Naveed Siddiqi, Novo Holdings 🇬🇧🇩🇰 | Sharp Macro View of EU Biotech | E16

We’re in San Sebastián 🇪🇸 with Naveed Siddiqi of one of Europe’s largest biotech investors, Novo Holdings.

We talked about how things really are in European biotech at the moment. We also talked about his upbringing as a second-generation immigrant in the UK and the importance of diversity. I’ve known him since he was a partner at Andera. He is a sharp investor, but also very friendly and open.

💎 ABOUT THE SPEAKER

Naveed has been a part of Novo Holdings since 2019 and splits his time between London and Copenhagen. He has over 25 years of experience in life science venture investment as well as investment banking. Before joining Novo Holdings he was a partner at Andera Partners, a venture capital and growth equity firm with over 2 billion euros to focus on life science investment.

🔗 LINKS MENTIONED


Transcript

[00:00:00] Intro

Naveed Siddiqi: My parents originally came from India, then briefly were in Pakistan, then they kind of came to the UK because they were both doctors. I’ve embraced change and diversity because that’s what I’ve grown up with. What we should be focused on, really, is making sure that diversity is viewed as a strength, not as a weakness.

Philip Hemme: We are lacking crossover fun. Yeah. We are lacking latest technology. This deal basically seems like a pretty good case study. I’m totally 

Naveed Siddiqi: with you. And it’s a very important point you pick up on there. Multiple billion dollar companies and then multiple billion dollar exits. We need more of that in Europe.

And that needs scale capital. I liked your robust discussion. Yeah, and I mentioned the more like heated discussion. You know we try to keep it not too heated. I always try to be respectful. 

Philip Hemme: Welcome

to a new episode. I’m your host, Philippe. And on this show, I’m interviewing the best Europeans in biotech. To help you grow. One of the largest biotech investors in the world is actually from Europe, and that’s Novoholding, the investment arm of the Novo Nordisk Foundation, who owns the pharma company Novo Nordisk.

So I went to talk with Naveed the head of European Venture, and I met him here in San Sebastian. Lovely environment. Actually, I’ve known Naveed for quite a long time. Several years when he was a partner at Andea and he’s a super sharp investor, but also very friendly and just open minded. So we talked about what’s really happening in biotech in Europe at the moment.

And we also talked about his upbringing as a second generation immigrant in the UK and the importance of diversity in biotech, especially in Europe. So, here’s my conversation with David, and if you’re enjoying it, please hit the like, follow, or donation button.

Naveed Siddiqi: Alright, welcome to the show, David. Well, thanks for having me here. I’m very excited in this beautiful setting of San Sebastian, can’t complain. Cannot do much better. It’s actually the first episode we do open air, so Oh, 

Philip Hemme: wow. You’re the first one. Fabulous. Great, so, 

[00:02:09] European biotech

Philip Hemme: I think You’re, you’re one of the best in Europe, I think, to, to really have the macro picture from from your role with, with Novo.

So I want to start there, and you told me, like, despite the current, like, market condition, you think Europe went from strengths to strengths, especially since, like, 2012. Can you expand on this a little, like, 

Naveed Siddiqi: yeah, what, what have you seen? Yeah, no, it’s it’s interesting because, like, I’ve been doing this now for 24 years.

So I’ve really seen how Europe has transformed over that period. I would say that I would characterize that period up to 12 as the, there was still, the jury was out about European life sciences and whether venture funds could make money here and whether biotechs could really kind of create transformational products and whether this model was viable.

Whereas since 12, what I’ve noticed is that we have now a really vibrant ecosystem. The returns which EIF has data on this when they look back. are very comparable to the U. S. And we have really a lot of capital now compared to what we, the early years, where it’s a transformational transformationally different.

So and then we have new entrants coming in not only from you know, our new funds here trying to raise, but also from the U. S. as well. Now I, I don’t want to belittle the fact that we are in a bit of a downturn, so it’s tough fundraising. But And these LPs, I, I think they, they in Europe are sitting on the money, their hands, and they could make more, more of a bet.

And we’ll talk about that. But but overall, it’s a, it’s a, it’s a dramatically different environment to when we’ve had, you know, previous financial crises, where I think it was looking very dicey at Europe. That’s good. Interesting. 

Philip Hemme: I, I see like even in the bear market now, it’s almost better than in 2012.

Yes. 

Naveed Siddiqi: Yes, yes, the mood is better. There’s more activity. We just saw some numbers from McKinsey this morning showing that you know, European investment activity in 23 was quite substantial on the private side. So I think that’s, that’s very good. 

Philip Hemme: On the, I’m curious on the, on the timing that’s current timing, especially from the, as a company’s fundraiser.

I mean, you mentioned like it’s a good time to invest. Yeah. Like good exits. Yeah. And, I mean, obviously you guys at Novo it’s a bit different, I mean, the funds, how it’s structured. But what I, and I hear a lot of VCs, they’ve closed funds, they have funds, but what I hear a lot from operators is like, no, everyone is waiting, and they are basically holding back or waiting for good terms.

So I’m wondering, I mean, I think for you guys it’s probably not the situation, but how do you see it? Like working with, or when you co invest, you know, the people 

Naveed Siddiqi: like, no. So I think you’re right. I think we were in a fortunate position because we have a lot of capital to deploy. We’re putting away, you know, last year we did 530 million.

So, so we’re very, very busy and we’re, we are using this as an opportunity to, you know, really add to the portfolio here because we see a lot of good opportunity. But having said that, I, I would totally understand and I see it myself in our portfolio companies, That is taking longer to get things done.

And I would contribute that to a few things. The most proximate to me is the, the fact that the U S public markets. Have mostly not been functioning. We had a little bit of a opening in december january where there’s a bunch of ipos went to high and x also Yeah, xbi made a big big move and then there was a a bunch of ipos that got done After february it’s been tough.

Yeah, so but it really shows that little window we had It shows it won’t take too much to turn. And I think investors are needing a little bit of that positive momentum to deploy. And I think as soon as it comes back, I think deployment activity will speed up and increase. So, I think it’s a little bit that aspect that’s holding things down, slowing things down a bit.

It’s not because of the pace of innovation or the differentiated valuations or data that’s being returned. I mean, the fundamentals of the industry are extremely strong. And I think my sense is that it’s really the macro picture which is holding things back a bit. Like the microwave in the macro non biotech picture.

Yes, it’s the great picture, which is slowing public market investment. And I think it’s, you know, when we even look at the data today from the McKinsey did, you know, there’s very little follow on finance. There’s almost no full follow on and finance things in the European public markets. And in the U.

S. Where we look at Nasdaq we’ve had strong quarters on the follow on side but it’s tapered down a bit again because interest rate cycle expectations are that, you know, they will be held higher for longer rather than, you know, the rate rate cuts the market was expecting. Yeah, 

Philip Hemme: that’s, 

Naveed Siddiqi: yeah, it’s, it’s good 

Philip Hemme: to hear.

Actually, it’s, it’s, it’s a really macro, thing. Significant macro factors that when this get better, the rest is actually there, it’s not like a more, not a deeper crisis. That’s right. That’s right. Yeah. Actually, I mean, you mentioned the, the 500 million figure which is crazy because I mean, you’re, you’re deploying this amount of capital basically every year.

Which is basically the, yeah, the size of a fund. Let’s say the, or Sofinnova or, or Ian’s last fund, but they deploy that over whatever, seven to 10 years. Yes. Yeah. So that’s, I mean, that’s a, I mean, you deploy globally, but that’s was, and, and actually I saw a picture or a figure for Oppenheimer, that’s Q4 2023.

And I will, I will show it that you were basically the most active, or most you have invested the most. Life science investor globally, even more than like, capital or whatever, like very busy fourth quarter. That’s crazy. Yeah. Yeah. No, we, I think I didn’t even know you were like, I mean, I knew you were big, but I didn’t know you were like bigger than like even us funds.

So it’s like, 

Naveed Siddiqi: yeah, no, I think it’s it’s interesting because we, we have a strategy which is quite broad, so we can do. Private and public and we can also invest in IPOs. And so when the markets are good and one we can, you know, increase our weighting to that part when the markets are, or if we think that something is very under priced, part of the market is under priced, we can increase our weighting there.

Gives us a lot of flexibility. So we can make smart decisions from capital allocation perspective. I think coupled with that, I think we could have a global kind of reach of deal flow. So we do see a lot and we have the resources to deploy. So. Those combination of factors enable us to be super active when we want to be.

We can be cautious when things are, you know, going bad all around, but I think because we don’t have any fund, to deploy, it has to be deployed at a certain point. It’s our own money. But in general, if you see our activity it has ranged between 400 to 600 million annually over the last several years.

And how much was that? In Europe? Yeah, so Europe ends up somewhere between 15 to 20 percent of that. So we end up with kind of a reasonable chunk going to Europe. And I think that sort of mirrors really the level of deal activity we see overall. And 

Philip Hemme: how is it? So, and the US I guess has a bigger chunk, I guess more than 50%.

Yes. Western us. Yes. Yes. And then I saw you were pretty active in Asia as well. Yes. Which I, I didn’t actually know you were that active in Asia. 

Naveed Siddiqi: Yes. So Asia, it’s not our team. There’s a separate team that, that deploys Asia. So these numbers I’m talking about are just US and Europe. Okay. So 80% 

Philip Hemme: us, 20% Europe.

Yeah, exactly. It’s about that much, which is basically roughly the market size. If you compare from a. market cap of the, let’s say, US biotech compared to your biotech, I guess would be 80, 20. It 

Naveed Siddiqi: tracks the overall ratios. I just saw a ratio today that 20 percent of FDA approvals are coming from Europe.

Okay. So it tracks. 

Philip Hemme: Maybe while we are on NOVA as well, what I find amazing is that it’s, it’s basically, I mean, in the NOVA holding is basically with tied to the NOVA NODIS Foundation. Which owns a big chunk of the world, all this and more and controlling right. I was, I think it’s 70 percent of the voting rights.

Yes. So basically a lot of the dividends from the world, all this cost foundation and it’s, it’s reinvested and has been, has been massively profitable over. Yeah. Many years. I mean, now even more, et cetera. And it’s, it’s almost too good to be true. I mean, it’s like amazing. I think, I mean, at least from outside, amazing research, amazing products.

Still keeping a market dominance, obviously a crazy expanding market, and then reinvesting, and even the reinvestment is very, I guess, the returns are there as well. Yes. It’s kind of very positive feedback loop. And I’m always looking like the little, like, I’m trying to look off. Okay, what, why, what’s like, not what’s wrong.

But if it’s too good to be true, like where, where is the chubby ointment? Yeah. And I, 

Naveed Siddiqi: I don’t find that much, like, or like, We, we have basically at, at Holdings level, just grown the overall investment activities. So now it’s venture growth, principal investments, bio industrials, Asia. We, we have expanded the overall activities, not just people’s health, it’s planetary health as well.

In the venture team, which is my team, which is just focuses on biotech. Separate from seeds. Yes. Yes. And it’s a little bit later than seeds. So seeds is in the name. And, and also their geographic fake is a little bit on the kind of making sure that could really grow the Danish ecosystem where adventure is kind of your broader Europe and the U S and later stage.

So I think the, the, the, the, the, the bit that I think, you know, where we have kind of been able to develop, I think, is. a way that we invest, which has been trying to basically make sure that this flexibility that we have being at Nova Holdings to, you know, play both private and public and to make sure that we understand how these companies evolve from an early to later stage and then be able to invest anywhere along that paradigm, I think is a, is a very powerful equation because it allows us to follow a story from its inception and have that ability to Consider it at that time or consider it a later point or a later point.

And, and sometimes that strength gives us a unique insight because we have this corporate memory of understand of understanding the development of something. And also a vantage point of understanding how this is developing against other stories elsewhere. So it has resulted us finding opportunities that have had a reasonably high hit rate.

From an M& A perspective, as well as IPO. So, for the last You mean this explains the performance Yes. novel holdings, basically. Exactly. So, up to 21, we were super active in the taking companies public. So, I think we had 17 NASDAQ IPOs between 2020 and 21. Wow. And and then since then, we’ve had in 21 six M& As.

Yes. And then we had another five last year and then we’ve had two already this year. So, so the M& A aspect has been a very important driver to our performance since then. So we run a portfolio about 80 companies, Europe, US, try to pick the best of the breed, follow them over a long time. And then having this flexibility in our model.

Is also, I think, a stray. Okay. Yeah. 

Philip Hemme: Yeah. And one of the exit I wanna, I want to discuss, yeah. I’m a little more, more in detail just after that. Yeah. One thing, still staying on this like too, too good to be crew. Yeah. The only thing I kind of see, and I kind of wanted to, I mean, challenge is a big work.

Discuss it . The only thing I see is that most of the cash petty comes from the Ronalds. Yeah. And comes from, from diabetes and now obesity. Yeah. Which obviously a very tragic disease and growing, but are mostly environmental caused, at least 80 90%. Yeah. And what I’m wondering is like, basically, is it as a society the best way to prevent or to treat or to manage the disease?

Yeah. To give pretty expensive insulin or GLP 1s versus preventing? Yes. Obviously it would be less revenues and less profits for Novo Nordisk. And I know Novo as a company, and I was surprised about this, that even 1 percent of your annual turnover goes to prevention actually. Yes. Which I think is one of the only partner company doing so.

Yes. But I’m still wondering, like, is it not, I mean, yeah, more like, cannot, is there not a way to do like better slash should Novo Nordisk revenues be actually, should they be a bit lower if we Prevented or fixed the problem? 

Naveed Siddiqi: No, no, no. Of course, there’s a huge societal question. I think prevention, of course, has to be the way forward as much as possible.

And I think the, the challenge is, you know, how does that get funded and how does that happen? And I think the way we could think about it is that basically the profits that Nova Holdings generates through what Nova Nordisk gives, ultimately goes to a not for profit and that prof, that not for profit.

The Nardis Foundation now is one of the largest foundations in the world. And they are really thinking about the prevention agenda and also how to kind of manage diseases and prevention better access, and trying to support that agenda as well as supporting, you know, what’s coming next in terms of, you know, moving the, you know, through quantum or stem cell research.

Right, moving to kind of really finding real cures for diseases, right, where there’s been a degenerative aspect to it and really supporting that. They set up a 200 million grant for a stem cell research center in Denmark. They’ve just done on quantum, just getting better insights into into disease. So I think that part of the equation, I think, is a foundation agenda.

And I think our job is the here and now because we have a lot of disease to treat as well. That has to be dealt with as much as the prevention agenda. So I think we’re trying to sort of see how can we could do it profitably. So the foundation has then the resources to then also advance the prevention agenda.

That’s, 

[00:18:06] Billion dollar exits

Philip Hemme: yeah. Yeah. I like it. Maybe going to the, you mentioned some exits. So yeah. Amulet. I think it was, oh, it’s end of last year or is it already this year? It was this year. This year already? Yeah. Yeah. In March. In March, yeah. In March or April. Yeah. Heh. So then it’s this, yeah, I mean, I wanted, I mean, it’s, it was Lyon, France, France based company.

Yes. Billion above billion dollar deal. Yeah. Total buy a box, 800 billion upfront, which is. Yeah. At least to my knowledge, I think it’s the largest upfront, I think ever in France, 

Naveed Siddiqi: or at least. Yeah. Maybe the second after advanced accelerator. Yeah. Okay. Yeah. Yeah. Where do you farm? Yeah. Okay. Yeah. Yeah. 

Philip Hemme: I mean, it’s, it’s impressive.

Yeah. It is impressive. Yeah. It’s and you guys, I’ve seen actually the Series C with Sophie Noah coming in, but I didn’t know you guys were leading the Series A, which is even more impressive. Can you tell a bit more on how you came to invest in the Series A? 

Naveed Siddiqi: So this is a very, I think this is a great story for Europe, by the way, and it really shows and why do I say that?

Because this is a story where the technology actually was initially came from the US from out of the MGH in Boston. And that’s why they were investors. Okay. Now yes, they, but the company got built by a serial entrepreneur in, in Leo Ance. Yes. And thi Abba, who has you know, sold to other companies previously.

And he brought his team on very quickly. Got it up and running very quickly, four and a half years ago. That’s when we came in alongside him around it with some mouse data saying they may have a differentiated product for hyperthyroidism to a. And be the second to market after another one that’s just ahead of them, which is up for approval now.

So I think, it was a European syndicate that significant series A invest financing. We co led it with the EQT guys and you know, lots of other investors came in, a mixture of a French investors would be there earlier at the seed, but then they came in, came in the A and then kind of really trying to Build a pan-European syndicate to kind of enable that advancement into the clinic.

And then successive rounds then dealt next one by end in France leading it. And then and then later on sofinnova coming in for the, for the series C leading that, last year. And then this year the company going was being sold in phase three, having delivered very exciting phase two data, but in four and a half years.

Superb execution moving something very early to, to that point and really backed by overwhelming majority of European investors. And, and I think that’s, that’s a great story. You know, we, we, we advanced the company from a very early stage, successive rounds of financing. All the way it was, we were funded to approval.

Now we were preparing to launch, you know, raise money, more money to launch it. And then we got the corporate interest. So I think it’s a, it’s a laugh, a nice message to show that we can do it here in Europe. Actually, I wanted to, 

Philip Hemme: to at least the syndicate picked my interest because I think you have managed to gather, I think almost all French VC in a single company, which Chick I’ve never seen.

’cause I mean, yeah, I mean, being French and knowing them, they are always like, they never like, I mean they don’t like each other too much. . They sometimes, but let’s say and andino it is quite rare to see them really on the deal together. Sure. So here, I mean, you had both of them. Yeah. Procure investors as well, think.

Yeah. And then EPI. Yeah. Yeah. DPI, but DPI in every French. Every French biotech. But, and then even like I’ll be mad invest. Yes. And Pontifex. Yes. Well, I mean, it’s pretty great. And obviously you guys plus some other investors. I mean, yeah. I mean, I want to ask like, how, how did you manage to get them all and all friends 

Naveed Siddiqi: on the board?

But I guess it, it worked well, I guess. Well, the asset matters. I think the asset was very interesting. The management team matters. Terry Abriba, if those who met him is you know experienced CEO. Yeah. Maintains great relationships with all the investors and has been really somebody who people can feel confident about executing well and then, you know, the investors who kind of came in I think also very experienced in later stage clinical stage opportunities because it became a clinical stage opportunity.

And they’re you know, the, the software growth folks, I think, you know, that’s their, their focus. It was, it was nice to have them there, with the capital available to, to, to participate and really you know, help us shape that last round. So I think those, those are all the factors that kind of come in to it.

And I think we, we’ve We’ve benefited from seeing that sort of early interest from, you know, the local French smaller investors and then the bigger group of was it the bigger funds kind of come in later? Yeah, that’s right. Actually, it’s 

Philip Hemme: the, what would your, your last comment about the crossover?

I mean, not just to, to, to compliment Sofinova, but I think it’s one thing that I heard a lot, let’s say from whatever, 2015 to 2020 is always like, Oh, we are lacking crossover funds, we are lacking later stage funds in Europe. And this deal basically seems like a pretty good case study for a 

Naveed Siddiqi: crossover fund.

I’m, I’m, I’m totally with you. And it’s a very important point you pick up on there. You know, we’ve had a, you know, people who’ve said, Oh, there’s a funding gap at the very early stage. True. But there’s also has been, and there is still a significant gap at the later stage. And when public markets are not active, in the U.

S., companies don’t have the access to raise and dream about being You know, raise those hundreds of millions and, and, and to kind of go public and maybe launch drugs, right? Like, we have many examples of U. S. companies who didn’t get bought, but they launched, and there have been successful launches, and they are now multi billion dollars.

Philip Hemme: After, 

Naveed Siddiqi: after launching, and then not launched, yeah, multiple billion dollar companies, and then, you know, multiple billion dollar exits. We need more of that in Europe, and that needs scale capital. And it’s great to see funds like you know, the, the, the software growth folks there, but also now, you know, we have the, the, the folks at at, at Gilda, we have you know, the, the folks at for beyond and, you know, they have, they’re, we have raised, they’re, they’ve been dedicated funds raised now to do kind of growth capital.

What would be classified as growth capital where there is kind of a POC and then you can sort of fund them to the next level. And Novo is very happy to kind of come alongside those groups and reach sort of help that piece. Now, have we got enough? I don’t think so. Because we don’t, we have a, as we just saw, and I mentioned, you know, there’s, there was nothing raised on for public European companies.

You know, in this last quarter, it’s absolutely terrible for their ability to raise money. And I think that’s a real gap that Europe has to address. And it’s been a, a real failure and we will not be able to succeed if we are totally dependent on all our companies attracting us investors, going to NASDAQ, we should be self sufficient here on the on the public markets as well, if we want to really create the future leaders of tomorrow.

So this is something that has to be, yeah, absolutely. And so we need some of this capital, which is not investing. And locked up in pension funds, insurance companies to kind of you know, somehow get directed or allocated, or maybe the EIB through its policy measures, how it’s helped with the venture funds, help create that sort of later group of specialist investors who can really carry these companies forward to the next level.

And then we can actually then have You know, the, the, the future Gilead’s here in Europe as well. So, yeah, that’s a 

Philip Hemme: good, it’s good. You mentioned the pension funds as well. I mean, when we, when we talked about this discussion, you said like, yeah, it’s mostly a policy problem and I’m curious about this. So what do you mean and yeah.

And how much is it a policy versus a, let’s say a return. I mean, why a pension fund? I mean, if the returns are so good, as you mentioned in funds, where, why are European pension funds not investing more in funds? Yes, I 

Naveed Siddiqi: think the problem is at sort of two levels. One there is a lack of understanding of what biotech is because I think it seems very risky.

All this funny jargon we use, the terminology is very hard for a non specialist to access and understand what he’s investing in. So that’s one serious problem, which is inherent to our sector, but it doesn’t stop the US guys doing it. Right. So US journalists do invest in the sector. Why is it that we don’t?

So I think there’s a risk problem as well. And I think Europe has still developing its equity culture and risk taking culture. Yeah. And I think there is a market failure. I would, I could only categorize as a market failure because now we’ve, this industry has been going for, you know, You know, 25 years plus 30 years, right?

And there’s a clear track record. EIF has the numbers and we had, we’re still having difficulty for LPs or you are, you know, institutional money really kind of allocating these very large pools of capital to, to risk. And so I think that market failure has to be addressed at a policy level. And that is where I think more work needs to be done in Brussels and, you know, in London.

And, and I think that there is some activity that happening there. I think there’s some recognition from the politicians that this has to be addressed. We have the TB initiative in France and we had a mansion house speech in London around it. I think we’re at that juncture where we need to build some momentum here and really see some real actions being taken, which will have a consequence for the next 20 years for European life sizes.

We would truly want to have life size as a core pillar of growth in Europe. We cannot do it without addressing the capital gap at the expansion level. So it’s crazy 

Philip Hemme: because as you said, as you said already throughout the interview, I mean, if, if you have like track records and you have return multiples, I mean, as a pension fund, that should be the most important metric.

Yes. Not like understanding how to do drug development. I don’t know. I mean, 

Naveed Siddiqi: yes, I think there’s some rules also that get in the way also around how they. Match you know, assets of liabilities and and all those things. So I think they are trying to evaluate their own risk level. Exactly. So, and how they allocate what water level risks are willing to underwrite.

So I think this is something that can be addressed. You know, we’ve had two big moves at a policy level, which had a profound impact on the shape of Europe we see today. One is the EBRD, you know, the EBRD setting up with Eastern Europe and integration into Western Europe, right? That was like a huge policy move.

Imagine if that hadn’t been put in place, right? What, what, what that would have what we would be living into now. And then I think the second part, which is more specific to our sector, Was the way that the EIB set set up to in the 2000 timeframe to support venture funds in tech and life sciences and seeded these VCs who were a cornerstone initiative.

They, they seeded almost 

Philip Hemme: every, you know, at least every biotech fund I think has 

Naveed Siddiqi: EIB. They have massive, I think they have a statistic around it. It’s like 90 percent of it or something like that. So it’s, and, and we have the venture industry. The reason we started off this conversation was around. How is positive today?

If they hadn’t done that, I don’t think the private sector, we know that for a fact, they would not have stepped in because they’re still struggling to step in now. So they certainly would have not stepped in then. We would have not, we would not be having this conversation here sitting in Europe. So it is important.

It is something that has to be addressed. And it is important that now policymakers really pay attention to this and fill the gaps and yeah, if some policy makers are listening, do 

Philip Hemme: your research or reach out, maybe one last thing, a bit on the, on the macro level, you touched on it when we talked about bringing all the syndicate and on the board level and that’s the people were like getting along and I think, and we discussed it before, I think, I mean, Biotech is still a people, people game.

Obviously you have scientific data and everything, but still, I would say. 50 plus percent of your success is people true. Can you like, I’ll talk, talk a bit about it of like, why, I mean, why is it the case how you, how you apply it in your investments? Like how you fill it out? Yeah. Oh, like, yeah. You mentioned even within the fund or within the company, there’s something you will work more with and not like, yeah.

Naveed Siddiqi: So I, you know, I was a great case. I think, you know, This was a story where there was an incumbent drug and you had to differentiate the drug and, convince investors that you’re going to have a differentiated story, right? Because you need to raise money. Investors are very focused on, do you have a differentiated drug?

Yeah. And then there are people who are willing to give you some backing and, and, and and there’s, and, and willing to give you lots of rope and others who want to see, you know, results today or tomorrow, right? Yeah. And you have that in all those discussions in a, in a, at a broad church like Amulet had of lots of investors, right?

