<![CDATA[REM Seed Grants Hit Their Targets]]> 28153 The Regenerative Engineering and Medicine (REM) research center, a joint collaboration of Emory University, the Georgia Institute of Technology and the University of Georgia, has awarded seed grants totaling $560,000 to eight teams of interdisciplinary researchers who are working to harness the body’s own potential to heal or regenerate in the wake of injury or disease.

REM research center leadership sees the annual program as a bridge-building first step to something larger.

“We believe the seed grant program is crucial to jump-starting new research activities between our institutions, and the awardees this year represent excellent examples of this type of interdisciplinary research,” says Johnna Temenoff, Petit Institute researcher, professor in the Wallace H. Coulter Department of Biomedical Engineering, and co-director of the REM center representing Georgia Tech.

Previous seed grant awardees have leveraged the funding into significant support from other agencies, like the National Institutes of Health (NIH) and the National Science Foundation (NSF), and the REM center’s three co-directors expect the same level of success from this year’s project teams.

“The goal of the seed grants is to create new research teams that can be successful in securing more substantive extramural funding,” notes Ned Waller, a Petit Institute researcher and the co-director from Emory, where he is professor of medicine and oncology. “These small grants are literally seeds sown into fertile scientific soil that will grow into robust research projects.”

The program targeted two research areas this year: optimizing cell-manufacturing and modifying the host environment to increase the biological effect of cell therapies.

“All of the submitted grants were highly significant and valuable,” says Steve Stice, co-director from the University of Georgia and also a Petit Institute researcher. “From advancing potential cell therapies for treating devastating eye diseases to a better understanding of how stem cells engraft in target issues, which could enhance the success of all stem cell therapies.”

Adds Waller, “We are excited by the proposals put forth by REM investigators and look forward to seeing the results from the new collaborations.”

Here’s a rundown of the eight projects: 

• Project Title: Single-cell epigenomics: Towards understanding the mechanisms regulating cell potency and epigenetic stability for regenerative biomanufacturing

• Principle Investigators: Rabindranath De La Fuente (University of Georgia), Yuhong Fan (Georgia Institute of Technology, Petit Institute researcher).

• Synopsis: The researchers plan to develop novel epigenenic sensors and single-cell epigenomics tools that could be adapted for high throughput analysis of potency in cell therapy products with potential clinical applications. This collaboration is part of a long term funding strategy to provide essential preliminary data and design a subsequent application to the National Institutes of Health (NIH), National Science Foundation (NSF) or the American Heart Association (AHA) to develop noninvasive diagnostic tools to predict cell potency and to improve regenerative biomanufacturing.

 

• Project Title: Hydrogels for Mesenchymal Stem Cells to Treat Graft-vs-Host Disease

• Principal Investigators: Andrés J. García (Georgia Tech, Petit Institute researcher), Muna Qayed (Emory University), Raghavan Chinnadurai (Emory University).

• Synopsis: The researchers’ objective is to engineer synthetic hydrogels that encapsulate mesenchymal stem cells (MSCs) and promote their survival and expansion in alternative transplant sites resulting in enhanced immunomodulatory activities for the treatment of GvHD. They hypothesize that these delivery vehicles will prolong MSC persistence and survival compared to intravenous delivery and will result in reduced GvHD activity in pre-clinical transplant models.

 

• Project Title: Transplantation of bioenergetics-enriched stem cells to boost muscle regeneration in ischemic myopathy

• Principal Investigators: Young C. Jang (Georgia Tech, Petit Institute researcher, Coulter Department), Luke Brewster (Emory University), Franklin West (University of Georgia).

• Synopsis: The overarching goal is to validate the importance of mitochondrial bioenergetics in peripheral arterial disease (PAD, a progressive degenerative disease) and to test whether transplantation of donor stem cells that are enriched for mitochondrial activity can rejuvenate muscle regeneration. The outcome of this work will have a broad and significant impact in the field of regenerative medicine.

 

• Project Title: Transcranial Direct Current Stimulation for Traumatic Brain Injuries

• Principal Investigators: Lohitash Karumbaiah (University of Georgia), Maysam Ghovanloo (Georgia Tech).

• Synopsis: Traumatic Brain Injuries (TBI) lead to a range of complex neurophysiological and functional deficits, severe long-term disability, and poor prognosis. There are no effective treatments for TBI, but noninvasive transcranial Direct Current Stimulation (tDCS) has shown promise. The researchers plan to test their hypothesis that tDCS in combination with low-frequency synaptic activation will enhance neuronal regeneration and improve synaptic strength of injured motor neurons in vitro, leading to functional recovery.

 

• Project Title: HDL-mimetic nanocarriers for miRNA-mediated direct cell reprogramming for vascular regeneration

• Principal Investigators: YongTae Kim (Georgia Tech, Petit Institute researcher), Young-sup Yoon (Emory University).

• Synopsis: Ischemic cardiovascular diseases are the leading causes of morbidity and mortality, afflicting approximately 26% of Americans. The underlying problems are associated with loss or dysfunction of blood vessels and/or impaired new vessel formation (neovascularization). Neovascularization is critical for tissue repair and regeneration and the progress depends mainly upon the functionality of endothelial cells (ECs), which are not easily obtained. If successful, this will be the first study of direct reprogramming of adult human somatic cells into ECs via miRNAs. Outcomes from this research could suggest a novel platform for ischemic tissue repair and regeneration and a source of cells for disease investigation and drug discovery.

 

• Project Title: Microfluidic technologies to rapidly collect stem cells for treatment of damaged cornea

• Principal Investigators: Todd Sulchek (Georgia Tech, Petit Institute researcher), James Lauderdale (University of Georgia).

• Synopsis: The researchers plan to apply a new microfluidic cell sorting technology to rapidly enrich stem cells from a damaged cornea for therapeutic replacement and regeneration of the cornea. The technology will enrich stem cells without labels through biophysical markers. The advantage of utilizing biophysical markers is that that sorting can be extremely fast and with no expensive or cumbersome equipment.

 

• Project Title: Enhancing transplanted MSC engraftment by selective opening of the bone marrow niche for hypophosphatasia renewal

• Principal Investigators: Luke J. Mortensen (University of Georgia), Ed Botchwey (Georgia Tech, Petit Institute researcher).

• Synopsis: Systemically administered mesenchymal stem cells (MSCs) are a promising therapeutic approach to prevent or ameliorate metabolic bone diseases, such as hypophosphatasia (HPP), which reduces bone mineralization and produces fragile bones. The goal of this study is to develop strategies to enhance carrying capacity for and engraftment of donor MSCs without damaging the bone marrow niche or compromising bone homeostasis.

 

• Project Title: Development of novel iPSC-RBCs engineered to tolerize recipients to alloantigens that complicate transfusion and other cell therapies

• Principal Investigators: John D. Roback (Emory University), James Dahlman (Georgia Tech, Petit Institute researcher), Sean Stowell (Emory University).

• Synopsis: Red blood cell (RBC) transfusion is a common therapeutic procedure, but its efficacy is diminished in recipients who have developed an immune response against the allogenic cells. By leveraging specialized models, the researchers have developed novel approaches to engineer RBC antigens and ultimately tolerize transfusion recipients to foreign RBC antigens, so these patients can continue to receive needed transfusion support.

 

Learn more about the REM Research Center

 

CONTACT:

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

]]> Jerry Grillo 1 1473428320 2016-09-09 13:38:40 1653584976 2022-05-26 17:09:36 0 0 news Eight interdisciplinary teams receive boost for research designed to harness the body’s own potential to heal

 ]]>
2016-09-09T00:00:00-04:00 2016-09-09T00:00:00-04:00 2016-09-09 00:00:00 Communications Officer II - Parker H. Petit Institute for - Bioengineering and Bioscience

]]>
574791 574791 image <![CDATA[human cell]]> image/jpeg 1473442436 2016-09-09 17:33:56 1475895383 2016-10-08 02:56:23
<![CDATA[Growing Back the Lymph System]]> 27195 A team including University of Georgia researchers has for the first time documented the regrowth of surgically removed pathways in the lymphatic system, a network of vessels designed to pump away inflammatory fluids and defend the body against infection.

Published in Nature Biomedical Engineering, the findings lay the foundation for a new class of treatment options for lymph-related disorders, such as chronic wound complications, and could even help prevent the spread of cancer.

Lymphatic dysfunction is connected to a variety of diseases, including both cardiovascular disease and cancer. For example, breast cancer cells break away from the primary tumor and travel through the lymphatic system. If left alone, they can spread throughout the body. Lymphatic vessels—which operate in similar ways to the cardiovascular system—are sometimes traumatized by cancer treatment or the removal of lymph nodes, which can lead to lymphedema, or the chronic swelling of a leg or arm.

“Right now, we don’t have a way to rebuild or reconstruct the lymphatic system. We hardly even understand how it works,” said Dr. John Peroni, a professor and large-animal surgeon in UGA’s College of Veterinary Medicine. “This study was one of the first in fundamentally addressing a basic scientific question that has been left unanswered: If lymphatics are injured, can they remodel or heal?”

Working with a sheep model, collaborators from Georgia Tech and the Regenerative Bioscience Center at UGA’s College of Agricultural and Environmental Sciences, removed one of two lymphatic vessels that run parallel to each other in the leg. These are as crucial to lymph flow as the heart is to blood flow, according to the researchers. Under these conditions, they were able to show the beginning of a lymphatic pump cycle and the start of the remodeling and repair stages.

As a result of the remodeling, the team concluded that molecular changes in lymphatic muscle cells enhanced oxidative stress, which typically occurs when the immune system is creating inflammation to fight off bacteria. After a period of six weeks, the team discovered that the remaining lymphatic vessel was working twice as hard to compensate for the oxidative stress.

“One would expect that when you remove the main lymph vessel, in the part that’s lower than the obstruction, it would swell. To our surprise, it only did so minimally,” Peroni said. “It turns out there’s a considerable amount of collateral lymphatic circulation that we were not expecting. At a microscopic level, there’s enough mechanisms by which the body can still recirculate and drain fluids out of the leg, even though the main ‘highway’ is removed.”

The findings follow the same researchers’ previous work which showed similar results in using the rodent tail, one of the oldest and most widely utilized model systems for lymphatic research.

“Perhaps the most important single feature of using a larger model like sheep, versus the historical benchmark of a rodent’s tail, is the gravitational benefits,” Peroni said. “Gravity makes it harder for lymph to be transported from the legs and the lower half of the body, and sheep provide a better gravitational model compared to the consistently flat position of a rodent’s tail. It’s almost identical to wound-healing issues in humans.”

Peroni worked closely with Brandon Dixon, lead author and associate professor of mechanical and biomedical engineering at Georgia Tech, who heads the Laboratory of Lymphatic Biology and Bioengineering.

“What distinguishes this study from others is that it focused on remodeling of the vessel that was not initially damaged during surgery, as it attempts to compensate for the segment that was removed,” said Dixon. “This is important for understanding secondary lymphedema in breast cancer survivors, since most onset of lymphedema occurs many months after breast cancer surgery, and the remaining intact lymphatic vasculature can no longer keep up with the demands placed on it.”

These findings are pre-clinical and further studies are required to confirm testing in humans, but they provide scientific evidence for lymphatic remodeling that up until now has been scarce.

“We’re excited because there is now an animal model that we can use to put the vessel under this state of prolonged stress that wasn’t result of the initial injury, but a result of the vessel’s adaptation to the surgery,” said Dixon. “It’s a good model for what happens to a human.”

This study was funded by the Regenerative Engineering and Medicine (REM) seed grant program. The Regenerative Bioscience Center is a unit of the UGA Office of Research, with generous support from the College of Agriculture and Environmental Sciences and its Department of Animal and Dairy Science.

Publication link: https://doi.org/10.1038/s41551-019-0493-1

]]> Colly Mitchell 1 1590671999 2020-05-28 13:19:59 1590672349 2020-05-28 13:25:49 0 0 news 2020-05-07T00:00:00-04:00 2020-05-07T00:00:00-04:00 2020-05-07 00:00:00 Charlene Betourney

]]>
635742 635742 image <![CDATA[Growing Back the Lymph System]]> image/png 1590672056 2020-05-28 13:20:56 1590672056 2020-05-28 13:20:56
<![CDATA[Developing a Better Model]]> 28153 One of the nasty potential byproducts of surgery to remove cancerous lymph nodes doesn’t rear its ugly head until it’s usually too late to fix.

“During the procedure, some of the lymphatic vasculature is taken out because the surgeon is almost operating blind. If the lymphatic system suffers from injury during surgery, the damage is often difficult to gauge, presenting as lymphedema maybe two to five years later,” says J. Brandon Dixon, associate professor of bioengineering in the Woodruff School of Mechanical Engineering at the Georgia Institute of Technology, where his research is focused on the molecular aspects of lymphatic function in the body’s dynamic, ever-changing mechanical environment.

