Absolute Gene-ius

Helping democratize access and use of mRNA technology

Episode Summary

The use of mRNA as a vaccine platform has been mostly derisked and its speed to move from antigen discovery to drug product is impressive. However, high-purity mRNA can still be a challenge to make. Dr. Christian Cobaugh, CEO of Vernal Biosciences, is on a mission to make high-purity mRNA available to more researchers and drug developers to help realize even more potential from mRNA medicines.

Episode Notes

The potential of mRNA medicines was postulated for years, but it took the COVID pandemic and emergency use authorizations for that potential to be demonstrated. By now, most of us have received at least one mRNA based vaccine and the platform has been mostly derisked. However, if you’re not one of the major players in this space, generating high-purity mRNA, let alone a GMP-grade mRNA-based drug product, can still be quite challenging. 

Dr. Chrisitan Cobaugh, CEO of Vernal Biosciences in Vermont, has been working in the mRNA field for more than a decade and is passionate about the potential of mRNA medicines. He’s also been in the field long enough to know firsthand the challenges of high-purity mRNA and lipid nanoparticle supply. Join us as Christian walks us through his story, the start of Vernal Biosciences, and their progress toward their mission of democratizing access to mRNA technology. Our conversation touches on the molecular biology of making mRNA, and the use of digital PCR and other methods in monitoring development and release of mRNA drug products, and the potential applications of mRNA as a platform (some of which you might not have guessed).

Whether you’re new to the technology, or have chosen mRNA as a focus area, you’re sure to find this conversation engaging and intriguing, and our guest insightful. 

Visit the Absolute Gene-ius page to learn more about the guests, the hosts, and the Applied Biosystems QuantStudio Absolute Q Digital PCR System. 

Episode Transcription

Jordan Ruggieri 00:00

Christina did your, did your dog in a hockey outfit like the episode today?

 

Christina Bouwens 00:04

He loved it, he loves all the episodes.

 

Jordan Ruggieri 00:09

Christina has a great picture of a dog in hockey gear on her wall for all the listeners who can't see.

 

Christina Bouwens 00:15

His name is Teddithee. Teddithee.

 

Jordan Ruggieri 00:17

Teddithee?

 

Christina Bouwens 00:18

Teddithee. Teddithee, like the very formal name of Teddy.

 

Jordan Ruggieri 00:35

Welcome to Absolute Gene-ius, a podcast series from Thermo Fisher Scientific. I'm Jordan Ruggeri. 

 

Christina Bouwens 00:41

And I'm Christina Bouwens and today we take a deep dive into mRNA research with our Gene-ius Dr. Christian Cobaugh.

 

Jordan Ruggieri 00:47

Christian completed his PhD in cell and molecular biology in 2007 and spent the following decade and more leading mRNA drug discovery and process development. In 2021, he became the founding CEO of Vernal Biosciences, an mRNA CDMO based in Burlington, Vermont. We loved learning about how Kristin and the vernal team use digital PCR in their work, and hope you enjoy our conversation. 

 

Jordan Ruggieri 01:14

Christina, I have another pun for you.

 

Christina Bouwens 01:16

Okay, I'm ready.

 

Jordan Ruggieri 01:18

Why did the mRNA drug discovery scientists go broke? 

 

Christina Bouwens 01:22

Why? 

 

Jordan Ruggieri 01:22

Because every time they tried to make a breakthrough, it translated into a hefty expense.

 

Jordan Ruggieri 01:33

Christian, thank you so much for joining us on today's episode of Absolute Gene-ius. We are thrilled to have you. Can you give us maybe a little bit of an overview of yourself and maybe Vernal Biosciences?

 

Christian Cobaugh, PhD 01:47

My name is Christian Cobaugh. I'm the founding CEO of Vernal Biosciences. Before I started Vernal, I got my PhD in cell and molecular biology and immediately started my career in monoclonal antibody engineering. But back in 2013, when I was working for Alexian Pharmaceuticals, I started in mRNA medicines as part of a team at Alexian partnered with Moderna, and I've been in mRNA ever since then. Back in 2021, I grew a little impatient working for innovator companies in terms of our ability to outsource mRNA manufacturing and lipid nanoparticle manufacturing. And, you know, because of COVID, there was a lot of enthusiasm and demand frankly, for mRNA manufacturing. And it was right around that time, in April 2021, when we launched Vernal Biosciences to sort of solve problems around high-purity supply of mRNA lipid nanoparticle formulation. And so Vernal is a research grade manufacturing company, but has recently added GMP manufacturing, which was part of the plan all along. I mean, our goal truly, and we live it every day, is to democratize access to mRNA. You know, Moderna and BioNTech have really solved for their own needs, but a lot of smaller companies have not. And those are the companies that Vernal is actively working with. So, a lot of science goes on here, but you know, we are a business as well.

