lung failure).

“We always thought it was due to the biophysical properties of leukemia cells, which become big and sticky and jam up the plumbing of our blood vessels in our brain and lungs, which happen to have the smallest blood vessels,” says Lam, who is collaborating with Todd Sulchek, associate professor in mechanical engineering and a Petit Institute researcher.

“We’re combining some of Todd’s microfluidic technologies and our microfluidic technologies, to develop more high throughput ways to address this issue,” says Lam.

He’s also collaborating with the lab of BME professor Krish Roy on developing a ‘lymphoma on the chip’ model, to study how new cell therapies can directly affect the killing of cancer cells, as a way to determine whether those therapies have what it takes to work in the patient.

It’s all part of the multidisciplinary, “basement to bench to bedside” approach that Lam’s lab, with its connections to Georgia Tech, Emory University and Children’s Healthcare, has become known for.

“Within our lab, we’re certainly interested in technology development,” Lam says. “But then, we’re also interested in the assessment of the technology and, ultimately, directly translating that to the patient. Our lab lives in that entire space.”

 

LINKS

Integrated Cancer Research Center

McDonald Lab

Georgia Tech Cancer Army

 

CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]> Jerry Grillo 1 1478277869 2016-11-04 16:44:29 1478700043 2016-11-09 14:00:43 0 0 news Integrated Cancer Research Center developing new weapons for war on cancer

]]>
2016-11-04T00:00:00-04:00 2016-11-04T00:00:00-04:00 2016-11-04 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
583539 583540 583539 image <![CDATA[Cancer Cells Nov. 16]]> image/jpeg 1478277701 2016-11-04 16:41:41 1478277701 2016-11-04 16:41:41 583540 image <![CDATA[John McDonald]]> image/jpeg 1478277830 2016-11-04 16:43:50 1478281061 2016-11-04 17:37:41
<![CDATA[Roy’s Roadmap Leads to the Vatican]]> 28153 Krishnendu Roy scanned the room, taking note of the people all around him in the Vatican, and thought, “what the heck am I doing here?”

There was Bill Frist, former majority leader of the U.S. Senate, and there was Tommy Thompson, former governor of Wisconsin and U.S. Secretary of Health and Human Services. There was billionaire philanthropists/businessmen Sean Parker and Denny Sanford, and Ron DePinho, President of MD Anderson Cancer Center, and Carl June, the pioneer of cancer immnotherapy, and there were the heads of the food and drug administrations (FDA) from Europe. And this being the Vatican, there was Cardinal Gianfranco Ravasi (Minister of Culture of the Vatican). Surely, Pope Francis was somewhere nearby.

“All in all, a very high-powered meeting,” says Roy, recalling the International Regenerative Medicine Conference at the Vatican in April.

In fact, it was the highest-powered meeting in a string of high-powered meetings for Roy, Robert A. Milton Chair and professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, whose travel itinerary the last few months has taken him from Rome, to the White House, to the Harvard Business School in Boston for the annual Business of Regenerative Medicine conference, and most recently, back to Washington D.C. as a member of the newly formed Forum on Regenerative Medicine of The National Academies. 

Frequent flying has become a bigger part of Roy’s job description now as director of the $23 million Marcus Center for Therapeutic Cell Characterization and Manufacturing (MC3M) in development at Georgia Tech, Director of the Center for Immunoengineering at Georgia Tech, and technical lead for the National Cell Manufacturing Consortium (NCMC).

Lately, he’s probably spending more time in boardrooms and conferences than in the Laboratory for Cellular and Macromolecular Engineering, where his team works on developing new biomaterial-based strategies for immunoengineering and cell therapy biomanufacturing. 

“There are only so many hours in a day, and I’ve taken on more administrative roles, so something has to give. What has given is the amount of time I can spend on my own research and my students,” says Roy, a faculty researcher at the Petit Institute for Bioengineering and Bioscience. “Fortunately, I have a really good group keeping up with the work and producing amazing results.”

Which means Roy has time to take his message about cell manufacturing to some of the world’s halls of influence, like the Vatican, where the Pontifical Council for Culture and the Stem for Life Foundation hosted the third annual International Regenerative Medicine Conference.

The council, Roy says, “actively engages in understanding what is happening in the scientific world and how that affects theology. The meeting was fundamentally focused on how to increase access to revolutionary cell therapies that are right now primarily restricted to the privileged. How do you increase access to common people? I think that was the fundamental question the Vatican was interested in – how do you distribute these life-changing, curative therapies to a broad mass of people?”

Accordingly, Roy made a presentation entitled, “Advanced Manufacturing of Cells: Making Cell Therapies Reproducible, Reliable, Cost-Effective, and Accessible (Challenges, Barriers and a Roadmap to Success).”

Roy and Fred Sanfilippo, director of the Emory-Georgia Tech Healthcare Innovation Program, were part of a panel discussion on “Facilitating Cellular Innovation and Distribution,” moderated by physician and CBS medical correspondent Max Gomez.