Dixon, needing an adequate model to study the effects of surgery on the lymphatic system, luckily ran into John Peroni, professor of large animal surgery at the University of Georgia’s (UGA) Regenerative Bioscience Center, during the annual meeting of the Regenerative Engineering and Medicine (REM) research center.

“We started talking about one of the things the lymphatic research community was lacking – a large animal model that could help us understand lymphatic injury,” says Dixon, who also has an appointment in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory. That conversation between he and Peroni has resulted in their recently-published research paper in the journal Nature Biomedical Engineering, “Lymphatic remodelling in response to lymphatic injury in the hind limbs of sheep.”

Dixon and Peroni, who are both members of the Petit Institute for Bioengineering and Bioscience at Georgia Tech, led a multi-institutional collaboration that also included the Albert Einstein College of Medicine (Bronx, N.Y.), and was supported by an REM Seed Grant. Dixon had previously developed imaging technology to capture in vivo lymphatic activity in small animal models, and was eager to test it on a larger animal model that could more accurately reflect the role gravity plays in opposing lymphatic flow in a human.

The main function of the lymphatic system in any vertebrate is to move lymph fluid (loaded  with infection-fighting white blood cells) through the body, via lymphatic vessels. There is no central pump, no heart; the lymphatic vessels themselves slowly pump the fluid, and the greatest force working against them is gravity.

One of Dixon’s graduate researchers drove equipment from the Lab of Lymphatic Biology and Bioengineering at Tech to Athens, so Peroni could perform surgery with the near infrared (NIR) image guidance system. The surgery team wanted to compromise the lymphatic vasculature of a sheep in one limb, “so they removed a chain of pumps in one leg and left the other leg intact,” Dixon explains. “Then we followed up with NIR lymphatic imaging on the animal for up to six weeks after the surgical injury. There was an initial decline of pump function, but eventually it started to recover.”

The researchers measured ‘pressure generation capacity’ in the vessels with imaging over the course of remodeling, and then carefully dissected out these sub-millimeter in diameter vessels at the end of six weeks for functional testing. They gauged mechanical properties, performed a proteomic analysis of the muscle cells, and found that the remodeled lymphatic vessel was working much harder than vessels from the control limb.

“Think of it like an elevated heart rate for a lymphatic vessel,” says Dixon, whose team noted, “significantly increased signs of oxidative stress.”

The team found, over the six-week period, that lymphedema hadn’t developed, and it seemed as if the affected lymphatic vessel could handle the load, “but it had to do the work of two vessels, and at the cost of oxidative stress, and that’s analogous to early signs of cardiovascular failure,” notes Dixon.

The stress on the vessel wasn’t the result of the initial surgical injury, “but a result of the vessel’s adaptation for the surgery,” Dixon says. “And it’s a good model for what happens to a human.”

Lead author of the paper was Tyler S. Nelson, former graduate researcher in Dixon lab. Other authors were Zhanna Nepiyushchikh (research scientist in the Dixon lab), Joshua S. T. Hooks and and Mohammad S. Razavi (former graduate researchers in Dixon lab), Tristan Lewis and Merrilee Thoresen (University of Georgia), Cristina Clement and Laura Santambrogio (Einstein College of Medicine), Matthew T. Cribb (graduate researcher in Dixon lab), Mindy K. Ross (former undergraduate researcher in Dixon lab), Rudolph L. Gleason (associate professor in Woodruff School and Coulter Department), Peroni, and Dixon.

]]> Jerry Grillo 1 1582828213 2020-02-27 18:30:13 1582828213 2020-02-27 18:30:13 0 0 news Dixon collaborates with UGA researchers to uncover potential early signs of lymphedema

]]>
2020-02-27T00:00:00-05:00 2020-02-27T00:00:00-05:00 2020-02-27 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
633057 633057 image <![CDATA[Dixon lab]]> image/jpeg 1582828018 2020-02-27 18:26:58 1582828018 2020-02-27 18:26:58
<![CDATA[Mediating from the Middle]]> 28153 When you nick yourself shaving, or clumsily slice your thumb while cutting sheetrock, you can thank goodness for integrins. These specialized proteins play a critical role in stopping the bleeding that ensues as a result of the aforementioned maladies (and other such breaches of the skin).

“Integrins are cell membrane receptors that mediate cell adhesion and mechanosensing,” explains Cheng Zhu, Regents Professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University.

They are vital to cellular biological processes, such as growth and development – cell attachment to the extracellular matrix (ECM) is a basic requirement in building a multicellular organism, for instance.

Following the mishaps described above, your platelets (which utilize the binding and signaling functions of an integrin called glycoprotein IIb/IIIa, or αIIbβ3) swarm the wound and clump together, forming a plug, or clot to stop blood loss. This first stage of wound healing is called hemostasis.

“Of course, the other side of the coin is thrombosis, which is what kills people who have cardiovascular disease,” says Zhu, a researcher in the Petit Institute for Bioengineering and Bioscience at Georgia Tech, describing what happens when platelet clumping runs amok, causing deadly clots.

So integrins basically facilitate how cells bind to and respond to their environment. They allow cells to cling to the ECM and to each other, and they are great communicators, transmitting bi-directional signals: inside-out, to activate the binding function; and outside-in, allowing the cell to sense and react to the extracellular environment. The integrin can instruct the cell.

But the mechanisms behind these important processes remain poorly understood, which is why Zhu and an international team of researchers are digging into integrins.

“This is a very important, dynamic biological molecule, a little nanomachine with hinges and moving parts,” says Zhu, co-corresponding author of a research paper in the journal Nature Materials, entitled, “An integrin αIIbβ3 intermediate affinity state mediates biomechanical platelet aggregation."

This is actually a continuation of research that began several years ago by former grad students in Zhu’s lab, where a major focus is on the biomechanics and mechanobiology of cell adhesion and signaling molecules of the immune system and vascular systems. The former Georgia Tech grad students – Yunfeng Chen and Lining Arnold Ju – are co-lead authors of the new paper, though they are now on opposite corners of the world. Chen is now doing his work at the Scripps Research Institute in La Jolla, California, and Ju is a researcher with the Heart Research Institute and The University of Sydney in Australia, working in the lab of Shaun Jackson, co-corresponding author with Zhu.

Other researchers on this multi-disciplinary project were: Fangyuan Zhou and, Jiexi Liao (graduate student researchers in the Zhu lab at Georgia Tech); Lingzhou Xues (Penn State University); Qian Peter Su and Dayong Jin (University of Technology Sydney, Australia); Yuping Yuan (Jackson lab at University of Sydney); and Hang Lu (Petit Institute researcher, and professor in the School of Chemical & Biomolecular Engineering at Georgia Tech).

The team used the dual biomembrane force probe technology developed in Zhu’s lab and also fluorescence biomembrane force probe, combined with microfluidic perfusion assays, and applied precisely-controlled mechanical simulations to platelets, identifying an intermediate state of integrin αIIbβ3, whose characteristics are all intermediate between the well-known inactive and active states.

The work reveals distinct integrin state transitions in response to biomechanical and biochemical stimuli, identifying a role for the αIIbβ3 intermediate state in promoting biomechanical platelet aggregation. Using a microfluidic channel that mimics blood vessel stenosis, the researchers could actually see thrombus formation as it happened.

“One of the things we were curious about is, will the mechanical activation of integrins change pathological situations in people with vascular diseases,” says Chen.

The researchers believe that their finding – that biomechanical thrombus growth is mainly mediated by an intermediate state triggered by a biomechanical activation pathway – will probably guide the development of new anti-thrombotic strategies.

This research was supported by grants from the National Institutes of Health (and under its umbrella, the National Institute on Drug Abuse), the National Science Foundation, the Australian Research Council, the University of Technology Sydney, the Diabetes Australia Research Program, the University of Sydney, the Royal College of Pathologists of Australasia,  e, the National Science Foundation, and the Cardiac Society of Australia and New Zealand.

]]> Jerry Grillo 1 1553536962 2019-03-25 18:02:42 1553604498 2019-03-26 12:48:18 0 0 news From the lab of Cheng Zhu: Integrin, vital proteins that play a major role in hemostasis and thrombosis, biomechanically facilitate platelet aggregation

]]>
2019-03-25T00:00:00-04:00 2019-03-25T00:00:00-04:00 2019-03-25 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
614034 619590 614034 image <![CDATA[Regents professor Cheng Zhu portrait]]> image/jpeg 1541705463 2018-11-08 19:31:03 1541705463 2018-11-08 19:31:03 619590 image <![CDATA[Yunfeng Chen and Lining Ju]]> image/jpeg 1553536189 2019-03-25 17:49:49 1553536189 2019-03-25 17:49:49
<![CDATA[Seed Grants Awarded]]> 28153 Congratulations to the interdisciplinary teams awarded seed grants by the Regenerative Engineering and Medicine (REM) research center. REM is a collaborative partnership building on the success of Emory University and Georgia Institute of Technology in cell therapy clinical trials and tissue engineering technologies, as well as stem cell research and large animal applications at the University of Georgia. REM seed grants are supported by the three partner universities in conjunction with the Georgia Research Alliance.

2018 REM Seed Grant Awardees:

Project Title: “Bactericidal Hydrogels to Treat Bone Infection in a Large Animal Fracture Model"

Principal Investigators: Andrés García (Georgia Tech) and John Peroni (University of Georgia)

Synopsis: The objective of this project is to examine the ability of synthetic materials (hydrogels) that deliver the potent bactericidal enzyme lysostaphin to eradicate bacteria infection in a sheep model of bone defect infection. Our hypothesis is that hydrogels with controlled delivery of lysostaphin will eliminate Staphylococcal infections thus preserving defect fixation. The establishment of these approaches using local antimicrobial engineering expertise developed at GT with the large animal modeling expertise available at UGA will generate critical data to establish the translational potential of this anti-infective material.

 

Project Title: “Comparing the effects of MSCs derived from pluripotent stem cells originating from normal and diseased joint chondrocytes on the progression of osteoarthritis"

Principal Investigators: Luke Mortensen (University of Georgia) and Hicham Drissi (Emory University)

Synopsis: Osteoarthritis is a degenerative disease of the cartilage with a significant societal burden both economically and emotionally. To date there is no cure for osteoarthritis. This proposal will specifically focus on post-traumatic osteoarthritis. Because patient specific induced pluripotent stem cells (iPSCs) are increasingly considered as a source of stem cells for cartilage regeneration, a question remains as to whether the cell source and disease state of IPSCs could influence their regenerative capacity. Specifically, epigenetic changes that occur during OA progression and perhaps are only partially reversed during cell reprogramming can dramatically impact the regenerative capacity of these cells. The project aims to: 1) Evaluate the epigenetic memory of inflammation in chondrogenic iPS-derived cells from healthy and arthritic clinical patients & 2) Trace the survival and differentiation of healthy and arthritic chondrogenic iPS-derived cells in vivo using multiphoton intravital imaging in a medial meniscus destabilization model. This will be a first step in elucidating some of the critical pathways required for iPSC-derived regenerative capacity for tissue cartilage regeneration.

 

Project Title: “Polycaprolactone Scaffold And Forearm Free Tissue Viability In Yucutan Pigs"

Principal Investigators: Scott Hollister (Georgia Tech) and Craig Villari (Emory University)

Synopsis: This REM seed grant proposes to develop and test a tracheal scaffold as the basis for a tracheal free flap in a Yucatan swine model. This tracheal flap is actually being developed for a current patient of Dr. Villari who has a long segmental tracheal defect. The scaffold design will be generated directly from this patient’s image data. The resulting scaffold will be 3D printed for testing. The goals of this project will be: 1) to determine the mechanical viability via finite element simulation and mechanical testing for a range of porous patient specific scaffold designs in comparison to published human tracheal mechanical properties and tested swine tracheal mechanical properties and 2) to test the four tracheal scaffolds in a Yucatan free tissue flaps with and without growth factors to enable vascularity. The resulting flaps will be mechanically tested, scanned using micro-computed tomography in the animal, and sectioned for histology to determine mechanical properties, resulting geometry and tissue ingrowth.

]]> Jerry Grillo 1 1536756486 2018-09-12 12:48:06 1536763046 2018-09-12 14:37:26 0 0 news Regenerative Engineering and Medicine Research Center supports three interdisciplinary, multi-university teams

]]>
2018-09-12T00:00:00-04:00 2018-09-12T00:00:00-04:00 2018-09-12 00:00:00 611350 611352 611351 611350 image <![CDATA[Garcia and Peroni]]> image/jpeg 1536755982 2018-09-12 12:39:42 1536755982 2018-09-12 12:39:42 611352 image <![CDATA[Mortensen and Drissi]]> image/jpeg 1536756081 2018-09-12 12:41:21 1536756081 2018-09-12 12:41:21 611351 image <![CDATA[Hollister and Villari]]> image/jpeg 1536756029 2018-09-12 12:40:29 1536756029 2018-09-12 12:40:29
<![CDATA[Dahlman in Elite Company]]> 28153 James Dahlman, assistant professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, and a researcher in the Petit Institute for Bioengineering and Bioscience, has been named to MIT Technology Review’s prestigious annual list of Innovators Under 35.