 

Christina Bouwens 03:24

Yeah, there's so much to dig into here. And I know, we'll definitely jump into more about mRNA, mRNA research but um, one that stuck out to me a lot that I think our listeners could learn a little bit more about is this idea of lipid nanoparticles. Can you kind of give a little bit of an overview about what lipid nanoparticles are?

 

Christian Cobaugh, PhD 03:41

Fundamentally, if you think about an mRNA drug product, which is the fully formulated material that's in a vial, the active ingredient as many people consider, is the mRNA itself. That's commonly referred to in our field as drug substance. In order for that, that drug substance to transit the site of injection all the way to its site of activity, the mRNA has to be protected. As many of us know mRNA is incredibly labile. Nature has designed it to be that way. And we'll break down almost instantly if injected without some type of shield around it. But the lipid nanoparticles do more than just protect it. They help the mRNA get into the cell type that they need to so they transit the plasma membrane and then they have this magical ability where upon the pH starts to drop in the compartment that brings them into the cells and this is an intracellular compartment called an endosome. The mRNA gets released into the cytoplasm and once it's in the cytoplasm the cell treats it like it does any other mRNA, including the ones that it makes itself and translates it into protein. So kind of many, many different roles there for the lipid nanoparticle. And generally they're made up of four different lipids. There's a cholesterol, phosphatidylcholine, pegylated lipid which provides stability, and then there's the ionizable lipid, which kind of gets does the magical thing of allowing the LNP to fuse with the endosomal membrane on that low pH and then the mRNA spills out into the cytoplasm.

 

Christina Bouwens 05:16

That's amazing. So I assume that part of in, part of this, this whole process is actually that payload design. And so that lipid nanoparticle creation is kind of the end part of that. But can you talk a little bit about that, that upstream process and maybe, you know, here at the Absolute Gene-ius podcast, we love to get into how dPCR is used and you know, is dPCR at all used in that that more upstream process? You know, before that lipid nanoparticle physical creation.

 

Christian Cobaugh, PhD 05:41

The answer to your question is dPCR is becoming an essential part of our workflows. If we go back a little bit to one of the original business problems that Vernal was set up to solve for, it was, you know, high purity mRNA. And not to bore the listeners with cute details around QBD. But you know, if you can't control your process, meaning if you don't understand what's going on with every step of your process, it's really hard to get pure product at, you know, high purity products at the very end. And so, you know, the mRNA process starts with a DNA template. Often, in our case, it's pDNA and the mRNA is made from there. And we're using dPCR, to evaluate the purity and the identity of the pDNA as well as the mRNA. So we do a cDNA conversion and we're able to use dPCR to confirm the identity, and in some cases the purity. And that's where we're really trying to innovate with dPCR.

 

Christina Bouwens 06:45

Amazing, and just to help our listeners here, the pDNA, is that plasmid DNA? Or, 

 

Christian Cobaugh, PhD 06:50

Yeah, so we mostly work with plasmid DNA. We bring it through E. coli, we do clonal selections, we bank our favorite, you know, or highest performing clones. And this isn't just the highest yielding, this is also clones that, you know, have other critical quality attributes. You know, we're using dPCR, you know, as part of the release package for those clones. And then, once the E. coli has taken up the plasmid, and we expand that, we do a plasmid prep, we linearize the plasmid, and then we move that forward once it passes the test to go into the RNA synthesis. And that's usually called an in vitro transcription reaction. But it all starts with, with pDNA.

 

Jordan Ruggieri 07:42

I want to go even maybe one step further back. A you know, mRNA therapy research has been a hot topic since COVID -19 pandemic. Can you discuss the research being done for mRNA therapies and why it's so promising and really a cool and innovative technology?

