mRNA: What did Covid teach us about drug development and distribution?

Invent: Life Sciences - Season 2 Episode 1 Transcript
Speakers: Stuart Lowe, Dave Sokolowski & Dan Peer
[Music playing]
Stuart: It’s hard to believe sitting here in 2023 that just three years ago, the world was fixated on a technology that few had heard of just months before. While vaccinologists were following multiple avenues to develop a tool to combat the threat of COVID, one approach stood out, Messenger RNA.
Messenger RNA or mRNA, seemed to offer a safe solution that was easy to develop and manufacture at scale, giving it a head start on other vaccine candidates. The main challenge quickly moved from would it work to how can we manufacture and distribute billions of doses?
Manufacturing processes were created using the most expedient means possible, which often meant redirecting existing capacity and all this culminated in the global vaccine rollout, which kicked off in late 2020.
Now, many scientists are seeking to deploy mRNA at the more individual level to create vaccines that target patient-specific tumour markers or respond rapidly to local infectious disease outbreaks.
One of the challenges we now face is that the methods that allowed for billions of doses to be manufactured might not be cost effective for much smaller batch sizes.
So, what are the new mRNA-based therapies under development and how can we make them?
Join me Stuart Lowe, as we plug in to Invent: Life Sciences, a podcast brought to you by technology and product development company, TTP.
Today we ask what did COVID teach us about the future for mRNA? To find out, I spoke to Dave Sokolowski from Cytiva. Dave is on the cutting edge of manufacturing technology, and he's kept a close eye on the RNA field and its potential applications that have emerged since COVID.
Dave: My name is Dave Sokolowski, I'm the global workflow manager for the Nucleic Acid Therapeutics business unit within Cytiva. So, Cytiva is a technology company that builds manufacturing platforms. We're remarkably well known for protein A and our mAb business and now we're really trying to focus on the new RNA business that's popped up due to mostly the COVID pandemic.
Stuart: I asked Dave to share some insight into Cytiva's decision to enter the mRNA manufacturing space.
Would you say that the decision to go into mRNA manufacturing or mRNA enabling technology is something that has been a long time in the brewing or is it been accelerated by what happened during the pandemic?
Dave: Advances in synthetic biology were our first trigger to start looking into RNA manufacturing. The pandemic sealed the deal.
Seeing new plasmid DNA technologies come around. So, moving from an E. coli-based plasmid over to synthetically manufactured RNA or DNA really, I think is the start of this all for us as a business, but the pandemic certainly pushed this over the edge for us to fully develop our strategy and our drivers in this space.
Stuart: And how about you personally, how did you end up in mRNA manufacturing?
Dave: I've been in this business for about 10 years so far. I came through both the biochemistry and the engineering route, so I've degrees in both of those. My graduate research prior to leaving a PhD program to pursue mechanical engineering was looking into RNA binding proteins.
So, I do have history in RNA manufacturing or at least early RNA discovery prior to entering the manufacturing space with my engineering degree. But this is near and dear to my heart. I would absolutely love to see therapies come out based on technologies that we develop as a team.
Stuart: So, obviously Cytiva was looking at the synthetic biology space. Was there any sense of okay, mRNA vaccines is going to be something massive or pre pandemic it was like, “Oh, this is something that might come along in the next, what, 10, 15 years.”
Dave: You're pinning the development time cycles exactly right, it takes 10 to 15 years and a few billion dollars to develop a new therapy. This was a small portion of the company thinking about which next generation therapies we support.
And I'm specifically saying therapies because we had no idea that it would be a COVID vaccine that would bring these technologies to bear. We thought it would be cancer therapy. That seems to be the drug that still hits at the top of the mRNA drug lists in terms of phase developments.
But it was really small team thinking about which new manufacturing modalities we need to come up with to support the molecules in the field. RNA was one of them sitting alongside exosomes, oncolytic virus, other technologies that are still waiting for broad adoption.
[Music playing]
Stuart: Speaking to Dave, I was excited to hear about the new wave of therapies and innovative manufacturing platforms on the horizon.
So, I wanted to find out how mRNA research is playing out now, post pandemic. I spoke with Dan Peer, a professor at Tel Aviv University. Dan is the director of the Laboratory for Precision Nanomedicine, as well as the Vice President for Research and Development at the university.
So, just to start us off, would you mind just introducing yourself and telling us a bit about your work in Nanomedicine?
