Skip to content

GSJ: Bringing Precision to Medicine

Georgia Science Junction

Nov 14, 2023

Speaker: Robyn Schlicher, PhD, Executive Consultant, fractional Csuite Executive for Biotechnology Start-up companies. 

Precision medicine is the use of information about a person’s lifestyle, environment, and genes to tailor medical treatment.  In this GSJ, Dr Schlicher talks about the history and development of medicine and therapies and how they can be personalized to help people. The very first-time precise medicine was mentioned was approximately in 1550 BC when it  appeared in the Odyssey written by Homer. 

Full Video

Full Transcript

okay welcome everyone to Georgia science Junction on Precision 

0:09 

medicine uh my name is Patrick Ryan I am the director of engagement at science 

0:14 

for Georgia uh science research has been uh a nonprofit since 

0:20 

2018 uh we came out of the March for science knowing that there was a need 

0:25 

for more communication of science between scientists and the public science, scientists and investors and 

0:32 

scientists and the legislators so our missions are 

0:37 

improving communication between science uh or scientists and the public we increase public engagement with science 

0:44 

through events like this and then get that science responsibly used in public 

0:50 

policy uh we do this through a lot of different events and organizations we do science communication training for 

0:57 

scientists and college students uh and businesses uh we have Outreach events 

1:03 

like at the Atlanta Science Festival this is the science of ice cream um we do round tables on Workforce 

1:09 

Development and other things sometimes even getting the lieutenant governor um 

1:16 

we host conferences about environmental justice and climate protection um this is one of our other 

1:23 

roundtables uh we’ve worked with literacy campaigns uh we worked with and 

1:28 

got uh March lamed Georgia reading 

1:33 

month um and we’ve worked with the science Community to somewhat oppose the 

1:39 

okoi mining operation because it is bit dangerous for the 

1:45 

okoi um we do a lot of our work with volunteers and other groups so if you’d 

1:51 

like to know more about our events you can join our catalyzer Network um we have done a Rebrand from 

1:58 

Atlanta science Tavern to Georgia science Junction we’re doing this to do more events around other parts of 

2:05 

Atlanta other parts of Georgia we’ll be next year we’ll be going to Augusta we’ll be going to Athens we’ll be doing 

2:12 

a lot of fun stuff you’ll also be able to see a lot of these hopefully on our streaming which those of you on stream 

2:18 

right now uh know of we’re on YouTube um we have our new events 

2:24 

calendar so you don’t miss any of our events um we do a lot of uh Outreach 

2:30 

um and in South D Cab and some other areas around 

2:36 

Atlanta got it okay um and if you’d like to help us out we do take donations we 

2:42 

are in nonprofit um we do this through your support um so thank you for mutation for 

2:49 

hosting I know it was a little bit trying to get set up but we’re good now and the Speak Easy is actually quite 

2:54 

nice so hopefully we’ll be able to be back here again um all right and now now 

3:00 

uh Dr Robin schlicker will be giving her talk on um bringing Precision to 

3:06 

Medicine uh she is an expert in many spots in this field I have not enough hands so give me one 

3:20 

second okay so Robin schlicker brings over 20 years of experience between 

3:25 

Precision medicine Diagnostics and entrepreneurial leadership she’s a recognized global expert in Advanced 

3:30 

Analytical techniques and consults with many up-and coming biotech companies uh in addition to working at well uh 

3:37 

recognize Global organizations like Abbot labs and Leica her publication on drug delivery by targeted cation uh 

3:44 

cavitation sorry has been widely cited and highly uh highlighted worldwide she 

3:49 

earned her doctorate in bioengineering from Georgia Tech and emmer University receiving National Institute of Health 

3:55 

and metronic fellowships and now without further ado here’s 

4:05 

Robin I think you’re going to have to click for me or I’m gonna okay you have to give me a second to understand 

4:11 

technology because I do a lot of Technology but you know 

4:17 

so all right I think I I think I have this down so thank you for the introduction Patrick and it’s lovely to 

4:23 

see everybody I’m glad that um you I like speaking to smaller audiences right 

4:28 

now in my life as Patrick said I’ve done this for a very long time and have a lot of strong ties to Georgia so I joke 

4:35 

about how I predate biotech as an industry but I I actually do I started in chemical engineering and then helped 

4:42 

Georgia Tech actually build the bioengineering Department program and have one of the first doctorates from it 

4:47 

in the laboratory of Dr Mark prut so I have a lot of ties here and I’m happy to 

4:53 

support bringing biotech further into Georgia I was also a person who part of 

4:58 

the reason I had to leave Georgia was because there wasn’t a good Corridor of Biotech here at the time that I was 

5:05 

working in it and I ended up going to San Francisco and that was where I got deeper into the diagnostic space but 

5:11 

we’ll talk about that a little bit we’re going to kind of go on a journey here um talking about what Precision medicine 

5:17 

actually is what do we mean by it and uh the evolution of it from ancient times 

5:22 

actually so you know whenever you hear about Precision medicine what does that really mean I mean have’t doctors as to 

5:29 

date at least in modern medicine try to make sure that you get the right treatment for your disease well there’s 

5:34 

different ways of thinking about it there was an evolution of it from what’s called evidence-based medicine which was kind of based upon population 

5:41 

determinance you know this is what it looks like on symptoms and this is how we’re going to try to treat people who present this way and that helps a 

5:48 

certain portion of people but those of us who have experienced rare diseases unu diseases uh that actually wasn’t the 

5:55 

best way to go about it and then that started moving into what’s called Precision medicine where Tech technology tools started to become more advanced 

6:02 

and we could like actually guide it usually people think about Precision medicine in terms of genomics but there’s actually a lot of different ways 

6:08 

you can think about how how to make it more precise for you so we’ll talk about that and then there’s this thing now 

6:14 

where we talk about personalizing it how does the medicine really help you as a person and then one of my last companies 

6:19 

like to say functionalize personalize Precision medicine because we could throw as many jargony words in there as 

6:25 

possible to say that we’re trying to make it about you because part of it is in our culture now we want to make sure 

6:31 

things are really about us but in medicine we really do want to make sure that the medicine is best for us and so 

6:38 

that’s one of the things that I’ve worked on for about two decades and we’ll talk about that a little bit more 

6:43 

a lot of people hear this term biomarkers and again a lot of the times right now we talk about it in terms of 

6:49 

genetics biomarkers are actually far more than genetics and biomarkers have actually been used throughout history 

6:55 

the respiratory rates the smell and taste of your urine all of these things actually indicative to portions of 

7:01 

disease patterns that you can think of as a biomarker and then um what has this 

7:06 

meant over time right so when did we start talking about 

7:11 

precise medicine I looked through some history and actually it goes all the way back into the BC times if you go to uh 

7:18 

the Odyssey written by Homer and I actually had the privilege of having a Classics education at a private school 

7:23 

before I got to Georgia Tech so I actually read all this when I was younger um but it talks about Telus the 

7:29 

son of adus visits menus and Helen to hear of news of his father and Helen 

7:34 

puts a drug into the wine to ease his grief and anger so she tried to use a specified type of medicine the wine to 

7:41 

help him ease his condition and so that’s the first time that you feel a mention of using something in a specific 

7:47 

way of treatment for a person to improve their status of health and then why this is important is even to the Egyptians 

7:54 

which were sort of The Originators of making medicine and medicinal improvements when you think of it through ancient times times they looked 

8:00 

to the Greeks and I thought that was kind of a cute fact that because Greeks were supposed to be um the descendants 

8:06 

of the god that or the the doctor to the gods so the Egyptians were looking to or the Greeks were looking to the Egyptians 

8:13 

I’m sorry the Egyptians and the Greeks had this tie together in this time and the Greeks actually followed their lead 