Dahlman is a bioengineer working at the interface of nanotechnology, gene editing, and genomics. His lab develops novel ‘big data’ technologies and applies them to the study of nanomedicine. One such application is the use of DNA barcodes to track thousands of nanoparticles directly in vivo; typically, labs will study a few nanoparticles in vivo.

His lab also has pioneered the use of DNA barcoded nanoparticles, and is using this powerful new technology to design nanoparticles that deliver genetic drugs to target tissues. He has designed nanoparticles that deliver RNA drugs to blood vessels; these nanoparticles have worked in more than 20 labs and are under consideration for clinical development. At the age of 31, he already has published in Nature Nanotechnology (twice), Nature Biotechnology, Cell, Nature Cell Biology, Science Translational Medicine, PNAS (twice), JACS, and other prestigious journals.

In addition to the Technology Review honor, Dahlman has won many national and international awards, and since 2014, has given dozens of invited talks at leading universities around the world on drug delivery and DNA barcoding.

For more than a decade, Technology Review has recognized exceptionally talented technologists whose work has great potential to transform the world. Previous Innovators Under 35 include Larry Page and Sergey Brin, the cofounders of Google, Mark Zuckerberg, the cofounder of Facebook, Helen Greiner, the cofounder of iRobot, and Jonathan Ive, the chief designer of Apple.

Gideon Lichfield, editor-in-chief of MIT Technology Review, said: “MIT Technology Review inherently focuses on technology first - the breakthroughs and their potential to disrupt our lives. Our annual Innovators Under 35 list is a chance for us to honor the outstanding people behind those technologies. We hope these profiles offer a glimpse into what the face of technology looks like today as well as in the future.”

Learn more about this year’s honorees on the MIT Technology Review website here and in the July/August print magazine, which will hit newsstands worldwide on July 3. The honorees are also invited to appear in person at the upcoming EmTech MIT conference, MIT Technology Review’s flagship event exploring future trends and technologies that will impact the global economy, happening September 11-14, 2018 in Cambridge, Massachusetts.

 

About MIT Technology Review

Founded at the Massachusetts Institute of Technology in 1899, MIT Technology Review is a world-renowned, independent media company whose insight, analysis, reviews, interviews and live events explain the commercial, social and political impact of new technologies. MIT Technology Review derives its authority from the world's foremost technology institution and from its editors' deep technical knowledge, capacity to see technologies in their broadest context, and unequaled access to leading innovators and researchers. MIT Technology Review’s mission is to bring about better-informed and more conscious decisions about technology through authoritative, influential and trustworthy journalism.

 

 

]]> Jerry Grillo 1 1530106443 2018-06-27 13:34:03 1530110698 2018-06-27 14:44:58 0 0 news Georgia Tech researcher named to MIT Technology Review’s Innovators Under 35 List

]]>
2018-06-27T00:00:00-04:00 2018-06-27T00:00:00-04:00 2018-06-27 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
607327 607336 607327 image <![CDATA[James Dahlman]]> image/jpeg 1530105505 2018-06-27 13:18:25 1530105505 2018-06-27 13:18:25 607336 image <![CDATA[MIT Technology Review ]]> image/jpeg 1530110671 2018-06-27 14:44:31 1530110671 2018-06-27 14:44:31
<![CDATA[Improving Gene Therapy]]> 28153 Last year, commercial medical history was made when the Food and Drug Administration (FDA) approved the first gene therapy treatment, Kymriah, in the U.S., opening the door to a new world of treatment for devastating diseases. As reports from clinical trials of tisagenlecleucel (marketed as Kymriah) became public – that it used a patient’s own T-cells to actually kill cancer – the news media called it a miracle cure.

Of course, it took decades of research, a lot of trial and error at the molecular level. The concept, transplanting normal or healthy genes into cells in place of missing or defective genes in order to treat diseases and disorders, has been under development since the Reagan administration.

“The idea of gene therapy’s been around since I was a kid,” says Wilbur Lam, associate professor in the Wallace H. Coulter Department of Biomedical Engineering at Emory University and the Georgia Institute of Technology and researcher in the Petit Institute for Bioengineering and Bioscience at Georgia Tech. “In the 1990s, gene therapy trials mostly failed and often caused deadly side effects like cancer. But the entire field has moved forward, and the field of gene therapy has now learned how to actually cure people.”

As it turns out, miracles are not only complex and elusive, they’re also expensive, to the point of being infeasible for large scale clinical translation. But with support from a new R01 grant from the National Institutes of Health (NIH), Lam’s lab is working to make gene therapies more efficient, and more economically accessible.

Kymriah, for example, has a price tag of $475,000 (or about $20,000 less than the value of Lam’s new four-year NIH grant).

“That price tag is largely a result of the difficulty and cost associated with viral vector manufacturing, which is, unfortunately, the key to these novel therapies,” says Reginald Tran, a postdoctoral researcher in Lam’s lab who is using microfluidic technology that he developed to incorporate basic mass transfer and fluid mechanics principles to increase gene therapy efficiency.

In gene therapy modified viruses often are used as vectors, or vehicles, to carry the good-guy genes into a human cell. It’s been proven to work, but it’s not a very efficient process, according to Lam. For one thing, “it takes about a billion cells to treat an adult for something like sickle cell disease,” he says.

Also, Lam notes that these engineered viral vectors have a half-life, “and they’ll deactivate if they don’t find their target cell in time. They will die. Excess virus is given to cells to ensure that enough cells get genetically modified, but this results in added costs and significant waste of the precious key reagent. ”

Tran, who earned his Ph.D. in the Lam lab, says he was amazed to learn about the capabilities of microfluidics, and how leveraging the differences in physics at the micro scale could unlock new possibilities for diagnostics and therapeutics.

“This project was really ideal for me since I could combine my backgrounds in mechanical engineering and microfluidics toward gene and cell therapy,” Tran adds. “I quickly found that microfluidics were the perfect platform to efficiently bring cells and viral vectors together to maximize gene transfer.”

Performing the critical gene transfer step in microfluidics dramatically reduces processing time and resources (up to five times less viral vector can be used). While many researchers continue to work on developing more potent genes or increasing viral vector manufacturing potential, the Lam group is focused on using less virus.

“At a time when virus shortages present a huge bottleneck in gene and cell therapy commercialization, we believe that our technology can help make these treatments more accessible and affordable for patients,” Tran says. “I feel extremely lucky to work on a problem that has the potential to make a meaningful impact on what could very well be the next frontier in modern medicine.”

 

]]> Jerry Grillo 1 1528137031 2018-06-04 18:30:31 1528145032 2018-06-04 20:43:52 0 0 news Lam lab using new NIH grant to make groundbreaking treatments more efficient and accessible

]]>
2018-06-04T00:00:00-04:00 2018-06-04T00:00:00-04:00 2018-06-04 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
606742 606741 606742 image <![CDATA[Wilbur Lam]]> image/jpeg 1528136697 2018-06-04 18:24:57 1528136697 2018-06-04 18:24:57 606741 image <![CDATA[Reggie Tran]]> image/jpeg 1528136659 2018-06-04 18:24:19 1528136659 2018-06-04 18:24:19
<![CDATA[REM Retreat Features Influential Guests]]> 28153 The Regenerative Engineering and Medicine (REM) research center isn’t a place. It’s more like glue with an agenda, binding together three research institutions – Emory University, the Georgia Institute of Technology, and the University of Georgia – and keeping them focused on the same basic target: enhancing the body’s ability to harness its own potential to heal, or regenerate.

“The goal is to create an infrastructure for three institutions in Georgia – Emory, Georgia Tech, and the University of Georgia – to work together to develop basic research collaborations in regenerative medicine,” Ned Waller told a packed room in the Miller-Ward Alumni House at Emory University for the REM’s annual retreat, on May 8.

“The goal is to bring people together and see how this program can continue to initiate and accelerate collaborations across our institutions,” added Waller, the co-director of REM from Emory.

The audience then received a fitting infusion of research and inspiration from cell therapy pioneer Carl June, who provided the highlight of the day, delivering the first-ever Dr. J. David Allen Keynote Lecture. June is the pioneering researcher who led development of the nation’s first personalized cellular therapy for cancer – last year, the U.S. Food and Drug Administration (FDA) approved Kymriah(TM) (formerly CTL019), a groundbreaking chimeric antigen receptor T-cell (CAR T) therapy which uses a patient’s own T cells to fight cancer.

He shared the story of his work in engineering a person’s own immune system to cure cancer, and it was exactly what REM researchers needed to hear at this gathering.

“Translational Medicine was a focus of this year’s retreat and is the next logical step for REM, working to move therapies from research to development and into the clinics,” said Steve Stice, the REM co-director from the University of Georgia (UGA), where he’s established himself as one of the world’s leading stem cell researchers.

June took his audience on a journey through what he called, “the complexity of the tumor microenvironment, to really solve the issues of human cancer.” And there wasn’t an empty seat in the room.

“I thought the number of new participants who were drawn to the Allen lecture was a particular highlight of this year’s retreat,” noted Johnna Temenoff, Petit Institute researcher and co-director of REM from Georgia Tech.

Speaking softly to the crowded room, June presented a number of slides to illustrate the story of his research.

“This is probably the most famous sight you will see at any cancer-immunology meeting now,” he said, presenting a rendering of the familiar cancer-immunity cycle, divided into seven major steps (circles on a screen, directed by clockwise arrows, beginning with the release of cancer cell antigens and ending with the killing of cancer cells).

June, named to the TIME 100 (the magazine’s list of the most influential people in the world) introduced his audience to the first patient to receive the CAR T-cell therapy, in 2010, Bill Ludwig, a retired corrections officer in New Jersey who was battling a lethal leukemia before the therapy from Penn actually cured him. The audience laughed as June presented a before and after photo of Ludwig, from 2010 and 2017.

In the after photo, he looks very sick as he begins the trial. In the after photo, he’s brimming with joy, wearing a t-shirt that says, “The clouds went away and there was no leukemia,” and holding up a handwritten sign that says, “I was patient No. 1 of CART-19 and all I got was this t-shirt and remission.”

In 2012, June and his team treated the first pediatric patient, Emily Whitehead, who has been cancer free for years. The technology was licensed to Novartis, in exchange for financial support, just six months after Emily’s trial began.

Part of the deal was support for a new on-campus cell manufacturing facility at Penn, something similar to what is happening at Georgia Tech, where the NSF Engineering Research Center for Cell Manufacturing (CMaT) is headquartered. “I applaud what you’re doing here,” June told the audience, referring to the REM state-wide partnership, including CMaT.

Though June was the main attraction at this year’s retreat, there was another honored guest – philanthropist J. David Allen, the retired oral surgeon whose generosity created the new annual lecture, as well as the J. David Allen Grant.

“Two years ago, Dr. J. David Allen made a very generous gift to accelerate collaboration, stimulate innovation, and enhance the reputation of Georgia’s three foremost research institutions,” noted Mike Cassidy, president and CEO of the Georgia Research Alliance (GRA), through which the grant is administered.

“Thank you for your gracious gift,” Cassidy said to Allen, who stood to be recognized. “It’s a source of inspiration, creating ripples that will result in a far-reaching impact that we can only imagine.”

The Allen gift to GRA (about $1 million) is being disbursed over a 10-year period in the form of seed grants, and is designed to bolster the partnership between the three universities – each seed grant team must have investigators from each institution. The first awardee is comprised of Todd Sulchek (Georgia Tech), Jim Lauderdale (UGA), and Young-sup Yoon (Emory). Following June’s presentation, Sulchek and Lauderdale took turns presenting their joint research.

Waller reminded the audience to check out the research posters from REM seed grant winners that were on display in the Miller-Ward Alumni House, and emphasized the influence and heft of REM to this point – the development of 17 biotech start-up companies, nearly $121 million in total funds granted to REM’s network of more than 170 faculty members.

Additionally, there were several success stories culled from the ranks of REM seed grants (which require interdisciplinary teams from at least two of the member universities). Muna Qayed of Emory, Lohitash Karumbaiah of UGA, and Temenoff all made presentations of their research, which has been supported by the REM seed grant program.

Officially dubbed the “Georgia Partners in Regenerative Medicine” seed grant, the deadline for submission of proposals for the 2018-2019 cycle is July 9. Winners will be announced in August.

“The impact of these seed grants on building the scientific community in our region cannot be over-emphasized,” said Temenoff, who shared the progress, novel directions, and new collaborations that have stemmed from her original REM seed grant project (“Biomaterials-based Strategies to Modulate Cathespin Activity and Promote Healing of Tendon Overuse Injuries”).