 

Christian Cobaugh, PhD 08:00

Well, you know, I think we can safely say that using this technology for infectious disease vaccines has been mostly derisked. Now will it be useful for all viruses? No, probably not. But neither is you know, most other vaccine platforms. The cool thing for infectious disease is the speed by which you can identify the antigens and then have a manufactured product. Obviously, as we mentioned, infectious disease vaccines, personalized cancer vaccines, the clinical data, it really is game changing. And then, you know, I'm also super excited about using mRNA, for in vivo gene editing. And I think more recently, we're starting to finally break through, or break beyond, what was commonly referred to as “gene graffiti,” I think George Church called gene editing that, in actual repair of genes. We're kind of in like version two and three of those technologies, where it's starting to feel less like a science project and more like a clinical application for mRNA. So the wheels are turning, progress is being made. And you know, the clinical trials are a little bit more mature than where I think they were pre COVID. 

 

Christina Bouwens 09:21

Yeah, I have a question kind of about where it is, where Vernal sits and the customers that you have and where you're interacting with. And my question kind of stems off of what you're just getting at, as not being so much of a science project anymore. We're getting into, you know, this, these are real, you know, real products really getting to market. Are you starting to get into the realm of where you're seeing criteria for, for testing and release and like, starting to really do in-depth QC testing before these products are leaving your lab. What kinds of tests are you seeing in your lab with customers and your workflows?

 

Christian Cobaugh, PhD 09:55

We're seeing all the use cases, you know, that I just mentioned, probably We have few more. We actually have some customers that are using mRNA as a research tool, and that's a very important part of our business and it's a very powerful way to use mRNA. You know, in terms of the release testing, if we look at all the major unit operations involved in manufacturing an mRNA, that there's five major ones, there's the cell bank, the pDNA, the mRNA, the drug product, and then the fill finish. You have release testing around all those. Across all those we've got roughly 50 different analytical tests. And within those 50, we probably have about 25 unique testing platforms. dPCR as an example, of a testing platform. Some are really common, widely utilized, and some are a little bit more cutting edge. We're finding things with dPCR, that we weren't able to find, you know, before now. And a good example of that is, you know, people generally will quantify RNA medicines using absorbance. It's pretty crude, because it gets all the RNA, including the product related impurities, and then you run a HPLC, or a CE to evaluate the purity. Again, you're getting a peak and you're kind of assuming that all the area under the peak is your target product. What we've been able to recently show with dPCR is that dPCR is able to show us what the active component of that is. And we're able to sort of use those other methods as orthogonal methods to say, hey, look, this is 95% active product or this is 90% active product. And so we're really excited about that. But understand that we're very much staying ahead of draft guidance, and we think that's actually a good thing. And so, while I think it's helpful to have, you know, forward thinking organizations like NIST and USP providing that draft guidance, I think it's also important to allow companies like Vernal and our sponsors to push the envelope forward, and the FDA, they want sound scientific principles for all these things. So, justify it, you know, qualify it, validate it, and off you go.

 

Christina Bouwens 12:27

I have a couple of deep dive questions that I had from our original conversation that I do want to get into. There's some keywords that stuck out that get my science brain excited, that I know, I know, our listeners would really like to learn more about. Especially because I do think that they tie really well with some of our, our dPCR applications that go hand in hand. Um, so I'd really love it if you could talk a little bit more about gene assembly technology. I thought that was a really nice key term that I'd love to have you give us a really brief overview of what that means and kind of tie that into you know, how dPCR is useful. I know, you mentioned that it's nice to not have to use a standard curve. I'd love to hear your thoughts on you know, why those two tie together because I think that they play really well together. And I'd love to hear your thoughts?

 

 

Christian Cobaugh, PhD 13:13

Yeah, I mean, it's it, it's important to know where all of this starts so that we can track from start to finish and it is the gene assembly. Vernal is using as many different approaches and strategies as we can to get those templates and to get them quickly. It often is the most time-consuming part about starting a project because traditional gene synthesis, it's error ridden, requires a lot of screening, and these days when done well it's there's a lot of automation involved. Thankfully, you know, we now have a lot of ligation independent cloning strategies. There's some that are sort of a hybrid, whether it's Golden Gate or Gateway. And then of course, Gibson has changed a lot of things. And so Vernal is using all of those both within our own four walls, but also with some of our partners to rapidly provision us with templates. I do envision a day where we start to move a little bit away from pDNA and move closer to enzymatic synthesis. It can be faster, because you avoid use of a cell bank. And this is where the molecular QC methods are going to be much more powerful than the methods that are reserved for E coli, which are a little bit more biochemical. Obviously, sequencing is a molecular method, but you know, we don't sequence at every step of the way and throughout, you know, so we were on, you know, gels and HPLC and CG which are a little bit more biochemical in nature versus what I think we're starting to see on a enzymatically produced template, which it's just easier to do and faster to do with, you know, either a q or dPCR based method. And like, as you stated, the game-changing benefit to dPCR is we don't have to get into this chicken and egg thing around reference standards, it's got single molecular resolution. And it's simple. It's not any harder for us to run than qPCR frankly.