Dan: So, my name is Dan Peer, I'm a professor at Tel Aviv University, the director of the Laboratory of Precision Nanomedicine here at Tel Aviv University. And I also happen to be the Vice President for Research and Development at the university, actively engaging in Nanomedicine in the past 20 years.
Predominantly one shop store basically for everything from synthesising lipids, special ionizable pH sensitive lipids, to creating targeting moieties that could be coated on lipid nanoparticles and help deliver RNA payloads to specific cell types.
Stuart: What about kind of the future for mRNA? Are we going to see many, many more medicines with an mRNA basis? I mean, in theory you could produce biologics.
Dan: So, it seems like this, in January 2020, there are only four or five clinical trials with mRNA, now there are hundreds of new things with mRNA in different variations, in different diseases, in different indications, not only for vaccines, but also for therapy, also for diagnostics. So, take into account all of them, this is quite remarkable.
Stuart: It's a big turnaround, isn't it? Do you think we would've got here without the COVID shock?
Dan: Well, most likely, no. I think it boosts dramatically the field. You also look at the amounts of funding that went into those developments.
Stuart: Yeah, you needed a spike, a big challenge in order to kind of marshal all of those. But actually, thinking about the impact beyond just that immediate COVID challenge, it has opened things up. What's the next step of the research? What are the next improvements that you see are necessary?
Dan: Well, I think that we need to show clinical proof of concept for the ability to target in a cell specific manner what we have doing in the past 17 years already in animals, it's time to do it in human.
Stuart: And is this in the field of oncology?
Dan: Predominantly it'll be in the field of oncology. I think the first one will be in oncology.
Stuart: Are you also thinking about neoantigens as well?
Dan: Yes, but I think neoantigens in oncology is a tricky area because tumours are modifying themselves and depending on treatment, it's an evolution that they're undergoing in real time.
Is it going to be a sustainable method or therapeutic approach? Maybe, I'm not sure. But I think that it's a battle against tumours and all the time they're changing, we have to come up with better ideas.
Stuart: Yeah, because it's like an arms race, isn't it? And the enemy is always evolving and coming out with other plans.
So, targeting is something that you are looking forward to seeing more targeted therapies.
Dan: I'm not only looking forward, I know that in the next two to three years, we will see a few clinical trials trying to reach this bar. Some of it will fail, but we will learn from those results and some of them will be successful, and it'll open up new avenues in this area.
Stuart: No, that's very exciting and the other exciting thing is the prospect of targeting solid tumours. So, you think that you've got real potential for using lipid nanoparticles in solid tumour.
Dan: So here, I'm less optimistic at this point, but I think it'll take more time. As monoclonal antibodies first being very effective in haematological malignancies, I would expect that in the case of LMPs, targeted LMPs will be effective in haematological malignancy first and only then we'll try to tackle solid tumours.
There is a lot of technical and biological barriers there. So, I'm not super optimistic in a short time, but I am more optimistic in the long run.
Stuart: Is there anything about the lipid nanoparticles in particular that lend themselves to solid tumours or is it just a different approach that may help?
Dan: No, I think in many types of solid tumours, there is an enhanced permeability and retention effect that could accumulate them in close vicinity of the tumour. But is it good enough and can they go deeply into a solid tumour? That's questions we don't have good answers right now.
And even ADCs antibody drug conjugates have issues in penetrating solid tumours or antibody by themselves, and there is also hypoxia inside the tumours. Hypoxia is a big issue as well.
So, I think that technologically and biologically we're not there yet. We might find some solutions to — it's like an onion take another layer and another layer until you go to the heart of it but I think we're still not there.
[Music playing]
Stuart: COVID vaccines were a great validation of the mRNA platform, but as potential applications go far beyond this single disease, these forms of treatments have the potential to give us new tools to fight some of humanity's most impenetrable diseases such as cancer.
In fact, mRNA that's been encapsulated in lipid nanoparticles has several benefits that allow it to overcome tumour biology, namely the ability to personalise therapies and target specific regions of the body.
Those who were keeping a close eye on COVID vaccine development will recall that the Pfizer vaccine needed to be stored and transported at -80 degrees, dramatically increasing the cost of the vaccine's deployment.
I asked Dave about this and other challenges he might face in manufacturing the mRNA molecules of the future.
I suppose necessity can often breed invention. What sort of surprising things came out of that urgent mobilisation?
Dave: So, aside from investing into supply chain, we rebuilt many of our internal product development systems and a lot of our new product introduction systems to help us move products into market faster.