And you do have people have different perspectives around that. Ten more people on the board. Yeah, yeah. It’s a big board. It’s a big board, yeah. We do, we, I mean, I was like, are we going to be able to navigate this? So I think but we did. And I think the rationale, the reason I think it worked was not because people held back and didn’t express their views.

I think what was you know, what observation for me from, from, from the AMLET case was the role that the CEO played and also the chair played, and also a little bit of sophisticated investors, making sure that they’re listening as much as they’re talking in, in the room. Two ears and one mouth.

Yeah. Yeah. Yeah. It’s, I think you need, you need both. You have two faculties. You should use both. And I think we, we saw a good example of that being used. And, and yes, we had to make some important strategic discussions decisions. It was a robust discussions at times where we were not fully seeing eye to eye.

And I think it needed but what was important was I think everybody was trying to aim to the same goal, right? Which was, we want to make sure that we can raise good money. We do the right experiment. And we could show the right differentiation and we have the best team possible that we, we recruit the best team possible to do that.

And we cannot do this on a, on a shoestring. You need to have a reasonable amount of resources to do this. And you need to build a story that will appeal to people. International audience, right? It’ll also appeal to U. S. investors, et cetera, as well as European investors. So I think we were looking for all of those things.

We had an opportunity maybe to think about going public earlier. There were sort of situations when the markets were very strong. I think there was a big push to try to take this public as part of that wave. I think it was quite a lot of discussion around that because some of us were a little bit weary about a French company that didn’t have, trying to go public on NASDAQ and what that outcome might have been.

I think it would have been tough if we had done that for various reasons that didn’t go as, as, as, as, as some people wanted it to, I guess the right decision was right and the outcome was right. So, so, but we had a robust state discussion at that time around it. And yes You have in management your robust discussion.

Yeah. And imagine like heated discussion, , you know try to keep it not too heated, but it can be. But I always try to be respectful. I very, very important that these are not never there. There’s no really ever right or wrong, wrong answer. These are judgment calls, right? And sometimes you get it right, sometimes you get it wrong.

And we just hope that we can build enough coalition to whatever we think is the majority view and kind of just get, get, get it there. Yeah, 

Philip Hemme: that’s cool. Maybe just before moving to your personal, personal background. Yeah. I just want to mention one thing, but which I think we’ve discussed and which I think illustrates also well the, where the European, ecosystem is now, and we have literally three, three biotechs now that are worth 20 billion 

Naveed Siddiqi: plus.

Yeah. 

Philip Hemme: In our GenX, GenMap, BioNTech. Yeah. And on our call you even said BioNTech was at like 80 billion. Yeah. Yeah. Yeah. I mean, and I think some people, first, some people don’t even realize or know, I mean, even myself. Yeah. I talk with Sander from Forbjorn and I thought, Oh, Argenix is, I thought it was still at 10 billion.

Yeah. Actually they doubled. Yes. So I think it’s also important to mention, like just looking at the market cap, I think it’s super, super, super impressive. Yeah. But even Bahidja from ImmunoCore. Yeah. I mean, I think they are pretty impressive. I would say number five in Europe, but I mean, three or four billion.

Naveed Siddiqi: We should celebrate our successes. We should, we should talk about them more. We should make sure people are aware because, you know, maybe as an industry, what we’re doing a bad job is actually talking about our story. And, you know, we’re Europeans. One of the things that we always criticize ourselves and we’re not good storytellers, right?

Actually, we have a fantastic story to talk about. And it’s, it’s something that we should sharpen our act with a great podcast to highlight it. Yeah. Thank you for doing this. 

[00:37:17] Diversity

Philip Hemme: Yeah. Little self promotion. I agree. Yeah. That’s great. Maybe. So moving to, to personal background and I have 10 minutes left.

Sure. The one thing that, that is. That also strike me, or as your personal background, you are second generation immigrant, your parents are from India slash Pakistan, am I correct? Yeah, that’s right. And you grew up, you grew up in the UK. Yeah. And you told me you, I mean, you’re, I’m curious, I mean, we discussed it, but I want to hear it again for the audience.

Like, how, how was it to, to grow up as a second generation immigrant? Yeah. How was it, especially in the lifestyle? Yes. Industries as ecosystem throughout your career. Yeah, maybe, you know, starting 

Naveed Siddiqi: with that. No, thank you. So look yeah, my parents originally came from India. Then we briefly were in Pakistan.

Then they kind of came to the UK because they were both doctors. And they so I had that early science and medical background. They were professionals and they came to really work in the national health system in the UK. I was born in, in the UK and the UK is my home and I’m European at heart.

So that’s kind of my, my sort of top level journey. I must say that, you know, the, the journey for me personally, has been where I’ve embraced change and diversity because I’ve, that’s what I’ve grown up with. My parents came from a very different culture. From a very different setting and still going into their world and their parents world as a young child, seeing how different that was to, to, to being in Europe was also very, you know, I, I learned that I had two parts of my brain, which I have to kind of switch gear, which I’m sure happens maybe if you’re French and German and on the border, you have to start thinking like a French person and a German person.

It was a bit like that. So I grew up with that sort of diverse as my sort of DNA in my upbringing. I had that, but yeah, probably less strong, I guess, in Europe. Yeah, I guess. Yes. Yes. A bit more differences, different, difference, religion, difference in in language, difference in culture, lots of differences and being able to, to do that.

But it may be very at ease with that. So I feel that’s a huge privilege I’ve had. The second privilege I have had, which I think is something that anybody who comes from a bit of a, you know, parents were professionals. So. We already have a a privileged position because we are, we are coming from a you know, that sexual middle class society, which has some advantages, has certain values, certain thinking.

So that’s, I think, is common. That’s not, whether it’s in India, whether it’s in Pakistan, whether it’s here, we have, that’s a common value that we have. So there’s not much diversity there. So, in a way, I had these two different countervailing forces. My career has been where I’ve felt very much at ease in and I’ve actually embraced diversity.

So I’ve worked for you know you know, Greeks Japanese French and now the, the, the Danes. And so, and then, you know, but UK is my home base and I’ve also done a lot of investing in the U. S. as well. So and now I work with a firm that’s a very U. S. European. Danish kind of, you know, UK, Dane and, and, and U S kind of culture.

So I have to sort of be able to navigate all of that. And we are, there are differences that we should respect our culture difference and what we’re good at culturally and what we, maybe our, our weakness spots are. And, and it’s, it, I think it is powerful to have that sort of vantage point. Now, the challenges and difficulties, I think, I, I would say that, In Europe we have, I’ve grown up in an age where, people have welcomed ambition, talent, skill, And where you come from and what the color of your skin is, has been something that’s been going in the rear view mirror.

And then something that’s been in the, in the, in, you know, how people define people. And the trend has been very Yes, very, I mean good, very progressive towards Yes. That, exactly. So I felt that has been a, you know, I’ve been fortunate to have that you know, as a tailwind. And I’ve never felt there’s been a challenge, but I can’t say that this is my experience is a definition of what everybody else has faced.

It can be that, and I’m aware that we still have issues where you know, discrimination or There might be equality, but lack of equity that kind of comes through in, in how people are promoted or how people are seen. So I don’t want to belittle other people’s experiences. And I think that, that, that is a factor and we should be striving to change that still.

So we have a more equitable society. But I think what we should be focused on really is the making sure that diversity is viewed as a strength, not as a weakness. And now there’s been some talk, quite a lot of political talk, which. Which tries to go away from that. And I would just urge people who are undecided to think about that and, and think that, you know, is it something that is realistic to think in a, in a world that’s so globalized, so interconnected, whether that’s the right way to think about the future.

Philip Hemme: Yeah, that’s a good one. I think I’m curious on, on, cause at this, From my view, I feel like the UK are a bit ahead Yes. Compared to the rest of Europe. Yeah. In terms of diversity and in terms of valuing ambition and Yes. And whatever strengths you have. Yeah. Do you, or did you feel it like, or do you feel it in your like, 

Naveed Siddiqi: yeah.

Well, I, I work for a you know, a French firm and they welcome me and I, I didn’t experience that so. In, in the UK and they’re definitely not in Denmark either. We have Kasim, who’s our CEO, he has you know, UK, but he also has you know that sort of same second generation background.

And then in the UK, I would say I haven’t really experienced it. You know, within the health service, within the research groups, I think there are lots of great role models and examples for the next generation that’s coming up to see that, you know, there are people can be successful in that environment and, and, and, and succeed.

And I think that’s very very positive. And I think that’s something that’s an achievement, which you can all pat ourselves on the back off for as society as whole. And in the rest of Europe, yes, maybe, you know, in some environments, I’d like to see more female participation. I think the numbers are terrible there.

We just had the statistic of 11 percent only of CEOs are female in life sciences. That’s, that’s not good. You know, we, why is that? I mean, we, we need to change that. So, so that’s not good. I think there are areas where I think our inherent natural biases are coming into play and which whether it’s probably subliminal, subconscious.

But how we’re making decisions, maybe there, there is something coming there. So I think we have to keep an eye on that one. Yeah. Yeah. And I guess as with every trend, I mean, there’s a time factor. 

Philip Hemme: Yes. Yes. This kind of change. Yes. So it’s like, yeah, yeah. I like it. I like, I mean, yeah, I liked your, the global message.

[00:45:04] Thanks for listening

Philip Hemme: I think we can, we can wrap up here. Yeah. Great. I mean, thank you. It was amazing. I mean, amazing, like story, personal story, company story, great successes. Yeah. 

Naveed Siddiqi: Thank you. Thanks for the opportunity. See. And really appreciate In this beautiful background, it was not too hot, it didn’t rain. It was not too hot.

Yeah. So so it was nice. Great. Okay. Thank you. Thank you. All the best. 

Philip Hemme: Thanks for listening to the end. I’m impressed by how Navid is a big believer of European biotech and how much capital he is deploying on how much Novo is deploying, but while still doing it in a, in a smart and effective way. I also really like the diversity part and think that European biotech ecosystem would benefit, and European society as a whole, would benefit from way more diversity.

So, if you have also enjoyed this episode, please hit the like, follow, or review button. Any of these actions would help a lot. And I would also be curious to hear what you think, so if you could just leave a comment, wherever you are, down below, or send me an email at philip at flott. bio. Alright, see you in the next episode.

Claudia Ulbrich, Cardior 🇩🇪 | Being the Founder of a Rare €1B Exit | E15

We’re in Hannover 🇩🇪 with the founder and CEO of Cardior, Claudia Ulbrich We talked about Cardior’s recent €1B exit to Novo Nordisk. We also talk about miRNA and Cardiovascular, and why she earned the funny nickname “Frau Biotech”.

Claudia has been founding biotechs since the ’90s. She founded her first biotech company, LipoNova AG in 1998 and went on to co-found many more, including Cardior Pharmaceuticals. During her time she has launched and brought public a cancer vaccine company, helped see through a multi-million dollar deal of over €20M, and much more.

🔗 LINKS MENTIONED


Intro

Claudia Ulbrich: You keep all options open and you have to go for a dual track strategy when you’re on biotech so that you are not being put into a position where you can only pick and choose one option. And that was also the case. Shando’s kids and my kids were running. The international school, Hanover region. And Schremmer said, Oh, I think I know this mom.

Oh, wow. Intriguing data. They really did amazing academic science and founded the company 2016. 

Philip Hemme: That’s your nickname was whole biotech. Yeah. The biotech woman.

Bienvenue to a new episode. I’m your host Philipp and on this show, I’m interviewing the best Europeans in biotech to help you grow. There are only very few billion dollar exits in German biotechs, including micromet. Ganymede and Morphosis. And the last one is Cardio. So I went to Hannover, pretty much in the middle of Germany, to meet with Claudia Albrecht, who is the CEO and the founder.

Actually, I didn’t know Claudia personally, but I’ve heard many great things about her through my network. One thing that is especially impressive that she has been a biotech founder since the late 90s and that’s especially impressive as a female. We talked about Cardio’s recent 1 billion euro firebox exit to Novo Nordisk.

We also talked about the micro RNA targeting space and cardiovascular and why she earned the funny nickname how biotech mrs biotech in english so here’s my conversation with claudia and please like and follow if you’re enjoying it all right welcome to the show claudia 

Claudia Ulbrich: Yeah. Welcome, Philip. Thank you that you are coming over.

So nice to speaking with you. 

Novo Nordisk deal

Philip Hemme: Yeah, it’s great. I mean, super nice place. I like to, and I want to start with obviously the recent exits and cardio to, to Novo Nordisk and congrats first. It’s a amazing deal. I’ve just made it to start just how do you feel? How do you feel about it? 

Claudia Ulbrich: Yeah, that, that’s really amazing.

So it was a tough ride on the one hand side. But also for, for me and the whole cardio team, it’s, it’s a big success. So we are all very happy about this outcome because as you know although a biotech journey is always yeah, dynamic we feel that this acquisition by NovoNordisk is a really yeah.

momentum and nearly, I would say best case, close to best case scenario for the transformative potential of our drug. And to bring in the company straightforward. 

Philip Hemme: Yeah. I’m cu, I’m curious what, what was. 

Claudia Ulbrich: Yeah. TAF is you always need to have as being a CEO, the vision to keep all options open.

So and also to execute on these strategic options. And there is a lot of R and D development, which we have brought through a classical GXP compliant development path over the last couple of years. We had in addition, several racings. Series A and B. So overall that, that is a tough run.

Philip Hemme: So you mean tough was the whole journey? The whole journey. More than the actual deal itself, yeah? Yeah. On the deal, I mean, when I saw the deal, and I think, I mean, it’s a 1 billion euro or 1. 020, 0. 25. I made me laugh about the 0. 25. But they didn’t was not disclosed how much was up front, how much was in milestones.

Obviously when I saw the deal, I was kind of the first thing I was wondering, okay, what’s, what’s the upfront, but at least I’ve, I’ve heard from some of your investors, they’re all very happy about the deals. I guess the, the, the amounts were very, very good. Even the upfront was pretty very good.

I’m, I’m wondering on, on this, on like, I mean, I heard also was a bit more from the NOVO side that that’s how they wanted to communicate, like, can you like, I don’t know, but disclose it, but can you comment on this, like, how, how did it like, how did it go, like? 

Claudia Ulbrich: I think NOVO NORDISK identified the transformative potential of our compound, our lead compound, CDR 132L.

And they valued also the whole technology with regards to non coding RNAs in the cardiovascular space. And therefore we would say the, the deal volume is a very significant one with that roughly 1 billion. On the other hand we have agreed to not disclose any kind of details of the deal.

So what, what I can say is that we have received a significant upfront payment and several milestone payments that, that are linked to, yeah, several R& D milestones as well as commercial milestones and overall it’s a very attractive deal. Yeah. Okay. 

Philip Hemme: Yeah. Yeah. That’s, sounds good. Thank you. I mean, I think you also, you mentioned more from a company looking at the different options.

I was curious about the deal as well as, I mean, you go into the clinical. I mean, you had phase one B results, I think was January or early 2021. We could talk, we’ll talk after reading details of what these results were, but I mean, you have your phase two basically ongoing. So I was also curious of, okay, why?

Why not wait for, let’s say, the end of the phase two, which is probably a big inflection point as well. Versus going to a deal before that, like, I guess you had the different options on the table or even raising another round. Yeah. Like why, why you made that choice? 

Claudia Ulbrich: I think it’s always that you, you keep all options open and you have to go for a dual track strategy when you’re on biotech.

Right. So that you are not being put into a position where you can only pick and choose one option and that was also the case. So last year we prepared also a next financing round and we We could have continued to also prepare for capital markets. We have always envisaged in cardiovascular to, to partner with a big pharma partner because as you know, phase three trials are larger, they are very expensive and at this point in time I would say Novo Nordisk with the acquisition was the best option to bringing our lead compound to as many patients globally as possible.

Because they have large resources not only in terms of funds, they have clinical and commercial expertise. They are strengthening their own cardiovascular footprint and a pipeline, so, and that was a perfect match. And they yeah, they, they saw the potential also in, in bringing CDR 132L.

Into a broader and late stage development and you asked why, why we did not wait for phase two data. So finally the trial has finished recruiting. So we are now in the observational period. And expect to see top line data, yeah first quarter next year. So it’s, it’s a little bit to go there.

On the other hand side we have a very good and robust data package that show shows safety, tolerability, and already some beneficial effects on cardiovascular disease and heart failure, especially. So, and this was, was identified and, and seen also by Novo Nordisk and we, we know the company since many years.

And yeah, they say they were, they were very closely following us and felt it’s now the time because we had also the plans already for the second phase two which goes beyond the myocardial infarction heart failure population into a more. broader chronic heart failure population, and this is a larger trial which we want to start in the U.

S., and it was the right point in time to step in for Novo Nordisk and to impact on this trial also. In terms of later potential phase three opportunities and bringing this drug to markets. 

Philip Hemme: So that’s also in the, in the press release or when the news was announced that Novo would start another phase two, 

Claudia Ulbrich: that’s.

That’s the phase one. Yeah. That’s the phase two. We already had planned for and had designed for, and now when Novo is in charge for this trial, which fully makes sense. This will be in close cooperation with the cardio team, and they are building already on our design for the trial, and they like the population where we wanted to go for, so we will run that together.

Philip Hemme: Sounds like a, sounds like a very good fit on, on many, many levels. Yeah. So it’s that’s, that’s good. I mean, it’s, it’s amazing to have so many options because, I mean, I talk with quite a few biotechs and Especially right now, for example, the financing option is definitely like less attractive or less stuck, especially on the public market, but even the private market is not that easy.

I mean, a lot of VCs say, okay, we, we still investing, but what I hear from more executives is like, everyone is kind of waiting to see. So it’s, it’s amazing that you had yet all the options. 

Results from heart disease phase1

Philip Hemme: Maybe we can go into specifically on the data and, or like, I mean, I saw the data that you announced from the, from the phase 1b that you presented.

Can you go a bit more in details on what are these data and how big was the need in this specific indication? 

Claudia Ulbrich: Yeah, I think overall heart failure, affects 65 million people worldwide. So it’s a large population that has an entire need. As we all know, we have seen over the last Twenty years, I would say, only symptomatic treatments, and the last 15 years were clearly driven by ACE inhibitors, beta blockers, diuretics.

Then we have seen some new kids on the block, like the SGLT two Entresto nowadays, also GLP one inhibitors. So, I think there, there is some movement, but on the other side this is all symptomatic treatment and it does not address the root cause of the disease. And that, that was a reason why we were driving our development.

In, in first instance to myocardial infarction heart failure patients. And in the phase 1b, and that was a, was a really special tea because. We had been running large PIC trials before, and these PIC trials were also reading out on efficacy. They have shown also that the, the drug is safe.

We have been running larger toxicity trials in two species. And the authorities, they have seen our PIC data in terms of PD data readout. And they said they would, they would expect that at least if we go into an affected target population with chronic heart failure, stable chronic heart failure, that was the population we have investigated in phase 1b.

That could bring also patients more beneficial effects rather than affecting them. And that was the reason why we could skip a healthy vulnerability study. That saved the company time and money. That was really, really good. That’s why 

Philip Hemme: you went into 

Claudia Ulbrich: the one. Yeah. And that was the reason why we went into phase 1B.

So we were not only allowed, we were also recommended to do that. We’re different. And had a very stable population. We had four different dose, doses in this trial. Okay. Signal and ascending dose, five on treatment, two on placebo. And what we could show was that we did not have seen any kind of liver or kidney toxicity, platelets were all fine.

Everything that you know from oligos that might affect patients, could not have been identified. And moreover, it, it, it showed beneficial effects in terms of yeah ECG parameters We, what, what, what we have that is specialty. We have the microRNA 132. This is not only found in a tissue, it’s also found in the plasma.

And you have a clear correlation between plasma levels of 132 and tissue levels. So that we could clearly monitor also how our drug affects people in different dosages. And we, we brought these microondate levels in the blood to, to normal levels in our PD active doses of this phase 1 P trial.

And that was, was the beauty. So you had a clear biomarker as well? It’s a clear biomarker and that built the rationale then to go into a phase 2, which really is the proof of the booty. Yeah. You, you need that. So the efficacy trial, and there we, we chose in our view, the best indication where we could show in a timely manner, proof of concept.

Thus, it’s, it’s, it’s, it’s a niche indication for sure, but we, we need to, to view that none of the current drugs, which, which are on the market or are being investigated in clinical trials. have shown any kind of difference in post myocardial infarction heart failure patients. So also the latest trials with, with the Gleiflocins they didn’t bring really significance.

And that, that is a niche where we felt we can make a difference. So the, this product, yeah, might act as a game changer. And now for sure for pharma, it’s the larger market, it’s the chronic heart failure market. And that was the reason Why we said after having now finished or finished on the recruitment for phase two and post myocardial infarction heart failure, we will now go into the chronic heart failure market, but also in a very specific subpopulation of heart failure.

Patients that have left ventricular hypertrophy this is one of the inclusion criteria. And that is independent of ejected fraction. So, and it, it affects also the half path population, for example, combined with left ventricular hypertrophy. This is really, really an, a dire need. So and, and that was really intriguing, also novel to buy in our development path.

Philip Hemme: I like that. I mean, I like that it’s really clear. Smaller, smaller indication, but very targeted, which you could prove and then scale, scale or expand. And I can imagine even for no, it’s like, there’s still a phase two readouts maybe next year that where they know, okay, how is efficacy and then they can also decide on, on how to scale that.

And right. So even for them, that’s, and it makes me realize also, I mean, cardiovascular is always think about whatever one disease, but it’s, I guess. whatever, hundreds or thousands of different, like, like in oncology, I can imagine it’s like, I don’t know if it’s as specific as like sub rare diseases in oncology, maybe a bit less, like, but it’s, I guess, similar with a lot of sub population and subgroups.

Claudia Ulbrich: Yeah. Chronic heart failure is a segmented population and there are for, for several populations also additional symptomatic treatments. So it’s not intended to replace everything also with regard to the very unique mode of action, it fits to many of the established drugs at the moment. And if there are obese patients they will get additional special drugs for obesity, others for diabetes, for example and the like.

So but, but we feel we, we have a drug in hand. That is able to also improve systolic as well as diastolic function. And that is something you want to see with a, with a new transformative treatment or in a, in a disease modifying modality. So we, we are able, or we have shown increased hypertrophy we have decreased hypertrophy being able to show.

We have increased contractility. We have a reduced fibrosis and also enhanced microcapillar density in these hearts. 

Philip Hemme: So, so at the end of the day, I mean you, from my understanding also you are You are preventing new episodes on your, from new episodes happening your heart failure is happening, but you’re also, say improving or reverse the progression or like, I don’t know if that’s the right term to correct me, but 

Claudia Ulbrich: yeah, I, I think if, if you look into these non coding RNAs, as we all know they are not coding for proteins.

But they are so called master regulators of the cellular function, and they can act down on several different path, streams. So and yeah, what, what they, they, they act like my, my colleague Thomas, he’s our scientist, he’s our brave founder yeah, and, and we founded the company together.

So he always explains that like micro on 832. Is like a conductor in an orchestra. Yeah. And if, if he gets out of control then you won’t get the right music in your cell. Yeah. And if you, if you bring your conductor back on stage, yeah, then it acts down on the several messenger RNAs in the cell and can normalize that detrimental pathways and reduce the.

pathological enhanced mitra on A132, levels in the cell and, and that is, that is a beauty where you have different angles where the drug can act and thus it’s more not only preventing, but also halting and reversing a disease. Yeah. That makes, that makes that so unique. Yeah. Yeah. 

MicroRNA

Philip Hemme: Yeah. I mean, I looked into, yeah, that was perfect transition to the next question on like about microRNA, I mean, from what I heard, like, they one specific microRNA can, like, impact, whatever, hundreds of different SNJ RNA, so kind of a massive cascade.

And if that’s also a thing where, I mean, a lot of drugs, where, where they are to target the microRNAs, because they’re just such a big impact and big, big impact. Whatever pathway could be impacted, leading usually everything is to, to pretty high toxicity, but so how, and I also, at least from what I’ve seen that you guys were basically one of the first three micro RNA targeting, like, at least in cardio where it really seems to work or work better versus other trials where, yeah, just what I said that just to high toxicity, so, like, what’s, like, Yeah, what, what specifically did you figure out basically, to make it work versus also?

Claudia Ulbrich: I think and first instance there was a different approach to identify the right microRNA. So in Thomas Lapps he did over many years. Structured vet lab experiments, robot based screening of hundreds, thousands of micro on ace. And finally found that one which, which he brought from the cell culture dish.

Yeah, in, in, into patients now over the course of a very contingent pharma GXP compliant process. But the difference was. We, we are sometimes asked whether there are other groups that have also been doing research in, in 132. Yes that has been done. But finally the, the whole IP that is around that, that was filed by the in 2012 with the first patent family by Thomas to, to the medical school Hanover and Cardio has licensed that exclusively.

And we have built a large protection around this asset, but what, what makes it unique, I think is also the chemistry. We have been working with different chemistries until we found that specific A locked nuclear acid chemistry. And that has a specific spiking pattern that stands for low toxicity.

So before we brought it into animals, we have been ensuring that the chemistry is a real optimal one. Yeah. And there were. And there were. Effective knockdown experiments the target is 100 percent conserved really from fish to, to worm, to, to mice, to pigs, to human that is, is a guarantee for the translation of the different results you get.

This is not the case in, in every micro A, but with 132 that that was successful. Okay. 

Philip Hemme: And also this, you, you’re downregulating it, right? 

Claudia Ulbrich: We are downregulating it. Okay. Yeah. So we are blocking this micro A Okay. So you have once the cardiac muscle gets into stress, being at a myocardial infarction.

Micro on 132 levels are increased. Yeah. And what we are doing is we, we target the micro on a by having antisense oligonucleotide. Yeah. That is a 16 R oligonucleotide based on a locked nucleic acid. 16 B 16 base. Yeah. With a Phosphol Hy Backbone. Yeah. And it’s very stable. Yeah, chemistry. So and, and this goes directly into cardiomyocytes.