8:19 

and that’s part of the reason that you hear about it even in the original sorts of documents to this long traval of 

8:26 

Homer but when did it really become precise so in Egyptian medicine they actually started to break it down into 

8:32 

categories of medical practice so how do we make this better like a doctor or physician was looking at how we practice 

8:38 

a specific type of treatment in Egypt and there was a specialization for a disease per body part so it’s starting 

8:44 

to be like let’s fragment this out and stratify how we can do different um ways to treat the body of an individual and 

8:51 

then it goes into what’s called Greek medicine it’s more of the Classical period And this is actually considered 

8:56 

the first use of real personalized medicine where they really took it to the body’s location and tried to use 

9:02 

understanding where it was located in the body and spewing specific sorts of study of that body and being specialized 

9:08 

as Physicians um to get a deeper understanding of each type of illness and so that they saw was getting better 

9:13 

therapeutic outcomes so instead of somebody who just sort of looked at you and tried to understand what humors they 

9:18 

looked specifically at how feet were different among people and treatments that actually helped your foot work better and how your heart was better if 

9:25 

you looked at its whole population so that’s kind of considered the first time that we starting to personalize 

9:30 

treatment to at least the individual body component not necessarily to the individual person 

9:36 

yet and then we get to the most famous doctor of all right any physician the Hippocratic Oath so we go to hypocrates 

9:43 

and he looks at this and he says okay this is really cool but you’re still saying a lot of things that are superstitious there’s a lot of this 

9:49 

pseudo science we’re s trying to understand like what humors do to the body we’re still trying to understand 

9:54 

how Gods affect it so hypocrates said you know we want to go more to rational 

9:59 

outcome and think about this is how diseases can be treated from their origin and that the treatment carried 

10:04 

out should be opposite to the cause of the disease so you know if you’re constantly being annoyed by the person 

10:10 

next to you maybe you need to get away from them kind of idea right if if you’re if you’re being constantly 

10:15 

exposed to something that you’re potentially allergic to maybe you should stop taking that so it’s kind of this first like input to what evidence should 

10:22 

be used to make your health better and then we’ve this created more of a 

10:28 

focused and person personized approach because again what is the thing that’s adding insult to an individual’s body 

10:34 

that maybe we should stop exposing them to so it started to become more personalized through hypocrates work and 

10:39 

that’s part of the reason that we you know glorify or I guess look to him as the originator of what we call modern 

10:45 

medicine he tried to individualize that medicine and he tried to approach it from an ethical and moral standpoint as 

10:52 

most of you know throughout history there was a lot of crossovers between doctors um and not such good players in 

11:01 

that space right so so he was trying to create an ethical framework under which we would make sure that we do no harm 

11:08 

that’s the most famous phrase that people think of from hypocrates and that’s actually really important these 

11:13 

days as we create more technology so I’m literally on the what they like to say bleeding edge of technology in San 

11:19 

Francisco I do not like that term but a lot of the questions that we ask ourselves are we going to do no harm 

11:26 

with this is this really something that needs to be done just because we can do it during my time here in Georgia I was 

11:32 

actually a stem cell scientist I was working on regenerative medicine products and the just to talk about some 

11:40 

of the politics of even that you know you guys are familiar with different levels potentially of stem cells 

11:45 

embryonic versus body cells adult stem cells and there was a lot of concern in that time so it was around the bush 

11:51 

Administration to the Obama Administration and Georgia in particular was not really fond of the thought of 

11:57 

using stem cells as a research tool particularly for embryonic and so many of us couldn’t actually continue a lot 

12:03 

of that research that’s part of the reason I ended up in California although I did switch then to Diagnostics so you 

12:09 

know going from how do you use body cells what’s the ethics of that and should we use those cells becomes the 

12:16 

next questions of should we continue to use the technology that have led to Gene editing so should we redesign your baby 

12:23 

you know these are things that you have to think about pretty often in my line of work um so this became the precursor 

12:29 

to kind of the thought process behind genomic medicine there are individual characteristics of you that are 

12:35 

potentially um treatable or indicative of your health State and how we can make that better so there were no genomics at 

12:42 

this time but he was talking about phenotypical potential outcomes is that leads to the idea that there’s something about you that makes you special that 

12:49 

makes you treatable in a new way so you know what is modern medicine 

12:55 

um I’ve read a lot of history of medicine books and one of the most interes ing things that I ever read was 

13:00 

actually about a serial killer so I like to tell this story um in the late and 

13:06 

early 1800s medical school was still kind of a free-for-all it was like can you get enough money from your parents 

13:12 

to get some training often it was based in Homeopathy and if you’re familiar with Homeopathy even today in its usages 

13:19 

it’s fairly questionable but it was particularly questionable back in the 1800s you’re adding more of the same to 

13:25 

do no harm those kinds of ideas and it wasn’t stratified or it it wasn’t I’m sorry it wasn’t solidified into a 

13:32 

program that was really necessarily based in pure science so often people would prefer to go see the barber 

13:38 

dentist right so you get your haircut you get your teeth pulled you get your surgeries done because they had 

13:43 

practical hands on training and people who even went to what were considered 

13:48 

top medical schools in the United States had a lot of theoretical training and often gave you things that had arsenic 

13:54 

in it so you know you would go to a barber if you were having you know obstetric like obstetrics kind of 

14:00 

question you saw your Midwife um so even though Medical Care was available in the United States it was still kind of shaky 

14:06 

now then they started to change that Paradigm and actually make like Johns Hopkins made a really good school and why this relates back to a serial killer 

14:13 

is that in Chicago Harvey mud actually trained as a homeopathic medical practitioner and was part of his killing 

14:20 

field need was to you know vivisect his his his victims right so this was all 

14:26 

part of this medical history book about how he was trained in medicine so I thought that was an interesting segue about the history of how training 

14:33 

happened in the United States uh so about in the 1870s we started taking more of a scientific approach and we 

14:39 

started to create medical programs that were actually based in treatment outcomes and again bringing in 

14:44 

hypocritic oath and saying we need to use ethical practices for our patients then around we jump up to about the 

14:52 

1950s because we’re starting to create more science and this becomes the evidence-based medicine I mentioned before we’re looking at evidence on 

14:58 

outcomes and this is where you get a lot of these general practitioner type of questions what are your symptoms how do 

15:03 

I match that to other symptoms of known patients and how do I give you treatments that will potentially alleviate those symptoms but the 

15:10 

problems with that again become often evidence medicine was based in data about men um and it was often based in 

15:18 

generalized population information that isn’t necessarily personalized enough for those of us with rare and 

15:24 

challenging diseases so then around the 1990s so we get into May era so that was around the time I came to Georgia Tech 

15:31 

and we started actually using the terms about uh prediction of drug responses and understanding things as personalized 

15:38 

medicine and this was kind of a new thought process in general we’re like well we can use technology and tools and 

15:45 

we can actually understand things about you at the genetic level and other things other things about you to help 

15:51 

make drugs that are actually tuned to these genetics and make things more efficacious on the end save your life 

15:58 

improve your outcomes and why did this come about well in part because we started thinking more about how biomarkers work so biomarkers are just 

16:05 

objective measurements about things of your body and again a lot of people think I mean genetics I actually thought 

16:10 

that for a while I was like oh a biomarker is a genetic marker outcome well actually it can be almost anything 

16:15 

I work with a company right now that actually looks at if you if you think about how cancer is formed in the body 

16:22 

it it reaches out with angiogenic angiogenetic factors to create vascularization to help feed the tumor 

16:28 

and you can look at the torsion and twistin and amount of those vessels and that actually is a biomarker that tells 

16:35 

you about whether or not that uh particular tumor is amenable to cell therapies this is a whole new line of 