“Looking forward, we want to continue to bring new researchers into REM, to continue building a vibrant forum for scientific exchange,” Temenoff added. “The recent emphasis on cell therapies within the state provides a new means to include even more scientists in the area of basic, applied immunology, translational medicine, and biologics manufacturing within REM.”

]]> Jerry Grillo 1 1527692445 2018-05-30 15:00:45 1527699030 2018-05-30 16:50:30 0 0 news Cancer research pioneer Carl June delivers first J. David Allen Keynote Lecture

]]>
2018-05-30T00:00:00-04:00 2018-05-30T00:00:00-04:00 2018-05-30 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
606592 606595 606594 606593 606591 606592 image <![CDATA[REM leadership]]> image/jpeg 1527691642 2018-05-30 14:47:22 1527691642 2018-05-30 14:47:22 606595 image <![CDATA[REM Carl June]]> image/jpeg 1527691835 2018-05-30 14:50:35 1527691835 2018-05-30 14:50:35 606594 image <![CDATA[REM Krish Roy]]> image/jpeg 1527691762 2018-05-30 14:49:22 1527691762 2018-05-30 14:49:22 606593 image <![CDATA[REM Ned Waller]]> image/jpeg 1527691706 2018-05-30 14:48:26 1527691706 2018-05-30 14:48:26 606591 image <![CDATA[REM June crowded room]]> image/jpeg 1527691018 2018-05-30 14:36:58 1527691180 2018-05-30 14:39:40
<![CDATA[Historic Regenerative Medicine Workshop Wraps Up]]> 28153 This year marked the beginning of one era and the end of another for the Regenerative Medicine Workshop, which wrapped up its 22nd edition on Saturday. For the first time, the workshop was held somewhere other than Hilton Head.

This year, more than 200 engineers, scientists, clinicians, and industry partners from across the planet gathered for the first time at Wild Dunes on Isle of Palms, just north of Charleston. And this year, Bob Guldberg presided over the workshop for the last time, at least for the foreseeable future.

Guldberg, executive director of the Petit Institute for Bioengineering and Bioscience at the Georgia Institute of Technology (the organizing body that launched the workshop in 1997), will begin a new post as vice president and the inaugural director of the Knight Campus for Accelerating Scientific Impact at the University of Oregon in August.

“Each year I wonder how we can possibly top the previous year’s workshop,” says Guldberg. “But this one was special. I’ve been coming to the Regenerative Medicine Workshop since it started. So this has not only been a place to showcase cutting edge research in regenerative medicine through the years. It’s been a community for me and my family. But I’m excited for the future of the workshop in a new location, and look forward to coming back as a participant in coming years.”

In fact, while other hands will guide the event next year, Guldberg is still expected to participate. So says Bob Nerem, founding director of the Petit Institute, who recruited Guldberg to Georgia Tech more than 20 years ago.

“Bob, let me assure you that even though you’re leaving Georgia Tech, and even though you’re not affiliated with the organizing partners of this workshop – you’re way out there in Oregon – we still expect to get work out of you,” Nerem quipped to a jovial crowded room during the conference dinner Friday night. Then he added, “so, Bob, from your friends, thank you for all you’ve done, and for your leadership in moving this workshop forward.”

This year’s workshop, branded ‘Synergizing Science, Engineering, and Clinical Translation,’ drew representatives and researchers (principal investigators, postdocs, and students) from more than two dozen universities and other institutions, including the organizing partners: the Regenerative Engineering and Medicine (REM) research center (a partnership of Georgia Tech, Emory University, and the University of Georgia), the Stem Cell and Regenerative Medicine Center at the University of Wisconsin-Madison, the Mayo Clinic’s Center for Regenerative Medicine as well as its Rehabilitation Medicine Research Center, and the McGowan Institute for Regenerative Medicine at the University of Pittsburgh.

Other partners/sponsors included the Advanced Regenerative Manufacturing Institute (ARMI), Biofabusa, MiMedx, WiCell, Biological Industries USA, ACell, and BioSpherix, Ltd.

Beginning on Wednesday (March 21) with a series of late afternoon presentations, the Sweetgrass Pavilion Conference Center at Wild Dunes was packed hour after hour over the next several days for keynote presentations, as well as other research, including rapid fire sessions for students and other trainees. The highlight on Thursday was a poster session and competition for trainees, won by Georgia Tech postdoc Woojin Han (who is co-advised by Petit Institute researchers Andrés J. García and Young Jang).

An annual highlight of the workshop has always been the Nerem Lecture, an hour long presentation by one of the world’s thought leaders in a particular field. This year it was delivered by Viola Vogel, professor and chair of the Department of Health Science and Technology and principal investigator of the Laboratory of Applied Mechanobiology at the ETH Zürich, Switzerland. 

Her presentation was entitled, “Unraveling the Secrets of How the Mechanobiology of Extracellular Matrix Regulates Cell and Tissue Functions,” but she devoted a portion of her talk broadly to the origins of the field of bioengineering itself. Coming from a background in physics, Vogel leapt into bioengineering in 1990 at the University of Washington, where she became founding director of the Center for Nanotechnology before moving to Switzerland in 2004.

Alluding to something Nerem said years ago, Vogel stressed, “how important it is for people to move from different disciplines into this field of bioengineering.”

Early on, as bioengineering was maturing from its infancy, she noted that engineers had a difficult time understanding, “the biology, the physiology, so they didn’t always pick the right questions to pursue. But I think what we’re seeing now is a younger generation that was educated with two backgrounds, the science and the engineering, and you’re seeing it in the presentations, in how they pick which problems to address, in how they package what they are doing, and that is absolutely crucial for this field to advance.”

There were 50 research presentations over the four-day workshop, including a wide range shared by this new generation of biomedical researchers.  The talks covered such timely topics as immunotherapies, cell manufacturing, engineered hydrogels, DNA barcoding, and organoid model systems and many others.   

So once again, it was a lot of deep diving along the Atlantic coast, into regenerative strategies to improve the human condition. And Guldberg, for one, came from the workshop encouraged and energized.

“The field of regenerative medicine is moving so rapidly both in terms of the science and progress towards therapies that are having a remarkable impact on patients with cancer, degenerative conditions, and traumatic injuries,” Guldberg says. “I love this workshop and look forward to continuing to come each year, because it is the premier meeting to hear the latest progress in regenerative medicine.”

]]> Jerry Grillo 1 1522252058 2018-03-28 15:47:38 1522252122 2018-03-28 15:48:42 0 0 news One era ends as another one begins for the annual gathering of leading regenerative medicine researchers

]]>
2018-03-28T00:00:00-04:00 2018-03-28T00:00:00-04:00 2018-03-28 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
604364 604373 604365 604372 604369 604371 604366 604367 604364 image <![CDATA[The Two Bobs]]> image/jpeg 1522249780 2018-03-28 15:09:40 1522249780 2018-03-28 15:09:40 604373 image <![CDATA[Wild Dunes]]> image/jpeg 1522250964 2018-03-28 15:29:24 1522250964 2018-03-28 15:29:24 604365 image <![CDATA[RMW crowded room]]> image/jpeg 1522249884 2018-03-28 15:11:24 1522249884 2018-03-28 15:11:24 604372 image <![CDATA[Krish discussion]]> image/jpeg 1522250897 2018-03-28 15:28:17 1522250897 2018-03-28 15:28:17 604369 image <![CDATA[Ed Botchwey]]> image/jpeg 1522250393 2018-03-28 15:19:53 1522250393 2018-03-28 15:19:53 604371 image <![CDATA[Woojin Han poster]]> image/jpeg 1522250613 2018-03-28 15:23:33 1522250613 2018-03-28 15:23:33 604366 image <![CDATA[Vogel speaks]]> image/jpeg 1522249960 2018-03-28 15:12:40 1522249960 2018-03-28 15:12:40 604367 image <![CDATA[Posters at RMW]]> image/jpeg 1522250306 2018-03-28 15:18:26 1522250306 2018-03-28 15:18:26
<![CDATA[Woojin Han Selected to Speak]]> 28153 Woojin Han, a postdoctoral researcher at the Georgia Institute of Technology, has been selected to present his research at the PMSE (Polymeric Materials: Science & Engineering) Future Faculty Symposium in Boston this summer.

Han, who is co-advised by two researchers in the Petit Institute for Bioengineering and Bioscience at Georgia Tech, Andrés García and Young Jang, will share his research on engineering synthetic material to deliver muscle stem cells to treat conditions like skeletal muscle injury, or genetic disorders like Duchenne muscular dystrophy.

The symposium, part of the American Chemical Society’s annual meeting (August 19-23), will feature oral presentations by more than 20 postdoctoral scientists working in polymeric materials fields, all of them planning to apply to academic positions during Fall 2018 for appointments beginning in 2019.

The Future Faculty symposium will be spread out among two full days and feature keynote speakers who will share their insights regarding the academic career path. Selected speakers are also invited to participate in a reception to meet each other, keynote speakers and invited guests from academia and other sectors.

“The symposium is a great opportunity for me,” says Han, who earned his PhD at the University of Pennsylvania. “This fall will be an exciting transitionary period for me, a time when I start networking more aggressively. I’m really interested in moving forward with my career in academia and starting my own lab.”

 

]]> Jerry Grillo 1 1521136272 2018-03-15 17:51:12 1521749295 2018-03-22 20:08:15 0 0 news Postdoctoral researcher from García lab invited to present research at PMSE symposium

]]>
2018-03-15T00:00:00-04:00 2018-03-15T00:00:00-04:00 2018-03-15 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
603855 603855 image <![CDATA[Woojin Han]]> image/jpeg 1521136148 2018-03-15 17:49:08 1521136148 2018-03-15 17:49:08
<![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[Guldberg Gets Top Georgia Bio Award]]> 28153 Bob Guldberg, executive director of the Petit institute for Bioengineering and Bioscience at Georgia Institute of Technology, took center stage Thursday night at the 2018 Georgia Bio Life Science Health Impact Awards Gala at the Cobb Energy Center.

Guldberg, along with James Wehenmeyer, vice president of research and economic development at Georgia State University, received the Industry Growth Awards, the highest honors bestowed each year by Georgia Bio, the state’s life science advocacy and business association, now in its 20th year. The award recognizes individuals in the public and private sectors who have made extraordinary contributions to the growth of Georgia’s life sciences industry.

“This award from Georgia Bio is a great honor and really a recognition of the efforts of the entire Petit Institute team,” Guldberg said. “It’s remarkable how many more start-ups are being launched now compared to 10 or 20 years ago. I am so proud of the collaborative entrepreneurial culture that we have built, where our students and faculty increasingly expect to successfully translate their lab work into commercial products and new clinical therapies.”

Georgia Tech and the Petit Institute were well represented at the awards podium as nearly 300 of the state’s life science industry leaders gathered to celebrate the contribution and achievements of people and organizations.

In addition to Guldberg, other award winners with Tech connections included Sherry Farrugia (chief operating and strategy officer of the Pediatric Technology Center, a partnership of Georgia Tech and Children’s Healthcare of Atlanta), who won a Community Award, and the NSF Engineering Center for Cell Manufacturing Technologies (CMaT) at Georgia Tech, which won a Deal of the Year Award.

“We are excited to recognize the individuals and organizations improving and saving lives worldwide through their healthcare innovations and leadership here in Georgia.” said Russell Allen, president and CEO of Georgia Bio.

Here’s a list of the 2018 Georgia Bio Life Sciences Health Impact Award winners:

 

GEORGIA BIO INDUSTRY GROWTH AWARDS: Presented to two people who have made an extraordinary contribution to the growth of the life sciences industry in Georgia.

Robert E. Guldberg, Ph.D., The Petit Director's Chair in Bioengineering and Bioscience; Executive Director, Parker H. Petit Institute for Bioengineering and Bioscience; and Professor, George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology

James Weyhenmeyer, Ph.D., VP Research & Economic Development, Georgia State University and Chairman, GSU Research Foundation Inc.

 

PHOENIX AWARD: Presented to two Georgia honorees who have forged academic and industry relationships that will drive translation and lead to new treatments and cures. This award is sponsored by the Metro Atlanta Chamber.

• UGA Center for Vaccines and Immunology / Sanofi Pasteur

 

DEALS OF THE YEAR AWARDS: Presented to one or more companies or institutions for the most significant financial or commercial transactions closed from November 1, 2016-October 31, 2017, based on the importance of the transaction to Georgia’s life sciences industry.

• Center for Biotechnology and Genomic Medicine at Augusta University

CryoLife

• Femasys

Georgia Clinical & Translational Science Alliance

• NSF Engineering Research Center for Cell Manufacturing Technologies (CMaT) at Georgia Tech

Vertera Spine

 

COMMUNITY AWARDS: Presented to a small number of individuals, companies or institutions whose contributions to Georgia’s life sciences community are worthy of special recognition.