Binds to the 132 micro RNA blocks it for four weeks or the like it does not destroy the target. It, it’s not like, like an si RNA, which destroys the target. So there is a difference. Yeah. And this binding which is repeated in, in our current trial for three times. In the more chronic setting, it will be repeated for six times.

So every month, the, the patient will receive an injection, and that, that should be sufficient which, which has been studied and showcased in these large pig trials. So we had two different animal models, one with the subacute setting and one with a chronic setting. Okay. Yeah. 

Philip Hemme: And that, I’m curious, just, do you know exactly, like, I mean, you say blocking, but, you know, I guess you can also quantify or like measure the amount of, of, of, of mRNA left.

Yeah. You’re blocking, or like, We 

Claudia Ulbrich: are 

Philip Hemme: blocking, We are 

Claudia Ulbrich: reducing the target with more than 50%, 55%. This is then also the PD active scenario if you, if you use a dose that is able to reduce your target. More than 55 percent than you are effective. And in, in our studies, we were also able to show a knockdown of 80 to 90%.

Okay. Yeah. 

Philip Hemme: But I guess you still won’t. Yeah. 

Claudia Ulbrich: But if you want to dive into more detail. Yeah. You need to ask. So see us all. 

Philip Hemme: Okay. No, I mean, I’m, I’m curious also on, I mean, as, as from the beginning of the discussion on the, on the microRNA. I mean. As it impacts so much. I guess there also some other function even in a 132 that has some functioning that it might be good to not knock down a hundred percent.

Yeah, just That’s true. Some, yeah. That was my last thought, but yeah, I, I like, I mean, I like that because, yeah. Yeah, so if I summarize really the Yeah. The target, but also the, the chemistry and how you attack the, let’s say attack you, you, yeah. Target the target, the target. Because I, I remember in microRNA, Abivax was on the show, and, but they target with some small molecules, also target microRNAs, but with small molecules, seems to work also pretty well.

But I mean, I’m running a phase three at least, we’ll, we’ll see how, how this goes. 

The history of Cardior

Philip Hemme: Maybe I’m curious on the On the, I mean, you touched a bit on the history of the company. Maybe can you share a bit more of like, I don’t know how you met Thomas? Yeah, I mean, I guess he was not far off already in Hanover.

How do you met him? How did you get started? Yeah, the whole like kind of founding story. 

Claudia Ulbrich: Yeah, you won’t believe it. So it is, it was really in 2016, my first advisory mandate with a biotech client in Hanover. So, so I’m doing now a business in the life science industry more than 25 years. So I have been working as an advisor.

Also for top tier consultants like PricewaterhouseCoopers and others I founded a known consultancy in 2008 helping biotech clients to spin out their companies, to raise funds. To bringing in IP and to develop the organization sometimes by participating in these companies or doing interim management, and the like.

So, so yeah, I have been an entrepreneur since yeah, I would say roughly yeah, yeah. Yeah, 1998, that was, that was the year when I founded my first biotech company that was a cancer vaccine company. And if you look into today’s business, also with Moderna, BioNTech and others now cancer vaccines are the hotspot.

Yeah, all old group 

Philip Hemme: back back, 

Claudia Ulbrich: but, but 20 years ago or 25 years ago there was pioneering a field Yeah. With all the pros and cons. And I think it’s a little bit the same also with these yeah. Non-coding RNAs. And it, it, it has really a tremendous potential. But back to Thomas. So I had never a client in Hanover, although I live in Hanover since 2000.

And 

Philip Hemme: you studied in Hanover, right? 

Claudia Ulbrich: No, I studied, I studied at the university in Lübeck in the North, Northern Germany. And I had a second study in Rheingau where I was at the European business school. So did health economics. I did my promotion, my, my thesis in, in Hanover. And yeah, I founded my first company in Hanover.

That was the reason why I moved to Hanover. Yeah, and yeah, I, I met Thomas after I had spun out a company together with other founders in, in Heidelberg. There was also a cardiovascular company they, they were doing gene therapy also for cardiovascular diseases, heart failure. It was very, very intriguing story and I was advisor to them.

Finally, we sold the company after, yeah, after its inception half a year later to Unicure. And Unicure as the gene therapy pope so to say, yeah also pioneers in the field. I, I was working then for, for 

Philip Hemme: Unicure. We, we had Sander on the, on the show. Yeah. I don’t know if you worked at, he was still there at the same period as you were.

Claudia Ulbrich: Yeah, 

Philip Hemme: Sander 

Claudia Ulbrich: van de Venter. And yeah, I, I, I worked together with Jørn and with, with Hans. And we sold one year later the whole gene therapy and additional nine targets in gene therapy and cardiovascular to, to BMS. Yeah, okay. That was 2015. And that was the time Thomas yeah was, was yeah, alerted and he found my name in the press.

And we have another co founder Shandor Batkay, so and, and they were doing research together at medical school. And Shandor’s kids and my kids were running, haha, the international school Hanover Region. And Shandor said, Oh, I think I know this mom, and I can, can ask her. And then they, they shooted an email.

Whether I would be able and willing to look at that data. So, and that was the story back in 2015 and I looked at the data saying, Oh, let’s see. I don’t think that your target is such a way better than what I sold a couple of weeks before, but curious to, to take a look. And then, then I felt, Oh, wow, intriguing data.

They really did amazing academic science. They, they had good data and yeah I felt we, we should develop a business plan together and build that upon a really thorough development plan. So scientists yeah, that they, they are not really familiar with R& D in the pharma industry, but we did that together also with one or two other people from my consultancy, yeah, and, and, and started through and founded the company 2016 and raised then funds in a series A 2017 this round was led by in these days LSP.

Nowadays equity, and we raised 15 million to bring the compounds through the preclinical stages, through the large animal models, toxicology studies, and the completion of the phase 1B. That was all the series A round, and we started with five people two academics three including myself from the business side I started as advisor, and then part time CEO.

And since the B round in 2021 I, I was responsible for the whole business as being a full time CEO for Cardiopulmonary. I had an active consultancy business, so I had other clients, but that was how we met. And Thomas said, Oh, I, I’m intrigued always by science, but I need somebody who is familiar with the, with the business and how to build a company.

And I think this, this is a very interdisciplinary approach and also some, some factor of success. So as, as we all know, you need good science. You need good patents, good, good people. You need a strong execution and all, always a little luck. 

Philip Hemme: Yeah. I liked it. 

Claudia Ulbrich: Yeah. 

Philip Hemme: I mean, the more, even the guests on the show, there’s always a luck component, which is, I mean, it’s just there.

It’s very hard to measure, but I find it very also, and there’s, there’s some fun in the luck element and it’s also humbling that there’s, there’s something that you cannot control like even in any entrepreneurial journey, I think. It’s, it’s amazing, amazing story. I mean, so basically almost, what, almost 10 years, basically.

15 to almost, almost, you know. 

Claudia Ulbrich: Yeah, the first point of contact is right things. Yeah, about 10, 10 years ago. 

Philip Hemme: It’s an amazing story. Yeah, it’s a I love this story. And about, about Hanover, I mean, yeah, I mean, actually, It’s not the biotech 

Claudia Ulbrich: hub itself. 

Philip Hemme: Yeah. Like, actually, somehow, I was like, I don’t know really why.

I think one of your, the VP business, I think he’s based in Munich, right? Peter. Our VP BDNL. And because I kind of saw the deals through him and I was like, ah, then I was like, I don’t know why I assumed you were Munich based. Yeah. And so I’m like, no, no. I think he is one of the only biotech in Hannover.

Just funny. And so, yeah, how, how, like, how was it to, to build here? I mean, like, like, yeah, how, how was it? I mean, you always hear, okay, it’s important to be in the ecosystem and whatever, like, But in Europe they are smaller than in Boston or whatever in the U. S., but it’s really important to be in the ecosystem, have things around if you are kind of the only one.

But how, like, maybe, how was it, like, how? 

Claudia Ulbrich: Yeah, I think it, it, it was straightforward because the roots. Not only of the disease, but also the roots of the company are clearly in Lower Saxony. So it was a spin out from medical school Hanover. And Thomas is leading an institute there with 50 people working in translational strategic therapies.

He has done several years of, of this RNA research. So and in, in first instance, we, we got some support by a local incubator. We closed a framework agreement with MHH having rent and lease agreements in the first two to three years. And yeah, we, we built the company from really five people to, to more than 20 and with And as you can see, we have attracted international venture capital and we had right from the start also pharma participation with the Bristol Myers, and also the second round of, of race.

Was done during the COVID time. Yeah. Not many companies got venture funds in, in that amount. So it was 64 million euros. So overall the company raised 80 in these days. And yeah, it’s, it’s, it’s not a biotech hub like Munich or like the Rhine area or, or Mainz with BioNTech, for example.

But overall, you can do it everywhere. So sometimes it, it, it was a bit challenging to, to grow the company with the right people. Because if you build more capabilities in terms of business finance business development, clinical operations, quality assurance, all what you need. To bring your compounds into phase two but overall we have been growing now to 32 people and we have people that are coming over every second week in our project weeks from Frankfurt, from Mainz, from Munich, from Freiburg yeah, Berlin, so, so.

This, this is not the reason. So if, if, if you have a good company, if you have a good culture and people like to working towards a common goal then they are also willing to commute. Yeah. And in, in our world after COVID, it’s also a hybrid world. And we, we have to find also new models how to work.

Philip Hemme: Yeah. And I guess also, I mean, it helps also in Germany that it’s, I think at least, I mean, I’m half German, half French living in Berlin, but Berlin, Hanover is like one half hour and Hanover is pretty central in Germany. So I guess you can get there from pretty much anywhere in Germany and whatever. What?

Three, four hours. Deutsche Bahn is not on strike and 

Claudia Ulbrich: it’s not on strike. 

Philip Hemme: That’s the harder part. I guess for flying, it’s a bit, maybe a bit harder from the, from airport wise, but 

Claudia Ulbrich: yeah, because you have to go in transit, 

Philip Hemme: I mean, yeah. And I think also maybe also from the business case, I just said, I mean, you grew the team, but it’s, you didn’t need like to grow a team to hundreds of people where I guess that it becomes a bit more challenging.

Yeah. If it’s a relatively small team, I like that. Actually, it made me think about what you say makes me think about our first guest was Antoine Papiarnik from Sophie Nova. And we talked a bit about location and he said, like, I don’t care where the company is, something like, if it’s in Dijon or in Saragosse or something, it, it needs to be set up as a global biotech company and on the, whatever.

Yeah, 

Claudia Ulbrich: but on the other hand, if you would then like to go for the US for, for preparing capital markets or the like. You, you can always open a subsidiary. This is, this is, this is not rocket science. Yeah. 

Philip Hemme: Yeah. I like that. But I think, I mean also from seeing how you, how, how the set up of the office, it feels like very like top level biotech.

Yeah. If you are in Hanover or not, you can. It takes a bit more effort maybe or something, but I think it’s one of the key. I mean, and you can compensate and whatever. But I think at the end, I mean, you, you proved it, that it, it works. I liked it. And I like also on the VC. I mean, I saw, and you mentioned LSP or equity.

I mean, basically I think top three VCs in Europe. I mean, I was fighting with Sofino and having worse, but let’s be polite as top three, and then. I saw that you even have, at least in the Series B, was, was Biomed with Markus. Yeah. And we talked a bit about, about you as well. 

Claudia Ulbrich: Markus was, was also in Series A.

Philip Hemme: Also in the Series A? 

Claudia Ulbrich: Yes. Right from the start. 

Philip Hemme: Okay. And then Sunstone joined. 

Claudia Ulbrich: Sunstone joined in B. Inkef and FunPlus joined in B. Koperion joined in B. Yeah. It 

Philip Hemme: was a large one. And it’s amazing that you had really a, like, full European, like, 

Claudia Ulbrich: Yeah, there were still Haitian, yeah, there were five new. 

Philip Hemme: But you had, like, really from all over, all over Europe, I mean, and I guess then you would do your board meeting here.

Claudia Ulbrich: Yes, that’s true. Yeah, that, that, that, that is true. That’s the boardroom. And yeah, we, we like it and you, you can run it in a hybrid setting and we have very good technical equipment and also flights, although Hanover is, is, is not the, the hub, if, for example, you go from Copenhagen to Hanover, you have an SAS flight.

Oh, yeah. Okay. Yeah, it’s, it’s, it’s not three times per day, but you have one direct flight per day. So it’s an hour. And that also makes our collaboration where we started with the transaction in the future very smooth. Yeah. It’s not far away. Yeah. Yeah. Yeah. Yeah. Yeah. Same time zone, support in any case yeah the conversations and integrations.

Philip Hemme: Yeah, yeah, nice. 

Working with Centerview

Philip Hemme: Maybe if we, if we zoom out a bit on like, I mean, we had some zoom out already, but zoom out. One on the BD side, I’m curious you. I saw that Centerview was one of your advisors on the deal. And I think, I mean, I’m amazed at least in the past few years, especially let’s say in, in 2023, I think they’ve advised like for whatever, 80 plus percent of the 1 billion plus deals globally in biopharma, which is which is crazy.

And I’m just curious on how was it to work with them? Like, why, yeah, why did you pick them? Like, like if you can, if you can share. 

Claudia Ulbrich: Yeah, I think, it’s a bit difficult to answer because, once it comes to such kind of evaluation of a transaction which we have been faced at the end of last year, it’s, it’s shareholder matter.

And as you know, we have 10 investors in the company. And they have also their networks and their relationships, and there, there came a recommendation, because typically we had always envisaged such kind of farmer partnership, but it was not that we were running a classical process, which we would, for example, have been setting up.

After phase two data, for example in the whole 

Philip Hemme: like bidding process, this 

Claudia Ulbrich: is a different story. But sometimes if you have as many investors and you have also management sellers, for example, it’s somehow valuable to have a neutral, body in, in the game that on the one hand side has the experience and the network does the valuations, for example, And yeah advises on the deal.

So, from a, from a BD perspective I would say there was no big need because we have the expertise in house. And 

Philip Hemme: as you said, you were in contact with Novo. 

Claudia Ulbrich: Yeah, we were we were already in contact with Novo. So that, that, that, that. that was established but finally the devil is always in the details.

Yeah. And yeah, I think it, it was helpful and valuable to have a neutral body into the game. 

Philip Hemme: Just what makes me curious also is that they are, I mean, from their position, they basically see every deal on both sides. So like, I mean, I guess you can rapidly have some conflict of interest slash flight.

Like, some, yeah, let’s say conflicts, but I guess, yeah, that’s like, for what you said, Sans. 

Claudia Ulbrich: Yeah, this is something you would need to ask them, have you? I cannot go on. I 

Philip Hemme: will ask, I will ask them. But it’s good, I mean, also that, that they, yeah, they have a reputation, they’ve done all the deals, so even as you as a, whatever, Hanover based biotech and you work with them, it’s also, I think, probably a good sign also to, for, for all the potential acquirers.

I mean, makes, makes total sense. 

Frau Biotech

Philip Hemme: Maybe, yeah, on the one thing we, we talked before, before starting the two that I heard. That’s, and you’ve been in this stream, you’re, you’re I think one of the few like top level or like C level women in this space, especially early on, I heard that your nickname was, was how, how biotech, the biotech woman, sound pretty funny.

How, like first, where, where did this nickname came from or who, like, if you can share the story about it. And more like, what was your experience like, from like, whatever, 20, 30 years ago? 

Claudia Ulbrich: Yeah, I think it was quite unusual that a biotech company is not only founded, but also run by a female leader, that, that was only in single cases.

So most of this world 25 years ago Was a man’s world and is still a man’s world, that we will be more, there will be more of us. And I think that, that has improved also in the biotech area, not only in, in terms of Being a leader in a biotech company, but also in venture, there are more female in doing venture business, being partner in a venture company and the like.

So I think that that will still take a while until this is, this is equal being recognized. But yeah, I really didn’t care about it, to be honest. So, I was confident in, in my skills and I wanted to do that. I have an entrepreneurial mindset and spirit. I’ve done it now for several times, yeah spinning out companies or founding or co founding companies and Yeah, I think it’s, it’s, it’s, it’s specific to have female in, in a leadership position but it’s more from a diversity perspective.

It’s not that you need to be, or you need to have a female or male CEO, so you should always go for. What is best for the company and what is best for the team? Then you should see that there is a balance, but what, what I would give at hand to every founder or co founder who wants to build a company or wants to start off.

Be curious, yeah, courageous, yeah, build in your confidence, and also face that you will make mistakes, yeah, so, and, and, in any case, you, you learn by failure, and that makes you more resilient, because on the other hand, you, you, you need for sure, tenacity, and the like, Yeah, and, and, yeah, I, I, I never had the feeling to not dare to do that.

So why not? So and my education was, was pretty clear, straightforward. My parents always. Yeah. Trusted on, on my skills and capabilities. You can do what you want to do. But if you do it, do it right. Yeah. And consequent . So, and yeah, there, there also needs to be some fun, but I wouldn’t say there is something specific.

Yeah. You, you, you need to grow together with your team, with your leadership team and hopefully have interdisciplinary skills not only in terms of finance or BD or science or whatever, but, but I liked always that, that overall architecture on managing, orchestrating, So that strategic thinking.

Yeah. And that is what, what I’m doing now for more than 25 years. And I like it. 

Philip Hemme: I like, I like what you, yeah. I mean, you have to like what’s, what you’re doing. I mean, I think for, for anyone, but also. Running a company in my had a mean a bit of experience, but it’s, it’s so difficult. It’s so tough that if you don’t enjoy it, one of the parts at least, it’s, it’s just very hard also to, to, to, to go through.

So, and I think any kind of job, it’s much better if you Mm-Hmm. where you enjoy it and have some fun in, in it. But I, I, I really like what you said at the beginning as well as, okay. I just have, I just. ignored it or whatever that it was on the only male world and I was just going for what I wanted to do and and what attracted me and just yeah kind of ignored and at the end it worked also like how like it worked I’m I think that I, I heard that from, so we had Agneta, for instance, from Lycote also against the vaccines and, and we talked quite a lot about, about that.

And she said something basically similar, I was in there, I just, I mean, was founding the science in the lab and then I had this technology like, and I really wanted to build a company, so I just went for it. And she mentioned some like. Challenges for sure, but also accepting some of the challenges and just whatever going through and 

Claudia Ulbrich: yeah But, but I didn’t answer your, your question on, on misbiotic.

Philip Hemme: Yeah. 

Claudia Ulbrich: When I founded my first company, Liponova, that was that cancer vaccine company, that was in 1998. So over the course of the early, century, 2000 millennium yeah, I, I, I was really, I would say an exception. Yeah. So yeah. And yeah, some people they, they always recognize me sitting on a panel or presenting the company or, yeah, and, and, and sometimes they forgot my last name.

And then they said, Oh, this is Ms. Lipunova or Ms. Biotech, so I always had that name. And I think in first instance, They, they said it in my back, so I, I didn’t hear it. But yeah once it came to a very funny yeah contact at, at one of the conferences somebody said, And I was like, are you kidding me?

But it 

Philip Hemme: sounds like you didn’t take it too personally either. Like, 

Claudia Ulbrich: yeah, 

Philip Hemme: no but Yeah. All fine. It’s funny in hindsight. Yeah. And actually, you’re the first, you’re the first woman on the show from Germany. And even in Germany, I mean, this is biotech founder, Sam. I mean, she’s definitely also in the, in the top level, but otherwise, really in like CEO position or even managing director, it’s, Yeah, it’s, it’s not that easy.

I mean, even for me, even to find GIST, it’s, it’s not that, it’s not that easy, I have to say. I don’t know, but it’s great. It’s great, really, and it’s great we managed to talk. Maybe if, or if some last thoughts or last lessons you want to share or something, just as a, as a wrapping up, like, you know. 

Claudia Ulbrich: Yeah, I think that was up to now an amazing story and journey with cardio and I think it will continue so, and we will try to bring this program to success now together with Novo.

Yeah, and yeah, I’m, I’m really keen to, to join the story a little longer and to see that the transition is being done very professionally and the next trial is on the fly. So that finally our drug hopefully reaches as many as possible patients globally. Yeah. Yeah. Great. 

Philip Hemme: And great, great story.

Thanks for listening

Philip Hemme: Yeah. Great conversation. Thanks a lot. 

Claudia Ulbrich: Thank you for your time and this great interview. Thank you very much. 

Philip Hemme: Thanks for listening to the end. I’m impressed by what Claudia has built with cardio and how she executed with so many like options, clarity, and how far biotech was open. If you’ve also enjoyed this episode, please hit the like, follow, review button.

Any of these actions would help us a lot. And also I would be curious to hear what you think. So if you could leave a comment. whatever platform you are or message me at philip at float dot bio. All right, see you in the next episode.

Bahija Jallal, Immunocore 🇬🇧 | Commercial, Diversity, and AI | E14

We’re super privileged to chat online with Bahija Jallal, the CEO of Immunocore. Immunocore is the largest biotech in the UK and the 5th largest in Europe with a market cap of just over 3 billion dollars. We talk about how Immunocore has helped thousands of cancer patients and has become a commercial biotech. We also talk about why she thinks gender and race diversity are a business case and how AI is changing protein and biologics engineering.

I’ve “known” Bahija for over 10 years, after reading her profile in a student pharma magazine when I was studying bioengineering in France. At that time, she was head of MedImmune and SVP R&D at AstraZeneca. Now as the CEO of Immunocore, with their pioneering T cell receptor platform, she has helped to bring incredible treatments for cancer patients to market.

🔗 LINKS MENTIONED

Bahija on the Breakthrough Labs podcast

Immatics builds on its PRAME Success (and competes with Immunocore)


Transcript

Bahija Jallal: We had one patient. She was the first patient, I think, to take Chemtrac. Now, seven years later, she is a CEO. She’s done so many things. She has so many birthdays. That’s really why I come to work every day. The company had a lot of rich history. Not always an easy one because it was the ups and downs.

Philip Hemme: Innovation is the sheen biotech stuff. It’s never a walk in the park. 

Bahija Jallal: I’m so excited to be in this era. Still, you know, be part of these, because we’re going to see a huge transformation with AI in the drug development process. 

[00:00:34] Welcome

Philip Hemme: I’m your host Philippe, and welcome to a new episode of the Flood Bioshow, where I interview the best Europeans in biotech to help you grow.

Immunocore is the largest biotech in the UK and the fifth largest in Europe with a market cap of just over 3 billion. And since early 2019, Bahija is the CEO, and we finally managed to connect I’ve actually known Daidja since over 10 years. I mean, known as I first read a profile of her in a pharma blog for students.

And when I was studying bioengineering in France, and at that time she was head of Medimune and the SVP R& D of Medimune. AstraZeneca and she just has a remarkable trichococcus. So in our conversation, we talked about how immunocore changed the life of thousands of cancer patients and how it became a commercial biotech.

We also talked about why biEJF things Gender and race diversity is mostly a business case and how AI is changing protein and biologics engineering. So, here’s my conversation with Bai Yichang and please like, follow if you enjoyed. Welcome to the show Bahija Jallal 

Bahija Jallal: good morning. Thank you for having me.

[00:01:46] Immunocore’s life-changing cancer drug

Philip Hemme: Yeah, it’s great, it’s great to have you. I, I want to start with with Immunocore. I, I find the company is really fascinating in, in many many ways, and I’ve followed it actually since, at least since 2015, since the massive Series A and one way that I find it fascinating is that, I mean, you really have changed the life of thousands of, of patients with a rare, basically, eye eye cancer thanks to an innovative drug, I mean, named Chemtrak, which is a T cell receptor, TCR, by specific fusion protein.

And what’s also fascinating that you managed to do that while owning basically the commercial rights, no farmer partner, at least sharing with a farmer partner. And you just reported revenues of around 240 million for the whole year, 2023. And I think that’s the second year the drug is approved if I’m if I’m correct.

So I’m wondering, like, I mean, that’s an amazing achievement. Can you share with how you, how you actually feel about it? 

Bahija Jallal: Okay. Yeah. No, thank you. I feel great about it. And the most important thing is really a drug that almost died to bring it actually to patients. What was extraordinary about it is that these patients actually you know, once they’re diagnosed with metastatic, UV melanoma, they literally have 12 months to live.

That’s what they tell us more and more, that when they are diagnosed, the doctor basically says you have to get your, your you know, affairs in order. And to bring that hope for people. And we had one patient that came and talked to us. She was the first patient, I think, to take Chemtrac. And now seven years later.

She is a CEO. She’s done so many things. She has so many birthdays and, and you know, and that’s really why I come to work every day. So it is, it is amazing what it’s doing for patients. I 

Philip Hemme: saw actually you reported I think this three year data as well and there was a Compared to standard of care. What was it?

Was it 25 per around there? 25%? 

Bahija Jallal: Yeah, it’s around 20, I think 27%. That’s what we saw. So we, we reported in New England Journal of Medicine, the three year survival, which is really amazing for a, a disease like this. And it’s still ongoing, but you can see the tail, what we call the tail, basically.

You know, at least 26 or 27 percent of patients still alive at 20 at three years, which was completely unheard of. I think what’s also extraordinary is the fact that in 40 years, that’s the first innovation in this space. And that is, really satisfying. 

Philip Hemme: Yeah. That’s like, it’s amazing. Actually, it, I was I listened to a podcast you gave on General Atlantic, I think it was in 2022.

And you mentioned actually also on, on the clinical trial that basically at the prime or the other intermittent readouts, the results were so positive that it was decided to basically stop the trial and go directly to the FDA for the approval, which actually, I’ve actually never heard any biotech companies have that actually.