16:41 

inquiry I would never have thought that you could look at vascularization as a biomarker so you know almost anything 

16:47 

can become a biomarker if you can measure it enough and get enough instances of it to create the data pools 

16:53 

that tell you about how to predict an outcome from it right and um so this this is very useful for again 

17:00 

Diagnostics you know are you ill or not based upon a particular profile of your 

17:05 

heart rate changing your genetics that tell you you may or may not have Huntington um those kinds of things and 

17:12 

again with the really um I think exciting utility of these Technologies 

17:17 

and where I’ve really worked the last few years is how do you match treatments to People based upon these particular 

17:23 

characteristics of their body so but why did we start thinking this way around the 1990s well because 

17:29 

this thing called the Human Genome Project happened it actually changed my life so when I was a kid I was thinking 

17:34 

about how I wanted to do something interesting with new types of technology and this was something that was kind of 

17:40 

what I was hearing about even before the 90s there were these things about how you could maybe use genes maybe use genes think about genetics and then it 

17:47 

became this massive Global or massive United States project in 1990 and we 

17:52 

created a lot of new technologies around it like being able to use faster sequencing actually came out of that and 

17:58 

to be able to try to understand more about the human body from the genes now um this is a way to try to understand 

18:05 

things that we were doing to try to tailor disease prevention again are you precursor genetically going to possibly 

18:12 

have Huntington like we can look at that um and then try to understand better ways to do treatment over time and it 

18:19 

was trying to take into account differences in your genes environments and your Lifestyles but this is actually 

18:26 

kind of of a question right now did we get everything that we promised out of the Human Genome Project and the answer 

18:32 

is actually no like when I was younger people were saying oh this is going to solve all medicine we’re going to 

18:38 

understand your genes we’re going to do the Genome Project but there’s a little piece missing even if you can look at 

18:43 

everybody’s genotype it doesn’t necessarily mean that you understand what it has to do with anything we had to get enough data and we still haven’t 

18:50 

had enough data to understand what each of those genes actually relate to in terms of not just disease Health 

18:56 

lifestyle longevity and treatment so it was really interesting because people were saying this is going to end 

19:02 

all things in medicine we’re going to have the Human Genome Project we’re going to know all your genes we’re going to make designer babies and we’re going 

19:08 

to make these you know these these cyborg humans and you know 30 years later we’ve done a 

19:15 

little bit of that but not really so so I I do think it’s still an important project but it’s not what people thought 

19:21 

it was and there’s a lot of that go goes on in biotech there’s a lot of this great big hype like the more recent one 

19:26 

was that crisper and it was come in and everybody was going to have the ability to have their genes changed um a little 

19:33 

bit you know but it’s it’s really not it takes a lot longer than people think when you first hear about the Star Trek 

19:40 

of it and I think that’s something that the population needs to understand a little better so but what did this 

19:45 

really do for us it helped us create a whole bunch of really interesting Technologies so to get to this place we 

19:51 

had to use all of these different types of tools that were being built in real time and you guys have probably all heard about many of these things PCR you 

19:59 

know like understanding more about DNA sequencing you know genomic outcomes like these are all very important 

20:05 

technologies that we use every day actually now not necessarily in their origin of the Human Genome Project but 

20:12 

every day to understand more about our characteristics of our body so in my mind it was actually a successful thing 

20:17 

because it created Technologies um and you know one of the interesting things I think that we really need to understand 

20:23 

is we’re all basically the same and I think we forget that and I’m not going to go on a big segue about politics and 

20:29 

religion but we’re all 99% the same the interesting flavors in US is actually a 

20:35 

pretty small part of our genomics but that’s really critical in understanding how to treat your disease so that’s 

20:40 

where a lot of the the dwell time is is what is that thing that makes us a little bit different the the Snips the 

20:46 

variable parts of our polymorphisms of our DNA like what does that actually Express in the proteins how are we able 

20:53 

to use that information to understand why my version of a disease is potentially different than my my friend 

20:58 

Sam over there and so the whole goal of this project was to get a better understanding of an individual’s genetic 

21:04 

makeup but I think we got a lot more from that actually we got a lot of interesting Technologies and a lot of 

21:09 

interesting tools and a new paradigm for how to think about things and I built my career on it we’ll talk a little bit 

21:14 

more about that now the Hope was that we would be able to very easily and rapidly predict outcomes using your genetics or 

21:21 

predict that you’d have a specific disease or match you and we do do that it’s just not as much or as easy as 

21:28 

people thought it was going to be so how do we get back to Georgia and we’ll talk a little bit about my life 

21:35 

because I think that it’s kind of interesting so my career um again started at this time and I was very 

21:40 

fortunate to be in a place to have access to some of the top Minds in personalized medicine as we were 

21:46 

creating personalized medicine so when people are like wow your career is really interesting I happen to be in the 

21:52 

right place at the right time but it was also really frustrating because you know I’m 19 years old in this small private 

21:57 

school in Oklahoma saying well I can take chemical engineering classes but I also need to take molecular biology classes and I can go build bioreactors 

22:04 

at this thing um at this National Lab and everybody’s like what is this woman doing well I was building bioengineering 

22:10 

um for myself basically and so it was part of the reason that I got into Georgia Tech Georgia Tech had it on the 

22:16 

road map to create the bioengineering program I came as a chemical engineering major and work with Mark prets and he 

22:24 

was out of Bob Langer’s lab Robert Langer who’s probably the only bioengineer that the average population 

22:30 

has ever heard his name if you familiar with madna he was one of the founders of that so at that time in MIT Robert 

22:38 

Langer Mark pret Samir mitoi a lot of very big names were building out these tools to create targeted drug delivery 

22:45 

and actually the things that later became mRNA vaccine like there’s a massive body of work that they’ve done because they looked at that and they 

22:51 

said well there’s got to be ways that we can look at things past at least in Mark’s lab when I was there let’s look 

22:57 

past standard pills injections and we can use that to alleviate the side effects and improve patient outcomes by 

23:04 

targeting drugs and therapeutic specifically to the region of Interest so we were building Micron needles and 

23:09 

others I’ll talk about him a little bit more in a minute but why that was important at that time was it was a 

23:14 

thought about using mechanical means to take drugs directly to the site of action we weren’t quite at the genetic level yet in this lab but we were trying 

23:21 

to get there now also at Georgia Tech I was working with a very well-known scientist named Robert naram Robert 

23:27 

neurum he was an expert in regenerative medicine and one of the key Pioneers there as well and so that way we were 

23:33 

looking at Technologies to create organs for your body to do organ replacement um 

23:39 

to actually understand more about anagen so the same genes that you turn on to make those cells create a new organ 

23:45 

could also be potentially cancerous genes so we were working together in some different ways to understand more 

23:51 

about how to use those Technologies Marie Chata um I mentioned that I was partly a stem cell biologist she’s a 

23:57 

well-known stem cell biologist and so she was looking at ways to use cells with specific specific characteristics 

24:03 

to improve health and that later became what we think of as cell therapy so if you think about some of these Cancer 

24:09 

Treatments imun oncology where we’re using specific types of cells to alleviate cancer those are cell therapy 

24:15 

methods and so she was on The Cutting Edge of that by using different cells of the stemness to understand how you can 

24:21 

improve outcomes using stem cells um and then I was learning molecular biology with a professor named Harish Krishna 

24:28 

who later actually went to Coca-Cola to focus in on I used to tease him that it was how to make your taste buds more 

24:34 

addicted to Coke products but he worked in the Statin line which is really understanding how to improve you know 

24:40 

those outcomes from drugs uh by using molecular biology so if you if you look at all this this kind of gave me a 

24:46 

background that I really understood a lot about cell biology um but I also understood a lot about how you could use 