We realised that for pandemic scale response, supply chain is one thing that needs to move but to create products and technologies that fit into the space accurately, we need to build them, launch them, and manufacture them much faster than we previously had.
Stuart: Looking back at how COVID accelerated the innovation in this space or the development of manufacturing technology in this space, is it fair to say that there's a bit of a silver lining because we now have a better outlook for indications like cancer?
Dave: And in addition, indications like malaria and yellow fever and rabies, which has been expensive to manufacture flow with relatively small population base, but quite a severe disease, nonetheless.
I think there is a shining silver lining that comes along with the approval of mRNA therapies. I think we're at the start of a new era of precision medicine. And when I say the start, I really mean the start. We're the very first references in a textbook that people will be quoting decades away.
But also, I think we're at the very start, meaning we really don't know the challenges that are coming up in front of us. For example, the incorporation of pseudouridine was thought to be the primary driver for low immunogenicity in the COVID vaccines or in mRNA vaccines.
But we're starting to see thoughts come about thinking that maybe it's just the presence of impurities in the manufacturing process that lend to the immunogenicity and the reactogenicity of these types of drugs.
So, while there's a silver lining, I think there's a lot of work that comes underneath the shadows of those clouds. And I think the whole industry is really excited to face those challenges because the tools we've been building over the last three decades in biotech are well suited to tackling them.
Stuart: It seems like there's going to be a lot that we can learn already from what's out there in terms of what the effect of impurities might be. Have you seen any tools or techniques that might be able to bring to bear on that?
Dave: Well, the tools are still under development and I don't really need to disclose what we're going to be releasing in the future, I would like to comment on the tools we currently have at hand for assessing impurities.
Since it is a fully synthetic process, we do know all of the impurities that come through in that process. Some of them are in such remarkably low concentrations and our immune systems are so highly tuned at recognizing them that we lack the capabilities to properly assess them.
For example, double stranded RNA is largely the most talked about impurity in an mRNA vaccine or an mRNA therapeutic. For a vaccine, maybe some double stranded RNA is required to act as an adjuvant for the immune system to uptake that genetic genomic material.
For a protein therapeutic where you're dosing 10 to a hundred times greater than that, I think a double stranded RNA profile of zero is acceptable, not 0.1%, not 1%, zero is going to be what's acceptable in the future.
How do you quantitate zero or near zero with tools that haven't been calibrated to quantitate zero or near zero? Those need to be developed, but they're not impossible to develop since we do have experience in the past to developing these types of analytics.
Now, the better question is can we do this inline or online? That remains to be seen, now we're stacking challenges on top of challenges.
Stuart: But that would be really exciting though.
Dave: That would be quite exciting. We can get there. I'm confident and that's blind confidence at this point. But I think with the amount of talent we have in the industry today; we can start to tackle the challenges of challenges and be wildly successful.
Stuart: And actually, that might open up things like perhaps we should be reformatting some of the existing vaccines made in animals or cells or eggs, because in the future we might understand the synthetic-based better than even the cell and animal-based vaccines.
Dave: We're going to be in a very exciting timeline of absolutely that, as you mentioned, new vaccines coming out that have increased durability, decreased risk to the patients, it's going to be a great 10, 15 years of RNA therapeutics.
Stuart: Yeah, well, it's a great time to be in the industry.
Dave: Absolutely.
Stuart: So, with RNA maturing as an industry, you say we're right at the beginning, who knows what sort of exotic flavours of RNA we might need in the future? We could talk about CRNA or self-amplifying RNA. How might the manufacturing processes need to change from what we do today in order to achieve those?
Dave: Well, they're different when we compare that to a standard linear mRNA, the linear mRNA process is pretty easily accessible, you can go do some quick googling on your own and find that manufacturing process.
These things change, however, so if we're talking about, for example, black boxing, that mRNA manufacturing process and turning it into a product that takes sequence in one end and spits drug product out the other, that's all well and good for linear mRNA.
But now we start to think about different forms of RNA, as you mentioned, circular RNA or self-amplifying RNA. The downstream purification processes look different. Let's pick up on circular RNA, for example.
Circular RNA probably doesn't have a cap in a tail as you would expect a linear RNA to have. So, affinity chromatography is a challenge. The manufacturing has to be tailored to that type of purification schema.
So, to take that black box and try to manufacture CRNA through it, poses a challenge. Your operations for purification are fairly fixed in a black box unless they've been designed to be modular in the sense that you can do a chrome step before a TFF step or vice versa and it really makes no difference to the therapeutic manufacturer or to the equipment manufacturer.