Sherry N. Farrugia, Chief Operating and Strategy Officer, Pediatric Technology Center, Georgia Institute of Technology; Director, Children’s Healthcare of Atlanta Partnership

Christopher D. McKinney, DA, MBA, Associate Vice President, Innovation Commercialization; Adjunct Professor of Political Science, Augusta University

• Center for Tropical and Emerging Global Diseases

• Suzanne Prichett, Field Sales Manager - Education & Medical Research Division, VWR International LLC

• Atlanta Center for Medical Research

 

INNOVATION AWARDS: Presented to the department, institution, company or individuals who are forging new ground by thinking outside traditional paradigms to create some unique technology.

• Aruna Biomedical

George Hsu, M.D., Chief Medical Officer / Interim CEO, Cathaid Inc.

James Ross, Ph.D., Chief Technology Officer, Axion BioSystems

• PanXome

 

EMERGING LEADER OF THE YEAR AWARDS: Presented to young individuals who have made a significant impact on the life sciences industry through their studies or employment.

Ashley Bohn, Ph.D, M.S., R.V.T., Georgia State University

Tami Hutto, MSPP, Program Manager – Emory University and Georgia Institute of Technology, Atlanta BEST Program

 

TEACHER OF THE YEAR AWARD: Presented to a Georgia biotechnology high school teacher who exhibits excellence in STEM teaching and support for the biotechnology pathway.

William E. Schuyler, Forsyth Central High School

 

For a list of past Georgia Bio Industry Growth Award recipients, click here.

 

 

]]> Jerry Grillo 1 1518807203 2018-02-16 18:53:23 1518807235 2018-02-16 18:53:55 0 0 news Petit Institute executive director among Georgia Tech winners at annual life sciences industry gala

]]>
2018-02-16T00:00:00-05:00 2018-02-16T00:00:00-05:00 2018-02-16 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
602484 602485 602488 602484 image <![CDATA[Guldberg Georgia Bio]]> image/jpeg 1518805410 2018-02-16 18:23:30 1518805410 2018-02-16 18:23:30 602485 image <![CDATA[Guldberg sign]]> image/jpeg 1518805470 2018-02-16 18:24:30 1518805470 2018-02-16 18:24:30 602488 image <![CDATA[CMaT student]]> image/jpeg 1518805903 2018-02-16 18:31:43 1518805903 2018-02-16 18:31:43
<![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[First Allen Grant Announced]]> 28153 Todd Sulchek, researcher in the Petit Institute for Bioengineering and Bioscience at the Georgia Institute of Technology, is a principle investigator on the team that has won the inaugural award of the J. David Allen Seed Grant Program in Cellular Manufacturing.

Sulchek, an associate professor of bioengineering in the Woodruff School of Mechanical Engineering, shares the $90,000 grant with his collaborators, Young-sup Yoon from Emory University and James Lauderdale from the University of Georgia (UGA).

The title of their research is, “Microfluidic Technologies for Improved Engineering of iPSCs and Their Applications to Treating Corneal and Lymphatic Diseases.” The goal is to analyze and validate a mechanical method to deliver reprogramming factors to human somatic cells, creating induced pluripotent stem cells (iPSCs) that would potentially be used, down the road, in the treatment of lymphedema, cancer, and eye disorders.

Earlier this year, Dr. Allen announced a $1 million gift that would be disbursed over a 10-year period, to the Georgia Research Alliance to advance cell-manufacturing related research and development at Emory, Georgia Tech, and UGA. The gift creates a unique partnership designed to accelerate collaboration, stimulate innovation, and help establish Georgia as a leader in cellular manufacturing.

Seed grant teams must have researchers from each institution, and the award is divided equally between the three schools. The award supports collaborative projects with a high potential for clinical or industry translation at earlier stages of development.

 

 

 

]]> Jerry Grillo 1 1504295603 2017-09-01 19:53:23 1513694554 2017-12-19 14:42:34 0 0 news Petit Institute’s Todd Sulchek part of research team to win new seed grant

]]>
2017-09-01T00:00:00-04:00 2017-09-01T00:00:00-04:00 2017-09-01 00:00:00 595446 595446 image <![CDATA[Todd Sulchek]]> image/jpeg 1504295247 2017-09-01 19:47:27 1504295247 2017-09-01 19:47:27
<![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[The Winning Team]]> 28153 Tennis elbow, a pulled hamstring, shin splints or an ankle sprain. Most of us have dealt with common sports injuries in an attempt to get in shape. Petit Institute researchers who are faculty at Georgia Tech’s College of Engineering are laser focused on providing sports medicine for even the most common injury.

Sports medicine today has become a specialized field with many facets. No longer just a study of orthopedics, sports medicine now encompasses new therapies and technologies that tackle all sorts of sports-related injuries and diseases, leveraging predictive analytics and wearables to keep athletes performing at their best.  

Tech’s sports medicine research program continues to grow, led by faculty such as Robert Guldberg, Omer Inan, Michelle LaPlaca and Johnna Temenoff, all leaders at the top of the field, all of them researchers in the Petit Institute. Guldberg is a professor in the Woodruff School of Mechanical Engineering. Inan is assistant professor in the School of Electrical and Computer Engineering. Temenoff is a professor in the Wallace H. Coulter Department of Biomedical Engineering, where LaPlaca is an associate professor.

Each of these engineers has made impactful contributions to sports medicine research, and their work is already seeing real-world application today. Each one of them is driven by a common desire to enhance the quality of life of athletes, both on and off the field. And even occasional exercisers can reap the benefits. 

Read the whole story in the CoE magazine right here.

]]> Jerry Grillo 1 1512158238 2017-12-01 19:57:18 1512162169 2017-12-01 21:02:49 0 0 news Petit Institute researchers from CoE leading Tech's sports medicine program

]]>
2017-12-01T00:00:00-05:00 2017-12-01T00:00:00-05:00 2017-12-01 00:00:00 599427 599427 image <![CDATA[Michelle LaPlaca DETECT]]> image/jpeg 1512161811 2017-12-01 20:56:51 1512161811 2017-12-01 20:56:51
<![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[The Next Frontier in Medicine ]]> 27513 Genetic testing today is mainstream, marketing to consumers who want to know where in Europe they came from or what types of hereditary diseases they could develop. For around $200 you can trace your family tree to learn your origins or identify genetic abnormalities that could signal disease. James Dahlman, assistant professor in the College of Engineering’s biomedical engineering department, specializes in genetics and believes these genotyping services can be helpful, as long as they are used responsibly.

“If you’re going to start making medical predictions, you have to be careful,” said Dahlman. “Most people are not equipped to interpret statistics correctly, which can lead to negative predicting and ethical dilemmas. In a few years, genetic counselors will be in high demand so folks can make better decisions about their health.”

Dahlman is fascinated by genetics, citing gene therapy as the most interesting field in the world. And it’s a field that he is revolutionizing through his research. Gene therapy is an experimental technique that uses genes to treat or prevent diseases, including hemophilia, Parkinson’s, cancer and HIV. It can help manage a number of diseases by leveraging genes instead of drugs or surgery. Although gene therapy shows promise, there are still risks involved, including unwanted immune system reactions or the risk of the wrong cells being targeted. That’s where Dahlman’s research comes in.

Dahlman’s lab focuses on drug delivery vehicles, which are nanoparticles. The nanoparticle delivers gene therapies to the right place in the body to fight disease. It’s critical that the gene therapies only target the unhealthy cells to avoid damaging healthy ones. Dahlman is laser focused on ensuring the nanoparticles know what paths to take to reach the correct organ to start the healing process.

“The issue with genetically-engineered drugs is that they don’t work unless they get to the right cell in the body,” said Dahlman. “You can have the world’s best genetic drug that's going to fix a tumor or eradicate plaque, but it’s not going to be effective unless it travels to the right organ. In my lab, we design different nanoparticles to deliver the genetically-engineered drugs to the correct location.”

Dahlman is redefining the field of genetic therapy with a testing system he invented called FIND (Fast Identifiable Nanoparticle Delivery). During the course of identifying effective nanoparticles for drug delivery, thousands of nanoparticles must be tested, which presents scalability issues. Mice must be used for the tests because a cell plate isn’t going to replicate organs in the human body. But ethically, researchers cannot inject thousands of mice for an experiment of this magnitude. So Dahlman developed a testing system that leverages DNA barcodes (a stand-in for the actual drugs) to label each nanoparticle. Once those are injected, researchers can see where the barcodes went in the mouse. For example, if a significant number of barcodes numbered 30 all went to the heart, Dahlman can deduce that the nanoparticle represented by barcode 30 is best suited for that organ.

“The barcode system is redefining our field because we are now able to do several really important things that we couldn’t before,” said Dahlman. “First, we can test thousands of nanoparticles at once, which has been a pipedream in our field forever. Second, we can now study the biology of drug delivery, understanding which genes affect how well a drug will work. And third, we can apply big data and artificial intelligence to drug delivery for the first time. With thousands of nanoparticles being tested at once, we can mine giant data sets for bioinformatics.”

Dahlman’s research and barcoding system has universal implications; he is designing testing systems that everyone can use. Labs across the country can leverage FIND to accelerate their studies. If the technology is used by more labs, Dahlman believes it will increase the rate that gene therapies are developed, advancing the entire field.

Non-liver gene therapy delivery is one of the biggest challenges today that Dahlman hopes to contribute to with his work. The liver has been easier to target with gene therapy because of its filtration system; larger blood vessels let the nanoparticles pass more easily into the organ. Diseases such as hepatitis and cirrhosis have responded well to gene therapies.

“All types of drug therapies have to be delivered,” said Dahlman. “Our field has had the best success with the liver, with 15 clinical trials already successfully running using the same nanoparticle delivery mechanism. The liver is responding extremely well to these therapies, and we are healing livers and curing people. The next frontier will be organs other than the liver, like the heart and brain with tighter blood vessel systems.”

After 12 months, Dahlman’s lab is coming into its own with nine graduate students and four undergraduates working in the lab. Much of the energy and excitement in Dahlman’s lab come from the fact that he is a young professor.  He’s only 30 and easily relates to the students in his lab. Dahlman’s had great experiences with faculty members too and describes senior faculty as extremely helpful and supportive.

“I’ve gotten carded at two different faculty events, and I’ve been mistaken as a student more times than I can count!” said Dahlman. “But there haven’t been any challenges here with being a young faculty member. That was one thing that really attracted me to Tech – the young faculty seemed truly happy here.”

Dahlman’s personal and professional goal is to be working on high risk, innovative science for as long as possible. He never wants to be in a rut, and he wants to have the courage to pursue interesting and risky science even if lower risk work is safer. In his lab, his goal is to produce great students. In 20 years, he wants to see his students as leaders in the field.

“I want people to say, ‘That person went through the Dahlman lab, we have to hire them,’” said Dahlman. “That’s what I want for my students, and I think we can get there.”

 

By Georgia Parmelee

]]> Walter Rich 1 1507748655 2017-10-11 19:04:15 1507898496 2017-10-13 12:41:36 0 0 news 2017-10-11T00:00:00-04:00 2017-10-11T00:00:00-04:00 2017-10-11 00:00:00 Walter Rich

]]>
597243 597247 597245 597243 image <![CDATA[James Dahlman, assistant professor, in the Wallace H. Coulter Department of Biomedical Engineering]]> image/jpeg 1507748416 2017-10-11 19:00:16 1507748749 2017-10-11 19:05:49 597247 image <![CDATA[James Dahlman Lab - microscope]]> image/jpeg 1507748519 2017-10-11 19:01:59 1507748519 2017-10-11 19:01:59 597245 image <![CDATA[James Dahlman Lab - students]]> image/jpeg 1507748482 2017-10-11 19:01:22 1507748482 2017-10-11 19:01:22
<![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[Testing Boosts Gene Therapies]]> 28153 Using tiny snippets of DNA as “barcodes,” researchers have developed a new technique for rapidly screening nanoparticles for their ability to selectively deliver therapeutic genes to specific organs of the body. The technique could accelerate the use of gene therapies for such killers as heart disease, cancer, and Parkinson’s disease.

Genetic therapies, such as those made from DNA or RNA, are difficult to deliver into the right cells in the body. For the past 20 years, scientists have been developing nanoparticles made from a broad range of materials and adding compounds such as cholesterol to help carry these therapeutic agents into cells. But the nano­particle carriers must undergo time-consuming testing — first in cell culture, then in animals. With millions of possible formulas, identifying the optimal nanoparticle to target each organ has been challenging.

Using DNA strands just 58 nucleotides long, researchers from Georgia Tech, the University of Florida, and the Massachusetts Institute of Technology (MIT) have developed a new evaluation technique that skips the cell culture testing altogether — and could allow hundreds of different types of nanoparticles to be tested simultaneously in just a handful of animals.