Bahija Jallal: Yeah, you know, the, the end point was overall survival and usually that takes a while to mature. So, you know, to just put it in perspective, we were still in complete COVID time, if you will. We finished the trial randomizing patients in June and the first interim analysis was in October. And we basically internally said, you know, the probability that that’s going to hit in October is very, very low.

And we were extremely surprised that not only it hit, but the hazard ratio was 0. 51, which was really amazing. And we did two things that I’m really proud of. One is, of course, we went to the FDA to, to have you know, to agree on the BLA and, and, and, and so on. But the other thing is, because this is such a deadly disease, Even as a small company you know, we opened the, the, what we call the EAP, so an, Patient Assistance Program, because we could not wait until it’s approved by EMA and everything to make it available for patients.

So we started helping patients right away, which I’m very at no cost at that time. Which I’m very, very proud of. 

Philip Hemme: Did you, can you, just from a context as other biotech companies who had that recently of like stopping a trial, like having a trial going to markets after interim data analysis? 

Bahija Jallal: I, you know, I don’t, I, I don’t hear too many of that and sometimes the interim analysis is not really public you know, you don’t, you don’t tell the market when you’re having the interim analysis.

So usually people don’t talk too much about it, but it’s, it’s not a, it’s not a usual thing. So. It was a powerful drug on the overall survival, which I think is a, is a, is a really amazing. 

[00:07:40] Commercializing Immunocore

Philip Hemme: Yeah, that’s, that’s great. And now I think one of the main, your main priorities for this year is the commercial rollout of Chemtrail.

I’ve seen that from my understanding you are managing most of the commercialization yourself. As I mentioned, you don’t have a pharma partner. But you do have a partner for the commercialization of medicine, medicine, pharma for some geography, Central Eastern Europe, Israel, Canada. But then you manage basically the biggest market, I guess, is the U.

S. That you manage yourself. And then, so how do you, what’s your plan for the, like, for the coming, let’s say months for the commercial? 

Bahija Jallal: Yeah. So I think, you know, so we chose to commercialize ourself for two reasons. You know, one is. You know, we believe we, we can. We have a great team and it’s not, it’s a, it’s an orphan drug.

So usually I, I think from the capabilities point of view, we, it was the first time and we thought we, we can do it. The second thing is this is a new platform and I am a firm believer that we continue to learn. We continue to learn in the clinic and we continue to learn in the, in the market. We to bring that learning back into the early you know, as we bring more more products.

So the goal this year, like you said, we had a real, a nice growth, so, you know, quarter on quarter. So the, ChemTrack is the, the standards of care in first line, which is amazing in the U. S. and in Europe as well. And so going into, into the year in the US, we still have some growth in the US to happen.

So we are now going more, you know, you start in the academic academic, setting first and then you get into the community and you know, the US is very big, so the community is very spread. So we need to continue, in the community and then in Europe is, as you know, it’s country by country.

So we had a really great success in Germany and France Italy, and I, the, we just announced the approval or the reimbursement discussions with Spain. So we will be launching in Spain this year. So we will get more countries in, in Europe and we’ll continue that way. 

Philip Hemme: I’m curious on this topic. I mean, yeah, market access in Europe is definitely tricky for, for innovative drugs on the, on the podcast with Mark, the Gary Dell from, from Abivax and formerly from Epson.

And he mentioned that most of his market was in the U. S. just from a reimbursement point of view. I’m curious for, for, for you guys, I mean, especially Kim Choik. I don’t know the exact pricing, but I guess it’s, pretty high, I guess, in the six figures, I guess. So how do you, how is the discussion there in, let’s say, in Europe?

Bahija Jallal: Yeah, so I think it’s notorious that U. S. is still paying for innovation, you know, so I would say that you know, we probably skim track for the value it brings because, frankly overall survival is the gold standard. A lot of several drugs that in the, in Europe, they don’t get reimbursed if, if you don’t have the overall survival, for instance.

So we have absolutely the gold standards. We We had two scientific evaluations, one in, in Germany and one in in France. And we got one of the, you know, the high marks for the scientific. Basically everybody sees the value of of Chemtrack, but it is, you know, the U. S. is still paying for innovation.

Europe is country by country. I think we, we got a very good, you know, price in Germany. And, but we are still in negotiation, country by country, that’s the tedious part, I think, about Europe. You 

Philip Hemme: disclosed the price you get with Germany? Is it public? 

Bahija Jallal: Yes, I think it was disclosed. It was 10, 000. Okay.

Philip Hemme: And how does it compare to the U. S. then? I mean, the U. S. is a bit complicated with all the insurance. 

Bahija Jallal: So the, the U. S. is right now is, is around 20, 000. 

Philip Hemme: Okay. So that’s actually,

yeah, no, I’m, I’m, I’m curious because I also read in, especially in, in France, there was a I don’t remember exactly, but there was a, a group of, of professors and oncology professor, I think, especially around the professor in Lyon and Clos d’Arnault, what they were. They were quite, like, upset about the access to innovative oncology drugs for patients and that there were some, or at least in their opinion, some reports where the scientific, evaluation was basically lowered, or let’s say lowered to influence then the reimbursement or to justify not reimbursing some drugs and that would be then the patient could not access it or even, even on a special request in the hospital could not access it.

So I was curious how it compares with, with you when you have really outstanding data. Does it translate then to really positive? But it sounds like it’s the case. 

Bahija Jallal: Yeah. So in France where we are, we, like I said, we had the assistance program, which is the there is a pathway for that in, in France and they pay for, for the EAP, EAP right away.

It’s called AP1 or something like that. It’s a new system. But we are still negotiating the price now with the, with France, so we’re not, we’re not done. This negotiations just take a little bit longer. Yeah. 

Philip Hemme: As, as things can do in France.

[00:13:46] Explaining the TCR technology

Philip Hemme: All right. Let’s maybe a switch a bit to the topic, to, to technology, I think. I mean, it’s a TCR, basically TCR receptor, as a, as a that you fuse of a fusion protein to some T cell activating, activating, at least for a vacuum chuck. I’m curious, can you maybe explain a bit more, and especially how it will, how it differentiates from, let’s say, a bispecifics, typically?

Especially, one thing that puzzled me was on the targeting of antigens on the cell. I mean, how, why specifically couldn’t target GP100 of the same antigen that, yeah. Curious. 

Bahija Jallal: Yeah, no, it’s a, it’s a very good question. So you know, the, the important thing in the, in the invention, if you will, is taking Really, what I would call always mimicking what happens in our body.

So we do have the T cell receptor and usually it’s really to differentiate between self and non self, you know, so. And we took that property the T cell receptor is membrane bound. So we made it soluble. Soluble, stable. Usually it’s also low affinity, so we increase the affinity a million times and with keeping the specificity.

And why this is important? Because the, well, the antigen is basically a peptides of nine to, or eight, nine, ten amino acids that’s presented by the HLA you know, so complex. So you have, that complex is pretty short to amino acids. And so you have to have. The specificity, and I’ll come to that because that’s where the differentiation comes from.

On the other side is the CD3 that basically will bring any T cell that’s you know, that has a CD3 receptor, any T cell will, will come. So we have in one hand, we are binding to the tumor cell. That’s basically our, our goal is to. Identify the, the tumor cell or, you know, make it visible. And on the other side is bringing the T-cells to kill the tumor cell.

So the differentiation, and to your point, why on GP 100 or OnPrem and things like that you can’t so, so far it’s not possible to, to, or there are, there are no monoclonal antibodies. Or antibodies specific to, to target. One of the attractiveness here is the fact that GP100 prime and all that are intracellular proteins.

And you know monoclonal antibodies have access to membrane bound or extracellular, you know, proteins. So the, these proteins, GP100 and, or PRAME or any other intracellular protein will be degraded and one, the peptides will be presented at the surface. So that’s how we have access to, to intracellular proteins.

Philip Hemme: So by specific, cannot, cannot like recognize it when it’s degraded and presented on the surface. 

Bahija Jallal: It’s so this is. It’s not specific enough. It’s not, so I’m not going to say never right? So at least to say TCR mimics, so we, we call them the TCR mimics, if they’re going to happen or not. But the difficulty, if you, if you will, the difference between monoclons, monoclons, basically they like, or they see antigens that are, non linear, right?

There’s a confirmation, and that’s how you have the binding of the monoclonal antibody. Here, we’re talking about very linear. You know, eight to nine amino acids linked to the HLA. And what we know is the T cell receptor needs to bind to both at the same time to recognize so the HLA and, and it’s very, very linear, very, you know it’s not a conformational change or anything like the monoclonal antibodies.

So, at least so far, the T cell receptor is made to, to recognize HLA complex. Antibodies less so, but it, I’m not going to say it’s impossible. 

Philip Hemme: Okay. This makes sense. And now I also understand, because I saw the, basically the slide or the figure of that you can target, let’s say 90 percent or the 90 percent of the antigens that.

by specific or T cells cannot target on the, the, because you can access basically all these kind of small antigens in different conformation. That’s okay. That makes sense. I have two questions or two, two things that comes to mind is one is what about basically alternative scaffolds or to have proteins or nanobodies.

I mean, we had, we had the CEO of XC of Abilene was just a previous guest. So I guess with the nanobodies, maybe you can target different conformations. And you can also do, basically, by specifics, I mean, 

Bahija Jallal: That’s why I said it’s not impossible. I think the best thing right now is the T cell receptor, right?

Because we understand it really well and everything. I think there is another feature that I, I, for differentiation that I didn’t talk about is the fact that we can target down to five to ten copies per cell. Monoclonal antibodies as we know them right now, you need a thousand, you need a much more higher expression, right?

And that’s really crucial. Like I said, it’s not impossible to have a TCR mimic. I, I really don’t believe that’s you know, I just, I just think the technology is not ripe enough, but it’s going to be possible. I, I just think it’s, it’s always possible, but it’s not as straightforward and you need a much higher expression of the target.

Philip Hemme: Cause that’s, that’s the second, I saw that for, I mean, also as a transition to PRAME that’s one of the biggest, let’s say, competitors or someone by the company exploring is, is Ematix also, also European company, German based, and they are, they are using by specifics, no? Correct. Okay. But they are, they are earlier than you, I believe, in the trial or still in, in phase one exploration?

Okay, so we’ll see in the results how, how well it works, I guess, and how specific they are as well. Exactly. 

[00:20:40] Targeting tumors with PRAME

Philip Hemme: And maybe maybe as also transition to PREEM, can you, like, talk a bit more about it? I mean, it’s basically the rest of your pipeline. And from my understanding, it’s a much I mean, it’s basically targeting solid tumors as a much larger patient group as well.

Yeah. Can you maybe talk 

Bahija Jallal: Yeah, no. So GP100 is very much a melanoma protein. It’s expressed only in melanoma. And we have now what we talked about, the whole data in uveal melanoma, but we have a trial ongoing in cutaneous melanoma. So there, it’s very much a melanoma specific protein. PRAME is really exciting targets for multiple reasons.

You know, one, it’s more broadly expressed. And in larger tumor types it’s also, not expressing normal tissues, which makes it very attractive, but only in tumor tissues. It’s also what we call a negative prognostic marker. So that means, you know, I, the way I look at it is tumors don’t want to get rid of it, basically.

And whenever it’s expressed, it’s more homogeneously expressed, which, you know, it’s a, it’s an advantage, basically. So yeah, so for all these attributes, I think PRAME is, is very attractive. We also showed, last year at ESMO, we showed that it’s it’s active it’s active in melanoma, uveal, and cutaneous, and also we showed activity in ovarian.

And we’re testing it in other tumors as well.

Philip Hemme: And you’re also in colorectal cancers, correct? 

Bahija Jallal: No, colorectal is on P Well. That’s another target. And it’s the third target coming to the clinic. 

Philip Hemme: Can you, yeah, I, I mean, I have a, I mean, a bit personal, but my, basically my mom, my mom had colorectal cancer, so it’s a bit also touching. And all the discussion we have is, is, I mean, Yeah, it’s amazing that, you know, I can relate to it and that you’re working on other patients and with, like, heavy, heavy cancer and you can have a transformative impact.

But yeah, so, but I hope to, I mean, I hope the rest of the trial goes well as well. I saw that you’re working on, on infectious disease as well, but I think, I mean, and looking also for functional cure, which, and HIV, which I think is also amazing. 

[00:23:26] Immunocore now and then

Philip Hemme: But maybe we can, we can switch a bit more to, maybe more to the team from looking at, I mean, obviously you’re coming with a MedImmune background, I was, what was it?

Almost, was it 10, almost 10 years, I guess I’ve had, 13 years, well, I’ve had a head of basically head of MedImmune and just from AstraZeneca. What I’ve seen is that quite a lot of, people in your team also joined you, or ex medium. Can you, can you share a bit about this? Like, was it, was it planned or was it,

Bahija Jallal: No, it’s never planned, of course.

Yeah, I think the, the, you know, to just close on, on what we talked about and the infectious disease and everything that, what attracted me to the company is The technology is novel. It’s a real pioneer in this area and also more transformative. You know, our goal is not to be just incremental, but to do something, you know, it’s, it’s high risk, high reward as I call it.

That’s why we’re going into infectious disease into. Not just controlling the viral load, but looking at, you know, can we actually get the functional cure? So every time you’ll see us in something, it’s really how can we, you know, can we be transformational, not just incremental? And so I would, I would absolutely believe that that’s part of what attracted several people from you know, from MedImmune or from AstraZeneca.

And you know, and it, it does help if you work with people before so they know, they know what to expect, right? 

Philip Hemme: I guess it’s, I mean, it’s, I guess they love working with you as well, and. 

Bahija Jallal: Yeah, and I love working with them as well. Yeah. 

Philip Hemme: Love is usually with Super, I mean, with Supercroft. That’s nice. I mean, I find it always a a great sign, for when, I mean, as, as a founder or CEO or manager, when, when your team wants to continue to work with you.

Even for me in the team now, Flood Bioteam, actually most of the people that joined me from the, from my previous startup, which I founded, which is also great. And they want to work with, I think it’s a great sense of, Oh, very nice. And maybe one word about also on your, on the chairman of Immunocore, I mean, John, John Bell, but what, what, what was.

What was pretty striking to me was that he is basically, he co founded the company, which dates back to like 1999 in some ways, as I think was Avidex at the moment, and he’s still chairman basically now, which I think is a pretty like remarkable duration of being chairman. I don’t know that that many chairman who last that long in companies who stay that long.

How, like, how, how is it to, to work with them and, and how is it, how, how make, make, I mean, that you co founded and it was chairman all the way along, how, how does it like play out? 

Bahija Jallal: So he’s, he’s very unique I would say because he is not only on the science and everything, but also really great entrepreneur, I would say, and great and understand You know the business really well.

I don’t think he was chairman all the time. I think the the The science started in his lab. He was, you know affiliated with the company but became a chairman I think just two years before I came or something like that. So j james nolan I think was the the chairman before but he is Absolutely a fantastic person and also extremely supportive and I I don’t think We have a great partnership and I absolutely, you know, the success of of Immunocore.

He absolutely should take a lot of, of, credit for it as well. 

Philip Hemme: Of course. Maybe on the, expanding a bit on the, on the history of let’s say on the history of immuno car, what was always also quite interesting is that. EZP was, there was like three different, basically it ended up being coming from three different companies with at least there was Medigene in, in Germany and, and Adaptimune also I think in, I mean, also in Oxford.

Bahija Jallal: Correct. They split at some point. 

Philip Hemme: And split it at some point, but basically it’s coming from the same lab, which is pretty, it’s pretty, pretty amazing. But at the same time, you know, I guess when fast surfing adapt immune is a bit, I mean, it was more in cell therapy and, and in solid tumors. Like, how, how, how is it today, the relationship with the different companies, like?

Bahija Jallal: So we don’t you know, we, after adaptamine and iminocor split, I think there’s, you know, after each one of the companies, one chose to go with cell therapy and we went with the soluble, you know, we don’t, we don’t have no, nothing special as a relationship, like, it’s completely independent. to independent companies right now.

So, as you said, the company had a lot of rich history. Not always, not always an easy one because it was the ups and downs, and I think that’s what happens in a new technology, when you’re pioneering something and it’s completely unprecedented, so there was, there’s a lot of time to be invested in, You know, bringing it to, to the level where it is right now, but in meanwhile, I think there was a lot of, ups and downs and, you know, for the, for the company, but hopefully right now it’s, it’s more stable.

Philip Hemme: mean, you know, innovation, especially in biotech. It’s tough. I mean, never a walk in the park, wash the 

Bahija Jallal: shares, and the patients. Yeah. 

Philip Hemme: And I, I also, because I, I read more on the financial side, I think, I mean, Medi Jean was basically main shareholder even of, of Unco, but at some point, I guess they were bought out.

So today they don’t have any share 

Bahija Jallal: connection or a minimum, I think. 

Philip Hemme: And then, I mean. I remember what, I mean, that’s one when I started really following it, when, what, you know, like kind of stood out on the map was in, in 2015 when they raised this like massive series a, I think was the biggest ever in, in Europe.

And even on us 10, that even today is still really big. If it was like 320 million, but at the same time, I heard you say on the podcast that it was a lot of money, but there was no strategy. Something like this and that it was. mismanaged. Can, can you expand a bit on this? I’m curious. 

Bahija Jallal: Well, I can, I can just say how I found the company, right?

So I think, you know and talking to investors when I took over, I, I think, you know, you raise money. It’s for me, the philosophy of raising money, you really need to have before you go to raise money, what you’re going to do with it. Right. And raising 350 in a series a where the company, you know, again, I, I would say it didn’t really have a plan and went through the 350, you know, more quicker than it should without the data.

And I think that’s the, that’s the issue. So when I came to the company, the, you know, there was no, the majority of the leadership team or the executive team has left. There was. Yeah, the company was just in, in not stable condition, I would say from the governance point of view, from the science, which direction it should go and, and so on.

But I really, the, the positive thing at why I came otherwise, you know, was the depth of, of the, or the innovation, the, the platform. I absolutely believed in the platform. And saw the potential in Kim Trac and, you know, I thought naively, you know, everything else I can fix. But it was hard, hard to fix.

It was not it was not easy. So thank goodness I was naïve, yes. 

Philip Hemme: Sometimes naivety is good, 

Bahija Jallal: it’s a blessing. 

Philip Hemme: Sometimes. Yeah, I, and also it’s yeah. I mean, I remember at that time also from, I mean, from the fundraising, I mean, I think what Ford was involved in, it had raised this like massive funds, almost private equity, also early stage, but massive fund.

And I remember talking with some investors, even some large French VCs, and they were all like super excited about the round and it’s one of the rare, but like basically everyone wanted to join in at some point. They were very excited also, I mean, from, also they were very excited by the platform as in, that it can be manufactured pretty easily, that has amazing data, this amazing potential, etc.

On paper, at least it could look very big, but I guess, yeah, it’s, need to, to deliver afterwards. Because, yeah, no, that’s, that’s what we needed. That’s great. I think, yeah. And maybe to, to wrap up on, on, oh, actually to, to, to, to wrap up on I immuno in the podcast you mentioned your dreamers to, to build, I think the largest or the largest fully integrated biopharma in the world.

I think it was a . 

Bahija Jallal: I think that start, I start with the, with the, with the Europe, and we’ll go from there, but yes. Why not? I think we have. You know, when I look at the, the platform, like the questions you ask, you know, how would you do that and why would you do that? Well, the first thing is we have, we have the science and we have the platform to do that, right?

So we have, we built everything from really strong research where we can bring more science and more targets. We have, the platform is modular so we can be in, cancer. Infectious disease, and then in the last earnings, we actually talked about the third, we’re at J. P. Morgan, the third possibility, which is in in autoimmune diseases.

You know, which is really an exciting area. So we have a big space to play in. We have a fantastic technology that’s, that’s not just, you know, the bi specific is the start, I would say. We can use, we used CD3, we can use any other effector. So when you have a vast you know, space to play in, if you will, then I think it’s, That has your ambition has to to match that as well.

So why not? I think we could why not? 

Philip Hemme: That’s great. I mean, yeah Guess you will try your try your best Absolutely. No, it’s good. 

[00:35:37] Why Bahija chose Immunocore

Philip Hemme: And maybe to go back on I mean that sounds super exciting also for you personally to join I’m curious on that decision said I mean As you, as you mentioned, you could have gone somewhere else.

I can imagine with your profile, you could have gone in many, many, many other places. Can you like, walk back a bit on the, on maybe the different options you had or the, how you came to that choice? Also, I mean, going for a European company versus US based companies or, or staying in Pharma, going maybe to a CEO position in Pharma.

I guess you had like many, many options. Yeah, 

Bahija Jallal: so I, I was, I was lucky that I had several options, including, you know, staying in MedImmune if I wanted to, but I felt like I had accomplished what I needed in MedImmune. We had a great vision there and we had, I had a fantastic time there. I, you know, the, I never, never had.

A development plan, as they call it in, in companies, you know, what do you want to be, where do you want to be after, and so on, because I’m somebody, if I put a goal on myself, I will try to achieve it, and I didn’t want to do that, right, so I wanted to just, sometimes great opportunities come from the side, right, when you don’t plan them.

But one thing that was very important to me, you know, while I don’t plan, you know, you know, in two years I want to be CEO or, or CSO or something like that, one thing I always know is I follow my heart and it has to be whatever I do, the science has to be really interesting. You know, because that’s my first motivation.

If I don’t like the science or if I cannot be passionate about it, I cannot do that. So yeah, while I had a lot of different opportunities, I was lucky that way. This one where people really didn’t understand at the beginning why you would go to this company, it was because of the science. And so I thought the you know, seeing that that platform and what it could bring when a lot of people didn’t believe I think is what, what attracted me.

Was it high risk? He was really high risk at that time, but it paid off. Yeah. 

Philip Hemme: It’s always amazing when you look back at such a decision and it turns out to go well. I remember actually when I, I mean I remember I was following and I got forced to step down as CEO, and then the CFO stepped down, like, okay, and stepped down without any replacement, or like, I will support the board, something, I was like, okay.

But then when you joined, I was also surprised, but I think, yeah, it turned out amazingly. A 

Bahija Jallal: lot of people were surprised, but yeah, I’m happy I did. 

Philip Hemme: Ah, great. And I’m curious about the development plan. I like that I am typically, I would not have many either. But I’m curious, now you still, like, I mean, you don’t have a development plan or, or how, what’s your, let’s say, or where do you see yourself in the next, I don’t know, 5, 5, 10 years?

Bahija Jallal: Well, you know, we have a lot to do at Ubuntu Core. I think we have a great platform. I think, you know, when I think about the next few years is you know, hopefully bringing, you know, still, ChemTrac to more patients. I think, you know, if we do the cutaneous melanoma, we have one, one thing that’s I’m really passionate about is, you know, how can we get earlier and earlier and earlier in cancer and, and help people.

So we have an adjuvant trial with the European organization that’s ongoing for ChemTrac. That, if we can, you know, bring these to patients even earlier, I would be, you know, really thrilled. You know, PRAME, if we can bring PRAME, you know, autoimmune infectious disease. So that’s all I’m thinking about.

And I think there’s, there’s a lot to think about, a lot of unfinished business. 

Philip Hemme: Yeah, it can keep you busy for a while. I guess maybe, 

[00:40:22] Top lessons from Bahija Jallal

Philip Hemme: No, that sounds good. If we can switch a bit to a topic. Curious on you, I mean, this podcast is also quite focused on, I mean, on European leaders in biotech, or at least He does have a pretty strong European connection.

I think you, I mean as you basically did your, your, like your, university studies and PhD in France. You grew up in Morocco, then, then PhD in France, postdoc in Germany. You moved to US for quite a while, but then coming, coming now with a UK company. Can you like, I don’t know how, what were some of your lessons there or how it, yeah, how it helped you, or it’s helping you now?

Bahija Jallal: Yeah, no, I, I am, I was extremely lucky, you know, to have done all my schooling in, in, actually in, in France and my PhD in France. It really, you know, France for me was not only on the academic side, but growing as a person, you know, the cultural side, everything, which was great. Then going to Germany was, I would say it was my first time seeing, it was a test for me, you know, can you take what you did in PhD?

And actually be an independent, somehow more independent researcher. And there I really fell in love with the whole cancer research I think was, was in Germany. And and then, you know, the US very much you know, I had the really this very strong idea that, you know, pharma companies or biotech or whatever, we didn’t know that at the time, they don’t do science.

If you go there, it’s, you know, because it was more chemical companies. And so, U. S. really showed me that you can, you don’t have to choose between the two. You can do science, great science to bring great drugs to to the market. I think I learned a lot about biotech and being entrepreneurial. Definitely that entrepreneurial spirit more in the U.

S. and that Especially in 

Philip Hemme: California, 

Bahija Jallal: I guess, where it’s strong. Yeah, completely at the hub of of all biotech. And, and not being afraid to take risks, you know, people do encourage you to do that, you know. And now full circle back, you know, I see it again, I think in, in the UK, is, you know, bringing this learning, all that I was exposed to, if you will, bringing it into, into the company.

And what I see as the learning, I think, for European companies and, and everything is that Europe, and I see it now in, in the UK have fantastic science, like absolutely fantastic science. I think you see in Germany started, few few decades ago or a few years ago. And then in the UK, you have the startup.

Arena is, is very strong, right? And you have more money to get a startup company. But then I think where it becomes less or more problematic is to mature these companies into a full fledged, you know, industrial, you know, company. And that’s because, you know, the, Yeah, the appetite for risk is really less than in the U.