We need to start thinking in terms of reconfigurable equipment that can lend to the exact manufacturing needs of a molecule. Exactly like the two you mentioned, circular RNA and self-amplifying RNA, which by the way is significantly longer than a linear piece of RNA.
So again, different manufacturing challenges and different manufacturing benefits to both.
[Music playing]
Stuart: We heard how keen Dan and Dave are to bring neoantigen therapies to the clinic. Indeed, from a technology-based perspective, mRNA has the potential to deliver a more tumour centric approach to therapy, but we'll need to continue innovating in pharmaceutical manufacturing if we want to bring the new treatments to people all over the globe, not just those in developed countries.
Other forms of treatment, such as gene editing, need to be manufactured differently in order to confer additional features and avoid unwanted side effects.
What are the drawbacks if you don't get it right?
Dan: So, it depends on your payload, if your mission is to edit genes and by accident you are going to the wrong cell, that could be disaster because you're going to edit something that is really needed.
I would say in many cases, if you have a level of specificity that the payloads deliver itself, you have this capability to have another layer of security. So, one layer will be your targeted approach and the other layer will be your payload.
Stuart: Okay. So, you can actually tailor the payloads to also only target the correct cells as well.
Dan: Exactly.
Stuart: And I suppose, maybe links to some things which are being promoted within that cell as opposed to other cells. So, you've got a double lock, right?
Dan: Exactly.
Stuart: That sounds really quite pertinent in the past, kind of three to five years. What benefits does your approach have for mRNA delivery?
Dan: So, first I would mention that our lab was the first to demonstrate systemic delivery of mRNA in a cell specific manner in an animal. And that was not many years ago.
I would say seven, eight years ago, 10 years ago, we were looked as outsiders, people in the field that in many cases even people do not really understand what we are doing and how we really can deliver RNA payloads from SRNAs to large payloads. How can we deliver them in a cell specific manner?
Stuart: And what are you hoping that gets picked up now? So, have you kind of dusted off some of these old research grants and said, “I actually, I think now the time is to look at these again.”
Dan: Yeah, so I think that the next wave will be therapy, using RNA for therapy, which the bar is much higher as we all know. And then in parallel genome editing in different diseases.
Stuart: So, maybe let's take that first one, first of all, so if you're thinking about therapy, why is the bar that much higher? What's different between a vaccine and a therapy?
Dan: Well, a vaccine, you are first dosing maybe once, twice, maybe three times in a period of time. In therapy, you'll need to dose much more. Is it once a week? Once every two weeks, once a month for a long period of time. So, there is safety issues that needs to be considered. There is efficacy that needs to be considered.
And then also, in vaccine you are vaccinating healthy people. They have an intact immune system, predominantly most of them, but in patients, they have other diseases.
In this field, we have two challenges. We have a technological or technical challenge. How to scale up, how to be less immunogenic, how to be more accurate, more specific.
And we have a biological challenge is how the body will react to this therapy. What will be the readouts that will convince us that this actually works. In vaccine is simple, you can measure titer of antibodies.
Stuart: And that gives you a good indication that yes, it's been incorporated, it's been expressed and there's an immunogenic response. But if it could be maybe a placebo or it could be that we've fought off the disease, but actually mechanism of action is not well known, right?
Dan: Exactly.
Stuart: So, is that what you are going to be looking at next? Trying to do a bit more mechanism of action, try to bring these through animal and then human trials?
Dan: Yeah. Since we are a very busy lab, I would say we have different projects and some of them are really doing discovery using those tools that we develop in vivo drug discovery, and we identify new potential targets for cancer and for other diseases. New vaccines, for example, bacterial mRNA vaccines, which I think is super important for antibiotic resistance.
And then there is the whole field of editing, genome editing, based editing, prime editing. There are lots of other new types of endonucleases that can help us in this.
So, I think that for us, there was a big challenge in the field of creating a bacterial mRNA vaccine because until now, all the mRNA vaccines are against viral vectors. And the notion is that extracellular bacteria brings its own proteins.
And they also know how to, I would say con the immune system. For example, there is very aggressive streptococci A that hides from the immune system by creating this hyaluronic acid coating.
Stuart: Okay, so it got a kind of a cloak.
Dan: Exactly. And that's coating protects those bacteria from being recognized and until they are, it's already a cluster of bacteria and it's already mostly too late. So, those very aggressive streptococci A strands that are of course antibiotic resistant cannot be even developed proper vaccines to those.