 

Jordan Ruggieri 15:38

Taking a break from our conversation for a quick message, if you are looking for absolute quantification for your gene targets, be sure to check out Applied Biosystems™ QuantStudio™ Absolute Q™ dPCR System, you can achieve precise quantification without the need for standard curves in only 90 minutes.

 

Christina Bouwens 15:55

You can learn more at www.thermofisher.com/absoluteq or visit the Absolute Gene-ius webpage.

 

Jordan Ruggieri 16:03

The Applied Biosystems™ QuantStudio™ Absolute Q™ dPCR System is for Research Use Only. Not for use in diagnostic procedures. And now back to our conversation. 

 

Jordan Ruggieri 16:15

Yeah, I do have one more actually, before we hop into the career element here. But can you actually talk a little bit about the interplay of q and dPCR in your workflow? And you know, do you find benefit in having both, and maybe where one might work better than the other? 

 

Christian Cobaugh, PhD 16:34

Well, I mean, a funny story here, we started by buying a couple of dPCR instruments here at Vernal. No qPCR instruments. And, you know, one of our staffers was, she was very comfortable with qPCR. But like I said, we didn't have it. And I think she suffered in silence for a while she was trying to both figure out how to operate the dPCR instrument, which is not terribly complicated, but you know when you're at arm's length from your vendor, they can't hold your hand every step of the way. There's a learning curve, of course. And then also trying to develop analytical methods on that. So, it's like two major learning curves going on simultaneously and she started going over to the University of Vermont to work at their core center on their qPCR in order to kind of vet, I would say, in a more cost-effective way, in a more comfortable platform to vette the dPCR assays. These are the primers and the probes. And, and so we were able to budget, you know, a Quant Studio to come in as sort of the foundational technology and to do some of the assay development work there. I think we're now sort of at a phase where we can do a great deal of our assay development on the dPCR platform. I think the cost structure around qPCR is still a little bit more favorable. So if it's, you know, sort of a frontier or a pioneering type of experiment, you might see those folks turning to the Quant Studio, or some other qPCR platform, in our case, it would be the Quant Studio. But to get those, you know, very new types of assays off the ground.

 

Christina Bouwens 18:39

I actually love that approach. To be honest, I think that's one of the ways that we've seen a lot of success in getting people off the ground. I think fundamentally, there's, there's so much comfort with how qPCR works. It's been the gold standard for a long time. Everybody's really familiar and comfortable with how it works. And that's been how we've been most successful in getting people comfortable with the dPCR is starting with qPCR. Starting with the basics and getting, you know, understanding for you know, how the assay works. What do the kinetics of the reaction look like? What are you expecting to see in your dPCR reaction? You know, if you're having problems and your dPCR reaction, get back to qPCR basics. So, I think that actually plays really well with how we've seen some of the most success and you know, in really, in selling the value of dPCR and understanding, you know, where qPCR fits best and where dPCR fits best. I love that story. I think it's actually a very good one. Is there a particular use case or application that you really see is very uniquely dPCR? Like where you really couldn't answer it was anything besides dPCR.

 

Christian Cobaugh, PhD 19:39

We're very excited about using dPCR to quantify the active mRNA. One of the issues with RNA synthesis is you get a lot of truncation products. It can only be maybe only, it’ll be 5% could only be 10%. And for a lot of folks, that's considered a success. But the current analytical methods don't do a great job sorting those out as separate from the main product. If we design dPCR assays to land on the right spot of the mRNA, we get a lot closer to the truth in terms of what is the active material. This is not something you're going to find in the USP, and I'm not sure you're going to find any other company that's doing this. And so we're super excited about that because, I mean, we've got again some orthogonal data, including sequencing data, that's showing us that we're heading in the right direction. We still need to move this into a qualified environment. But, you know, this is very differentiated and honestly, I'm not sure of any other technology that can be that accurate in its ability to evaluate the active component of your DS in an mRNA medicine.