That was April 30, the last day of a three-day event that featured a lineup of big-shot media moderators, like Gomez, Katie Couric, Sanjay Gupta, and Robin Roberts. They presided over discussions with researchers, clinicians, university presidents and also a group of people that Roy doesn’t often have exposure to.

“There were a lot of patients there, patients who have benefitted from cell therapies by having their cancer cured or another debilitating disease completely cured,” Roy says. “That was very different for me, because I never get that perspective. Clinicians do all the time. But as an engineer, we don’t get that perspective, and that was very touching.”

Before the weekend was finished, Pope Francis addressed the conference attendees emphasizing the need for access of breakthrough medical therapies to all citizens of the world, regardless of their socio-economic status or religion or which country they live in.

Also, U2 guitarist The Edge became the first rock musician to play at the Sistine Chapel, U.S. Vice President Joe Biden flew in from Iraq, and Roy’s wife and daughter met Pope Francis. “Yeah, it was all pretty amazing, not a typical conference,” Roy says.

In mid-June, Roy went to Washington, D.C., for the White House Organ Summit, and the official unveiling of the National Roadmap for Advanced Cell Manufacturing. The 10-year roadmap was developed by the NCMC, the industry-academic-government partnership created by Georgia Tech and the Georgia Research Alliance.

Last week, Roy was in Boston, where he was part of a panel discussion on “National Manufacturing Programs” (moderated by Petit Institute Executive Director Bob Guldberg) at the annual Business of Regenerative Medicine conference.

The bottom-line theme to Roy’s message these days as a spokesman for and leader in the cell manufacturing movement is this: The aspirin you buy at one drugstore is the same as you might buy from another, but cell-based therapies are a different story: they can vary from one center to another based on how the cells are isolated and processed (i.e., manufactured). There are established ways to quickly assess the efficacy and safety of small-molecule drugs like aspirin, and Roy and his fellow researchers want to develop and establish similar processes for therapeutic cell manufacturing.

“I think, at the end of the day, what matters is getting these high quality products to the people who need them most, at an affordable cost,” Roy says. “That’s the motivation of the Marcus Center, that’s the mission, that’s what I’m passionate about.”

 

CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for Bioengineering and Bioscience

]]> Jerry Grillo 1 1469783622 2016-07-29 09:13:42 1475896932 2016-10-08 03:22:12 0 0 news Petit Institute researcher stresses need for reproducible, reliable, affordable, accessible cell therapies

]]>
2016-07-29T00:00:00-04:00 2016-07-29T00:00:00-04:00 2016-07-29 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
556561 556561 image <![CDATA[Krish speaking Vatican]]> image/jpeg 1469797483 2016-07-29 13:04:43 1475895355 2016-10-08 02:55:55
<![CDATA[ImmunoEngineering Seed Grants Announced]]> 28153 Cutting edge research is not a solo act. Successful results are acquired through an ensemble effort, like the Georgia ImmunoEngineering Consortium, a collaborative partnership of multidisciplinary researchers from the Georgia Institute of Technology and Emory University.

But even the best collaborations require nurturing if they are to blossom into world-changing discoveries. That’s where the ImmunoEngineering Seed Grant program comes in.

“These seed grants allow us to pilot new ideas, gather data and be more competitive for large federal grants,” says Krish Roy, director of the ImmunoEngineering Research Center at Georgia Tech. “It also builds new bridges – collaborations across Georgia Tech and Emory, by providing ways to work together and generate new ideas, gather new data.”

Five new “bridges” – teams of Georgia Tech/Emory researchers – have just received important foundational support through the seed grant program for 2015-2016. These research proposals, each receiving a $50,000 award, are:

• “DNA-barcoded peptide-MHC tetramers for profiling antigen-specific T cells.” Researchers: Gabe Kwong (Georgia Tech), John Altman (Emory).

• “Modulation of early inflammatory response to prevent muscle degeneration in massive rotator cuff tears.” Researchers: Claudius Jarrett (Emory), Johnna Temenoff (Georgia Tech).

 • “Development of novel immune enhancing microparticle-conjugated RIG-I agonist.” Researchers: Mehul Suthar (Emory), Krish Roy (Georgia Tech).

• “Engineered mesenchymal stromal cells for enhancing lymphangiogenesis as a therapeutic for osteoarthritis.” Researchers: Nick Willet (Emory/Georgia Tech), J. Brandon Dixon (Georgia Tech), Rebecca Levit (Emory).

• Human mesenchymal stem cell-driven immunomodulation for enhanced engraftment of human pluripotent stem cell derived cardiomyocytes.” Researchers: Young-Sup Yoon (Emory), Satish Kumar (Georgia Tech).