So, we were asking ourself, can we really find some uniqueness in bacteria? Can we understand the biology of some bacterial strengths and maybe can develop an mRNA approach for vaccination?
And one of the things we ask ourself, can we get a model bacteria that is highly aggressive, highly resistant to antibiotic and work with this?
And one of the things that we came up was basically Yersinia pestis, which is the bacteria that is causing the bubonic plague or the black death in Europe. And you cannot really work on this in an academic environment.
Stuart: Yeah, I'm not sure they want to let that out too easily, right?
Dan: Exactly. But you can collaborate with some places and some research institute that do have those special BL 4 labs.
Stuart: And there's a few in the U.S. a couple in the UK maybe.
Dan: And I think every country do have at least one of these. And we did locally also in Israel and with the IIBR, the Israel Institute of Biological Research, which basically have those labs.
And we both understood the biology of this bacteria … and it took us about three years to create this vaccine from mRNA, because initially it did not produce antibody and it's not good enough because then you cannot do any challenge trials because it won't take the bacteria out. So, you don't have enough antibodies, or you don't have antibodies at all.
And so, we had to work on this both from an mRNA standpoint and for changing the sequence, modifying it, creating areas that are more rich by some of the nucleotides and it took time, but at the end we found it to be outstanding.
So, we had optimised this, created the challenge trial and even from a single administration, we can get protection, which is quite remarkable.
Stuart: So, you've might degenerate the antibodies and then in the trials you get them attacking the-
Dan: Yeah, exactly. We get tons of antibodies, and we are very happy. But it was not a trial and error, but a learning situation where we have to learn from biology. And at the end, I strongly believe that we as scientists need to understand nature better. And bacteria offer a lot, but on the other hand, you have to tweak some of the technology to fit the biology.
And this is what I always say is that we have those two challenges in parallel, we have the technological challenge, which is mostly manageable, and we have the biological challenge that is unfortunately unpredictable.
[Music playing]
Stuart: So, mRNA isn't only improving targeted therapy, it's also opening the door for bacterial vaccines. Could mRNA provide us with new tools to fight antibiotic resistant bacteria? Will these treatments ever leave the lab, and can they be manufactured cost effectively?
The COVID vaccine had to be produced by the billions. How can we scale down to non-pandemic production volumes? I asked Dave for his thoughts.
What's the high-level manufacturing process and where my kind of difficulties or innovation points be needed?
Dave: So, RNA manufacturing is quite simple. You start from an enzymatic reaction where there's no cells present. You're transcribing your DNA into RNA and simply purifying it through downstream operations, like TFF, Tangential Flow Filtration, chromatography, either using affinity or size exclusion and then packaging that into your delivery vehicle, which we're positioning in LNP. That's what both of the COVID vaccines were packaged in.
Aside from that manufacturing the COVID vaccine is about raw materials and about speed.
Stuart: And it's useful to think about that being a cell-free process because that opens up opportunities to sequence your manufacturer in a slightly different way.
Dave: Absolutely. And we're seeing that in droves. Our end users are coming up with a new and clever ways of one, building new RNA molecules that simplify purification, and two, coming up with new purification technologies that, for example, reduce the overall cost of manufacturing to help lower the cost of a dose.
Stuart: Given that we understand the process, we've certainly road tested it at scale. What are the kind of exciting innovations that are going to come out next from RNA manufacturing?
Dave: Yeah, so let me answer that with a small pivot. So, when we manufactured COVID vaccines, we did these at scale. We knew we had to vaccinate the world. So, we had repurposed protein manufacturing equipment, which had been primarily large format, a mAb fermenters, usually about 2000 litres.
Our end users would use six of those. We would call those a six pack to manufacture large amounts of mAb therapeutics, because those are typically dosed quite high.
Pivoting to a COVID vaccine, we're talking about a 50-microgram dose across the board. So, to do a 10-litre in vitro transcription reaction, we're coming out with hundreds of thousands of doses from a single facility per run. This is a remarkably concentrated product.
So, one issue that we noticed with our current manufacturing setup is that is remarkably outsized for the indications we'll see coming down the pipeline.
We still have our large indication populations like yellow fever and malaria to go through but after that, our indications start moving into oncology and personalised medicine, where our large format manufacturing may not fit appropriately.