Read the entire story in Research Horizons.

]]> Jerry Grillo 1 1501613215 2017-08-01 18:46:55 1507554750 2017-10-09 13:12:30 0 0 news Researchers use DNA "barcodes" to develop new technique for rapidly screening nanoparticles

]]>
2017-08-01T00:00:00-04:00 2017-08-01T00:00:00-04:00 2017-08-01 00:00:00 593916 593916 image <![CDATA[James Dahlman]]> image/jpeg 1501612748 2017-08-01 18:39:08 1501612748 2017-08-01 18:39:08
<![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[REM Nurturing Collaborative Research]]> 28153 Thirty years ago, long before there was a Petit Institute for Bioengineering and Bioscience, biomedical research in Georgia experienced a big bang with establishment of the Emory/Georgia Tech Biomedical Technology Research Center and a seed grant program that nurtured faculty interest in collaboration.

The seed grant required researchers from the two different institutions to bring their respective strengths together on projects designed to improve human health.

“That seed grant program was absolutely essential and its influence on success of the Emory-Georgia Tech partnership can’t be underestimated,” said Bob Nerem, founding director of the Petit Institute, which launched in 1995. “It provided a foundation for everything else that has taken place since.”

That spirit of partnership has evolved in many ways, one of the most significant being the Regenerative Engineering and Medicine (REM) research center, which brought the University of Georgia (UGA) and its research strength into the mix with Emory and Georgia Tech, and the seed grant program remains as critical as ever, sparking multidisciplinary collaboration between the three institutions.

Recently, the REM research center awarded this year’s seed grants, totaling $700,000 for eight teams of interdisciplinary researchers working to harness the body’s own potential to heal or regenerate in response to injury or disease.

Here’s a rundown of the projects:

• Project Title: Using Pluripotent Stem Cells to Treat Male-Factor Infertility: Towards a Potential Regenerative Medicine Strategy.

• Principal Investigators: Charles Easley (UGA) and Anthony Chan (Emory)

• Synopsis: The researchers are developing a novel approach for differentiating human embryonic stem cells and induced pluripotent stem cells into advanced spermatogenic lineages. If successful, this proposal would show that functional male gametes can be derived from no greater starting material than a skin biopsy.

 

• Project Title: Identification of Molecular and Epigenetic Signatures of Cell Potency and Enhanced Embryonic Stem Cell Reprogramming for Regenerative Biomanufacturing.

• Principal Investigators: Rabindranath De La Fuente (UGA) and Yuhong Fan (Tech)

• Synopsis: The researchers propose to use their recently-developed episomal sensor of histone H4 acetylation to conduct a quantitative analysis of changes in genome-wide chromatin structure and the dynamics of epigenetic reprogramming in real-time. Their studies will provide a genome-wide map of the chromatin regulatory landscape and transcriptome profiles with predictive value to evaluate potency for neuronal differentiation.

 

• Project Title: Hydrogels for Mesenchymal Stem Cells to Treat Graft-vs-Host Disease.

• Principal Investigators: Andrés J. García (Tech), Muna Qayed (Emory)

• Synopsis: The objective of this project is to engineer synthetic hydrogels that encapsulate Mesenchymal stem cells (MSCs) and promote their survival and expansion in alternative transplant sites, resulting in enhanced immunomodulatory activities for the treatment of Graft vs Host Disease.

 

• Project Title: Elucidating Natural Killer Cells as a Cell Therapy for Parkinson’s Disease.

• Principal Investigators: Jae-Kyung (Jamise) Lee (UGA) and Levi Wood (Tech)

• Synopsis: The research team’s goal is to obtain new data determining if Natural Killer cells have the capacity to protect neurons and modulate microglial activation in the context of a Alpha-synuclein.

 

• Project Title: Exploring Mechanosensitive Cues to Enhance Mitochondrial Structure and Function During Regeneration.

• Principal Investigators: Jarrod Call (UGA) and Khalid Salaita (Emory)

• Synopsis: The primary goal is to determine mechanical signals that influence mitochondrial structure and function. The central hypothesis is that mitochondria structure and function are sensitive to mechanical stimuli.

 

• Project Title: Pro-Regenerative Immunomodulatory Therapies to Repair Volumetric Muscle Injuries.

• Principal Investigators: Nick Willett (Emory) and Edward Botchwey (Tech)

• Synopsis: The researchers’ proposed research will investigate whether engineered immune modulatory nanofibers will enhance regenerative capacity in rodent models of volumetric muscle loss (VML).

 

• Project Title: Condyle Regenerative Medicine.

• Principal Investigators: Scott Hollister (Tech), Shelly Abramowicz and Steven Goudy (Emory)

• Synopsis: The researchers want to develop a patient specific, 3D printed biologic pedicled flap approach to temporomandibular joint (TMJ) reconstruction. Specifically, they’ll design and optimize a porous mandibular condyle scaffold, fabricate the condyle from polycaprolactone (PCL) using 3D printing laser sintering, adsorb bone morphogenetic protein 2 (BMP2) to the scaffold and implant the scaffold in the temporalis muscle. After attaining vascularization and bone growth in the construct at 1 month, the locally pedicled tissue engineered flap will be surgically positioned to function as a mandibular condyle.

 

• Project Title: Skeletal Muscle Stem Cells are Novel Regulators of Collateral Blood Vessel Formation.

• Principal Investigators: Laura Hansen and Robert Taylor (Emory), and Young Jang (Tech)

• Synopsis: This project will determine if satellite cells are crucial to collateral vessel formation in a murine peripheral arterial disease (PAD) model as well as investigate if the positive effects of exercise therapy are dependent on satellite cells. The potential ability of satellite cells to restore blood flow through vascular growth and repair muscles damaged from ischemic myopathy makes them a promising and novel therapy for patients with PAD and critical limb ischemia.

]]> Jerry Grillo 1 1506700922 2017-09-29 16:02:02 1506700922 2017-09-29 16:02:02 0 0 news Seed Grants awarded to eight interdisciplinary teams from Georgia Tech, Emory, and UGA

]]>
2017-09-29T00:00:00-04:00 2017-09-29T00:00:00-04:00 2017-09-29 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
596683 596683 image <![CDATA[REM leadership]]> image/jpeg 1506700523 2017-09-29 15:55:23 1506700523 2017-09-29 15:55:23
<![CDATA[Georgia Clinical & Translational Science Alliance Receives $51 Million NIH Grant]]> 27303 After a decade of research collaboration, the Atlanta Clinical & Translational Science Institute (ACTSI) will welcome a new partner and change its name, reflecting a new statewide focus. The University of Georgia will officially become the fourth academic partner, and ACTSI will now be known as the Georgia Clinical & Translational Science Alliance (Georgia CTSA).

This alliance is celebrating 10 years of research advancement by expanding across the state through a five-year, $51 million Clinical and Translational Science Award (CTSA) from the National Institutes of Health (NIH). The Emory University-led Georgia CTSA will focus on transforming the quality and value of clinical research and translating research results into better outcomes for patients.

The Georgia CTSA unites the strengths of its academic partners: Emory University, Morehouse School of Medicine, the Georgia Institute of Technology, and the University of Georgia. Emory is a national leader in health care and biomedical research as well as an outstanding leader in clinical and translational research training and education. Morehouse School of Medicine is a nationally recognized historically black institution that brings ethnic diversity to biomedical research, addresses health disparities through successful community engagement research, and serves as a pipeline for training minority researchers. Georgia Tech is a national leader in biomedical engineering, bioinformatics  and the application of innovative systems engineering to health care solutions. The University of Georgia has a proven track record in outstanding basic and translational research and, as the state’s land grant institution, offers a robust statewide network that enhances community outreach, service and research.

“Continuing such an alliance and involving these leading state institutions is extremely important and in line with Georgia’s goals for the promotion of clinical and translational research, innovation and development,” said Georgia Governor Nathan Deal. “Having an active Clinical & Translational Science awardee in Georgia has brought our citizens cutting edge cures and the latest in clinical and translational research.”

Georgia CTSA is one of 64 Clinical and Translational Science Awards (CTSA) at major academic medical centers across the country, funded by the National Institutes of Health’s National Center for Advancing Translational Science, and it is the only CTSA in Georgia. The award will fund cores focused on improving quality, efficiency and collaboration of the research process; provide consultative support and new tools in informatics and biostatistics; pilot funding for new research projects, training and workforce development, while integrating special populations and focusing on participant interactions, and creating local centers tackling clinical trial inefficiencies.

The Georgia CTSA welcomes contact principal investigator (PI) at Emory, W. Robert Taylor, M.D., Ph.D., and a new multi-PI leadership structure: 

W. Robert Taylor, MD, PhD
Contact Principal Investigator, Georgia CTSA
Interim Chair, Department of Medicine
Director, Division of Cardiology
Marcus Chair in Vascular Medicine
Professor of Medicine and Biomedical Engineering
Emory University School of Medicine

Elizabeth O. Ofili, MD, MPH
Principal Investigator, Morehouse School of Medicine, Georgia CTSA 
Professor of Medicine, Cardiology
Senior Associate Dean of Clinical and Translational Research
Director, Clinical Research Center 
Morehouse School of Medicine

Andrés J. García, PhD
Principal Investigator, Georgia Institute of Technology, Georgia CTSA 
Rae S. and Frank H. Neely Endowed Chair and Regents' Professor, Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience
Director, Interdisciplinary Bioengineering Graduate Program
Georgia Institute of Technology

Bradley G. Phillips, PharmD, BCPS, FCCP
Principal Investigator, University of Georgia, Georgia CTSA
Millikan-Reeve Professor and Head, Clinical & Administrative Pharmacy, College of Pharmacy 
Director, Clinical and Translational Research Unit (CTRU), Office of Research
University of Georgia

Henry M. Blumberg, MD
Principal Investigator, KL2 and TL1, Georgia CTSA
Professor of Medicine and Epidemiology, Division of Infectious Diseases
Emory University School of Medicine

"The Georgia CTSA creates a unique opportunity for synergy among historic partners in health care, education, and cutting edge research, and has emerged as an innovative and integrated environment where clinical and translational researchers can flourish," said Taylor. "The Georgia CTSA is a catalyst and incubator for clinical and translational research across the state, with impacts throughout the Southeast and nation."

The Georgia CTSA has improved health care and research for Georgia citizens through collaboration with the Grady Health System, Children’s Healthcare of Atlanta, Atlanta VA Medical Center, Georgia Research Alliance, Georgia Bio, and multiple community medical groups throughout the state. 

"Georgia CTSA continues established, strong clinical and research partnerships by leveraging the infrastructure support of the NIH-funded Research Centers at Minority Institutions (RCMI) at Morehouse School of Medicine. We will continue to implement innovative patient centered and participatory care delivery models, toward the elimination of health disparities," said Ofili.

“Georgia CTSA’s innovative support of discovery and collaborative partnerships help to rapidly translate scientific discoveries and new technology, which positively impacts patient care in Georgia. This is an exciting story for the state,” said Garcia. “The new alliance is improving health care and clinical research for the citizens of Georgia and continues to create synergies that foster and accelerate new and emerging technologies and discoveries.”

“The addition of the University of Georgia provides the Georgia CTSA a statewide footprint to connect with every county in the state to address health and wellness needs, particularly in rural and underserved populations; opportunities for continued excellence in research by strengthening existing and expanding new research collaborations; and the ability to enrich interprofessional education to include students and trainees from pharmacy and other disciplines so that they can learn how to work together as a team to discover new approaches and treatments that improve health and patient care,” said Phillips. “As a new member of the Georgia CTSA, faculty and students across our campus will have unique support and infrastructure that builds upon current capabilities and increases our trajectory in fostering clinical and translational science in the state and beyond.” 

Written by Georgia Clinical & Translational Science Alliance.

For more information, please visit www.GeorgiaCTSA.org  

]]> John Toon 1 1506305224 2017-09-25 02:07:04 1506603302 2017-09-28 12:55:02 0 0 news After a decade of research collaboration, the Atlanta Clinical & Translational Science Institute (ACTSI) will welcome a new partner and change its name, reflecting a new statewide focus. The University of Georgia will officially become the fourth academic partner, and ACTSI will now be known as the Georgia Clinical & Translational Science Alliance (Georgia CTSA). This alliance is celebrating 10 years of research advancement by expanding across the state through a five-year, $51 million Clinical and Translational Science Award (CTSA) from the National Institutes of Health (NIH). 