S. Hopefully it’s changing. Hopefully with the success of BioNTech in Germany and, you know, hopefully with Iberocorp, you know, in, in the UK and things like that, that will encourage people to invest. Because then it’s, it’s like a vicious circle. If you don’t have that, capital to, to grow the company, then you have less you know, talents on the clinical side and so on that’s exist in the U.

S. So, I think that’s the learning and the comparison between the two, that 

Philip Hemme: I bring. Ah, that’s, that’s that’s great. And talking about, I mean, what you mentioned in, in Europe also now you have, I mean, luck, I mean, not luckily, but I mean, luckily, but it was not only luck, but now a few commercial biopharmas or biotechs, I mean, you know, obviously, but you mentioned BioNTech, GenMab, Argenyx, CryDoing which are still, like, Independence for the product on the market and the product on the market that makes good, like what makes an impact.

And in this case of whatever, hundreds of millions of revenues in the billions of revenues, which I think also brings, I guess, what’s your, I mean, other people who have that experience that you just mentioned, and the bring them to, to the rest of the industry. And I think it’s, it’s great to see. And it, it happened actually pretty.

Obviously with the delay compared to the U. S., compared to the Gen Zines and the Gen Techs, but still, yeah, great to see. 

[00:45:48] How is risk appetite connected to innovation?

Philip Hemme: And also maybe just connected to the discussion we had previously, I’m always curious also, I mean, risk appetite for sure, capital appetite. I’m also always curious on how much is it connected that the market in Europe for innovative drugs, as in reimbursement, is so less good than in the U.

S. How much is that, like, how much is that connected, basically? 

Bahija Jallal: Well, it has to be connected, right? So if you look at the U. S., we always argue that that pays for the innovation because, you know, we don’t have, in this business, The cycle for innovation or the cycle for R& D is much longer than the tech, you know, in the tech the R& D is much shorter and here you invested when I think about ImmunoCore, and we talked about the Series A and everything, that’s been 20 years in the making, you know, so how do you 

Philip Hemme: And in the billion, over, probably over a billion dollars 

Bahija Jallal: invested for?

It’s completely 20 years to, to come up with something that’s completely novel. So yeah, it takes, it takes much longer and you have to recoup that at, at some point, right? 

Philip Hemme: Yeah, but I, I, yeah, definitely, I mean, but, but really compare, I mean, because I, I’ve seen, even from inside some European biotechs and, I mean, going to the U.

S. is basically mandatory, but it’s still a big challenge and big friction and you have to hire a different team, different company culture, et cetera, et cetera. And that’s, I think, really not easy for, for European biotech. I mean, it’s always hard to quantify these things, but, 

Bahija Jallal: I agree. It’s not, it’s not easy.

And I would say you cannot do it from Europe. Like it’s the same things, like if you get it commercialized, you have to have a presence in the U. K., in the, in U. S. You know, if you are dealing with more investors here, that was my, my thing with ImmunoCore. We needed to have a presence in the U. S. because, you know, I, as I call it, out of sight, out of mind, you really need to be, have access to talent as well.

So we have, you know, and a lot, most of the clinical is here, most of the commercial, we have a team in, in Europe. So, I think we, We had to have the presence in Europe and in, in the U. S. to accomplish what we needed to accomplish.

Philip Hemme: No, but it’s, yeah, I remember we talked a bit with, with, with Mark Duggeridoo about, about this and about access when I mentioned him before. And then, and he was also a bit on the political comment and saying, okay, I mean, it’s, you should also not be surprised too much as a, let’s say, as a government or the governments or the union.

If you don’t finance innovation, there’s no market for it. That you don’t have companies developing it and trying to develop it or that you have champions in the space It’s you cannot have both Like it’s harder to have both. So yeah 

[00:49:10] Diversity in biotech

Philip Hemme: Maybe on another personal topic as well, on, it’s on diversity. I heard you talk on the, on the podcasts and I found it actually pretty striking that you’re very open about it and very direct, like putting the words, like switch, switch.

I found, I found really like, not a bit surprising, but also it really resonated, like it really resonates. I really liked it. Maybe you said something. Especially for gender diversity, it’s not only fair, but there’s a just, there’s a business case which I find it’s like, I mean, for a business, I like that, but you didn’t really expand on it.

Could you, could you expand on, on the business case and especially, are you measure, measure? 

Bahija Jallal: Sure. Yeah, no, I feel, I feel really strongly about this because I think when we talk about diversity, we usually talk about, it’s not fair, we have to, you know, We have to be fair, we have to do these kind of things and, and my view is for everything we bring a business case, why this is good for business, right?

So instead of bringing it to the business as a guilt, you know, and making people feel guilty because they’re not, we are actually not doing ourselves a service by not explaining why it is important. 

Philip Hemme: Because you bring it as an opportunity. 

Bahija Jallal: And to say why, why, why it is important for, for your business to have the, the diversity of thought in general when I look at where we are, we have, it’s in the business of innovation, right?

And to be innovative, it’s not having 10 Moroccans working with me that are women are all the same that went to France and stuff like that. We all have the same, you know, the same reference, the same cultural reference and everything. But if you bring people from different, if you look for that you know, diversity of thought, you automatically going to have, you know, a diverse, slate or diverse, employees.

My thing is, you know, You know, there were studies actually in nature showing that labs with more diverse you know postdocs and, and and and PhD students produced more, more articles and more research than, you know, measured by the number of articles published than non, you know, diverse lab.

And I don’t think we do enough of those kind of studies. So whenever you see me in talking to academics, it’s like, you know, actually bring data. Because if you bring data, you’re going to influence the change a lot more than if we just, you know, make people feel, You know, it’s about fairness or guilt or something like that.

It’s really about why this is good for business and I absolutely believe it is good for business. It’s not easy. It doesn’t mean it’s easy, but it’s good for business.

Philip Hemme: Yeah, I, I, I, I like that I, where, where I am also, where I would not, not challenge you, but where I would like, where, where I’m also like, what’s a bit hard is, is really making that case and having evidence to back that case, that business case. I mean, you mentioned the study, but how do you translate, I mean, academic output to, let’s say a company like Immunocore.

It’s got a different setting and also from a personal experience, I really believe in this. I mean, I applied pretty much the same to my first startup and having with diverse hiring. And I mean, more from a feeling and seeing that this brings us better products and we could live a better product for, for our users and, and, and that leads us to grow as a company.

But it’s, it’s very hard to like quantify and like put into, you know, like into really precise or more precise, like let’s say bullet points or whatever of, and, and then especially in the hiring because you rapidly become and like being having to favor. certain diversity in the hiring process to, to get this diversity.

And I found that also not that easy to, to have a, like, let’s say, more quantifiable approach there, or having some a bit more specific things. How do you, like, How do you go more granular? 

Bahija Jallal: I think there is more, I think there is more to it. I think you, you point your finger to something that’s important is we don’t do enough of the studies because I think it’s really possible to actually quantify that, quantify everything.

I think for me is in, and we did that in, in my previous you know, life. Is we said, okay, we get it. I want to have a really diverse because it’s so important for, for our business of the innovation that we can be more productive, that we can be actually more bring more, you know, drugs to the market.

And the first thing was not to be counting how many or not, but to focus on the environment. Right? Because if you want to attract different people and everything, they have to be in an environment where they can feel themselves, because that’s really the difference. You know, can you be, you know, in an, in an very diverse, setting.

Most of the important thing is what we call the, the inclusion. Right? So to have a diverse is to have everybody can be themselves. And you cannot tell me when we don’t feel ourself that we are productive, right? When we have to be somebody else and, or try to not be ourself, you know, it’s stressful. It’s not, it’s not great.

But if you are in an environment where you can be yourself, you’re going to produce your best work, right? How to quantify it? I think that’s, that’s my, my always you know. Work for or appeal to, to the academics, you know, we need to focus on that. Instead we’re focusing on numbers, you know, how can we quantify that, that innovation?

I, I’m pretty sure, you know, I quantify it here. I quantified it in my, in my previous life. How did you quantify it at the Renault Kwong? Is how, is how productive we are and how, you know, the, the quality of the people we’re attracting. Which is really important, you know, if I can tell you, you know, some of it’s the diversity here is, is fantastic and we started from the board on to my leadership team.

You know, as you have to lead there by example, but I can tell you, I quantify it by the quality of discussions we have, the challenges we have, each other, you know, the the innovation that I see every, every day, right? But you have, it’s not easy. I’m not saying it’s easy, but you have to, to produce an environment where people can be themselves.

And then. And then it’s a vicious circle, like when people are feeling, you know, it’s a great place to be, then they attract others and, and so on. And that’s, I’m not saying we have all the metrics and how to, to measure it, but I can see the end product. You know, can we be productive in what we do? In what we do, can we be better in what we do?

And I’m still appealing to academics to, to focus on that versus the numbers. 

Philip Hemme: Yeah. But even, I mean, even when you can, you, it’s hard, you cannot, basically A, B test, you cannot really compare. A less diverse immunocore, what would you have delivered? I mean, I’m just challenging and playing the devil’s advocate here, but 

Bahija Jallal: No, no, no, it’s good, but that’s the kind of discussion we need to have, right?

That’s the kind of discussion that we need to have. It’s not See, the discussion would have stopped very quickly if I would just say it’s not fair not having women, you know, 50 percent of the, of this and that and whatever. But that’s the discussion we need to have. 

Philip Hemme: I find it actually personally it’s pretty difficult with a discussion, at least me as a, let’s say, white male, is that, like, I would have no legitimacy in, in, in bringing this topic of discussion, you know.

At least I’m perceived as having no legitimacy and it’s, I find it, but yeah, it’s, 

Bahija Jallal: but see, I don’t see it this way. So this is where I feel like. We are all part of it, right? So part of how, why we believe diversity is good, right? And not it’s, it’s for you white male to solve this problem. No, it’s all of us, right?

So I don’t think I, I, I want, I don’t want the, the haves and have nots, you know, the have that feels guilty and the other ones is not. If we believe and we focus on that diversity of thought. That is important. It’s your, it’s your goal should everyone is responsible as all of us to make it possible because if we believe that that’s the best for society in, in general, frankly, and, and for the work in, in particular.

Philip Hemme: I like that a lot. I mean, and very, I liked that a lot and I can see him on. US also, I mean, more US mindset than US, and what you’re saying there. And it’s something that I experienced. I mean, I finished my studies in Boston and that’s where I really could feel this like meritocracy. We don’t care really where you’re coming from, what’s your color, whatever, what’s your accent is about really more about your mindset and being aligned with everything and what you deliver.

Which, yeah, I found really good and, and I, I remember in the, in the interview you said when you came to France, you discovered how people were judged on the color of their skin and that was pretty shocking. That’s, 

Bahija Jallal: yeah, because I, I come from a country where we’re all very different actually. You know, if you think about Morocco, you have people who came from Andalusia, they were chased from Andalusia.

You have the Nolats in the South, you have the They all look differently and they’re all very different. So that was completely normal. Yeah. And in Paris it was very, it’s, yeah, it was very shocking actually that people were labeled with, the color of the skin, you know, what I learned in Midwell.

Philip Hemme: Yeah. I think it’s yeah. It’s, I guess it’s changing in France, but it could yeah, there’s, there’s a lot of work to do in general in Europe. 

[01:00:59] Biological innovation

Philip Hemme: Maybe to, to finish on the, on the last, on the last topic, a bit more on the, let’s say, looking at the, at the industry and overall. And you, you mentioned when you came to us, you restrict how, how you could do more biological innovation, not just chemical also at pharma.

I remember you were saying in, I think in one interview you said that when Astra bought Medimune, I think there was 5 percent of the pipeline was biological, in 2014 I believe it was 50%, I don’t know what’s the percentage now, I guess. 80 percent plus,

like, but at the same time lately, at least in the last one, two years, but was also what, curious to me was that quite a lot of small molecules brought amazing results. Also a lot of yields. I find like, I mean, if I, if I looked at it, it was a kind of small maker was kind of. going down and replaced by biologics and now there’s a, I mean, still biologics, how do you feel about it on, on all that topic?

Bahija Jallal: Yeah, no, I think, you know, so you, you start with the small molecules, the, the degraders and other things. So the innovation, you know, started there as well. So I, I think, you know, the, I still, I’m still passionate about biologics. I think there is still, you know, we’re just mimicking what’s happening in our body.

And I think there’s still a lot more to discover there. I think, you know, you’ve seen the antibody direct conjugate, for instance, 

Philip Hemme: Say, no, it’s merging. 

Bahija Jallal: Yeah, I think what I, what I like and that’s what we do in ImmunoCore now is really, you know, my message to everyone is we need to start from a biological problem and have the tools to solve that problem.

Right? We should not be, you know, we, our technology is the start, but how can we take all the learnings and stuff to solve a biological problem? And I think what’s fascinating in this era and fascinating in this in where we, where we are right now as an industry is that there is a really a huge toolbox.

of technology and to, to solve biological problem. And I think we are in an amazing era, frankly. So yeah, I’m, I’m still passionate about it. 

Philip Hemme: Does it mean also that you are not closing to getting other technology to move? If it never, yeah, never, 

Bahija Jallal: never. I think, you know, we, But you always, you know, go where, where your expertise is and, and collaborate and things like that.

But we are really trying to tackle some, some difficult problems and, and hopefully have the tools to, to solve them. 

Philip Hemme: That’s, I think, I mean, well, one, what comes to my mind as a, at least as a person, company that applies that pretty well is, is is BioNTech actually which always surprised me that they didn’t start with mRNA at all and, and now they have, I mean, I think they’ve even a prime, a prime by specific if I’m not mistaking, and then they have ADCs.

I think they have pretty much every biological technology in the pipeline possible, which is, especially as early biotech is pretty surprising. Yeah. Yeah. Yeah. Yeah. 

Bahija Jallal: Yeah. Yeah, I’m not, I’m not advocating to be in everywhere, because I think, you know, there is a saying in in, in the U. S. at least, which says, how do they call it, master, you know, Jack of all trades and master of none you know, you have to develop some expertise and depth in, in the areas that you can you can master basically but you have to be open.

And I think to look at how to solve again, biological questions, I think that’s the most important thing. 

Philip Hemme: That’s I think that’s it, sir. It’s where’s the balance and how to prioritize it. That’s the half. That’s the half. I guess, I mean, not to defend BioNTech, but I guess they’re not trying to be everywhere to be everywhere.

But it’s probably opportunity based. And I mean, on mRNA, they were super good in mRNA. 

Bahija Jallal: I’m super tense. Yeah, no, I’m not making any judgment upon them. I think they also have they, they were successful and had a lot of money, you know, so they can afford these kind of things with the COVID. So, and we’re all very grateful that they’ve done that, right?

Philip Hemme: Yes, absolutely. 

[01:05:49] AI in protein engineering optimization

Philip Hemme: And one, I mean, one hot topic, but I guess we, we, we kind of not have to talk at it, but a bit about AI. And I heard you talking about AI already two years ago. And one thing you were focused on was really on the protein engineering Optimization. I’m I’m curious how, how do you translate it to, as, at, at Immunocore if it’s specific examples and what’s the scale of that example thing?

Bahija Jallal: Yeah, no, I, you know it’s a really I’m so excited to be in this era still, you know, be part of that. of these because we’re going to see a huge transformation with AI in the drug development process, right? So you see it today with monoclonal antibodies that a lot of the, a lot of the monoclonal antibodies starts in the computer, basically, because we understand a lot more about monoclonal antibodies.

We are in a very lucky situation where, you know, I told you, we’ve been at it for 20 years. So we know what works and what doesn’t work, and we have tons of data. And you know, when we did the when we developed the, to look for specificity and how it works, We did everything with co crystal structures.

So we have a resolution of maybe, we have resolved maybe 500, something like that, 500 co crystal structures. So you can imagine the, the richness of that data. To actually start looking at predicting, you know, what kind of TCR you need to have before even doing experiments in the, in the, in the lab.

That’s what we’re focused on. And I think that’s going to cut a lot of time. To, to bring you know, that, that’s one part where we can actually have, make a difference there. 

Philip Hemme: And how, how are you doing it then? Are you like training some models yourself? Are you partnering with some? 

Bahija Jallal: So we have one collaboration, so we’re partnering with the, with the one company and then we have internal efforts as well.

Because we’re sitting on that data, if you imagine not only the data that I, I talked about for the crystal structure and everything, but we also now, treated 2000 patients or something like that from clinical trials and everything. So there’s a lot of information there as well. So I think we are just at the beginning, but I’m really, really excited about that.

Philip Hemme: That’s good. Cause I, I think, I mean, the, And the potential is, is huge and can be really transformative. I think, Bernhard Lantau, that was a pre CEO of Eurotech on, on the show, and he mentioned that the gains, if you look at the industries, drug development would cut by 30 to 50 percent the costs. That was the whole process.

And I mean, what, as we discussed, and you think in billions and in years, time and money, it’s huge. Yeah. Absolutely. Yeah. But what I’m, I mean, that’s huge on one hand, but I’m also always like curious is what you just mentioned, having like some concrete, let’s say, examples of now, how can it help, especially biotech companies, like as of today or in the, in the shorter term and how they like integrated or work with it or profit from it.

That’s always, I find that. less easy to necessarily see, or not, not this easy to, to translate, and that’s what also curious, like, was very interesting to see. 

Bahija Jallal: Yeah, there is something really concrete now, like if you look at some companies going after monoclonal antibodies, nobody does, or at least these companies are not going to to immunize llamas, or immunize mice, or immunize whatever.

Look at, you know, or have a big phase display library and things like that, you know in small molecules, it started the same thing, you know, to do the design before. But in monoclonal, it’s really I think is showing the way there that basically you do all that modeling in silico, basically. And that’s AI, that’s basically, you know, based on the information we have about monoclonal antibodies.

Yeah. That’s what we’re trying to do on the TCR, because if you can cut all that empirical way of finding, you know, doing the testing and finding and all this, these are months. When you, when you cut, you know, a year or something of the drug development, that’s a lot of money already to start with. And that’s just the beginning.

I think once you get, the more you do of this modeling, the more you, you know, educate the system, the better it is and, and so on. And then you start, I think there is a company doing that in monoclonal antibodies in a, in a finding a different way of by specific, just all with AI. That’s completely fascinating.

So I think we just at the beginning, you know, 

Philip Hemme: the, the company, 

Bahija Jallal: what’s, what’s the name of the. You know, biologic, I will say, but they’re doing that. Yeah. Yeah, that’s it. 

Philip Hemme: And maybe do you have other names?

Bahija Jallal: I think Abcelera, I think, as well. Abcelera. 

Philip Hemme: Yeah. Do you have other, other, use cases, at Immunocore where you use AI?

Bahija Jallal: Yeah, we just, we just basically the, the, the area where we are focused on first is taking all that learning on how we can, before going experimental, find the right TCR. You know, just through that modeling and knowing. Because it’s not just finding the TCR. If you find the TCR, does it bind correctly?

Does it do and all this stuff? You can, you can do all that now in Silicon. Like I said, this is the beginning, but we are really working hard on that. Ah, sure. 

Philip Hemme: Well, one thing, I’m curious, also one thing mentioned Thomas Clausell on, from Oaken was on the show and he, I asked him actually, okay, what, what should a biotech platform how should a biotech platform use AI today?

Because obviously it’s very, I mean, tricky what we just discussed, too. Should you partner, should you do your own things, where to apply it? One thing he said, he also is on, on really on, on the clinical trials, on certification of patients or finding the right patients where you have massive potential gains as well.

Because do you, like, are you also using AI on, let’s say, on optimizing clinical trials? No, 

Bahija Jallal: not, not to the same extent, you know, but I think there is potential there. Good work. You know, is where to get the, the data. Like I said, we have 2, 000 patients or something, those, so we know a lot more and we are trying to use the data to to inform us.

But you know, I think where hopefully it will make more of a difference is basically, you know, clinical trials take a long time, finding the right patients take a long time. If we can cut from that time. You know, and bring the right patients to, to the trials and, and so on. You know, it’s, it’s not an easy thing and if we’re gonna do it properly, we’re gonna have to really collaborate in that space.

Philip Hemme: You’re not collaborating at the moment, you’re, you’re looking at it, you’re looking at it, I guess, collaborating. Right. Nice. That’s, amazing discussion. I could continue for a while, but I know we, we are, we are, we have a, we have a time limit. Like, yeah. I think, I think it’s a great wrapping up.

I think we can finish here. Thanks a lot for joining. 

Bahija Jallal: It’s my pleasure. Thank you very much. 

Philip Hemme: And please continue amazing work and like keep my fingers crossed that everything works out well and on the day 

Bahija Jallal: it comes out. Thank you so much. I really appreciate it. All right. Thanks. Bye. Bye. 

[01:14:42] Thanks for listening

Philip Hemme: Thanks for listening to the end.

I have to say it was one of my favorite episodes so far. I’m impressed by what Baija has been building at Iminacore and how she is doing it with focus on the science, on care about the patients, following her heart, while at the same time she’s super down to earth. And, and having fun. I hope you enjoyed as well.

If you did, please hit the like, follow, review button. Any of these actions would, would help us a lot. And I would also be curious to hear what you think. So please, leave a comment below or send me an email to philip at flot. io Alright, see you in the next episode.

Edwin Moses | How to build large biotech platforms | E13

We’re in Brussels with Edwin Moses, the former CEO of one of the most successful platform companies in Europe, Ablynx, now part of Sanofi. We talk about how he feels 5 years after the exit from Ablynx and his lessons. We also talk about the parallels between industrial biotech, AI bio and cell therapy, and how a chairman can help CEOs grow.

Edwin Moses is Chairman of Achilles Thx (cell therapy for solid tumors), Avantium (bioplastics) and LabGenius (AI biotech). He is the former CEO of one of the most successful platform companies in Europe, Ablynx, now part of Sanofi. I interviewed him back in 2018 when he sold the company.

🔗 LINKS MENTIONED


Transcript

[00:00:00] Intro

Philip Hemme: And the company’s still subsidiary, is it? 

Edwin Moses: Yeah, it sits there in Ghent. Approximately four or five hundred people they have. I think they put more people in, not less. The platforms, you know, they’re a means to an end. They’re taking you somewhere. 

Philip Hemme: It is the recipe for a platform company to focus on assets.

Why they don’t manage it then? 

Edwin Moses: They’re very loyal. They take a considerable amount of time. They have a view on confidentiality regarding investment. Okay, I’m, I’m cool with that, but the quality of the interaction with the company is very high quality. 

Philip Hemme: How many times have you been proven wrong in the last years where you, you thought the answer was this and it was not this?

Edwin Moses: I can’t remember the exact numbers.

Philip Hemme: I’m your host, Alip, and welcome to a new episode of the Flodd Bioshow, where I interview the best Europeans in biotech. Building a platform company with a new technology can be extremely powerful, but also has many challenges such as validating the tech and prioritizing your own assets through technical trials.

Edwin Moses has built one of the best platform companies in Europe with Ablinks, which is now part of Sanofi. So I went to Brussels to talk with him. I’ve actually interviewed him back in 2018, just when the deal closed. And he is since then chairman of three different companies, one in biosolutions for bioplastic, one AI in biotech, and one cell therapy for solid tumors.

So we talked about how he feels now that it’s been five years after the deal. And what were his lessons. We also talked about the similarities between biosolutions, AI biotech, and platform companies such as cell therapy. And also how a chairman can help a CEO grow. So, here’s my conversation with Edwin.

[00:01:52] After the Ablynx acquisition

Philip Hemme: Welcome to the show, Edwin. So I want to start with with what happened basically after AdLinks acquisition by Sanofi, which was, I checked, I think, 3. 9 billion euros at that time, euro was like 4. 8 billion, almost like euro, euro billion dollars, euro was a bit stronger. It was January 2018 and I think it’s still the second largest acquisition ever in European biotech.

And I looked, I think the lead product in Cablivy, the brand name was, was approved and it was launched. Some of the programs I think were cut, so how, I’m curious, how do you feel now that it’s been a bit more than five years looking back on the deal, like how do you feel? 

Edwin Moses: I think the deal was, came as a bit of a surprise at the time because I think, as you remember, it started with a hostile bid from Novo.

We’re just moving towards approval of our first product and it set up a sales team and a commercial team. So we were all planning for the future and had raised hundreds of millions of dollars on the NASDAQ to fund the commercialization of the product. But when I look back on it, I think, positive things most certainly are that Sanofi brought an enormous amount of resources.

So as you take that lead drug that was approved. They now sell it all over the world. We would only be able to sell it in certain specific countries. We wouldn’t have had the capacity to do that. They have a enormous broad disease knowledge, so they can apply the platform more broadly than we could have applied it ourselves.

So I see that as very positive. If you look at the way the company is developed, I see it from the outside in. So I don’t have a very special information flow out, but just talking to our ex colleagues. What you see is the, the entrepreneurs moved on because for them, it’s all about fast moving, about quick decision making but the people with a lot of content knowledge and, and really understood deeply the technology have stayed and contributed that very well.

So, 

Philip Hemme: and the company is still, oh yeah, as is like independent subs, I mean, subsidiary physically. 

Edwin Moses: Yeah, it sits there in Ghent. Approximately four or five hundred people, I think they brought people in, not less. What’s, what’s a big difference I think is, a biotech really prospers by having low walls between different functions.

So it’s easy, the functions work, development, discovery, production, all of them, they’re only ten minutes walk away from each other, so you can always go and talk to somebody about what’s going on. I think what you saw very quickly there with Sanofi is that the Sanofi research people talk to the research people and have links that many faffing for their man, many faffing some silos.

Yeah. And, and when I talk to my colleagues, I think that’s one thing that makes me sad, it’s not the right word, but you, you, they don’t know what’s going on in the rest of the organization. I mean, for us, one of the things was as a biotech was communicating to everybody what the key things that were going on everyone was.