24:52 

different techniques thought processes and the work of these really great key leaders in Geor 

24:58 

um to make things different and interesting in My Chosen field later of Diagnostics so from here I went to um I 

25:08 

went to California and fell down a well called omix and so omix is actually 

25:13 

tools that give you information about all the parts of your body that we’ve been discussing genomics transcriptomics 

25:18 

which is the transcription of your genomics proteomics which is the proteins that come from that translation 

25:24 

of your genomics um and metabolomics so what is your metabolism doing and we’re talking a lot more about metabolism 

25:31 

these days particularly in the context of coid because there’s all this fatigue there’s other things that are happening 

25:36 

with how you’re struggling to you know live your life basically and that kind of comes down to impart your 

25:42 

metabolomics um so from there I started thinking more about what’s called 

25:47 

pharmacogenetics and pharmacogenomics and I worked with a company called the Gina biosystems and 

25:53 

they were able to use mass tect trometry at the nuclear level and we were able to look at at specifically how your genes 

26:00 

react to drug medication so one of their most famous products um is in at the VA 

26:05 

where they’re looking at how to better use Mental Health drugs to um Target the right mental health patients so if 

26:11 

you’ve ever like had this experience or worked or had someone in your life with the experience of trying to take 

26:17 

medication for mental health effects you kind of tight trade on your own and you kind of try things until you see if they 

26:22 

work but with the Advent of things like pharmacogenetics you can actually look at how that pH tical that drug is going 

26:29 

to be acting upon specific parts of your genome based upon the data and evidence and see that you could potentially have 

26:35 

a better outcome from it now the most useful thing that happened for me with this is there’s a thing that actually 

26:41 

measures the amount of your metabolic uptake of drugs and what’s called Sip 2d6 and some of us have more copies of 

26:47 

it than others and so I have way too many copies of this so it was like why can’t I stay under anesthesia why do I 

26:53 

get why am I no longer numb so you know very quickly from a dental work like my dentist used to say I have to give you 

26:59 

four 10 12 times as much anesthesia I give you as much anesthesia as a linebacker and you’re aund you know 40 

27:05 

pound 5 foot 6 woman and it’s because I’m metabolically upright up upregulated 

27:11 

and I personally think this is actually one of the most important things that you need to know about yourself is your pharmacogenetic status and I think it’s 

27:17 

really important to understand that in Pediatrics because somebody like me if I were to take the wrong opioid which is a 

27:23 

Time release drug I will over metabolize that and some of the things that you see with bad outcomes come from the fact 

27:29 

that we just don’t know someone’s pharmacogenetic status and I’d love to talk more about that it’s kind of a hill I will die on okay so um what I really 

27:38 

think that one of the most important things I’ve kind of alluded to data data data data data nothing that we were able 

27:44 

to build would have been possible without really having good pools of data and being able to utilize that data so 

27:50 

we’re talking about the uprise and Advent of data scientists bioinformaticians I actually said that 

27:57 

right I’ve almost never been able to say that word right and the use of the data that we’re getting from the omix tools 

28:03 

to understand how to make potentially predictive modeling so you know it’s really about being able to use those 

28:09 

data that we’re able to access and look at them in new ways to understand outcomes better and from that we can 

28:15 

then create a personalized approach based upon data about that particular genetic status or is this example here 

28:23 

um a cardiac example in the 60s where we looked at different ways to understand how biomarkers and behaviors could 

28:28 

actually affect outcomes um of your heart so I think that you know none of this really would be possible if we 

28:34 

didn’t have good data management so where medicine really started to meet science fiction in my 

28:40 

life was when I went to Abbott Laboratories and we started using the genetic characteristics of patients so 

28:47 

using information about their genes to match them with a treatment that their genes will recognize or that can use a 

28:53 

person’s gen to improve their health and one of the best examples of this is a drug based B upon the genetics of cancer 

28:59 

cells that’s highly used across the board but was first in my world in breast cancer and this was a really 

29:05 

interesting time um in everybody’s life this was early OTS and we started going 

29:12 

into Physicians and saying chemotherapy of this so this is a particular um 

29:18 

stratification of type of breast cancer when we first started using it and what we saw was that these patients were not 

29:24 

really getting well from the standard of care and chemotherapy but if you added in a drug and the first one we were talking about was a vastin and from that 

29:30 

they used her septin if you added this additional drug on then their survivability went way up and we could 

29:37 

prove that using fish probe so path vision is one of the first ivt tests on 

29:42 

Market um that was owned by Abbot labs and we were using fish and you can see here like normally you have two Al right 

29:49 

so you should have two copies of each of these genes but if you’re what’s called upregulated you got this whole bolus of 

29:54 

these things and this is potentially a candidate who would be responsive to to her septin in her breast cancer and so I 

29:59 

was going around teaching this at hospitals to cytologists and going out and basically evangelizing the use of 

30:06 

your genetic status to help give you better treatment and Care from a specific type of medication and this 

30:12 

then became um one of the first what’s called companion Diagnostics so if you’re familiar with how Pharma 

30:17 

companies and genomics companies are trying to work together now you’re looking at a potential biomarker again 

30:23 

like genomics and matching it to a drug outcome and you want to build those two tools together so you can identify the 

30:29 

patients that will be the most what we call responders to that drug in real 

30:34 

time so that you attach a CDX a companion diagnostic test to a drug and 

30:40 

you use that diagnostic tool to understand the patients that should receive that drug it’s a very expensive 

30:46 

process that fails a lot but when it works it’s remarkable so another thing that I was 

30:52 

working on this goes way back to Georgia Tech um targeted drug delivery I mentioned it earlier and I thought I would give another another nod to my 

30:58 

good friend Dr pret we were building things that weren’t the rule of our lab we called ourselves the drug lab even 

31:04 

though we didn’t make drugs and we were the delivery lab even though we didn’t make pills or injections you couldn’t do 

31:09 

those two technologies of Mark lab but what you could do is build Micron needles and Micron needles are everywhere and he actually in this lab 

31:16 

in Mark’s hand has the first patents on Micron needles so um it’s accepted that all micronal technologies that come down 

31:22 

from this kind of originate here at least that’s that’s Mark’s argument so we’re going to we’re going to go with that one um but yeah so one of the first 

31:29 

ways that we designed that was in what was called lithography to make metal tips metal needles and try to pass drug 

31:36 

through try to put things on the skin and that actually wasn’t the best concept because you’re up against volumetric measurements and trying to 

31:42 

get enough load into there and so these are actually dissolvable needles so that you can put the patch into the skin they 

31:47 

dissolve and do a transfer rate and this is to develop vaccine so one of the things that we were working on in conjunction with the CDC was trying to 

31:54 

create vaccine potential that doesn’t have to be in the cold C chain and that works specifically um for you as a 

32:01 

Target on your skin and isn’t in injectable and so that’s he’s gotten a lot of um interest in that work and it’s 

32:06 

been really great to see that progress over 20 years my work was in using um my 

32:12 

my friend used to say I used to play music to cells so this is actually a very this is the famous image that he 

32:17 

alluded to earlier in my publication so I was using ultrasonic cavitation to 

32:22 

expose transfer portals in the cell so that you could have their itics transfer 

32:27 

in at a faster rate or to make sure that the therapy only goes to those cells that are targeted by the cavitation for 

32:34 

transfer because in the use of typical chemotherapies it’s given in through you know your vascularization it goes all 

32:40 

through your body and kills a lot of your fast growing cells you get all these side effects Our Hope and there’s still people who work on this to this 

32:46 

day is to take cavitation tools um expose the drug expose the cells to the 

32:52 

cavitation and the transfer only happens in those cells of interest and I um was 

32:57 

up against electroporation there were a lot of theories about how this worked I said it actually literally causes 