“We congratulate this year’s class of ImmunoEngineering Seed Grant recipients,” says C. Michael Cassidy, president and CEO of the Georgia Research Alliance (GRA), the lead granting agency for the program. “The collaborative work of Georgia Tech and Emory researchers is developing innovative approaches for groundbreaking research.”

The Georgia ImmunoEngineering Consortium brings together engineers, chemists, physicists, computational scientists, immunologists and clinicians to collaboratively explore the inner workings of the immune system in a quest for breakthrough solutions to improve the lives of people suffering from cancer, infectious diseases, autoimmune and inflammatory disorders (such as diabetes, lupus, multiple sclerosis, arthritis, fibrosis, asthma, inflammatory bowel disease, etc.), as well as those individuals undergoing regenerative therapies (think of transplantation, spinal cord injury, bone and cartilage repair, etc.).

“In addition to fundamental immunology studies, the program also supports studies aimed at improving our ability to predict, measure and manipulate the intensity, quality and durability of the immune response,” notes Ignacio Sanz, a GRA Eminent Scholar and director of the Lowance Center for Human Immunology at Emory, where he also serves as chief of the Division of Rheumatology.

While GRA provides most of the funding for the seed grant program additional funding is provided by Emory and Georgia Tech. Roy likens the program to an early stage venture investment.

“If you find ten really good ideas that are already pre-screened and peer-reviewed, the chances are, when those apply for large federal grants the success rates will be high,” says Roy, professor in the Wallace H. Coulter Department of Biomedical Engineering, a joint department of Emory and Georgia Tech. “Even if only two or three of those 10 gets funded and becomes successful, that represents a manifold return on investment.”

In other words, the ImmunoEngineering Seed Grant program and the winning proposals are a research community example of smart money aimed at a good bet.

 

CONTACTS:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

 

]]> Jerry Grillo 1 1445593567 2015-10-23 09:46:07 1475896787 2016-10-08 03:19:47 0 0 news Five teams of Georgia Tech-Emory researchers awarded $50,000 each

]]>
2015-10-23T00:00:00-04:00 2015-10-23T00:00:00-04:00 2015-10-23 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
461811 461811 image <![CDATA[ImmunoEngineering]]> image/jpeg 1449256373 2015-12-04 19:12:53 1475895206 2016-10-08 02:53:26
<![CDATA[A Better Understanding of Lymph Node Remodeling]]> 28153 Susan Thomas, a faculty member of the Petit Institute for Bioengineering and Bioscience, is at the forefront of research into the targeted delivery of drugs to battle cancer. 

Thomas and her lab team have designed lymphatic-seeking nanoparticles to promote anti-tumor immunity and hinder tumor growth, a strategy supported by a three-year grant from the Susan G. Komen Foundation. 

But, before this form of immunotherapy can be deployed, Thomas and her team are gathering important intelligence about the enemy, to ultimately improve lymphatic targeting ability.

“One thing that’s not really understood is how lymph nodes in healthy individuals are different from those in someone with a disease. That information can critically impact our ability to target therapeutics,” says Thomas, who led a team of mostly undergraduate students in research that was recently published online in The FASEB Journal

“This is a first step toward a better understanding. But it’s not just what are the cellular differences or histological differences – the things that clinicians think about. We’re starting to define more the way engineers think of systems,” says Thomas, main author of the paper, entitled “Lymph node biophysical remodeling is associated with melanoma lymphatic drainage.”

Thomas describes the research as a basic analysis of how tumor lymphatic drainage affects the lymph nodes. 

“We detect a lot of tumors or cancers when the patient detects a stiff growth,” says Thomas, assistant professor in the Woodruff School of Mechanical Engineering, who recognizes that there are mechanical changes in the tumorous tissue, which make it different from healthy tissue.

“We wanted to see if the same kind of change is happening in the lymph node,” says Thomas, whose co-researchers on the project included one Ph.D. student (Nathan Rohner) and seven undergrads (Sara Tuell, Alex Warner, Blair Smith Younghu Hun, Abhinav Mohan and Manuela Sushinitha). “And what we found is that there actually are stiffening responses in the lymph nodes, and it was associated specifically with tumor lymphatic drainage.”

A better understanding of the biophysical changes associated with tumor lymphatic drainage will inform future studies and, ultimately, lead to better ways to diagnose and treat cancer patients.


CONTACT:

Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

 

]]> Jerry Grillo 1 1442225744 2015-09-14 10:15:44 1475896773 2016-10-08 03:19:33 0 0 news Undergrads drive new cancer-related research from Thomas lab 

]]>
2015-09-14T00:00:00-04:00 2015-09-14T00:00:00-04:00 2015-09-14 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
447231 398621 447231 image <![CDATA[Lymphatic system]]> image/jpeg 1449256246 2015-12-04 19:10:46 1475895187 2016-10-08 02:53:07 398621 image <![CDATA[Susan Thomas]]> image/jpeg 1449246371 2015-12-04 16:26:11 1475895115 2016-10-08 02:51:55