We respawn our NPI, NPD or New Product Introduction, New Product Development processes to account for faster turn products that this industry now needs because our protein size manufacturing equipment is far outsized for our rarer indications or our low middle income countries that need in-country for country size manufacturing.
Stuart: What would that look like if you were trying to do in-country manufacturing setup?
Dave: We're not talking personalised individualised scales yet, but for in-country for country, we're seeing the most interest in 2-gram to 20-gram manufacturing scales. We're talking tens of thousands of doses at this scale already.
So, our large format pandemic scale responses are 350 grams, so coming down about an order of magnitude brings us into accessible and rarely entertained indications for vaccinations.
Stuart: But if you would take that to the limit and think about individualised therapies, what were we going to have to change beyond that scale?
Dave: Individualized is a very unique challenge in this space. When you see a patient coming through the clinic that needs in-depth treatment and that isn't accounted for by currently commercialised therapies, when you see a patient that really, really needs a personalised therapy, they typically need that fast, they need that remarkably fast. And those products in the industry don't exist today.
How does one build one single dose of vaccine for one single person in a week or two? We don't have that infrastructure built yet and we don't have those products built yet, but we need to build them because the technology is at hand.
We have all the tools on the table to deliver vaccines or cancer treatments to patients that desperately need them. We simply need to build the products to facilitate that transaction.
That's where we're focusing in part today, is helping bring small scale, small volume, rapidly analysed drug product to the patient as fast as possible. That's where we're focusing a lot of our effort now.
Stuart: How long do you think it will be before we could make an individualised therapy?
Dave: We already can make them, it's just remarkably expensive. We, as both technologists and therapeutic developers need to see the long-term viability of that operation. If we can't start to serve a community of patients, then what's the end goal in developing technologies to manufacture individualised therapies?
The business case to build a new, for example, machine needs to be there. We need to see the industry largely adopting individualised therapies for us to build this. Fortunately, we're seeing that and we're doing so, but the cost pressures are vastly different for a global vaccination call versus an individual therapy.
[Music playing]
Stuart: The development and manufacturing of individualised therapies is clearly a costly endeavour, which is a potential threat preventing more widespread adoption.
Thankfully, people like Dave are putting their efforts into reducing these costs so that these treatments are available to those who need the most.
And while there is much we can learn from COVID, there is a massive difference between producing for the global market and producing for the individual. But what did we learn from providing vaccines on the global scale and what can we implement into the post pandemic era?
So, we were talking a little bit about scale up. Why did people think it would be challenging to manufacture mRNA in such quantities?
Dan: From a scale up standpoint, I think that people did not have any experience in large scale manufacturing. And the technologies that were available, were available for small scale still.
And small scale meaning you can do 3000 doses, but not 3 billion doses or 2 billion doses, 1 billion doses, that was the goal for the first round, was to create 1 billion doses. And I think that, that was a huge challenge back then. Creating something with the same industrial process for 1 billion is different than 3000.
Stuart: What innovations did you see that needed to be implemented in order to overcome that challenge?
Dan: Well, it depends. I mean, the mixing technologies like T-line or microfluidics have been very helpful. In fact, most of them have been scale up using T-line, very, very simple approach. Microfluidics is a little bit more sophisticated, maybe even more accurate, but definitely, it's the next wave.
So, we now know that there are companies that are producing everything with microfluidics, but they don't need 2 billion doses.
Stuart: That's the kind of thing that could be deployed quite quickly as well, right?
Dan: Yeah, basically, you need maybe a high school diploma, maybe even less to operate this. So, it's very simple, straight forward and in the future, probably, I'm just giving here a free idea, you can operate this on your cell phone.
Stuart: So, then it's kind of almost on demand.
[Music playing]
Dan: Yeah, exactly.
Stuart: COVID certainly pushed humanity into a corner. We were forced to innovate across development, manufacturing, regulatory and distribution to create a safe and effective vaccine.
Manufacturing mRNA vaccines on a global scale of billions gave us many valuable insights about vaccine creation and distribution and highlighted gaps in our ability to provide cutting edge treatments to those in less developed countries.
The pandemic showed the scientific community the power of mRNA-based treatments, including the development of targeted therapies, which could provide us with new avenues for treating cancer.
However, individualised therapies operate on a wildly different scale to that of the global distribution networks that were needed during COVID. And so, more innovation is needed to navigate these scaling issues.
Thanks so much for joining us, and a huge thanks to both Dave and Dan for sharing their knowledge on this fascinating story.

mRNA: What did Covid teach us about drug development and distribution?
Broadcast by