33:02 

cavitation damage effects and took very high level microscopy images because I will take it I will take it to the mat 

33:09 

to prove something and so I became famous for these types of images um and showing how Electro or son operation you 

33:17 

know worked so and it’s interesting because that actually became a big part of my career 

33:23 

um the technology that I’m most famous for is my ability to do microscopy so I worked for like a Microsystems and help 

33:29 

launch uh super resolution Grand slate depletion Technologies where you break the defraction limit and look at things 

33:35 

very very small that you can’t see typically light microscopes so it was to take those sorts of images so that was 

33:40 

electron microscopy so when you talk about looking at viruses and very very small things in high resolution you 

33:46 

usually have to go to electron microscopy and so this is using like a light based tool ground state depletion 

33:52 

to look at what’s called super resolution to try to get you into that same level of resolution without having to to go to electromicroscopy so some 

33:59 

explation um one of the other things that I think is really interesting to talk about that’s how to Resurgence is what’s called functional medicine so 20 

34:05 

years ago when I was working on these Technologies and trying to show that you could improve Cancer Care um by treating 

34:12 

these cells directly we were actually kind of making a bit of what’s called a functional assay where you take the cells or you know parts of the patient’s 

34:19 

body expose it to the drug and show that it would potentially make the patient better but 20 years ago we didn’t have 

34:25 

the compute power and you’re talking about taking cells from the body and trying to pretend that they’re like the body and so those outcomes are really 

34:32 

not well predicted but what’s happened in the last year or two really um there’s been a complete Resurgence in functional medicine so I was working in 

34:39 

this London based company and we were able to take cells from patients that had been diagnosed with specific types 

34:45 

of cancer and then we’d have them come back in and we’d take some more cells from them or we would take part of their 

34:50 

surgical resection we would reconstitute it in a hydrogel and then we would inflex that hydrogel with the therap Y 

34:57 

and then we would use computer vision and my favorite thing can focal microscopy so we would take three-dimensional microscope analysis 

35:04 

after they’ve been exposed the drugs do live dead staining and then we would tell if that drug was going to be good 

35:10 

for the patient and that’s in clinical trials right now in the UK so um but 

35:16 

it’s been interesting to see that come back as a Resurgence a lot of things that we were trying to do 20 years ago like gene therapy other you know this 

35:23 

functional medicine that we really didn’t have enough technology or enough understanding to do well back then are 

35:29 

are definitely having a Renaissance now and then this is something that I’m 

35:35 

pretty proud of um we hear a lot about machine learning AIML artificial intelligence and uh this is one of the 

35:43 

first products on Market um based upon that one of the first panels on Market that uses a machine learning algorithm 

35:49 

to do predictions about matching patients to specific drugs and this is the the product that we built so that 

35:56 

you can kind of reproducibly do that so again you need massive amounts of data 

36:01 

and you want to create a biomarker based around the data that you get from patients that were responding so that 

36:07 

you can understand more about how to find those patients who will respond in the future and this is based in a IML 

36:13 

principles and it’s actually doing pretty well these days so um pretty pretty proud of 

36:19 

that I think one of the things that people were most afraid of and it is actually happening in real time now is 

36:25 

how I keep talking about those designer babies so when I was at a company called Nera um we were working on what’s called 

36:31 

nip non-invasive pregnancy testing and the goal of that is to take a pregnant Mom’s blood sample and tell you about 

36:38 

the status of the fetus without having to necessarily do the amnio yet is the pregnancy affected with Down Syndrome 

36:45 

for example the most the most common triome and the one that most people recognize Down syndrome so um when I was 

36:50 

at Abbot we were actually doing kype staining amnio sorts of staining which are much more invasive so this is trying 

36:56 

to use or this uses blood samples from a mom and is able to tell you about the 

37:02 

status of the fetus quite early on like early stages of the pregnancy now again or a lot of moral and ethical quandaries 

37:10 

that come from this kind of testing um especially in countries where you are time and rate limited on when you could 

37:17 

potentially end an affected pregnancy so we’re trying to get these tools that can tell you in the first nine weeks in the 

37:23 

first 10 weeks but you’re up against Precision ISS isues and you’re also against what’s called reportable issues 

37:29 

this is a screen so you should not be using this as a diagnostic tool to make 

37:34 

a decision about a pregnancy unfortunately it does get used this is something that I think we should talk 

37:39 

about like so using these kinds of Technologies we are understanding more about the pregnancy but this is supposed 

37:47 

to be a screen to tell you to take the next level of testing to really understand you need to go to kot typing 

37:52 

right but again time and rate limiting steps here make it very challenging about this process the other thing that 

37:58 

we’re understanding more is like looking in IVF at the embryo status you can kind of pick an embryo potentially that has 

38:03 

characteristics or non-characteristics that you’re interested in um and then carrier screening the the parents 

38:09 

themselves and this is really important in specific populations like ashaji Jewish populations right so again we 

38:15 

have the Technologies now to kind of designer baby a bit and in some places 

38:22 

they do make some claims um not in the United States but I work globally they do make some claims about telling you 

38:29 

more about the potential baby that you’re going to have so you know um 

38:35 

we’re already kind of doing it now and then this is a very well-known company I really appreciate everything the CEO 

38:42 

does it’s called in Silicon medicine and they you know made a promise to each other a while back that they were going 

38:48 

to work really hard to use AIML tools to create new drugs and Therapeutics and they actually put one of the first on 

38:54 

Market they focus in on some long vity but longevity and oncology are very tied into each other and they do a lot of 

39:00 

different types of work so they’re they’re literally you know kind of changing the future right now by using 

39:06 

these tools and AIML to help make more effective drug therapies from the outset and I have you know strong feelings 

39:12 

about the use of AI tools I personally think that it’s good for my work because we’re able to look at lots of data 

39:18 

faster get better resolution from the data I think it’s going to be useful to help people more and so I I pretty much 

39:25 

embrace it in my work work and I’m I’m very proud of the you know inroads that I’ve had so far to use 

39:31 

it and then uh getting back to how can we really really change people well 

39:36 

there’s this technology again mentioned called crisper so we could potentially go in and edit and really design you to 

39:42 

be somebody different are we anywhere near that not really um but that there are some Physicians overseas who claim 

39:48 

to have affected fetuses and the babies have been born with some some use of crisper generally crisper though is used 

39:55 

for other things um in Diagnostics or in understanding you know how how to make better Therapeutics so there’s therapy 

40:02 

cell therapies that are using some crisper but in terms of actually affecting people and we said this 20 

40:08 

years ago gene therapy we’re going to go in and change your genes so I don’t think that’s a really an issue right now 

40:14 

but it does get back to the moral quander of how do we really use these tools that I think about all the time how are we really doing no harm are we 

40:20 

really making sure that the drugs therapies AI all of that is really doing the best for the patient and you know 

40:28 

that’s a question that I ask myself every day that I work on it um one of the other things I really like to say is 

40:34 

that in you know one man’s lifetime if we look at Watson cck and rosn Franklin 

40:39 

and how they understood what DNA looks like um he was a they were able to 

40:45 

elucidate the structure and then we were able to elucidate what it looks like in your body and then we were able to 

40:50 

Target drugs and things to make you better and now we’re able to potentially change it and so that’s how fast this is 

40:58 

changing and this kind of walkthrough tells you just about my career in it since the 90s and this tremendous the 

41:05 

amount of changes that just I’ve seen and I really appreciate the time you’ve 

41:10 

taken to listen to me talk about one of my favorite topics and if you have any questions I happy to talk about 

41:18 

[Laughter] 

41:25 

it 

41:30 

what are some hurdles that you’re 

41:53 

facing oh 

41:59 

[Laughter] we should talk more because I’m always interested in microscopes and I I consult for a lot of companies that have 