Now they know exactly what’s going on in their departments very well, but they don’t really know what’s going on with here or whether there’s a problem. It’s just the way it is. It’s not a, it’s not a criticism. It’s just an evolution. So I think the, the entrepreneurs moved on the people who want to see things happen very quickly or like working in small environments have gone on to find new environments.

So I think one statistic that I love is that at present count, I think there are 12. CEOs now who were ex Ablynx employees. So I think, you know, if, if I look at the legacy and the thing that I’m proud of, I know 3. 9 billion is fine and Michelle doesn’t make money and those things that I like that for being one of the most things as much as much of the products is that what it was, was an environment where people could learn and have the confidence they could go and do this themselves.

And I, I think that’s really, that makes me sure that that’s a really pleasurable result. 

Philip Hemme: It’s amazing. Yeah. Especially in a European environment, which is typically lacking of qualified executives. 

Edwin Moses: Exactly. 

Philip Hemme: So I’m curious, 

I think the, what’s mostly the Sanofi Genzyme team was, at least if I look to a marketing the product, so I guess there was also more, let’s say Boston.

I don’t know how, I mean, I guess now it’s, it’s very integrated. 

Edwin Moses: It’s very integrated. But it was certainly live from the Boston side. That’s 

Philip Hemme: for the, okay. But did that have a. Impact, as in like having a bit more of the biotech 

Edwin Moses: ginseng, right? Not really, I don’t think that would be fair to say. No, I think they’re a very professional, large organization that really just works as a machine and then sell.

Philip Hemme: I’m curious, I didn’t, I didn’t find, well at least I didn’t really find the sales numbers. Yeah, I just found this, like, there are some estimates when the FDA approval came on in 2019 that was, could be peaked at 500 billion. Are you aware of what the numbers are? 

Edwin Moses: Yeah, I’m aware of what the numbers are, but I’ve, I’ve, I’ve been told that Privately but I think that peak sales is very achievable from what I can see.

I think they’re well on their way to achieving that from what I can see from the outside. 

Philip Hemme: Okay. Yeah. I mean, I was wondering if there was really private, public, public data. 

Edwin Moses: I’ve been a little bit of public data, but I think basically it supports that original assumption. And that’s, 

Philip Hemme: yeah, I mean, it’s, and actually, you mentioned, you mentioned Novo, which was, which is, which is quite funny. Also, this was a, was a show in the podcast because basically Novo lost that deal to, to Sanofi. And then they went to acquire Corvidia, which where Marc, Marc de Geidel was on the show a few episodes back. It’s funny how, I mean, how biotech is a, is a really small world.

I just kind of, wanted to, to mention it. I also, I mean, what’s, what you mentioned, I’m curious also. I mean, you mentioned that you were prepared basically to roll out. You already had, you already had submitted for the European market approval. You’re already like, I’m curious when you look back, do you think they could have been.

Basically more value creation, as in, I mean, if you look at neighboring gains, let’s say RGNX just now, basically in the 20, I think 20 billion market cap. I guess in a comparable situation, did you see that this could have, I mean, could have happened? 

Edwin Moses: Yeah, absolutely. I mean, you mentioned the, the pipeline behind the product, the V program and the time, the acquisition, we had about 45 programs.

Now, a number of those upon, but the RSC program, which you point to, which was discontinued by Sanofi eventually, because they also let RSC programs. But we were very enthusiastic and you can see the products that have been launched lately for RSV and the interest in it. And of course the, the Ablynx program was an inhale product, which had special advantages because of the nanobody characteristics.

So. Yeah. There was a, there were a lot of products coming behind. I think that the, could it be a, I think the algenics strategy is very, very clever. The the pipeline within a product idea, you know, you go through all the development and the safety issues around the product and then you prove it’s safe.

And then if you can then apply that to multiple indignations, that’s a clever, clever thing to do. And that’s what, what they’ve done. But yes, the entire, that having said that, I think when I last looked. To get that valuation, ar genix had to raise about 5 billion in equity. Okay. We raised about half a billion in equity to get our valuation, so, 10.

So yeah, it’s, it’s not as straightforward seemed. Then of course, as we discussed at the time, we didn’t set out to sell the company, but once someone initiates a big process, it’s very difficult to say no. And I think that I may have mentioned to you last time when, when that initial process or the Novo offer was announced.

Only one big shareholder called me up and said, you don’t have to accept any of these offers. We’ll back you to stand alone. Everybody else said, get the very best price that you can. And no, that big shell was, was fascinating that they said it was, I’m happy to say it was Baker Brothers, very famous North American investor.

They had taken ages to build up confidence in the company. We’d been to see them for years and years and years, and then they made big investment in the Nasdaq. But by that point, they didn’t need anybody else to tell them that they believed in the company. And so they were prepared to see us stay independent for a considerable period of time, but everybody else was very keen that we maximize the price set up.

And that’s what you’re. You know, you hear a discussion about whether CEOs should sell the company or shouldn’t. It’s not their job to decide that. They have to do it ultimately, so. Yeah, they do it in the best interest of their shareholders. And if, if you get an offer, or the last time that was, our share price was around 20.

If you get an offer that ends up at 45. It’s very hard to argue that it’s in the best interest of the federalists, you know, you have to convince them you’re going to get to 45 60 within a year or two. And we all know the risks in doing that. So I think at the time it was it was not what we planned, but we were also very lucky.

Sanofi, being a French company, was very committed to Belgium, maintaining the site in Belgium. And then the research site. We spoke to American companies. At the time, and one of the heads of R& D owned you very well, so look, of course I could buy the company, but the minute I retire as the head of R& D, the American company will close you.

And I don’t want to see that happen. So in a way, getting a local, local pharmaceutical company was a huge advantage for maintaining the amblinx territory and activities. I mean, 

Philip Hemme: and for watching, I mean, sounds like a very fair I mean, fair, even premium fair, the info for everyone at the end, like, I mean, of course you can always think, oh, yeah, I mean, there’s some missing out and what’s the upsides, but if you look overall, I mean.

Edwin Moses: Exactly. And I think you, you know, we talked at the last time that you, you, in clinical trials, you know, a few months before that, we’d completed the phase three trial and you turn over the card to say are the results good or bad. And and I still remember the Saturday morning when I got the call from the chief medic officer.

And you don’t know what the answer is, and if it’d be negative, we’d be in a different place altogether. And he said, I think we’ve got a winner here. 

Philip Hemme: I think, because we still need to look at it. 

Edwin Moses: And he just, and you think, and, so I mean you, every time you do another trial, you, you’ve found, and recently Argenix for the first time has had a few little failures, nothing very dramatic, but you know, just reminds you, this is not, Easy to plan your way forward.

So no, I’m, I’m, I’m, I’m satisfied. I think the other thing I like, if I can just mention it, which I think is important is people talk to me, this is current employees and ex employees as it being the best time of their lives. That they learned a lot that they enjoyed working the, all the extracurricular activities.

We did a lot of stuff outside of work that the company promoted parties sports activities, all sorts of things to make it a whole, whole experience. And I’m just, but I’m good. The company benefits from building a relationship between the people in the company and the way they think, but they look back and say, I don’t think we’ll ever have a time like that again.

And that, again, I I’m pleased as well as the products and the technology. And of course the other thing that happened when Capitalism Kali was approved was suddenly people believed, and it was always a shock to me. They hadn’t believed before that nano bodies could be real products. So now you’ve seen, I dunno if you follow Moon Lake for example which has got a.

A product that they, that we licensed to Merck Sirona and they’ve licensed it at Antique Alsantein AF which is doing fantastically well in phase two studies now. So that’s another nanobody. The nanobody for TNF was approved in Japan by a Japanese company who licensed the product from Albin’s. So now you’re starting to see more and more nanobody products approached, how they get onto the market, approach the market.

That technology is now being accepted as real. There is a, a big pipeline in Sanofi and antibody products, so it’s, that’s exciting than the technology one. 

Philip Hemme: It’s good. Yeah. I, I wanna jump on the technology before that I wanna mention. Yeah, we, we, I mean, we talked in basically 2018 in the video is online.

I will, I will put, I will put a link. There’s definitely details on, on the platform and what happened, and I think it’s great also to compare. I listened to it again. It’s, it’s, it’s great to, to to see, and 

[00:14:49] Baker Brothers investor secrets

Philip Hemme: I want to jump on, on the Baker brother comment you made because, I mean, they are, they are very, I mean, they’re super, super successful first and you’re probably one of the most successful and they’re very famous for being very long term investors. I think with Siegen, they were there for 15 plus years and they made I don’t know how much the return was, I think a hundred fold plus.

But they’re also very famous for being very secretive, and, and actually I heard about them not that long ago, I mean, I think maybe, it was two years ago, I was talking with a VC and they stopped mentioning it, I was like, no, I mean, okay, I don’t, I will check it out, and then the funny thing is that they I started like, checking, and the only person I saw talking publicly about them was Yurik Shushan in the article.

I think it was some of like, Fears or, or Stardust or, and you were, you were quoted there. As in because they invested and I think you were saying that it took them a long time to invest in that from a judicious point of view, they were like very, very, very knowledgeable and doing it really. But once they were convinced that they will go, yeah, like all in, can you yeah.

And yeah, I mean, I also, one funny thing when, I mean about secretive is that they are so secretive that they don’t even put their name in the press release. It’s always. Unnamed healthcare, like a, like, what is it, oh, a significant healthcare investor. It’s really, really funny at the end, but I mean, but I know that choice, but can you talk a bit more about like, yeah, well, like, I don’t know what makes them, I mean, how was it, what makes them so?

Edwin Moses: Yeah, I think from the, from a, from a C, if you take it from a CEO’s perspective, I remember going there early on in my career in AppLinks and it was probably the most challenging experience of my career because the CEO, you go into the room and you might be used to meeting two or three people on the other side and they would come off as quite often with eight or 10 people.

And they would all be very expert, deep, deep science, and you know, you would come in and you show the CO with a sort of quite high level picture, and they’re suddenly asking you, so what is the color of the eyes of the patient in this trial, are you, idea? Now I’m just being facetious, but it’s sort of really detailed questions, and I think that the.

The amount of pre research they did about every discussion, you know, you go into a lot of investors, particularly here in Europe, but also sometimes in the US, where they haven’t really done much preparation and they haven’t, they’re not using their time wisely and they’re not using their time, your time wise, never the case with Beta, but they, they’ve gone through it and then they called.

KOLs in the area. So they’ve already talked to key opinion leaders to ask them, well, this drug, they claim it’s going to do this. Do you think it really will? Yeah. And so they’ll actually say, well, I talked to a hematologist consultant yesterday and you go, Oh dear. And so you, but what it does is it makes you raise your game.

You said, you know, as a CEO, you suddenly realized, no, I need to be better than this. If I’m going to do, you also realized you need to take more of your colleagues with you. But you can’t do all that on your own. You really can’t manage it, but it’s fantastic. But also challenging your story. And I’ve been invested in other companies I’ve been involved in and they are.

They’re very loyal, they take a, they take a considerable amount of time. You’re right, they have a view on confidentiality regarding investment. Okay, I’m, I’m cool with that. But the, the quality of the interaction with the company is very high class. They are very loyal to the company and to the CEO once they buy into you.

And I’m like, I think this is worth probably saying the, the NASDAQ flotation that we did, and they, that was when they became a very important investor in the company. One of the things that was going on, as you can imagine, is you have a pricing discussion, but you’re just trying to set the price for the flotation.

And the banks involved said to me, oh, well, there’s a lot of pressure from all the investors to lower the price. And then I said, I was grumbling to myself. And so I thought, well, I’ll pick up the phone to Baker Brothers, who I know are one of the biggest tickets in here. And I said to him, are you really putting a lot of pressure on the Pratt?

No, no, no, no. You fix the price of whatever you think’s right, Edwin, because we’re bad at that. So again, all about long term. Not, it’s not naivety. It’s all about not trying to screw the company or the existing investors for the short term. I mean, how do you build a company properly and feel you can trust them?

And that’s why they’re so successful, because they have a long term view of the, those things. So I was able to go to the bank and say, well, biggest investor coming in is, is very happy with the price. And we were stuck at, we stayed at the price. So no, I think all the, all the, the type of people, they, they employ top class and very, very good too.

Philip Hemme: It sounds like, it sounds like this really long term view. On the public market is, is not that common. And I would see it, I mean, but this is from my view and from my experience with investors, I would, I would see that kind of behavior much more like really high level VCs of angels who really bet on the, on building trust, super transparent.

And actually, I mean, from my little experience with investors at La Biotique, which was what I liked a lot with some of our investors, we were super open, transparent, even the valuation, we never really like negotiated super hard, was like, Hey, what’s kind of, what’s the fair price. And, and then working with them for long term.

And even when we sold the company, it was quite, I mean, relatively, obviously, I guess for you, I mean, same when you’ve paid off money on the table, it’s always. It makes the dynamics a bit, a bit different, but still it, it’s, yeah, it’s, it’s, yeah, it sounds, it sounds very like, yeah, very, I mean, very, I mean, very good, but I guess it takes longer to show that it’s, that it works as a, from an investment strategy as well.

I mean, I guess now with the CGN stuff, it now looked up and, and it, it’s proven to work, but it took them quite some years as well. But yeah. 

Edwin Moses: Yeah. I think they’ve had a lot of big successes and they, they proven that business model wouldn’t it? I think you’re right. It’s got to be a long term view.

They, what I like about them, which is a bit more prevalent in the U S and H C in Europe, if they don’t need anybody else to tell them whether this is a good idea or not. So actually when we walk into them and we talk, Oh, I’ve just got a big collaboration with Merkle, with Pfizer, and they really love our technology.

They sort of, it’s a long blink. They don’t really trust it because. They, from their point of view, they will evaluate whether technology is good or not. That they’re making a major call and that’s, that’s refreshing actually. You know, people who, who need other people to validate the idea, that’s not such a strong endorsement of what you do.

So you know that if they’d gone through your story and your equity story and what you promised and they’d like it, it’s a good story. I mean, they really will shake it to them and make you think. 

Philip Hemme: And you, are you still working with, or like, working with them on certain 

Edwin Moses: the company, one of the companies I’m involved in yesterday, they I understand.

Philip Hemme: And it makes me think, talking about also investor makes me think about the, the Strongman brothers and in Germany, which are, I mean,

investing earlier stage, I would say, but a bit in the similar, like, I mean, the, the, the investors behind I mean, now it’s, it’s Biointech, but many other than he made and many other successful, I mean, not most, but I would say most of the really successful stories, biotech stories in Germany. Yeah.

And it’s also amazing where, like, to just come in and invest and buy and take is crazy how much they invested in, I think they still hold like 40 percent of something like other company and we’re talking about 20, 30 billion in their company, which is crazy. But that’s, I mean, I think it’s also very, I mean, you say refreshing, but I think it’s very valuable for the ecosystem and we need, I mean, we need.

Investors like this, like, 

Edwin Moses: yeah, I think you need, I mean, you’ve got people who are more go with fashion. So, you know, today everything is obesity. They’re all pining into obesity and then something else. 80s thing people, you know, you did the people who followed and then the people who, who stay longer with the story and then they, they.

From a company point of view, what, what’s good is they don’t flip in and flip out all the time, you know, that you get a bad result that that doesn’t mean that they, they think your technology is wrong. They can see through that. Okay, that didn’t work. But the other things will work. And that’s, you need people like that who’ve got that, who are going to be with you for the, 

Philip Hemme: and why would you not work only or mostly with people like that?

Edwin Moses: Like, cause there are, there are not many people out there like that. I think we’re in reality. They’re, they’re quite a, a small number of that sort of quality and exist. 

Philip Hemme: But you as an executive, you would like, would you like prioritize those kind of assets? But it’s not like a limited. Yeah. And you just, you would be, I mean, you need to work on getting them.

It’s, 

Edwin Moses: I think it ends what your proposition is. I think if you have a very strong proposition, it also needs quite a lot of money. I see European propositions here where, The ambition is never quite the same as a, in my own company, so then these, well, they come to me and say, well, I need to raise 20 million.

Will you introduce me to U. S. investors? And I said, well, you know, the ticket size just, they, they can’t, someone like Baker Brothers, our capital, it’s. It’s hard for them to want to spend the time to do the dividends to only get 6 million of the 20 million ticket. It’s not the way they think about the world.

So if you’ve got a big proposition where they can really put money to work hard and it’s a, it’s a proposition that will stand up to their dividends. Yeah, absolutely. I would, I would take those companies to them straight away. And then people like R. A. Capital and Perceptive and, you know, in the U. S. that, who I think’s all had very good relationships with.

I’ve got, I’ve gone to them again where the proposition was very similar. 

[00:25:12] How to build a platform company

Philip Hemme: Ah, sir. Okay. Maybe on the, just on the talking a bit about the nanobodies, let’s say, like, I mean, you mentioned that, I mean, the, the, I mean, the platform was basically using fragments, I think from alpaca or fragments that you can then combine together that a lot of properties, I mean, in hiding will be one of them on many, many other benefits.

But what I’m amazed is always, I mean, there’s, there’s like, you know, there’s like, okay, you pitch the platform and there’s a lot of possibilities, but at the end of the day, you also need to prove and you, you mentioned, okay, you approved it in a phase three and it works in patients and then it kind of validates the rest of the platform, which I think is always, yeah, I mean, I would be curious, I mean, on, on there on like how, okay.

I think what’s really tricky and also it comes a bit from my experience, I worked with molecular partners with I think, I mean, a similar platform, obviously they would, I mean, I mean, it’s similar as in it’s, you could also have, you could, you could link several, like let’s say, they were not antibodies, they were alternative scaffolds, but darpins, what is it called?

But I think at the end of the benefits were, were, were quite similar. They were struggling as in the, as in to to, to really move the pipeline forward and have and validate the platform. I’m wondering for you, like, or some of the lessons maybe you’ve seen of, of how do you like, prioritize, like I think that’s really tricky as in like prioritizing, having your lead asset pushing really forward to the lead asset or.

Or doing more partnerships and what, like, what’s the best way basically to, to build such a, like, let’s say, platform company. 

Edwin Moses: I think it’s a key question. It’s something I get asked a lot, broadly, the platform companies to say, what should we do? I think that the, if, if you go back to the Amplink’s experience, there’s just one, one data point.

But the thing that triggered the sale was the lead compound. I’ve heard since that, let’s say at around the number of the 4 billion euros that Sanofi paid, I’ve heard, said to me that essentially when they did their calculation, they thought about half of that was for the lead compound, about half of it was black hole.

And I only have that information, I have no evidence for that. That’s what I’ve heard around. I think, and then I’ve heard that they, they think that was a hell of a deal getting that whole platform for two billies, you know, that was, that’s not bad. And, and so I, but I think that the, the trigger is often the lead compact, because that’s the thing where you can actually see the asset.

You can see what it can do, and you can see that it’s validating. 

Philip Hemme: Cash, I guess, from the revenue benefits also. Yeah, more palpable. 

Edwin Moses: Absolutely. Platforms, you know, they’re a means to an end. They’re only, they’re taking you somewhere. And, and so you’ve got to show, where’s the end journey taking you? Take you to that?

Oh, that, to that product. Okay, that’s an example of what it can do. Now I love the platform. But when you just wave your arms with platforms, that’s harder. And I think that one of the things about the nanobodies, which was, and you mentioned the origin. The origin comes that there’s a group of animals that come, there’s camels, llamas, alpacas, gormakas, vikuras, which all have this thing that when you put an antigen into a bloodstream, they produce normal antibodies against that antigen, like we do, but they produce these little VHH molecules as well, much more than that, so you can take those out and you can actually make, now the disadvantage of those compared with the example you gave me is that those have been produced naturally in nature So they’re already stable, they’re already buying to their target.

Liquid water formation. Yeah, so they give you a lot of the properties straight away that you really want. So artificial approaches, you have to overcome some more hurdles. So these aren’t Nature. Now the disadvantage of coming from nature is, as you could have mentioned as a CEO, kind of, Ah, I’ve got this product that comes from llamas.

Well, you see, some investors will just twitch off as you finish saying that. Because, of course, the previous 20 years, everybody had been getting humanized monoclos, stopped getting them from mice, you know, and we’d gone backwards in the mind of some investors. Oh, you were not humanizing them? You can humanise them, but the other source came from slender llamas themselves, and so there’s always the idea, Oh my goodness, I, I put a, an antibody from a llama into somebody, and it’s gonna cause them to grow ears, or horns, or forelegs.

And it sounds ridiculous, but there’s a bias, and a, and a, a concern that there are going to be unknown disadvantages with it. 

Philip Hemme: But I guess it’s coming from the EMAOFU as well. 

Edwin Moses: There’s always going to be challenges. You know, when we did our first clinical trial, there was the concern that you were going to get responses because it was immune responses, because it was coming from a foreign source.

Now, in fact, they, they are very, very nice molecules to work with. And we, at least in the experience I’ve had with them, you, we, we didn’t see any toxicity at all. I think then to your point about. How do you balance platform against assets? I think, I think one of the mistakes I see in platform companies today is that they.

They’re not taking, they’re not investing enough in their assets earlier, no, so they, they present themselves for rounds of funding to investors and they go in in a candidate and optimization and they’re going to be in the clinic in four years time or five years time. And that’s not quick enough. You have to start getting close to the clinic, starting getting clinical studies.

And I think we were lucky because the product that we chose, we, we chose it because they was found that the nanobody could bind to the target, the von Golovan factor, which was the target. better than any monoclonal antibody. So people have tried to develop antibodies to it, and they couldn’t get it to bind strongly enough to do that, and particular reasons that antibodies could.

But then we went after a different indication, a bigger indication that didn’t work out. And so we said, oh gosh, what do we do? And then we found this indication, TTP. Very rare blood deceit where suddenly there were no, there was no drug product available for it. We could see how our product could work in that environment.

And it was a market that from both the clinical trials and a commercial point of view, we could manage, we could realistically develop the product ourselves to keep it under our control. Because I think that’s the, the, the struggle for a platform company is turning into assets. If it at some point has to part of that, that asset of phase two, because it’s in RA or whatever it may be.

It loses control of that. And that, and that’s so key. I mean, back to the ambling story, the whole trigger was us getting phase three product success. And this, if the big pharma company is taking longer to develop your product because you partnered with them earlier, that’s very difficult for you. So it was, it was a lot of luck along the way, you know, you know, luck and being opportunistic being saying, ah, that’s important to see.

And I think that that that’s where the platform companies have also got to protect their investments. So they, it’s all, it’s super to do collaborations and invest in the platform, but you then have to ring fence your asset development and not always that it get dragged into another collaboration or something like that, because otherwise your assets won’t move forward.

And that’s what worries me with some of these platform companies. If you see them, they’re at too early a stage and particularly in this financing environment, where everybody’s looking for later stage assets. It means that they struggle to get refinanced going forth. 

Philip Hemme: I, I’m, if, I mean, if I bounce back on, I mean, the, it sounds also like, like, I mean, that it, it is the, the recipe for a platform company to focus on assets.

And I’m wondering, oh, it’s, it’s, but I’m wondering maybe, like, why do biotech not manage that? I mean, for all, like, let’s say. Even if platform got it, no, we have to move our assets forward. Why? Like, they don’t manage it that way. 

Edwin Moses: I think two things one of them is, is cultural. I think in, in Europe, again, the investors really learning the validation of partnering platforms.

They like it when you have a, a pharmaceutical partner comes in and partner with the platform because it gives them reassurance that the platform is strong. The American investors we discussed a little bit more independent. So they, they don’t really need that validation, but if you’re European biotech, that’s what your investors are pushing you to do, to do that.

The other thing is cash generation. So nowadays. When equity file, 

Philip Hemme: they push you, but, but from what you said, it’s basically not necessarily the best way to go as in the best 

Edwin Moses: long term solution. So what it means is, and I think this is where you talking about companies, but generally you’d have to realize that each venture capitalist around your board, has their own agenda and has their own boss.

They have to go back to. And so if they’ve been invested for one or two or three years in a company and nothing much is happening, they get beaten on the head by their boss. What’s going on? If things go back and I say, Oh, we just had a deal with Merkle. We just had a deal with Pfizer. That gets them, everyone says, Oh, well, and that’s fantastic.

That must be a, just show what a good investment we’ve made. So I think that there are those pressures. And it’s one of the reasons why I think CEOs have to own the business plan. Yeah. I see CEOs. I’m not where I’m working formally, but people come to me for advice, and they, I had one the other week, where he said I’m going to present the business plan going forward to my board on Friday.

I don’t believe in it, but the VCs are making me do it this way. And and, and, and that’s not uncommon, where they feel, pick it for your first time as CEO, and you’re surrounded by, Very strong, very experienced VCs who say this is the right way. Well, it may well be the right way from their portfolio point of view, but it may not be the right way for you as a company point of view.

So I think that’s, so why do they make those decisions? Do the collaborators can’t do that? But also partly when, when equity is more scarce, they raise cash through that. To, to raise cash through upfronts from pharma companies. And if you remember, you have Bingster, it’s 500 million was from England, 500 million was from collaborations.

And if we’ve had to raise that directly, it’s our lead, it has to sell some more to do that. So that, and I think that the thing, the other thing that I tell the companies I work with it, you learn so much from pharma, good and bad. You can see what, and, but you learn, they know an awful lot. And the other thing that you see is it helps you figure out how you should position your platform because.

You might mention some features that you think are really important, and they might be totally uninterested, and then they suddenly find, oh no, this is what they’re really interested in. And I’ll give you one example, it’s not long ago. So, we, we were filing patents at Amplink on everything that moved, basically, and we, she found that nanobodies could bind to ion channels, which are very interesting.