]]>
2017-09-25T00:00:00-04:00 2017-09-25T00:00:00-04:00 2017-09-25 00:00:00 John Toon

Research News

(404) 894-6986

]]>
596398 596399 596446 596398 image <![CDATA[Andres Garcia and diabetes research]]> image/jpeg 1506304217 2017-09-25 01:50:17 1506304641 2017-09-25 01:57:21 596399 image <![CDATA[Andres Garcia and diabetes research2]]> image/jpeg 1506304606 2017-09-25 01:56:46 1506304606 2017-09-25 01:56:46 596446 image <![CDATA[W. Robert Taylor]]> image/jpeg 1506363789 2017-09-25 18:23:09 1506363789 2017-09-25 18:23:09
<![CDATA[REM Leveraging its Success]]> 28153 When the Center for Regenerative Engineering and Medicine (REM) gathered for its annual retreat and workshop at the University of Georgia (UGA) in May, there were the usual conversations about interdisciplinary research between the organization’s three partner universities (Georgia Institute of Technology, Emory University, and UGA).

But this time, there was some added excitement over future potential with the introduction of an innovative, additional granting mechanism from the Georgia Research Alliance (GRA). The Allen Fund, a $1 million gift (from Dr. J. David Allen and family), to be disbursed over 10 years to the GRA to advance cellular manufacturing research and development at REM’s three member institutions.

“The Allen gift represents a significant milestone in the partnership between Emory, Georgia Tech, and the University of Georgia, in that this is the first gift that was given to the Georgia Research Alliance specifically to be split between all three partner institutions to support joint projects,” said Johnna Temenoff, Petit Institute researcher and REM co-director from Georgia Tech.

“I believe this is reflective of our collective leadership in the field of regenerative medicine as a whole, and cell manufacturing in particular,” Temenoff added.

Since 2011, the REM has fostered the fundamental transformation of treatment options and outcomes for human disease and injuries through the development and translation of new technologies that boost the body’s ability to heal itself.

REM’s roots actually go back 30 years, to 1987 when Emory and Georgia Tech forged a historic alliance with creation of the Emory/Georgia Tech Biomedical Technology Research Center. That partnership evolved in 1998 with creation of the Georgia Tech/Emory Center or the Engineering of Living Tissues (GTEC, a National Science Foundation Engineering Research Center).

GTEC evolved to become REM, an Emory-Georgia Tech initiative until 2014, when UGA and its vaunted Regenerative Bioscience Center (RBC) joined.

“The purpose of these retreats has always been about getting people together from the three institutions to continue the discussion of how we can collaborate on our research,” says Steve Stice, the REM co-director from UGA, where he is a professor and director of the RBC, and a researcher with the Petit Institute for Bioengineering and Bioscience at Georgia Tech. “We’ve been doing this for some time now and some fantastic things that have come from it, and we expect more of that going forward.”

Participants in this year’s retreat also set out to demonstrate examples of commercial and academic success as a way to highlight the impact that traditional REM seed grants have had on fostering collaborative research and commercial translation across the state.

“We then tried to capture that momentum to establish more intra-institutional collaborations through interactions at the poster and speed dating sessions, and make next year’s grants even more successful,” Temenoff said.

Several accounts were shared from fledgling companies that either emerged from REM collaborations, or were assisted by REM interactions and grants, or exist within the realm of regenerative medicine in general. The companies – Sanguina, ArunA Biomedical, and Cambium Medical Technologies – represent the kind of success stories that REM, and its seed grants program, was built for. Since 2010 (a year before the actual launch of the REM center), the seed grants have resulted in nearly $18 million in leveraged funding (a return on investment of over 3:1).

In her presentation on the ‘academic path to success,’ Petit Institute researcher Susan Thomas emphasized the importance of the REM seed grant she and Emory researcher Ian Copland (who passed away suddenly in July 2015) received in 2014-2015. “That grant has helped us expand our research,” she said. “It opened up doors to new directions and additional funding and to new avenues that we’re still exploring.”

Representatives from each REM institution also had a chance to present their distinct SWOT (Strengths, Weaknesses, Opportunities, Threats) analyses before adjourning for a poster session featuring research from previous seed grant winners. Then they moved onto a ‘research speed dating’ networking session. (“Now is the time to practice your elevator speech,” quipped Stice, who wore a referee’s whistle around his neck and used it to move the conversations along).

That was followed by a ‘wrap session,’ to discuss the REM seed grants and how best to utilize those going forward. Seed grant applications for the coming year are due in July.

Ultimately, the goal is to keep the momentum going, and that may include expanding the partnership that currently comprises REM. Present for last month’s gathering were representatives from Augusta University (AU), including David Hess, dean of AU’s Medical College of Georgia.

“It’s great to see this become a broader group,” said Ned Waller, REM co-director from Emory. “The spirit of REM is to encourage collaboration, and a Georgia-wide initiative is to the benefit of everyone.”

 

REM Center

]]> Jerry Grillo 1 1498137850 2017-06-22 13:24:10 1498248856 2017-06-23 20:14:16 0 0 news Annual gathering of regenerative medicine researchers looks to new opportunities

]]>
2017-06-22T00:00:00-04:00 2017-06-22T00:00:00-04:00 2017-06-22 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
592874 592875 592876 592874 image <![CDATA[REM Athens intro]]> image/jpeg 1498135554 2017-06-22 12:45:54 1498135554 2017-06-22 12:45:54 592875 image <![CDATA[REM directors]]> image/jpeg 1498135785 2017-06-22 12:49:45 1498135785 2017-06-22 12:49:45 592876 image <![CDATA[REM poster session]]> image/jpeg 1498136948 2017-06-22 13:09:08 1498136948 2017-06-22 13:09:08
<![CDATA[Regenerative Medicine Workshop, Part 21]]> 28153 The Regenerative Medicine Workshop at Hilton Head began its third decade with a long and diverse lineup of researchers who presented their latest work on a spacious range of topics, from DNA barcoded technology to strategies to reverse tissue degeneration in rotator cuff injuries.

In other words, the usual dizzying array of up-to-the-minute research from some of the world’s leading scientists and engineers.

But if there was a topical theme to last week’s 21st annual workshop (March 1-4), it was immunology.

“The Hilton Head summit has always been a place where you can learn about the great, late breaking innovations in regenerative medicine,” says Ned Waller, professor in the Emory University School of Medicine, and a researcher with the Petit Institute for Bioengineering and Bioscience at Georgia Tech. “What's striking this year is, half the talks are about immunology.”

And that suits Waller just fine. He is director of the Division of Stem Cell Transplantation and Immunotherapy at the Winship Cancer Institute of Emory, where he also directs the Bone Marrow and Stem Cell Transplant Center. And his research presentation at Hilton Head was entitled, “Another Arrow in the Anti-cancer Quiver: VIP Immunotherapy.”

Waller also is one of three co-directors of the Regenerative Engineering and Medicine (REM) research center, a consortium of research institutes in Georgia: Emory, Georgia Tech, and the University of Georgia. REM is one of four organizing partners of the workshop, the others being the Stem Cell and Regenerative Medicine Center at the University of Wisconsin, the Mayo Clinic’s Center for Regenerative Medicine, and the McGowan Institute for Regenerative Medicine at the University of Pittsburgh.

Accordingly, faculty, post-doctoral, and student researchers from those institutions were well represented. But the workshop also drew researchers from across the spectrum and the planet. Among the speakers were Ronald Germain from the National Institutes of Health, Molly Stevens from Imperial College in London, and Rolando Gittens, who earned his Ph.D. in bioengineering at Georgia Tech in 2012 and is now a research scientist at the Institute for Scientific Research and High Technology Services of Panama.

 

Deep Roster of Research

There were also deep-dive presentations from researchers based at Duke, Harvard, Tufts, and Yale universities, among others, and Jeff Hubbell, the Nerem Lecturer from the University of Chicago (who delivered a talk on “Biomolecular Engineering in Regenerative Medicine and Immunotherapies”).

Steve Stice, as co-director of the REM from the University of Georgia (UGA), the newest member of the consortium, appreciated the geographic range of work that was presented.

“One of the nice things this year is that UGA and other institutions are well represented,” says Stice, professor and director of the Regenerative Bioscience Center at UGA and a Petit Institute researcher. “So it’s not just Emory and Georgia Tech, it’s also Mayo, and Wisconsin, and Pittsburgh, and we’ve brought in speakers from all over. It’s really grown and become a highly recommended event in the regenerative medicine community.”

Trainees – postdocs, grad students, and at least one undergraduate – had a chance to present their work, also. First there were rapid fire presentations (5 minutes) on Thursday afternoon, then a research poster competition that night, featuring 65 different projects on display.

The winning poster came from Daniel Hachim, a grad student at the University of Pittsburgh, whose project is entitled, “Unveiling Macrophage Populations and Mechanisms Driving the Better Remodeling Outcomes Associated with Shifting Phenotype in the Host Response Against Biomaterials.”

 

Going Live

Cheryl San Emeterio, a Ph.D. student at Georgia Tech, has presented posters the last three years at this event, but this was her first rapid fire presentation.

“I thought it was flattering and inspiring, to talk among so many distinguished scientists here,” says San Emeterio, who does her research in the lab of Ed Botchwey, associate professor in the Wallace H. Coulter Department of Biomedical Engineering (a joint department of Emory and Georgia Tech).

“It’s great to get my work out there on this scale, and I hope that people are interested and want to discuss it further. And maybe we can form some sort of productive collaboration,” adds San Emeterio, whose research is entitled, “Age-dependent immune Dysregulation during Repair of Volumetric Muscle Injury.”

Standing near her poster for most of the evening was Madeline Smerchansky, a Petit Undergraduate Scholar from Georgia Tech attending her first Hilton Head conference. She saw the opportunity as something of an investment.

“This is practice for the future,” says Smerchansky, a third-year student.

At least one researcher during the four-day workshop offered a glimpse into the future from a perspective that did not include biomolecular science or immunology. Aaron Levine, associate professor in the School of Public Policy at Georgia Tech and a Petit Institute researcher, delivered a presentation called, “Regenerative Medicine in a Time of Policy Uncertainty.”

“We haven’t seen a lot of clear signals yet with how the policy environment is going to play out from the current presidential administration,” says Levine, who focused his Friday morning talk on, among other things, potential policy drivers for regenerative medicine, such as the 21st Century Cures Act (will it be implemented by this administration, and if so, how much of it?), and the appointment of a commissioner for the Food and Drug Administration (FDA).

The future of the Cures Act may be largely dependent on who the next FDA commissioner is, noted Arnie Caplan (of Case Western University) during Levine’s post-talk Q&A session. 

Later that evening, it was Caplan’s turn to take center stage with Chris Evans of the Mayo Clinic.

They were the main event, you might say. With a backdrop of Caplan and Evans as photo-enhanced boxers the mood was light for their Friday night debate, entitled, “MSCs are Not Stem Cells.” Or, as Nerem put it, “is an MSC a mesenchymal stem cells, a medical signaling cell, or a mediocre scientific concept.”

By all accounts, they verbally fought to a draw. But who knows. Maybe there will be a rematch in 2018, when the Regenerative Medicine Workshop returns to Hilton Head (March 21-24).

 

CONTACT:

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

]]> Jerry Grillo 1 1489155325 2017-03-10 14:15:25 1498248809 2017-06-23 20:13:29 0 0 news Annual gathering of researchers at Hilton Head begins third decade with heavy focus on immunology

]]>
2017-03-10T00:00:00-05:00 2017-03-10T00:00:00-05:00 2017-03-10 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
588565 588562 588563 588566 588564 588567 588568 588565 image <![CDATA[Hilton Head clubhouse]]> image/jpeg 1489152512 2017-03-10 13:28:32 1489152512 2017-03-10 13:28:32 588562 image <![CDATA[Nerem Lecture]]> image/jpeg 1489152086 2017-03-10 13:21:26 1489152086 2017-03-10 13:21:26 588563 image <![CDATA[Packed room]]> image/jpeg 1489152272 2017-03-10 13:24:32 1489152272 2017-03-10 13:24:32 588566 image <![CDATA[Cheryl San Emeterio]]> image/jpeg 1489152777 2017-03-10 13:32:57 1489152777 2017-03-10 13:32:57 588564 image <![CDATA[Madeline Smerchansky]]> image/jpeg 1489152351 2017-03-10 13:25:51 1489152351 2017-03-10 13:25:51 588567 image <![CDATA[Winning poster]]> image/jpeg 1489153230 2017-03-10 13:40:30 1489153230 2017-03-10 13:40:30 588568 image <![CDATA[Arnie and Chris]]> image/jpeg 1489153350 2017-03-10 13:42:30 1489153350 2017-03-10 13:42:30
<![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[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[Regenerative Engineering and Medicine Retreat]]> 28153 Researchers from Emory University, the Georgia Institute of Technology, and the University of Georgia gathered for the annual Regenerative Engineering and Medicine (REM) Research Center retreat, but this time, something was different. 

It was a few things, actually. For one, the rotating event returned to the Petit Institute for Bioengineering and Bioscience on the Georgia Tech campus after touching down at the other two universities the past few years. And this time, participants gathered earlier than ever before with even more focus, thanks to a new format.