42:07 

microscope tools so we should we should talk more technical hurdles or business 

42:13 

hurdles or social hurdles um I actually talk about it in 

42:19 

terms of all things so one of the interesting things about my career is I started as a technologist got led into 

42:25 

being a sales person and then ended up becoming a business leader it was not 

42:31 

necessarily by Design I thought I’d still be sitting in a laboratory you know I I just had a a knack for 

42:37 

understanding a global view and and demanding money from people I actually am very good at negotiating things but I 

42:44 

I actually see a lot of different hurdles now my personal hurdles sometime can be that I Mentor a lot of early 

42:51 

stage companies and whenever you come in 

42:57 

young um so there’s a lot you know there’s this the rise of of of a young crew of CEOs they have these great ideas 

43:03 

and they are a founders of these great ideas but there’s not foundational knowledge of what it really takes to get 

43:08 

all the way through to a therapeutic so sometimes like they’re just up against their own challenges of understanding 

43:14 

that it’s going to take 10 years and that’s going to take half a billion dollars so one of the biggest challenges 

43:20 

is that getting anything on the market is extremely expensive requires significant 

43:26 

amount of data lots of patient participation like there’s a lot of things that come into play you can we 

43:32 

can start a diagnostic company tomorrow and I could probably get some money for that but to actually get all the way out 

43:38 

to something that’s going to affect patient care that that’s a you know like best statistic s to 10 years and lots 

43:45 

and lots of money so the first hurdle is everybody’s got a great idea um how do you actually apply that idea in a way 

43:51 

that makes sense and then from a social standpoint um there’s a this constant thought again should we actually be 

43:58 

doing it so some people don’t want to know if their pregnancy is affected some people don’t want to know if they’re 

44:04 

potentially going to have huntingtons and so there’s different things about do we really want to give Diagnostics to 

44:10 

people that don’t want it and then there’s a a lot of questions about the quality of the Diagnostics so FDA versus 

44:17 

CMS I there’s a lot of like alphabet soup in here but when you want to do a reportable diagnostic to actually affect 

44:23 

patient care there’s levels of evidence in there most things in the united states are under what’s called an ldt 

44:28 

where you have to show practical outcomes and and CLE there’s all the things that go from it and IBD is a very 

44:34 

high level of stringency that most companies cannot afford and it would really block down on progress if 

44:40 

everything had to be IBD but there’s a lot of talk right now um at the federal level president’s office should we 

44:47 

change that stringency requirement to make sure that the things that get on Market are better than what’s on the 

44:52 

market right now as all of you probably saw with like Co the the emergency use authorization put lots and lots of tests 

44:59 

on the market how many of those were actually that good so you know that’s one of the biggest challenges there as 

45:05 

well so right now what are regulations going to say and how do we get these on the market and then from a personal 

45:11 

level um and from a technological level it takes time so like I said 

45:18 

there’s a Renaissance of gene therapy there’s a Renaissance of these functional things so even if you had a 

45:23 

great idea it might not be matur enough for prime time we might not have the compute power we might not be able to 

45:29 

get enough data about it you might not find enough people with that disease to get enough instances to even make the 

45:36 

model work um when you’re looking at rare trimes of pregnancies and you want to get it from actual patients you’re 

45:42 

talking about people at one of the worst instances of their life so you’re having to make like potentially artificial you 

45:49 

know things to measure so there’s just a lot of different factors that get involved but I’ve seen many many many 

45:56 

things in on Market that do really great things for patients and I’ve really only 

46:02 

in in the time scales that I’m talking about 20 years actually isn’t that long considering how many things I’ve actually seen come to the market does 

46:08 

that kind of answer I mean that’s big question with lots of thoughts but I’m I’m happy to talk with you more about 

46:13 

your 

46:22 

technology so what you do is you take a a samp from the mom and then you take it 

46:27 

down to the plasma level and you look for what’s called cfdna cell-free DNA and you can separate out by different 

46:33 

Snips things that are most likely the mom’s DNA versus things that are most likely the baby’s DNA and then you can 

46:40 

weight average that and look for a trim in what would be the fetal fraction of the 

46:47 

DNA yeah right and so that’s interesting too because if you look at self-free DNA companies then they all start switching 

46:53 

over oncology companies so nari helped them become an oncology company because I came in when they were nip but I had 

46:59 

this big background in oncology and a lot of self-free DNA there’s a very common thread between a tumor and and a 

47:06 

baby in some ways right you’re talking about DNA that isn’t necessarily the host DNA and so you’re identifying 

47:14 

unusual bits of genetic information and so you a lot of these companies that were anpt companies also have cancer 

47:22 

studies and cancer products too because self-free DNA is a very power F thing to 

47:41 

study it it’s a weight of how many instances you have so the mom’s going to have more and the baby’s going to have 

47:46 

less kind of thing right so so you look at it from a position where it would be on the genome and then you wait it out I 

47:52 

mean they there’s other ways to look at it so the original companies were looking at nothing methylation status because you’re upregulated in 

47:58 

methylation when your body is changing which is cancer again and pregnancy and certain types of diseases so part of the 

48:05 

reason that they started thinking about this in terms of cancer is they were actually predicting some cancer in maternal instances they were like well 

48:11 

this looks really strange and they’re like oh that’s actually cancer you know so you you can wait the different Snips 

48:18 

and find things that are probably not from your body and that’s how these liquid biopsies work so nera’s product 

48:24 

that I helped with is a what’s called a liquid biopsy so we can look in your blood and understand about your tumor 

48:30 

status and whether or not your tumor is going to recur or um more about like the specific genomics of your tumor to 

48:36 

understand how to do better treatment on it so liquid biopsy is a a very big 

48:42 

thing right now there’s many many liquid biopsy companies and so it’s based upon the self-free DNA concept and where my 

48:49 

my use of it started out in the nip companies yeah a lot about 

49:16 

so that’s I think something I’ve thought about for a very long time I stayed in Georgia longer than a lot of my 

49:21 

classmates because I belied so much in the power of you know Georgia Tech and Emory Mercer and all of you Georgia 

49:27 

State there was this beautiful infrastructure in it um and we were hoping to create this Corridor of new 

49:33 

biotech startups but part of what happened then was in 08 and the economy started failing we all actually lost a 

49:40 

lot of our investment and one of the things that was trying to come in were large companies were trying to create a 

49:47 

footprint in Georgia like I believe dendreon was one muuk cor was another 

49:52 

one of the reasons that San Francisco has such a good startup environment is 

49:58 

in part because they have major players there as well so you kind of need to have both I think it would help if there 

50:05 

was more infrastructure from either large pharmas or large diagnostic companies because it is very hard to 

50:11 

sustain an ecosystem that is a bunch of early stage startups that are hoping for their next VC round just keep their 

50:16 

doors open uh I can give a whole other talk about what it’s like to keep hoping that you get that next fundraising round 

50:23 

to try to not just pay your rent but to make make sure that your technology goes forward so one of the instabilities in 

50:28 

early stage companies is can you keep getting enough evidence and can you keep getting enough Data before you run out 

50:34 

of funding and it’s really really challenging to do that I think there’s still a 90% failure rate on biotech 

50:40 

startups so I think as much as I appreciate all of the biotech startups 

50:45 

that are here in Georgia there’s some there’s some really good ones that I I know um to really get the attention and 

50:51 

money and funding that it should be at I think you’re going to have to get a few more larger players in but that’s you 

50:58 

know just personal opinion um but it it would be good I think too with the way some things have changed with 

51:04 

infrastructure here one of the things that we were up against in part was some of the politics of trying to build 

51:10 

biotech in Georgia it it was challenging so I think that there’s more openness 

51:16 

now because in part the movie industry is here and the population is a bit more 

51:21 

open-minded about Technologies and tools and a lot more demanding I think um I I know with Hollywood here there’s 