But we, we really didn’t know quite what to do with that. You know, see, an interesting group, they’re quite difficult proteins to deal with. And then one day we got a knock on the door from Merck and they said, yeah, we’re interested in your ability to bind an ion charge. And we said, we started to sort of do the story and dance about and they said, oh, shut up.

They said, look, we’ve read your patterns. We’ve made the nanobodies. We’ve tested them out. They do work. You are right. So we want to collaborate with you because you, you are the experts in nanobodies. So they said all that word before they even come to talk to us. So But then they taught us what their opportunities they could see with their therapeutic knowledge and their knowledge of the, the target base.

What can you do and how could we work together for success? So I think you can learn a huge amount from the pharma companies and that, that shouldn’t be forgotten. And I mean, one thing also, when you look at the way the nanobody is developing, can be here in, in Brussels, but we’re talking Proserix. So they’ve taken antibodies, and one of the asbestos antibodies, they’re very robust molecules.

I mentioned you can take them orally and they can survive the patch of the gut. But they’re also great for tying radionuclides to it, because they don’t get destroyed straight away. So they’re great at delivering radionuclides to targets. But also, they have quite a short half life, so they actually disappear out of the system quite quickly, which is exactly what you want.

Hit the thing, and out. So there are all sorts of companies taking advantage of nanobodies, and there’s one here also in Belgium using nanobodies in agricultural applications. Because they, they can do all sorts of things there, so they, they, it’s getting broader and broader, the application. 

Philip Hemme: Nice.

It’s, it’s good. Yeah. Yeah, I mean, we, yeah. It, it, it’s, it’s, yeah. It’s, it’s, it’s funny to, I mean, it’s, it’s interesting to hear your, your view and then when I connected to what I’ve seen at Micro Partners where you’re also amazing properties in the platform, but. Yeah, it’s, there were, I mean, at the end, you still need to, to prove it in the clinics and finding this balance is just, and as you said, also, I think it’s, and it’s, it should, I mean, we should not forget, but it’s like, there’s luck, there’s a luck factor and there’s a, just science factor of if things work or not, and every platform company, if you look back, I mean, at Dublinx, probably the science worked, as you said, like worked remarkably well and you know, No problem in the clinics and the good results and you found a good application, I mean, the good indication, everything.

I mean, that’s, that’s a lot of things lined up as well. I don’t know. I mean, we could talk. Yeah. We could talk longer about about nanobodies and, and radio, radio nuclear, I mean, I think there’s, there’s a lot of things happening at the moment, 

[00:38:57] Getting onto the board of an industrial biotech

Philip Hemme: but, but I want to, I want to switch a bit gears and you, you mentioned agriculture, which.

It’s great because I wanted to talk a bit about, like, let’s say, non healthcare or industrial biotech. And yet, so you’re a chairman of Aventium, Aventium? Yeah. And I followed this company for a while, actually actually from a very long time ago for almost when I started Labiotek, or it was not even a company.

And I met Truquin at Sofinova, and he talked about Avensium and showed a plastic bottle. I said, okay, that’s, that was his like, flagship, non, non healthcare investment. Which is crazy is that now they, they really, I mean, really worked. And so they opened like a massive factory and starting to roll out, like, scale production and, yeah, I think some kind of replacement PT from, from Plants in the Arthritium.

I, I was a bit, I mean, a bit surprised to see you on, on, on the board, on this board, which, I mean, okay not, not healthcare, not the platform, but can you maybe talk a bit more, like, how, how you came, how, how it came to be and, and what’s your view on, let’s say what their product and industry by taking an alternative.

Edwin Moses: Now I said, it’s a really, something I’m very, I really, really like one thing to say is that I was involved in Vantim even longer than you were by the sound of it because I was chairman back in 2000 and four originally. Okay. So after I left my first CEO job in the UK, I set up a portfolio of Vantim was one of those.

And at that time, Vantim had a number of activities, including activities in healthcare. So I was part of that and I was chairman of that company at the time. And I only left them because when I moved from being chairman of Outblinks to being CEO, I knew we were going to do IPO. And Avantium was thinking of an IPO as well.

I, I couldn’t be involved with two companies doing an IPO in the bank at the same time, it was too complicated. So I very regretfully parted away, but I knew Denny very well all that time. And then they came back for me. In 2019, I think it was, after I stopped with Applings, in which you consider being involved again.

It’s meant to know that I can do as the CEO was someone I appointed when I was chairman of the first time around. So I, it’s a very incestuous sport and it was Danny Lukan knocking at the door saying he’s now retired from the board, but he said, you know, be friends with me a lot. Now, why on earth am I there?

You’re quite right. I look, I look a little bit odd in terms of profile, but if what I love about the company, first of all, it’s like a biotech, so it’s full of very smart people, about 200 people, a lot of PhDs, they do. Very, very clever science, because the whole science is about taking sustainable sources and converting that into building blocks for plastics that are truly biodegradable and have advantages compared with current plastics.

So as you know, a lot of plastics now claim to be biodegradable, but they take 200 years to biodegrade. That’s not really. Ours can biodegrade in one to two years depending on the conditions, so it’s, it’s real. But it’s all about from sustainable sources. What’s fascinating is that you can develop these chemistries on the lab bench, just like in biotech, but in biotech, the next step is to do a clinical trial.

In this world, the green chemistry world of Amantium, the next step is to build a factory. Because the chemical companies don’t believe you until you made You can make 5, 000 tons a year at 10 a kilo or whatever the price is. So you have to show the economics work, that you can scale it. It’s safe to scale.

And that’s exactly what a manatee had to do. The, the challenges from my perspective are quite similar. And in Pusil you’ve got a whole bunch of very clever scientists and you have to think about how you’re going to monetize and commercialize that, which is what people already worked on. But then it’s raising money to build the factory.

So this factory that we’re building has cost. A couple of hundred million euros more, and we had to raise the money to do that. It will be completed this year and it will make them 5, 000 tons of this intermediate called FTCA, which is then turned into a plastic called PEF. And that plastic is already being used by big brand names like Carlsberg.

There’s a Carlsberg bottle with the names on this. You know, they did many, many industries. We could get Crona also. Yeah. Major supermarket stores are looking at it for their own brand products and getting rid of normal packaging, you know, producing it. And so the business model there is you build your 5, 000 ton factory, and then that proves that you can make it in scale.

And then people take licenses for that technology and build their own 100, 000 ton factory somewhere else in the world. And you get a license income based on that. That’s the way in which the commercialization works. It, it’s so similar because you are pre profit quite a long time. You’re, you’re burning money just like biotechs are.

So I sit around a board table of people from, we constructed a board, which is fascinating. So we have the CEO of Castrol Oil, for example around the table. Someone who was very senior in Tetra Pak someone who was very senior in Ineos. So huge, huge companies with very different perspectives of the world.

But the thing that makes them nervous is when the cash flow is going down with the PC, none of their gyms do that. And when the sign sticks longer, I’m very reluctant because I’m used to seeing cash running out all the time. That’s life. So, I bring that side of it, and they bring this huge knowledge of the end market and what people really need and, how to position the products going forward.

So, it’s a lot of fun to be involved in it’s still challenging because in contrast to biotechs. You have benchmarks in biotech. So isn’t it? Let’s say fabrics or whatever, you’ve got people you’re comparing yourself with, and the analysts were looking with for advancing the year, there really is hardly any benchmarks.

So the analysts. When we raise money announced that we’re going to raise some more money. It’ll be recently just raised to 70 billion rights issued quite often. Some of the analysts are, oh, they’re raising money again. What did you think we were going to do? You know, we, we don’t have a product, a revenue stream or a sizable revenue stream yet.

That is the nature of it. So, but they don’t have any benchmarks to compare it with. So we, we are a pioneer in that. Having said that we benefit certainly in the Netherlands from. very strong political pressure to improve the environment and improve the way in which we source and make plastics, not from coal and gas anymore.

So the government puts a whole lot of pressure. By the, by the economy. And, and so that means that we have a consortium of the major Dutch banks who support us with debt facilities in a way that a biotech complete. I mean, you would never get that sort of support from banks. So there’s a, there’s a real benefit in that.

You’re a pioneer, things are hard because you’re at the front, but it’s fascinating around being involved. It’s amazing. 

Philip Hemme: I like it a lot. And I mean, it’s, it’s crazy because it, I mean, you said you were involved at the beginning, but it’s really, it took them 20 years to reach, to reach that. And, and what you mentioned that it’s one of the only, I think, I mean, it is, it’s funny because I was always super like, actually really curious about this field of green biotech or environmental biotech from, from the beginning.

And When I was, when I was, I mean, when you said, 2004, I was still in high school. But when I started studying bioengineering, I was really fascinated by this and it was the phase when you had all this like, biofuels, microalgae, I was thinking of global bioenergies in, in, in France and, and like things were starting to, to cook, but at the same time, a few years down the road where things quite collapsed, at least in the U. S. for all that I can think of. By a few then still today hasn’t really like I mean, it was not a renovation phase, it was really just like, I mean, I guess maybe it will take off at some point. But it’s, it’s amazing that Aventium, I think, global IDGs are still running and, and seeing. But otherwise, there was always not that many in Europe that have managed to survive and grow.

Edwin Moses: They have other technologies. I mean, they have one which is a carbon dioxide. 

Philip Hemme: I mean, from like that’s industrial biotech at scale. They’re not competing, but yeah. 

Edwin Moses: So it’s, it’s, it’s, but I think one of the things in Mentorage I’m, I was fascinated by, if you look at good ideas, a lot of the best ideas take a long time to germinate.

I mean, if you look at Ampling’s, well, it first, it first began in the mid 1990s. The VIB supported it here in Belgium with the surge, but it couldn’t really get any funding. It took four or five, six years of them giving it soft money until 2001, the company started. And it was 17 years later that the first product hit the market.

Genyx, it’s been going around 2007, I think they started. Now it’s fantastic, but it’s taken And they were quite fast, weren’t they? I thought such good things take a long time to generate. I’m, I’m involved in a, I’m called Lab Genius, which is in the UK and people, some investors were asking me about it and they said, Oh, but it took quite a time to get there.

Well, yeah, because when you’ve got an idea of the technology, it’s not always obvious what you do with it. So you’d have to try and fail and try and fail. But those are the special people who they’d carry on and find the right thing to do with the technology in their life. But it’s, it’s not obvious from day one.

None of the big ideas are wrong. 

[00:48:41] What makes an entrepreneur

Philip Hemme: That’s the, yeah, I guess the fundamental of innovation and, and also on, I mean, innovation and also on doing things that have impact. I always have this, this quote that comes to my mind is a quote from Obama, like nothing worthwhile is easy. And I think there is, I mean, in life, but also in, let’s say biotech or in innovation, there’s some truth to that.

Like, 

Edwin Moses: it’s a lot of truth. And I think that one of the things that fascinates me, you know, you know, what makes an entrepreneur, what’s. If I take Onov Andostopov, who was the smoothie business CEO of Goanpinus, my favorite stuff, because I know him really well, quite well, is, is the way he came back from disaster time and time again.

You know, the, the, the product that is now on the market that forgotten him. When he did the original, I think it was preclinical, he did a deal with a major pharmaceutical company, I don’t know the name, and they, they ended up giving him back. Came back three or four times or something. And they, well, yeah, they said.

Never put this into people. They didn’t only give it to him back, they sort of gave him a curse at the same time. And I just think, when I looked at him, and I, I think the lesson to all entrepreneurs is you are going to have horrible days, things are going to go terribly wrong, and you’re going to fight back and say, that’s it, I’m going to have a glass of wine, go back in the next morning and start all over again, and that’s, that’s what makes him, that’s what made the great success that he developed, and if I take Time Genius, the, the CEO there, went down some path that didn’t work out, and And then figured out the way forward and that was, you know, and he’s, he’s a good example of what he figured out this machine learning AI business approach that he wanted.

He couldn’t get any European investors to pay attention because he talked about algorithms and things like that. And that switched live science and this stuff because they wanted to talk about the end of Robin. So what did he do? Got on a plane and went to the West Coast. I found a tech investor who understood exactly what he was talking about and invested in it.

That’s a real entrepreneur. I don’t sit there in Europe and go, Oh, I can’t get any investors, but go on a plane and go to the right place. 

Philip Hemme: I find that solution. Yeah. That’s amazing. Actually, it’s amazing that it works so well with James. I connected with you guys. It’s crazy. 

Edwin Moses: Well, I’m doing some due diligence calls, investors at the moment with him, and they say, well, how did it happen?

I said, well, I didn’t realize at the time, but James had always, was always going to make it happen, so he identified somebody who had a, a connection with me. And he’s He’s very clever in this whole idea of warm introductions. I mean, you know how that works. You know what, it’s one thing just calling someone up and say, hello, it’s me, but if you get someone who has already got a connection and an established relationship, and that’s what he used.

I mean, yeah, you, you were the heart of that. And it was very weird when he told me, yeah, well, I, I picked you out as the person that I wanted Ed with. If I needed to get to you and I knew him and I asked Philip and he got the introduction going. So here I am. 

[00:51:36] How AI biotech can help R&D

Philip Hemme: That’s amazing. And I did, actually, it’s, what’s, it’s funny also with you, what I think you’re just saying is that I think he was basically, I mean, one of the first, let’s say, AI biotech or whatever, machining biotech, I mean, they started in 2016, I think, and so there’s kind of Ahead of the curve.

Yeah. But I think now, and, and through our conversation, they’re facing, I mean, at least to my view, similar challenges of a platform companies. And it’s great to have a tech and to license the tech, but assets are basically where, where you make also, where, where the value question is. Absolutely. They seem to be now more, I mean, at least building a pipeline and or versus having a user tech.

Edwin Moses: Exactly. I think that having said that, that’s completely correct that the first important collaboration we had was with Samofi. blahblahblahblahblah and That, that went very well to the point that Sanofi came out and said, I think it’s also figuring out what, what can your platform do. And this is back to what I’ve told you before with the nanobodies.

And, and with, when I first went to, to James and one of the things he, he emphasized was how his technology could speed up R& D and that’s absolutely true. But when you look at the whole R& D process, it’s taking a few months out of R& D. We had to say a three or four or five year process. But you’re probably not gonna get ahead of r and d at Roche or head of RD at Pfizer.

Jump outta their seat to talk to you about that. But when you see how, 

Philip Hemme: maybe now a bit more, now that they’re old, they, I didn’t know it, but, 

Edwin Moses: but how are you gonna get, really get their attention? Well get their attention by was now the, the key USP of of ness is we will find things that you would never find yourself.

So that’s exactly what Sanofi said after we did the collaboration. We have found nanobodies that we would have never found ourselves because of the internal biases we have. And of course, if you look at the way pharma’s structured, who runs the departments? Very clever people who spend 30 or 40 years doing discovery science.

So they, they think they know, this group here never works, that group they must have, they’re whatever. So they have, and with machine learning, you take that all the way. You don’t know that come, come in the Discussion at all. You just put data in and allow the machine learning, the AI to do the projections.

So then you find things that the human would’ve been biased and said, oh, we can, we can never manufacture something with that. Then, well, you can, because you can use machine learning to, to teach you how to do that. So I, I think that that whole concept now of finding things that they would never have found, that is a very powerful concept.

And finding them quicker as well. 

Philip Hemme: Then yeah, from a value proposition, instead of saying. I, I cut down by one month out of five years and it’s a few percentage of, and you said, okay, I find you a product that’s where they’ve never found. Makes sense. And, and then how, how does it balance with doing the internal pipeline?

So then they apply the same thing to find things that, oh, that was building the pipeline 

Edwin Moses: and they look at certain particular indication areas. They’re interested in T SUN engages where they can actually use the, the properties of. Machine learning, which is to, to evaluate in parallel, different characteristics of molecules.

So instead of sort of developing and how strongly it binds and then looking at stability and then looking at something else, you can do two or three characteristics, optimize them in parallel. And that’s a really big advantage of machine learning. So yes, they’re applying, they’re trying to pick the best targets out for the application of their technology.

And the other thing I think is interesting in that company, which the learning for me, but. Is that you, you are also then combining tech investors with life science investors, and they are different types of investors. So it’s, it’s an interesting, it’s the same as in the company itself. You have life scientists, but you have software engineers, you have robotic people, and when you look at what they do, you know, honestly, could Big Pharma do it?

Yep, they could. But. The trouble is, the walls in Big Pharma are such that getting those sort of disciplines to interact in a cohesive way is really quite challenging in very large companies. But in a 50 company, you get them all in the coffee room together. And then the robotics people realize the life sciences people are normal, and that they can actually talk to them, and they can talk to each other and understand.

But it was interesting with COVID, for example, it just had meant mugged me too, to speak. But some of the disciplines wouldn’t come into work in COVID as some of the disciplines would. So they reacted differently to COVID as a, as a, so they behave differently in the way that they think and you’re not, that probably won’t surprise you.

But it means that you have to, but it’s a lovely environment of getting these different disciplines to interact and respect each other and learn from each other, which I, I, one of the reasons I like them.

Philip Hemme: I mean yeah, it makes me, it makes me smile, but I mean, I totally understand and agree. I was a bit in, I mean, myself a bit in that scenario.

I mean, in the media content space, but basically tech company, digital company applied to biotech and kind of, I’m talking a bit to the two worlds and yeah, it, but what’s, what’s quite amazing is that let’s say five years ago was two different worlds. And I, I’m amazed by how quickly it’s, it started like that, or how the, the, the, the overlap like grew pretty fast over the last years.

Obviously, Gen AI, GPT, everything helped, I guess, especially make farmers people understand that AI and there’s something coming and they should look and try to understand it a bit more, but it’s quite amazing. And even. But still, yeah, in investors, I mean we had Tomas Klosal from Okin on the show.

And he was saying that finding investors who want to invest in tech, in biotech, was like finding a panda in the zoo, a baby panda in the zoo. And it’s true that, I mean, especially when you try to raise that, let’s say a billion valuations. Then it’s, it’s really hard to find. And he also has most of the investors in the US and, but I find it quite, I mean, it’s a good, it’s, it’s, it’s, I think it’s very, also very beneficial for.

Edwin Moses: Oh, I understand. And I think that what you do is it. Just as I managed to say with the Avantium thing, when you get people from different worlds around the table, you get different perspectives. So you get the tech investors who think about the world in a different way. They, their, their appetite to risk, risk is different.

You know, it’s hard for them when you say, I’ll tell you what, we’ll put 10 million into an IND and it might fail when we get to the end of it. Now for your licensing investor, that’s their daily. That’s what they’re used to doing. That’s not how the tech investor would generally think of the world. And the idea that they, it fails or, or succeed, it’s really quite a hard concept for them to, but they, they bring their own special way of thinking to the idea as well.

I think you’d get, I like these mix ups, these sort of potpourris of people coming at it from completely different directions. We are in Lampedusa, I think, at the time where we, we’re going to successfully bring the life science investor together with the tech investors, and that’s fantastic because they can learn from each other and we can learn from them and everybody benefits.

Philip Hemme: Yeah. No, it’s, I mean, I mean, it’s, it’s funny what you mentioned about the IND, but also the In the tech world, like optimizing something by significant 10 percent is basically what most digital companies do. And it’s very valuable. I mean, whatever productivity tool, like a note taking app is just improving something, but if you make it 10 percent or, I mean, there’s a saying thing, make it 10, 10 X better than than people with switch.

But yeah, basically I was making better versus taking a huge risks. It’s, and I find, I think it’s, I mean, what you, what you mentioned is a key thing, and I see it, I talked with a few tech, let’s say, tech bio, or biotech companies, and When they start from the tech angle, they always start to optimize and reducing cost.

And, but I think what you said is very true. It’s not necessarily the value proposition or how it, what, what people are buying in, in the biotech products, that’s industry pharma industry, or maybe it’s not the best value proposition you can have. 

Edwin Moses: Key is, you know, the, the, the old story of rubbish in rubbish out in terms of a model is that what you have to do is generate data fit for purpose to put into your model and the idea that you can just.

For this type of work, just go onto the internet and, and find some data and stick it in your model, probably it doesn’t work. And so the key that James got, you know, that genius got around was defining careful, quite complex experiments to produce functional data that could be loaded into the, the models.

That would give you the ability to predict much more interesting spaces of activity rather than just straightforward binding data or something like that. And making sure you do that yourself, you think about the design of the experiments themselves. So you generate your own proprietary data. And, and it’s a similar thing with Achilles, the other company I’m involved in.

So it’s not the way it presents itself, but it’s based upon the fact that it’s got a license, Tracer X, which is a data set. It was created in the UK around lung cancer patients. So because the NHS treats basically all patients in the same way, you can actually combine these data from different hospitals in a way that’s much harder to do in a lot of other countries where different hospitals have different approaches.

So there’s 700 plus patients. which have been followed over the long term in terms of their lung cancer. And those data have been collected, and it allowed the company to develop, then, a model for predicting what would be the neoantigens, so the cancer associated with them, but more importantly, what would be the clonal neoantigens, which are targets, which appear on every tumor in the body of those patients.

So you can then take a sample of the tumor. You can actually then predict what these clone on the edges, the ones that stick all around the body and develop therapeutic approaches to deal with those. So it all starts from data. It all starts from very, very high quality data. 

Philip Hemme: And you can, and you can have the neurointelligence for all these patients in the same.

Edwin Moses: You do this individually. So this is individually, 

Philip Hemme: but you can compare all the patients. And 

Edwin Moses: yeah, yeah, yeah. So it builds up that thing. So it’s all built on really high quality specialist data, which then gives you very powerful predictive methods. 

Philip Hemme: It’s an amazing parallel between data and machine learning to data in a. self therapy company, but they have similar dynamics. 

Edwin Moses: Very similar, exactly. 

Philip Hemme: It’s crazy. You make the perfect transition to Achilles, which I wanted to mention as well. One thing that comes to mind is like, it’s amazing how on the surface it looks like different. I mean, machine learning to industrial biotech, to Abilene’s platform company, nanobodies.

What, what fascinates me is how below or like below the surface the dynamics. There’s a lot of similar Yeah, similar dynamics issues. Yes. Similar overlaps of technology 

Edwin Moses: be almost, at least for me, I have to be in it to release even the theater. I was, it was a while before I was in Achilles, in in Loves Jeans.

I thought, you’re not a similarity period. You know, and I hear James going on about why the. Quality of the data was so important. And then I realized that’s exactly what I’d heard from the Akiming’s founding scientist. But I just, it was presented in a slightly different way and I was hearing it. But then you suddenly know there, it’s identical.

I mean, completely right. But it’s identical. And if you start off with unique, high quality data, and then you’re in good shape. 

Philip Hemme: And it makes me connect to actually what we mentioned, Mark DeGaridelle. And he mentioned about the, the quality of the clinical trials, and that, I don’t remember, one of their phase two, they didn’t meet the endpoints, and it was because how they were, basically, how the, the, how the Sierra was running the trial was not at all about the drug.

Yeah, yeah. And he emphasized, I mean, or shared his experience in that basically on how to run, let’s say, properly the character and not trying to rush too fast, but having actual great data and, I mean, similar story. 

[01:04:22] The lowest market cap for Achilles

Philip Hemme: Maybe quickly on, on Achilles, then I want to finish on more on the personal note, but on Achilles, I mean, it’s basically on, on, on solid tumors.

Recently IO events got approval for the first solid tumor cell therapy. So tumor car or what is it? What’s til, til, til til therapy approved, which also helped, helped I guess some help on the, on the market cap. One thing that I was like, when I looked at the company, I think this is like the lowest.

Market cap to cash balance. I’ve actually seen, which is quite sad to see, but, a little bit, a bit surprising. I mean, it’s, it’s hard for all the, I could say small cab, especially platform bio companies at the moment. But he, I mean, I think was what, what, what, something like on 50, 60 million market cap and 150 million in cash, like It’s quite surprising, actually, that it gets to control the market, I guess.

Edwin Moses: Yeah, I mean, the company, it’s just public knowledge, so you can see it. The company went public in 2021 at about 18 a share, and it’s currently about 1. 50. It’s just gone up quite a bit because of the AI events discussions. The company hasn’t done anything wrong. It hasn’t had a fail. I looked at the news and I’m like, well, you know, nothing goes as quickly as you’d like it to do, but we’re in phase two studies.

We’re waiting for the data. We don’t know what, what that’s going to come out with, but it’s, but what happened is I think you saw a combination of events, the markets fell. New companies on the market got into trouble because of that. Cell therapy got a bit out of favor, personalized cell therapy, and the Ivan story dragged on a little bit.

People weren’t sure whether that product was going to get approved or not. And so we, we suffered a little bit from that. I think, but you know, it’s, it’s a thing used as a, as a board and as a, I feel particularly sorry for the management team in this because they haven’t done anything wrong, they carried out the plan.

They weren’t like crazy. And everyone says, don’t look at your shared price, but as A-C-O-X-C-O-I know you can’t help the, the be of shared breast, and you see it as a reflection of what you’re doing, and it isn’t a reflection of what they’re doing. I think the ref effectual will be when the data are available.

We see how that all works out. But the, the basic premise of what they’re doing, which is when you compare with iban, so IBANs basically take a tumor from a patient, then find the, the pills which are the tumor infiltrating. Yeah. So the, the way in which the body actually attacks the tumor. But the tumor has ways of controlling that, so what you do is, you pull that out from the tumor, and then you expand the tills, make lots, billions of them, and put it back in, to basically help the patient heal themselves.

The patient 

Philip Hemme: is not actually engineers they don’t express any thought or anything. 

Edwin Moses: So that’s how that works. The difference then with the Kelly’s approach is to say, hang on, there are some tills in those, in that big mix of tills, which, target the clonal neoantigens, the ones that I said are special in neoantigens and exist on every tumor.