 

Christina Bouwens 21:04

And if I can ask this is probably tied to the ability to not just look at at total length, but the ability to interrogate single molecules in their state? 

 

Christian Cobaugh, PhD 21:13

Correct. Yep. And you know, you can tile assays, because it's, you can multiplex, eventually we see a way to do this with just a single assay on the RNA. So that multiplexing right now is there to kind of develop the method. But ultimately, when we go to qualify this as a release assay, we think this will be a singleplex approach.

 

Jordan Ruggieri 21:40

That's awesome. Oh, that was a good question. All right, Christian, let's move to the career corner portion of the podcast where we get to talk a little bit about you and your journey. You had mentioned earlier on that you have a cell and molecular biology educational background. Can you elaborate a little bit of that pathway? How did you come to that particular concentration and focus and take us a little bit back on your educational journey?

 

Christian Cobaugh, PhD 22:11

You know, as I mentioned, I got my PhD in Cell and Molecular Biology from the University of Texas at Austin. I kind of wandered around for the first year through an RNA structural biology lab, a lab that studied group two introns. But I wanted to migrate more towards clinical applications. And that ended up being a lab that developed platform technologies to discover monoclonal antibodies. You know, I just kind of felt like that would get me into a career in the commercial sector a little faster than had I done something a little bit more fundamentally based in, you know, RNA biology. I will say at the time, antibodies were hot and I did a relatively brief postdoc. It was an industrial postdoc. and then I rolled forward into monoclonal antibody discovery with a couple of companies. The most successful company was Alexian Pharmaceuticals and they were rare disease focused first. And that eventually brought my lab and another side function that I had in search and evaluation for the business development team, into the Moderna partnership, that Alexian signed back in 2014. So, I diligenced that partnership, did the technical diligence for the BD team, we made a recommendation to the board to partner with Moderna and we signed that deal. And next thing I know, they asked me to lead the research for that partnership, and I moved up to the Boston area from Central Connecticut, which was nice. And we opened up a lab close to Moderna. And had a really wonderful time collaborating with them. It was highly collaborative and they had great science going on. And then we built out drug discovery capabilities and drug development capabilities at our lab. And again, I've been in mRNA ever since then, but I stayed scientifically curious, read papers you know constantly, and got kind of lucky. It wasn't about following a particular path. It was just about, you know, the curiosity, working with urgency, those types of things.

 

Jordan Ruggieri 24:38

We hear that a lot actually is you know, it's not a path that you would have picked right away where you're from the get-go. But being open to those opportunities. Staying curious is a critical component of building your own career and going where you want to go. Another question for you that's a little bit more open-ended is, why do you love what you're doing now? What makes you excited to get up in the morning and go to work every day?

 

Christian Cobaugh, PhD 25:04

You get to a point as a scientist, where, at least in industry where you have to start paying attention to other things. The hardcore financials, there's your investors, there's, you know, building and managing a team and a culture, you know, there's customers. So, one of the things that really excites me is like learning new things and doing new things. I was pretty comfortable as a scientist. And I think I probably felt like I had a lot more to achieve in life. But getting into that can also be kind of a rut and I think this sort of achievement mentality, you'll never be fully satisfied. And so what I've told people, at least in the startup environment is like, don't be afraid about moving forward and moving beyond what you're already doing quickly. If you're allowed to do it, take advantage of it. Because leaders need to be kind of well rounded. It's not enough just to be a really good scientist, and to understand the scientific method. You know, you need people skills, you need to be able to negotiate, you know, you need to be able to motivate people to show up on time, but also to take care of themselves, and those are sometimes opposed to one another. And, and so you just have to continue learning and experiencing new things. And getting feedback as well, that's, that's really important because you just, you can't see it yourself. So it's like listening to your own voice on a recording, you're like, you can't believe that that's you. And same thing is true of your career growth and development.

 

Jordan Ruggieri 26:54

That's another important reason, I think, to have a mentor. Yeah, excellent. Okay, now for the juicy question. We asked this of everybody on the podcast. So it's not just you. Ba dub a dub a du. We'll add a drumroll in. Drumroll, Matthew. What is your most embarrassing lab moment? And on the flip side, what are you most proud of?