51:27 

going to be more interest in potentially longevity technology so a technology I didn’t talk about that’s got a lot of um 

51:33 

interesting players in it and again in silico is one of them is longevity how do we study your genes the length of 

51:39 

your tea mirror things like that to understand how much longer can we make you live and that actually I think is 

51:44 

interesting even to talk about women in menopause there’s some data now saying that if you could understand more about 

51:50 

um menopause and arresting menopause maybe that would increase your health outcomes so you know it’s a long answer 

51:56 

to a short question but right now I just think there needs to be bigger names larger brands or somebody here in 

52:03 

Georgia with an early stage startup needs to get a great big check from an 

52:24 

investor well it’s it’s looking at like the the different Pathways of the mutational 

52:30 

burden of as a whole but but yeah so if you look at again the liquid biopsy 

52:36 

Technologies you can measure out the different polymorphisms that are potentially from the tumor but even if 

52:42 

you look at um if you’re looking at like regular biopsies the h& staining and 

52:47 

then looking at the genetic component of it in I IHC and then to fish you’re 

52:53 

still looking at specific genes and we’re like given with um the herun new it’s an upregulation of those particular 

52:59 

genes so so you’re still looking at how the genes are changing or there’s more of those genes of a particular type that 

53:06 

shouldn’t be there in the body so it is still kind of a statistical measure if if that makes sense um but yeah it’s 

53:12 

looking at those instances that make it different from you as a person really I I don’t I’m not if I I can send you some 

53:19 

papers if that would be helpful 

53:34 

combin well you you could but that’s actually not what they’re doing but they they are purely lucky of the self free data I you guys want to have more 

53:41 

information yet it is based again on on using that data analytics and the upregulation of these different changes 

53:48 

in the polymorphism 

53:54 

snips 

54:00 

back in the day we were using some kind of uncontrollable Len viruses at times 

54:05 

but there there is better ways to deliver the gene therapy there is more understanding of what those genetics 

54:11 

look like and I think there’s more stable constructs for doing that transfer but it’s not something that I do a great deal of these days so you 

54:19 

know and and again I’m on the business side more than the technical side so that’s kind of my high level standing at 

54:25 

this stage about that condition and if I am wrong I will be happy to come back and correct 

54:42 

myself yeah I I’m happy to go back because I realized I was afraid I was 

54:48 

running low on time so this is an sem image scanning 

54:55 

electron microscopy with the coating over the cell so these are cell surface features in microvilli and this is what 

55:00 

normal cell substrate looks like the normal surface features of the cell here you can see this kind of grainy area and 

55:06 

that’s where we’ve basically kind of peeled away the normal cell area and this is cytoskeletal infrastructure that 

55:12 

actually makes it more porous and so you can have an easier transfer of the drag 

55:18 

or therapeutic into this cell and it this is like a one in a million shot kind of thing if you look at the statistics on how it would be defined 

55:24 

this was I was purely lucky even find this thing um but but yeah like this happens at higher levels of instance if 

55:31 

you because of the nature of electron microscopy you’re looking at a cell per cell and because of the nature of 

55:37 

cavitation you’re looking at a variance of effects and so it’s like if you think about when a bomb goes off everything 

55:43 

closer could be decimated everything out here is probably more where you want it to be for good transfer and everything out here is probably not really affected 

55:50 

and so if you look at like again fluorescence microscopies and light microscopies and Even Flow citometry was how we originally measured this um you 

55:58 

can get more statistics and more instances of the cells of interest but using em techniques but I didn’t know 

56:04 

when I took this on because I was going to prove something you’re looking at each individual cell and you’re hoping 

56:11 

to see a feature and so I remember when I first saw this with Robert EP carry and I was like what is going on with 

56:16 

that cell that’s so strange what is that going on there and I actually have a mirroring tem images which is uh the 

56:22 

cross-section where it’s a cut and you can actually see it’s not of this cell it’s of a similar cell and you actually 

56:27 

see the outside of the cell membrane and a great big hole in the cell membrane itself and so when I was looking at the 

56:33 

temm I was like that’s a hole in the cell so cell side exactly so I was very 

56:41 

lucky where I did way way way too much of this microscopy because TM is the same you’re looking at one so one cell 

56:47 

and since once I used these kind of ultra structural measurements then I was able to go back to confocal microscopy 

56:53 

which uses fluorescent dieses and I could do like a wide scan and so in fluorescent microscopy you see these fluorescent rings with a hole in it and 

57:00 

so I have lots of those images but these these images are actually really special because like they were on NIH website 

57:07 

they’ve been used in um series like movie series that talk about it because 

57:13 

they’re they’re really such such a good instance of looking at a cancer cell in EM so yeah yeah is that cool like I I 

57:20 

did this a long time ago and I still get notes from people who are like can I can I use these cell images is um Robert 

57:26 

ataran was a a professor at Emer who tragically died in 2006 and they named the micro the 

57:34 

microscopy the Emi Center actually over him and they named a forest in Armenia for him and so I think every time 

57:41 

somebody uses these images again you know a little bit of him carries on so 

57:46 

but thank you for that question I I I I think it’s be it is it’s it’s beautiful work and it’s it’s it was on the cover 

57:52 

of the Boston Globe it was actually highlighted in Georgia tag like it kind of made my career and then made me 

57:57 

realize I didn’t want to do research anymore I know and I really feel like I 

58:03 

need to go back and study some more about liquid biopsy because I’m just like well unfortunately that was four companies ago and I’ve done 18 

58:09 

techniques since so um but I really appreciate those kinds of questions because it is important to understand 

58:16 

how they’re doing that especially these days because every time I turn around there’s yet another liquid biopsy 

58:22 

company and so I actually IM met kind of the limit of my knowledge they might be doing it a little bit different now I 

58:27 

helped launch sigera which is nara’s liquid biopsy back in 

58:32 

2016 and that’s a funny story like I was going around the world doing Tech transfer of nip and the CEO was like we 

58:38 

really want to put this cancer product on the market we had a Ted fellow Dr Jimmy Lynn who’s one of the most famous 

58:45 

oncologists in the world actually um who was the chief science officer and I was 

58:50 

a commercialization expert and they came to get me and they’re like so you’re the person who understands I’m oncology and can help bring this to the market so I 

58:57 

got on a plane to London and then that the bridge burned down in Georgia so I’m in London and that was when the bridge 

59:04 

Georgia um and and so yeah so we started talking about this to drug companies as an nip company saying that we could help 

59:12 

you understand more about disease in oncology and it’s really taken off like that’s actually kind of Nara signature 

59:18 

product now signatera um and it it really does a lot of great things not just for mrd now like they’ve got really 

59:25 

great research collaborations potentially to do what we call Patient stratification so as we keep talking 

59:30 

about this in drugs what we want to find is a person who’s a responder to the Therapeutics you call it responder and 

59:36 

that’s why the my genal product is called responder id id does responders and so they’re actually using these 

59:42 

tools um like signatera to try to understand more about those potential responders to a therapy and 

59:50 

stratify patients before they go into clinical trial so kind of the Holy Grail of improving your clinical end points is 

59:57 

frontloading it by pre- identifying patients who would probably be the ones 

1:00:02 

to respond so that you can get good clinical end points to get that out to the 

1:00:11 

market some successes to demon yeah because one of the biggest challenges in getting drugs on the market is like 

1:00:17 

making those clinical end points a lot of drugs fail in phase three and it’s actually one of the biggest challenges about those companion Diagnostics you 

1:00:23 

build a diag ostic tool that’s going to help get this to the market by saying these are the right patients and you’re 

1:00:29 

trying to get the drug to the market and you still have to take it all the way through the process and this is again 