“We decided this year to have a more dynamic, interactive environment by adding breakout sessions, where we can get smaller groups of faculty together to talk about the themes of interest to us in regenerative medicine,” says Johnna Temenoff, professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, and one of three co-directors of the REM.

The retreat was planned this time to coincide with the 2016-2017 Georgia Partners in Regenerative Medicine seed grant program, announced a day before the retreat, which was held on May 13. The seed grant program is intended to stimulate new, collaborative research among the REM partner institutions.  The grants are due July 11, 2016.

“The timing was important, because it gives people an opportunity after the retreat to discuss grant ideas and put things together in a more focused way,” says Steve Stice, UGA professor and an REM co-director.

The preferred research themes are:

• Modulation of Immunity and Host Responses to Improve Regenerative Therapies (local and systemic means to modulate the host environment to create more effective regenerative therapies);

 • Optimizing Regenerative Biomanufacturing (scaling up and/or predicting the quality/potency of regenerative therapies during processing, for example, new assays for potency prediction, or new bioreactor or material technologies to promote scale-up of regenerative therapies).

While all proposals for the seed grant are welcome, those responsive to the identified themes will be given funding priority for 2016-2017.

“You want the seed grants to have a return on investment,” says Ned Waller, Emory professor and REM co-director. “You want to advance the science that will lead to external funding, and by bringing people together now in a focused way, really getting them to start collaborating today, we hope that by July they’ll have a seed grant, and that a year from now it will lead to an R01.”

The RO1, or Research Project Grant, is the NIH’s oldest, most commonly used grant program, generally awarded for three to five years.

The retreat began with presentations from two researchers. Art Edison from UGA spoke about his lab’s focus on the use of ‘omics’ data for a variety of biologic questions. Muna Qayed of Emory spoke about the clinical uses of stem cells, based on her work in developing a treatment against graft versus host disease (GVHD).

Then, participants split up into breakout sessions that focused on the seed grant themes. These were open discussions that took place in morning and then later in the afternoon, with each group reporting back on the main discussion points from each session.

Meanwhile, as researchers talked about their ideas among themselves, a group of trainees gathered with Andrés García, professor in the Woodruff School of Mechanical Engineering and director of the interdisciplinary bioengineering graduate program, to pick up some networking tips. 

The theme there was to make connections with peers and P.I.’s (at events like the REM retreat, for example) and learn the communication skills to augment the academics and the research, basically, “to stand out among the 400 applications we typically get for a single faculty position,” García says.

Most of the retreat attendees were particularly interested in the seed grants, which are intended to stimulate new, collaborative research among the three institutions. Each seed grant team must have at least two investigators and an equal partnership of faculty from two of the participating institutions. And this year, researchers have a better idea of which types of projects could receive funding.

“That was something we really wanted to do, pair the retreat with the call for seed grants,” Temenoff says. “This is a chance for researchers to come together and actually begin the grant planning process today.”


CONTACT:

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

]]> Jerry Grillo 1 1464288560 2016-05-26 18:49:20 1475896909 2016-10-08 03:21:49 0 0 news Annual meeting features new format and focus on upcoming seed grants

]]>
2016-05-26T00:00:00-04:00 2016-05-26T00:00:00-04:00 2016-05-26 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
540691 540701 540691 image <![CDATA[REM leaders]]> image/jpeg 1464710400 2016-05-31 16:00:00 1475895329 2016-10-08 02:55:29 540701 image <![CDATA[REM speakers]]> image/jpeg 1464710400 2016-05-31 16:00:00 1475895329 2016-10-08 02:55:29
<![CDATA[World Stem Cell Summit Comes to Atlanta]]> 28153 The Regenerative Engineering and Medicine (REM) research center, a collaboration among three of Georgia’s top research institutions, will not only have a front row seat for the World Stem Cell Summit, Dec. 10-12, in Atlanta – it is playing a lead role in facilitating the planet’s largest interdisciplinary gathering of professionals engaged in stem cell science and regenerative medicine.

REM, a sponsor of the summit, is a research partnership including Emory University, the Georgia Institute of Technology, and the University of Georgia (UGA), and is focused on transforming the treatment of diseases and injuries.

Scientists, students, clinicians, venture capitalists, investors, industry leaders, philanthropists, policy makers, experts in law and ethics, patient advocates – an array of stakeholders in stem cell science and regenerative medicine – will convene at the Hyatt Regency in downtown Atlanta for the summit.

"This is a fantastic opportunity that brings together different viewpoints to share the latest in research and commercialization of regenerative medicine therapies," said Johnna Temenoff, associate professor in the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and the co-director of the Regenerative Engineering and Medicine research center.

The summit brings more than 200 international speakers and 65 hours of in-depth programming arranged around thematic tracks that include: “Discovery, Translation & Clinical Trials,” “Regenerative Services & Restorative Medicine,” “Innovation Showcase for Cell Manufacturing,” “Regenerative Engineering & BioBanking,” “Hot Topics & Emerging Trends,” and “Ethics, Law and Society.” 

In addition to compelling keynote speeches, plenary sessions and focus sessions, the three-day event includes the “Conversations with Experts” luncheon, roundtable discussions, a centrally located exhibit hall, the poster forum, and an awards dinner. 

One of the honorees is Dr. Robert Nerem, who led the launch of the Georgia Tech/Emory Center for the Engineering of Living Tissues, which has evolved into REM. Nerem, founding director of the Petit Institute for Bioengineering and Bioscience at Georgia Tech, is being honored with the Leadership Award. 

Nerem, is slated to offer comments during the morning session of the first day, Thursday, Dec. 10, leading off the list of speakers from the three REM institutions, including REM directors Temenoff, Steven Stice (who also leads the Regenerative Bioscience Center at UGA) and Edmund Waller (Emory Winship Cancer Institute).

Attendees of the World Stem Cell Summit also have an opportunity to attend the RegMed Capital Conference, a co-located meeting committed to advancing commercialization and investment opportunities for companies targeting cures. This could be particularly useful for the 18 start-up companies that have emerged from the research of REM lab members.

Currently, REM has more than 70 faculty members from its three institutions working to develop innovative treatments for a variety of diseases in the areas of cancer, neurology, cardiology, orthopedics, and pediatrics. Since 2000, REM has garnered almost $121 million in total funding and its researchers have licensed 25 technologies.

The World Stem Cell Summit and RegMed Capital Conference serve as the flagship gathering of the international stem cell and regenerative medicine community, with the aim of accelerating the discovery and development of lifesaving cures and therapies and bringing together stakeholders to solve global challenges.

In addition to REM, other organizing partners are the Genetics Policy Institute/Regenerative Medicine Foundation (producer of the event), the Mayo Clinic, the Kyoto University Institute for Integrated Cell-Material Sciences, BioBridge Global, the Wake Forest Institute for Regenerative Medicine, and the New York Stem Cell Foundation.


CONTACT:

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

]]> Jerry Grillo 1 1447862505 2015-11-18 16:01:45 1475896803 2016-10-08 03:20:03 0 0 news REM plays lead role in annual gathering of global stakeholders

]]>
2015-11-30T00:00:00-05:00 2015-11-30T00:00:00-05:00 2015-11-30 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
471401 471401 image <![CDATA[World Stem Cell Summit]]> image/jpeg 1449257176 2015-12-04 19:26:16 1475895220 2016-10-08 02:53:40
<![CDATA[Guldberg Named TERMIS Fellow]]> 28153 Petit Institute Executive Director Bob Guldberg recently was named an International Fellow of Tissue Engineering and Regenerative Medicine (FTERM) at the TERMIS World Congress.

The honor was established as a way to recognize individuals who have shaped the global field of tissue engineering and regenerative medicine (TERMIS stands for Tissue Engineering and Regenerative Medicine International Society).

“It’s a great honor to be selected as a Fellow of TERMIS,” says Guldberg, professor in the Woodruff School of Mechanical Engineering. “I must say I felt a bit old when I realized I had attended many of the early meetings during the formative days of the society.”

TERMIS was created through the consolidation of several smaller societies, says Guldberg, who credits the organization’s founding fellows for their vision in aligning their interests, “to create TERMIS as a global organization.”

The TERMIS World Congress is held every three years. At each of these meetings, five or six new Fellows are selected. Guldberg received his honor at the World Congress held Sept. 8-11 in Boston.

It was the second consecutive World Congress in which someone from the Georgia Institute of Technology was so honored. Bob Nerem, founding director of the Petit Institute, became a Fellow at the 2012 event in Vienna, Austria.

“Probably the greatest benefit that TERMIS has provided to me, in addition to being an intellectual home for my research, is the huge number of people from around the world that have become friends and colleagues through the society,” Guldberg says. “For that I will always be grateful.”

CONTACT:

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

 

]]> Jerry Grillo 1 1443113083 2015-09-24 16:44:43 1475896776 2016-10-08 03:19:36 0 0 news Petit Institute executive director honored for leadership in tissue engineering and regenerative medicine

]]>
2015-09-24T00:00:00-04:00 2015-09-24T00:00:00-04:00 2015-09-24 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering and Bioscience

]]>
452031 452031 image <![CDATA[Guldberg TERMIS]]> image/jpeg 1449256280 2015-12-04 19:11:20 1475895194 2016-10-08 02:53:14
<![CDATA[Reading the Signals]]> 27195 New study, published in Hepatology, shows the regenerative capacity of liver cells

Patients suffering from chronic liver disease often develop liver fibrosis (the accumulation of scar tissue), which frequently results in cirrhosis, which means a loss of liver function, which often comes with a choice between a liver transplant and certain death.

It’s a perfect storm of terrible things as sustained fibrosis dampens the regenerative capacity of hepatocytes, thwarting their ability to make a therapeutic response, resulting in a grim prognosis and high mortality. At its essence, this is a communication problem, based on a study by a team of researchers in the lab of Chong Hyun Shin at the Georgia Institute of Technology.

Their findings explain how signaling pathways and cell-cell communications direct the cellular response to fibrogenic stimuli. But they also identify some novel potential therapeutic strategies for chronic liver disease. Results of the study (funded by the National Institutes of Health, the Emory/Georgia Tech Regenerative Engineering and Medicine Center, and Georgia Tech’s School of Biology) were published recently in the journal Hepatology.

“We aim to understand the molecular and cellular mechanisms that mediate the effects of sustained fibrosis on hepatocyte regeneration, using the zebrafish as a model,” explains Shin, assistant professor in the School of Biology, with a lab in the Parker H. Petit Institute for Bioengineering and Bioscience. Her fellow authors are Frank Anania (Emory), Mianbo Huang, Angela Chang, Minna Choi and David Zhou.

In their fibrotic zebrafish model, they studied the effects that different levels of signaling have on the regeneration of liver cells (hepatocytes). Specifically, they took note of the relationship between ‘Wnt’ and ‘Notch’ (signaling pathways). They discovered that lower level Notch signaling promotes cell regeneration (the proliferation and differentiation of hepatic progenitor cells, or HPCs, into hepatocytes), while high levels suppressed it. And they discovered that antagonistic interaction between Wnt and Notch modulates regenerative capacity: Wnt signals can suppress Notch signals, or, in other words, when Wnt is up, Notch is down, and hepatocyte regeneration can happen.

The data, says Shin, “suggest an essential interplay between Wnt and Notch signaling during hepatocyte regeneration in the fibrotic liver, providing legitimate therapeutic strategies for chronic liver failure in vivo.”

Inducing tissue regeneration via stem or progenitor cells, while delaying fibrosis, has been on the rise as antifibrogenic strategies of great potential, according to Shin, whose studies offer a clue of how to guide the differentiation of HPCs into hepatocytes in patients suffering from chronic liver failure. “Overall,” she explains, “employing the in vivo-based hepatic regeneration strategy may allow us to complement fundamental drawbacks in stem cell therapy, opening up new avenues of endogenous cellular regeneration therapy.”

This research is supported by grant number K01DK081351 from the National Institutes of Health (NIH), the Regenerative Engineering and Medicine Research Center Pilot Award (GTEC 2731336), and the School of Biology, Georgia Institute of Technology.

Read Hepatology journal abstract here


]]> Colly Mitchell 1 1406198738 2014-07-24 10:45:38 1475896608 2016-10-08 03:16:48 0 0 news A new study, published in Hepatology, from Chong Shin's lab shows the regenerative capacity of liver cells. 

]]>
2014-07-24T00:00:00-04:00 2014-07-24T00:00:00-04:00 2014-07-24 00:00:00 Jerry Grillo
Communications Officer II
Parker H. Petit Institute for
Bioengineering & Bioscience

]]>
310421 310421 image <![CDATA[Chong Hyun Shin, PhD - Assistant Professor, School of Biology at Georgia Tech]]> image/jpeg 1449244726 2015-12-04 15:58:46 1475895020 2016-10-08 02:50:20 <![CDATA[Read publication in Heptalogy]]> <![CDATA[Chong Shin]]>