 

Christian Cobaugh, PhD 27:18

Oh, my gosh, my most embarrassing lab moment, I'd say, pretending that I knew how to do phage display for a solid year. And I built beautiful antibody libraries, which were well QC'd, and I had no idea how to QC my phage display experiments. And so almost literally a whole year's worth of effort as a PhD student had to be repeated. It was fine because I enjoyed being a student. Fine. But I think on the back end, you know, I had a wife and a child, and they're like, you know, like, “Hey, like, let's, let's move on here.” Like it happens. That's one of the reasons why a PhD takes so long to get because you make mistakes, and you learn from them. And the key is honestly, learning from that it's having the right controls, and not, you know, not taking shortcuts, because it's worked so well for somebody else in the past, therefore, it has to work well for you. That's not the way it works. So in terms of like embarrassing ones, I would say that's the best. And by the way, I, when Vernal started, I did work in the lab, and I occasionally will go in and do stuff, small stuff for people on the weekends. But I do also warn people, like, you know, at this point in my career pipetting and that, like it's going to take me a lot longer than if somebody more skilled, more talented, were doing it.

 

Jordan Ruggieri 28:46

That's hilarious. And what about what you're most proud of?

 

Christian Cobaugh, PhD 28:48

Oh, that's a good question. I take a lot of pride in the work of my teams. Maybe that's kind of like a parental pride. But yeah, I mean, I love to see people, you know, that we've hired, come in and learn something new. And then after learning something new, and they kind of master it, they become an SME. I mean, we've got people at Vernal that I often introduce them as “the world's leading expert in whatever,” a lot of cases, it's the newer technologies. They'll be friends with the field application scientist, and the next thing we know the field application scientist is calling them up for answers. And so people get really excited about that and so it's sort of this self-perpetuating cycle. That's probably, workwise, the thing I, I just feel the best about it just gives me the warm and fuzzies to see that happen. 

 

Jordan Ruggieri 29:51

That's amazing and awesome. I know Christina has been very excited to ask a question from you. So I actually want to pass it back to Christina to ask this question.

 

Christina Bouwens 30:02

I have a very silly fluffy question, because I was looking at a Vernal's website over the last couple of weeks, and I noticed there's some pretty intentional coloring on the logo. So I have to ask, is there a story behind the name?

 

Christian Cobaugh, PhD 30:16

Yeah, so the name itself, obviously, it incorporates RNA sort of consecutively. And so that was part of this. We're also based in Vermont, and Ver, and you know, Vermont kind of work well together. It is sort of short, in terms of French for green. And right now, as I gaze out of our picture, windows, all I see is green outside, and this is the Green Mountain State, green and Vernal are both they both evoke images of rebirth, of life, and renewal. And those are images that I think are really important for a business that's, you know, trying to play a small but important part in bettering human health. So you know, it's a great time to be in Vermont, but even the winter when it's cold and white is also a great time to be up here. And so we really do value our roots up here, because of some of the, the imagery that it evokes.

 

Christina Bouwens 31:25

Well, I love it and I thought it was very intentional. So I'll give our our fellow marketing colleagues over there good shoutout. I thought it was a beautiful logo. 

 

Christian Cobaugh, PhD 31:32

Thank you very much. 

 

Christina Bouwens 31:34

I want to call it out. 

 

Christian Cobaugh, PhD 31:35

Thank you.

 

Jordan Ruggieri 31:36

And even in the snow, you know that green is coming right? So it's, it's an awesome story. Christian, thank you so much for being our guest on Absolute Gene-ius today. We really appreciate your time and we look forward to having our listeners listen to all these wonderful stories and insights and thank you just a few so much for all your time.

 

 

Christian Cobaugh, PhD 31:56

My pleasure, guys. It's been a lot of fun.

 

Jordan Ruggieri 32:00

That was Dr. Christian Cobaugh, founder and CEO of Vernal Biosciences in Burlington, Vermont. Thank you so much for joining us for today's episode of Absolute Gene-ius. It was produced by Sarah Briganti, Matt Ferris, and Matthew Stock. With more great science around the corner in future episodes, stay curious and we'll see you next time.

 

Christina Bouwens 32:20

All right, Jordan. Do you want to hear my joke? It's a good one.

 

Jordan Ruggieri 32:23

Oh, I love jokes.

 

Christina Bouwens 32:26

Why was the RNA so good at making music? 

 

Jordan Ruggieri 32:29

Why? 

 

Christina Bouwens 32:30

Because it always knew how to transcribe a hit!