1:00:35 

millions and millions and millions of dollars are invested in this and often drugs will fail in phase three and I’ve 

1:00:41 

seen that take out to to Sam’s question the these kind of things are what takes out like destroys these companies is you 

1:00:47 

make the wrong bet on a drug or you make the wrong bet on a tool and so you get 

1:00:53 

to this certain level of evidence that you need to get to the market and and it just doesn’t work so a lot of these 

1:00:59 

tools now are being used to do what’s called rescue drugs so there’s drugs with millions and millions of investment 

1:01:05 

in it they didn’t quite make a clinical end point and if we can look at what the data tell us about the patients in 

1:01:11 

particular that did actually respond or that the likelihood of those responses and then again upload Freel you know 

1:01:18 

preload a set of patients with a disease type that are going to respond to that drug um maybe we can rescue that drug 

1:01:26 

and get it to the patients NOP starting again potentially 

1:01:31 

um to rescue it now I because again the clinical trials have to be built in designation with end points but you know 

1:01:37 

there’s there’s drug candidates that had either good efficacy not but not enough 

1:01:44 

and so one of the thoughts is to take that drug that had great potential and 

1:01:49 

maybe even use it in a different indication because again if you’re looking at what was actually practically working for that or if you’re thinking 

1:01:55 

about how you can use that drug in different ways um you know you can use some of these tools on predictions and 

1:02:01 

try to say well maybe this is a better patient population so you know it’s become a really Rising thing in the last 

1:02:09 

I want to say less than 10 years but it’s definitely become a rising thing in in in my uh experience especially for 

1:02:14 

for companies that have have large investment in these drugs and so there’s companies now that are Incorporated 

1:02:20 

around buying rights to these and trying to restur them using these 

1:02:41 

tools I I really love this question because it gets back to one of the basic tenants of 

1:02:47 

some some Farm of things about Blockbuster so when you asked your question what are challenges that you 

1:02:53 

say rare drugs drug or rare disease does not get nearly the attention that it 

1:02:58 

really needs if you have a small subset of the population that’s going to really really benefit but isn’t going to be 

1:03:03 

what’s called total adjustable Market it’s not going to be a high T that it’s going to be harder for them to get 

1:03:10 

enough investment to make that actually come to fruition because the stringency doesn’t change whether you have a drug 

1:03:16 

that’s going to help 20 million people or 200,000 people you still have to 

1:03:22 

support it all the way through all these trials and so um drug companies are you know the is again the Paradigm is their 

1:03:29 

their commercial entities that want to make money so it’s it’s hard to get 

1:03:35 

something that isn’t necessarily going to Blockbuster why there’s lots and lots of diabetes treatments right because 

1:03:41 

that’s a big Blockbuster of a growing Market what I’m seeing happen now is that smaller companies are trying to 

1:03:47 

step in like there’s a lot of small companies that really just focused in on rare disease and they’ve just made that 

1:03:55 

what we call their thesis so a thesis for the company and a thesis for an investment arm and they’re you know 

1:04:01 

positioning themselves as with the benefit of this will be so high to the people that need it that we’re going to 

1:04:07 

you know make this our way of life and so I think it’s probably a better 

1:04:12 

Paradigm to do it that way than hope somebody like a major Pharma company is going to take the time and attention to 

1:04:19 

look at rare disease now that being said there’s a very large company called blueprint medicine that actually took 

1:04:24 

the time to look at a disease I struggle with um it’s a masto m cell disease maso 

1:04:30 

cytosis and they actually which is considered fairly rare but is actually really probably not that rare and and 

1:04:35 

they spent a lot of time making some some drug options on that so you know but they’re not fizer abbid or that 

1:04:40 

they’re still relatively small but they’re they’re a pretty large company um so you know it it really is one of 

1:04:46 

those ethical questions you know about the Do no harm or due best effort versus 

1:04:51 

the cost of drugs and capitalism and investors now um that being said too one of the 

1:04:57 

things about treatment of patients particular in something like oncology is that there’s you know these interesting 

1:05:03 

biomarkers and cdxs her to her septin all that kind of stuff but you don’t 

1:05:08 

have as many big Blockbusters that really show great strong effect in 60% 

1:05:14 

of patient populations that often you’re more like five% 10% and they will still prescribe that 

1:05:22 

drug even if there’s like a 5% % chance of saving that patient’s life and one of the things that we use to motivate 

1:05:28 

investors and to get money in into this and to get two Physicians who um one of 

1:05:35 

the challenges is you’re up against standard of care and liability right so if you want to substantially change a 

1:05:42 

physician’s opinion especially if it’s going to cause a change to an outcome of a terminal patient or a patient that 

1:05:47 

will be terminal if you don’t give them the right care is you’re really going to have to give them sufficient evidence to 

1:05:54 

convince them to change that pathway and that’s not easy at all like one of the hardest things I’m up against actually is the liability of deviation from 

1:06:00 

standard of care and the concerns and I I always understand this when I hear us from an oncologist because they are 

1:06:06 

making that decision about saving somebody’s life so technologically are you really showing enough evidence to 

1:06:13 

change what’s an accepted standard of care and so when we talk to the investors about how to make these tools 

1:06:20 

better so you can get more people you can actually use a financial argument there and so you can say well I can actually 

1:06:26 

you know make this more than 5% of an expensive drug 5% of treatment option of 

1:06:33 

a very expensive drug I can like help these tools and maybe you’ll hit 50% of people who need it kind of thing but 

1:06:38 

also the drug companies you know they’re still getting paid for use so even if it doesn’t help 95% of patient potentially 

1:06:44 

they still want that prescribed because they’re still getting that money in right so these are the constant things 

1:06:50 

that you have to ask like am I doing no harm on a fi % chance that I might help 

1:06:55 

them with a potentially toxic medication or am I doing no harm by telling them no 

1:07:02 

when it might actually be something that can help them and so that’s again why these tools are so important and why it 

1:07:10 

is very very difficult in some ways to make people change practice because if 

1:07:18 

you’re talking about fragile patients if you’re talking about terminal illness you are 

1:07:25 

harming somebody no matter what you do in some way and that’s actually I think one of 

1:07:32 

the biggest take-homes of of my entire career you know am I waiting the things I’m helping to get to the market do the 

1:07:39 

best kinds of impact or am I just making a capitalistic way of 

1:07:46 

making people pay for more drugs right I think about that all the time 

1:07:53 

well thank you very much 

1:08:09 

Robin so once again thank you to mation for having us we’re probably do this room again hopefully there’ll be a 

1:08:15 

little less crowd outside um and a little easier to find the Speak Easy which is already a hidden room um but 

1:08:23 

yeah thank thank you very much for coming to this science Junction we really appreciate it our next event will 

1:08:28 

be for science Junction will be January 25th we’ll be at highcard brewing in Tucker and uh our executive director Amy 

1:08:36 

Sharma will be talking about advocating for better beer laws in Georgia um like how to advocate for 

1:08:45 

anything environmental science beer laws is an 

1:08:52 

interesting navigation of politics and the legislator here in Georgia being in 

1:08:59 

Atlanta we definitely have the capital so there’s a little bit more access so that’s going to be part of that talk um 

1:09:06 

hopefully then you’ll be able to buy more than just a couple packs from your breweries might be able to buy a couple 

1:09:11 

a little bit more uh so thank you again for coming out we do this event for 

1:09:16 

people like you and we get support from people like you um if you would like to 

1:09:21 

scan this QR code and let us know what she thought really appreciate it and thank you very much good 

1:09:34 

night 

Was this article helpful?

Science for Georgia is a 501(c)(3). We work to build a bridge between scientists and the public and advocate for the responsible use of science in public policy.

Back To Top