If we can pick those out specially and then expand those, expand a specific population of the TILs, we may, we hope, we may have a more effective medicine. So that’s the idea. So it’s, it’s a little bit like IVANS 2. 0. It’s going along the same line, but it’s trying to pick out a special subgroup of TILs, which it then expands.

Based on that, Tracer X dataset of lung cancer patients, which is able then to make predictions from. 

Philip Hemme: And the first group, if you have a subgroup, then you still have enough quantity. 

Edwin Moses: Yeah, well, that’s what’s got to be proved, that you expand them and So I mean, it’s not easy to expand. Yeah, the mental sites are there.

For them, and for us, it’s always going to be challenging because each tumor you do, you know, the starting material is different everywhere and everything else, but the, you know, the prospect of a patient designed drug is specific for the patient’s neoclonal enchant is very exciting, I believe. 

Philip Hemme: And why don’t you why can’t you engineer a bit more the title?

I mean once you have extracted it and that’s I think something 

Edwin Moses: we wouldn’t see a reason why you would need to do that because it is already specific use of the body’s already created that they’re the right drug it just needs more of it and the local environment stops it the body producing that so you take it out and make millions or billions of the of these cells and put them back in so you know you’re still giving the body a helping hand you’re letting the body create pharmaceutical but then you’re making more of it yeah okay 

Philip Hemme: yeah compared to to a car I mean to a car seat where you will take the lymphocyte out, we engineer it to recognize the antigen of the tumor, and then we inject it.

Edwin Moses: And obviously the potential advantage with this is that you’re not doing any engineering, so you’re putting back in the natural molecules that were extracted from the patient. These are not new molecules for the patient. 

Philip Hemme: Yes, you limit the risk of immune response and of, and of what, stone, what, what’s the chemo ah.

Yeah. Yeah. On it. Okay. Well, let’s see. Shows, it shows I think how, how biotech can be sometimes it’s, yeah, the whole story, but you need to keep on the long term. And also, I mean, what you said keep on the long term, 

Edwin Moses: what I, this sort of pro proposition, which is the same as the other ones I get involved in.

It’s, this is gonna sound very facile, but it, the, the elevator pitch is obvious. You know, I’m taking a molecule that body’s developed. Specifically to fight this cancer and it may have lots more of a give it back to you and make you better. That’s going to work. Now whether it works for Achilles or whether it works for somebody else, I am sure in 10 years time we’ll look back and say, Well, of course, that’s the right way to do it.

That’s a patient specific medicine made from molecules that exist in the patient. That’s, that’s a great way to do it. So it’s, it’s, it’s so simple, it’s, it’s obvious. We, it’s LAMPGENIUS. What they’re doing is, concept is simple at a high level. Will they be the ones who make it successful? Yeah, I very much hope so.

But somebody will make it successful using this technology to design new therapy proteins very effectively and find ones that people can’t find anywhere else. That’s the exciting part about it. It’s sort of obvious. It’s gonna, it’s gonna work. 

Philip Hemme: But yeah, the execution of it and the timing. When is it ready to?

Edwin Moses: Well, you can’t predict that. Certainly. 

[01:10:53] The draw of being the Chairman

Philip Hemme: That’s great. Maybe if I’m just switching a bit more to, to a personal note or finish on a personal note. I’m curious, you seem to like a lot the chairman role. You have three of them, but I guess you had many other options from whatever other regular board membership on bigger biotechs of pharma.

Involved with VCs, whatever, like, can you like share a bit more on what’s, what’s the rationale behind this, this like, Sweetball 

Edwin Moses: isn’t that always safest? Although I don’t have to be chairman in the company. It’s just happened like that. I think probably where the driver for that comes from the fact that I, I genuinely like to get involved.

All I’ve found in a small to medium sized company is that as a. Just a non executive director, it can be difficult to provide, to get access to allow you to leverage what you know and what you do, it’s a little bit more tricky because the chairman CEO access is so critical and that’s got to work very well and that’s key to the company so that’s why I enjoy being part of it because I want to be part of that and off my services.

So I like, I like to get involved. I’m talking to the companies all the time in in different ways. So that it’s, it’s part of my level of engagement in this, in this size of small to medium sized company. What I’ve chosen not to do, go off poignantic, if you feel involved in sort of short term consultancies to look at projects I, I, you know, people will say, well, we’re thinking of buying this company where you look at this company for us and make an assessment talks.

Private S3 companies do that quite a lot. I did a little bit of that. I don’t sound funny, very satisfying because I’m involved just at a point of content and I, I’m, I’m like a reporter. I tell them, I think this or that about the management of technology, and then they go off and do whatever they do.

I’m much more interested in operationally running, how do companies develop? How do they build, how do they hire people? How did they write the business plan? And, and being with them as they go through those transitions, just sort of giving them some advice and then going on to the next job. I think it’s, obviously, you know, that’s what consultants do and they like doing it.

It’s not for me. I want to, I want to see how this all works out. If you look at the training involved in that, I’ve already been involved with about, I think it’s like the shortest time, four years. These are projects you really get involved in. I have to learn. 

Philip Hemme: I guess it’s more entrepreneurial. I say more.

Edwin Moses: Yeah. And, and that involvement in, in the whole aspect, you know, raising money, hiring people, firing people. What sort of business do we want to be? How do we, how do we develop that and and going through the different transitions from being a private company to a public company and that. Those things I really enjoy and I want to be part of that.

Philip Hemme: It, it makes, it’s, it makes connect to, what you know, that cause I’m sure in crystal therapeutics and, you know, I forgot his last name. And they mentioned it and I think they mentioned it quite a lot about, especially with founder CEOs who are maybe a bit less experienced having the experienced chairman and that this duo can be like, can, I mean, can work very well and bring kind of this like entrepreneurial energy, naivety sometimes can be beneficial, but same time really this experience and when they work together, Do you see that?

I mean, maybe, for example, is that genius in you? 

Edwin Moses: Yeah, absolutely. I think you can see that. I think what from the experienced chair point of view, if I can call myself that, where you have to be very careful is, is not always coming with what you think are pre baked solutions. Because it’s boring for the CEO to be told this is the way to do it.

It’s also wrong because it’s not always true. So what you 

Philip Hemme: You’re really going into deep understanding. Deeply understanding. 

Edwin Moses: And I think what you have to do is, is More likely provide options, say these are the sort of choices you can consider or challenge some of the conclusions, rather than saying this is the answer.

It can be frustrating because as a CEO for 20 years, I always thought I knew what the answer was. And, so you, you get to, you think, I know the answer, this is obvious. You, you realize that’s not the way to be. 

Philip Hemme: How many times have you been proven wrong in the last years where you, you thought the answer was this and it was not this?

Edwin Moses: I can’t remember the exact number. Not that you’re used to them. But I think what you do, I assume, with it, Unless the CEO gets there in their own way, you haven’t got a solution. If you try an input, it’s, it’s your, you’re working different ways and at different speeds and in different ways and different sort of shades of gray in terms of what the right solution is.

It’s got to be their solution. So you, you really just got to try and you’ll put a red flag up that you can see something that’s a dead end. Don’t go down that dead end. That’s horrible. But the other, the other thing they’ve got to find the right thing that suits for them. So I think it’s, it’s giving those ranges ranges.

And I think more. Also, quite often when I’ve been talking to new companies, they say, well, watch your style as a chairman, you know, and people have been through what I have, but I’ve got a tendency to come in and say, this is the way I work. Now, I, I actually say, well, I think I should adapt to the way that James wants to work or, you know, I should have kicked these and not be imposed upon them the way I work.

I’ve certainly, I’ve got certain sort of styles and certain quirks and so forth, but it’s best to fit, it’s more fit, I’ve got to fit and try and, I don’t want to say gaps, but I’ve got to support the areas where they don’t have strengths. And the area where they’ve got strengths, let them, you know, carry on, right?

So that’s what I find interesting is figuring out, you know, some of them like a lot of contacts, some of them like to, to check things with you quite regularly. And I think with, with James, a good example is that. Early on, he, it was probably my fault thought. There were things that were too small to bring to my attention.

And I, and it took me a little while to realize that. And then I, I said to, well, you know, there’s nothing to talk. I can, we, you know, we are, we’re friends, we are colleagues. You can, you can come to me with anything and if I think you are, you are working at the wrong level, I can always tell you. So this, he then felt much more open and nothing was too slow, because if it’s important enough to be bothering him.

Talk about it. And, and so I think 

Philip Hemme: And also the small thing gives you a deep understanding of 

Edwin Moses: Exactly. Exactly. And, and there can be, you know, for first time CEOs, there are things they just haven’t done before. That’s the nature. We’ve all been first time, whatever. You know, you would first time you interview someone, there’ll be things you learned from that and you go on and you’ll have done things then that you would never have done again or whatever.

And so you can just see some of those things and you see them coming. And you can Exactly. You could help them with that but it’s, I think it’s got to be, you mustn’t overwhelm because it’s all about, at the end of the day, they are the boss of the company, not you. 

Philip Hemme: I was about to say, yeah. What you mentioned also is owning the business plan.

I mean, at the end of the day, they are executing and they have to, I mean, even if the solution, you propose a solution there, it’s settled. The management team or CEO who is actually trying to 

Edwin Moses: Let me say this quick. I think one of the things that, when I, in, in, when I’m talking different CEOs, not just the, the, ones that I’m talking about that I’m directly involved in, I think sometimes what they find hard They’re very happy to take the credit when things go well, but when things, particularly out of their control, don’t go well, then they say, well, that’s not my fault.

And I say, no, it’s not your fault, but your candidate would be. It’s a joke. You can’t have it both ways. And is it, you know, if the clinical trial were to fail, then the CO is at the end of the day? the way you’re going to look at it, even if it wasn’t their fault. But that’s what you, you know, that’s the positives and negatives.

You can’t, you can’t just want to have the good times. So it’s one of those learning things and giving them support. In times like this, I think is, is really important and then having the feeling that I ain’t in perspective, I guess, someone that, yeah, that today’s, today’s a bad day, but tomorrow’s going to get better, you know, and then just helping them and look at what are the good things that you can see here, because sometimes it’s, When things are going wrong, it’s easy to focus on just the negative, you know, what, what’s happening.

Yeah, but let’s look at the bigger picture. What have we got here? We’ve got a company that’s got these investors. It’s got this technology. It’s got this great group of people in the company. That’s pretty good, isn’t it? That’s, yeah. It’s amazing. 

Philip Hemme: I mean, it’s, it’s almost a bit like a coaching, I mean, role, but this is also super valuable.

I mean, I can imagine as a CEO founder. I mean, who else would really give, provide this kind of value and, and even from the, then from a company building and shareholder, it’s just an amazing role. I mean, 

Edwin Moses: I think the other thing that they, that become a sign of myth that they use, you’re doing it not from a theoretical, this is an MBA, I’ve read an MBA.

They know you’ve been there. That doesn’t mean that, that you, you’re right about everything, but they, they recognize they, you know, if they have a hard ball beating. And he’s not gone their way. And, and I can go to them and say, yeah, I sat in Har I I’ve been the one who didn’t get the picture right or did people didn’t agree with me and that’s frustrating.

And, but they didn’t have what we have to do and I liked that. So you can help them bounce back from the more difficult things experience. 

Philip Hemme: I like, actually, it’s funny through to wrap, but one thing that came out of my mind and out of memory from this Twin Reach talks in 2018 was actually the slide of like as a CEO you receive credit.

When everything goes well, but then you get hit on the back when things go badly. And I found this clarity about it, which I found pretty, like, I mean, first I could relate to it as you, myself, and I think, yeah, it’s, I mean, I think it’s very true, but I think you’re very, like, approached to it of taking full ownership, especially when things are bad, and that, I think that is very difficult to do.

Yeah. But once you embrace it, then 

Edwin Moses: You race it and once you, it’s easier said than done, but yeah. The way you depersonalize it that you, you, you don’t take it in your heart if you, you know what I mean? Which is a little bit harder to do, but that’s 

Philip Hemme: very hard when you found a company that’s basically, yeah.

Edwin Moses: But you, you have to sort of say, yeah, okay, this didn’t work out. And the main thing is, yeah, I think one of the lessons, you know, talked to James about quite a lot, is that boards don’t want to be confronted by problems. So if you’ve got to fail a clinical trial which is not what James has experienced, but one of the eight William or Willam clinical trials, and you go to the board and say fail, you will get massacred by that board.

They will, they will tell you what you did wrong and everything else. So what you’ve got to go to, and this is what we’re going to do next. Everybody wants to know what you’re going to do next when something goes wrong. And that’s what the CEO. That’s the great thing about bassoon. You are in a position where the guy who’s managing the orchestra, you can point the violins and say they’re gonna play for 20 minutes while we figure out what to do.

So you can, you can actually Lay out that forward plan, but that’s what you could, I think sometimes people think on the board wants to hear what, what happened. Well, they do, but they want to know what’s going to happen next. Is this a Bush mask? Yeah. Well, and even good news. They want to sit up here. Right.

That’s good news. How are we going to build on that good news? What are the, how, does that mean we’re going to raise money or we’re going to have a big collaboration or whatever it may be? So you’ve got to, that’s what you, you always got to be forward thinking, just. The reporting bit of what happened is the least important part of the theater’s job.

It’s got to be done very well and very clear, but it’s the creativity is saying what’s going to happen. 

Philip Hemme: Sounds like it’s you’re fitting in this role. Like, I mean, it sounds very balanced and it sounds like you’re enjoying it. And that 

Edwin Moses: it’s never the same as being the member of a single team. And 

Philip Hemme: I was about to challenge you on like, why not being executive some like another? But 

Edwin Moses: yeah, I looked at a few you know, never say never. I think the other thing I, I like a lot about this role, which is, which you’ve got the hint of, is that being a steel and pickle company, you get very expert in it, you know what we’re doing, but you can get a bit lazy because you are expert in it.

You know, when, the day you told me about Applings, I knew everything about Applings. I knew everything there was about nanobodies. But now I’m thrown into machine learning, I’m thrown into environmental chemistry, software. I knew nothing about those things when I went there. So I had to learn. So each day it made it look so wonderful.

And I, that, I like that. As, as you get older, the challenge of not being relaxed about what you know. Yeah, I know some stuff, because if I didn’t know some stuff, I’d be no use to anybody at all. But, but I’m, you know, I’m just in awe at some of the things I learn every day from the different interactions with the companies, remember?

The technology, the end markets, all those things. And that, that’s what gets me excited. 

[01:23:35] Thanks for listening

Philip Hemme: Great. I think it’s a great it’s a great wrap up to, to all, to our whole discussion. I mean, I really enjoyed it. I mean, that’s, I really love these like longer formats because we can explore deeper and we can see how things are so connected and how the content, I mean, it’s fascinates me from our discussion, just to summarize a bit, but yeah, and how, how the story of links and building a platform is connected to other platforms. How the dynamics are similar, how the experience, how everything it’s great. Thanks a lot. 

Edwin Moses: Thank you very much indeed. Great. 

Philip Hemme: Thanks for listening to the end. I was amazed by Edwin’s really deep experience and it also fascinates me how things can look so different in life sciences on the surface but if you dig a bit actually have so many similarities.

If you have enjoyed this episode please hit the like follow button and if you are listening on apple podcast or spotify You can leave us a five star review, any of these actions would help us a lot. I would also be curious to hear what you think so I can improve further and help you grow even further.

If you could, comment wherever you are, if you’re on social media or on YouTube, or send me an email to philip at flut. io Alright, see you in the next episode.

Jens Nielsen, BII | How to make translation of life science research work | E12

We’re in Copenhagen with Jens Nielsen, the CEO of BioInnovation Institute. We will talk about what works best to translate academic research in bio into innovation for humanity, especially how everything is connected. We also talk about how BII is solving some of the main challenges scientist entrepreneurs face and the drawbacks of Novo financing most of Denmark’s biotech ecosystem.

Jens Nielsen is one of the best in this space in Europe. Not only is he the CEO of BII, he’s also a professor of systems biology at Chalmers University of Technology in Sweden, and is the most cited researcher in the field of metabolic engineering as well as industrial biotechnology. He’s the founder of the International Metabolic Engineering Society and has also founded many biotechs.


[00:00:00] Intro

Jens Nielsen: What is of course important for creating an entrepreneurial culture in a lab is the open mindedness. 

Philip Hemme: It gets down rapidly to that of what’s the role of academia. 

Jens Nielsen: That’s a, that’s a difficult question. That is why I think that many biosolutions companies failed because they were inspired developing new pharmaceuticals.

There you’re not pivoting. 

Philip Hemme: In innovation especially it’s all about people. Yeah. Works also. Yeah, 

Jens Nielsen: yeah.

[00:00:29] Welcome

Philip Hemme: I’m your host, Philippe, and welcome to a new episode of the Flot.bio Show, where I interview the best Europeans in biotech through how to grow. Innovation in biotech mostly comes from translating academic research into products, but it’s far from easy. Jens Nielsen is one of the best in this space in Europe, so I went to Copenhagen to talk with him.

And I’ve known Jens for many years, and I met him when he was running one of the largest academic research group in bio, and he’s now the CEO of the Bioinnovation Institute or BII. We talked about what works best to translate academic research in bio, especially into innovation for humanity. And especially how everything is connected.

And we also talked about how BII is specifically addressing some of the main challenges that bioentrepreneurs and the biotech ecosystem has. And we talked about the drawbacks of Novo, especially Novo Nordisk Foundation, financing most of Denmark’s biotech ecosystem. So here’s my conversation with Jens.

Audience. Yes. Welcome to the show. Thank you, Philippe. Great. Everything works now. Yeah. Okay, good. Ready to go.

[00:01:43] Translation of science

Philip Hemme: All right. So I want to start with talking with you about translation of science, especially in life science, biotech. I think you are at least one of the best I know in that space from whether, when you were as professor, as PI, entrepreneurial lab, now as CEO of BII.

Maybe the one to start with is what were your lessons of what works and also what doesn’t work? 

Jens Nielsen: Yeah, thank you. That’s a, that’s a difficult question. You know, because it’s, it’s hard sometimes to define when you have a good case but Let me rather try to start by saying a little bit about, you know, my, my perspective and my view of, of running my research group.

And then, you know that resulted in a number of cases that work and some that, that did not work. So, when you, when you have a research group you know, you, you of course are focusing very much on. On, on educating students, PhD students postdocs doing research that are, you know, of, of addressing scientific challenges and so on.

I’m trained as an engineer and so therefore I always had the kind of the perspective of trying to see that things we do should have some impact on society and they should, you know, address some challenges that eventually could lead to products or services for, for the society. And, and so of course when you then begin to look at, at, you know, a research problem, you can find things that are big challenges in society and you start to do research in that area.

And, and here you immediately come into certain things that will not really work for translation because there are simply some barriers, either regulatory. Or simply because if you are going to do that, you will need, if you’re going to pursue a new business in that area, you would have to transform so many things in, in society and so on.

So, so that I think I learned. Quite early on, actually, that, that starting a new venture, can be difficult and you need to be able to find where you can actually address a certain market and where you also can, you know, and, and, and of course there will always be barriers when you make a new venture, but, but, but, but they should be, it should be possible to pass them.

Philip Hemme: So it is finding the sweet spot where. Your technology or your science is basically solving a problem and then where it’s actually there’s a market, not just a market need, but where it fits with the market, investments, markets. 

Jens Nielsen: Let me take a good example, something that we did a lot of research on in my group was, you know, new biofuels, advanced biofuels.

Which is something that you could say that it’s obvious we need this in society. And I I worked very closely also with some of the large oil companies in that area. They were also very active in it you know, 10 years ago. They all stopped because this is extremely sensitive to the oil price and this is something you do not as an entrepreneur have influence on yourself.

And, and so unless there is, you know, you could say there’s made some overall, and maybe we’re beginning to move in that direction now, but at that time it was just impossible to make really you know, a viable startup company in, in that area. So, so, so there is where you can say. Do we need it? Yes, we do in society, science wise.

Can we do it? Yes, we can, but it, it, it will not, and it could not work. 

Philip Hemme: How, so I mean, yeah, I guess, I mean, the, the timing also has everything. And I mean, startup wise or even translational innovation wise, the timing always is a fine line and you don’t want to be too early. I mean, biofuse, I guess if you were like 10 years ago, even like.

The early 2000s with that kind of biofuel bubble, which completely bursts and still today it didn’t take off or it’s still not solved. But how does it translate to you, maybe your lessons of what works, maybe what works and also what works when you’re running your lab of like, where you may, may question, where you trying to take it like backwards by knowing that, There is a need of timing and fit and a certain type of problem can be translated and then you move it backwards and you define the research project.

Jens Nielsen: Yeah, so, so I think so, so as I said, we did a lot of research on, on making advanced biofuels and that was based on, on, you know, chemicals that could be derived from fatty acids. So therefore we. had a quite a large you know, research activities on producing fatty acids, and that was using yeast as a, as a production platform.

And and as I said, we, you know, we’re looking into whether there could be a, a opportunity to make a startup addressing biofuels, but, but, you know, that I could just begin to see that, that we would never come through with that, we would never get financing and so on. So then, of course, you, you start to evaluate what, where, where else can we use this technology?

And, and you then you know, maybe begin to look into some speciality fatty acids and stuff like that. And, and, and in, in each case, you need to dig a little bit in order to evaluate you know, is this potentially interesting and, and, and, and key metrics that comes out as, you know, of course, market size, because it needs to have a certain market size in order for it to be interesting.

But also, can you produce it with the technologies you have now at sufficiently, you know, quality and cost? And so we iterated actually quite a around, but then an interesting thing happened is that you, you know, with this whole plant based food movement that you started to see, there was suddenly a need for capabilities in producing fatty acids that had, you know, unique properties for, for that application.

And so the company eventually pivoted into that space and has now raised you know, several millions of euros to address that market. And this company, we also changed the name. It was originally called BuPetrolia and then it, it pivoted and changed the name to now Melt and Marble to address that, that market segment.

So, so I think that’s, that’s an you know, I often say. If you have a very strong research base, there is a good chance, and you did have an application in mind, that You can, you can find an outlet for this, but you may have to pivot on the business model several times. 

[00:08:48] Being flexible is key

Philip Hemme: That’s, I’m thinking back to my question of like, what, what’s the lessons here, what works, but I guess, yeah, yeah, being flexible.

Jens Nielsen: I exactly, I think a key lesson from this is being flexible and not, you know, and being open minded, to, to continuously adapt and learn from, from what feedback you get, And, and, you know, it can sometimes be hard as a scientist because you get really committed to the, to your course and think that this is exactly the right thing.

But when you want to make startups, I think you, you have to be more flexible and, and be willing to, to scrap your ideas and actually go in a different direction. 

Philip Hemme: Yeah, I think, I mean, even I’m making, thinking about the parallel between life science startups and more digital startups, or let’s say any startup, but I think I don’t know many startups even.

Mine that haven’t pivoted. Yeah. It’s very rare that you start with a, whatever projects market and that this is the right fits product market fit and that it goes big. I mean, it’s very rare. So yeah, it’s interesting to apply the two life sides where, I mean, I think you have probably a bit limited or more limited to, I mean, Your patents and your technology is there, but the application then is, is usually you have several markets you can tap into and you can go 

Jens Nielsen: No, exactly.

No, I and, and I agree. I mean, and I actually, I’ve reflected a lot on this pivoting, you know, because if, if you take tech for example, or, or it, you know, there’s very typical that you, you, you pivot along, you know, and and you know, you create mv MVP, you know, and you evaluate that and so on.

In life science, it’s hard to make an MVP because, you know, alone to make a product is, is, is a difficult task. But I think also where and, and, and this Melton, where Melton Marble is operating is what we call biosolutions space. But what I think that space had suffered a little bit from was that it had had the mindset of, you know, biotech and when we talk about biotech, that’s often pharmaceuticals and trying to apply that business model, whereas actually the bio solution should more look at tech.

For, you know for in terms of inspiration in, in, in, in changing and modulating the business plan along the way, but still pivoting, of course, around a very, you know, solid scientific route. Yeah. And, and, and, but that, that is why I think that many biosolutions companies failed because they were inspired, of course, from, from the, the, you could say the, the, the more advanced scientific analogy, namely developing new pharmaceuticals.

But there you are typically stick there. You’re not pivoting. We see that also at BII. They come, these companies, maybe they adjust a little bit what, what disease indication to do approach first, but, but, but, you know, that’s, that’s, that’s not really a major pivoting. It’s just adjustments, you could say.

[00:11:58] When to stop being flexible

Philip Hemme: Yeah, I like, I mean, the whole MVP lean startup. Yeah. I think it’s, I find it, I mean, I think it, it works. I mean, text for sure, but also in biotech and, but I like your point. And I think that’s probably where it’s really tricky as well as finding. How much does it really apply? How much does it not apply?

How much can you do MVP? How quickly can you do MVP? How quickly can you pivot? I mean, in tech, maybe, yeah, okay, in 3 months you can test a product in the market and then decide, yes, no, biotech will take, whatever, 9 months, 12 months, even more. I mean, how much runway do you have? I mean, that’s also very hard to apply, and I think also What I can see there, and I think with BI you’re addressing this, is that I think that also takes some skills that are typically a bit more, let’s say, strategy business skills than typical scientific skills.

And yeah, maybe you have some scenes, I mean, I guess you have seen what works there from the, like, making the connection. 

Jens Nielsen: Yeah. You know, I think it’s a, it’s a good point. Let me first try to address this about, you know, pivoting still, you know, because of course you, you, you also need at, at some time stick to a certain plan because otherwise you will not be able to raise money.

And, and as soon as you begin to raise money, it’s important that you maintain the same narrative and, and actually address that market. So at one time you need to, of course, really dig in and, and, and, and push towards that direction and so on. So I think, and that is also more because you need to have a pretty much laser focus on what product are you aiming for, because as soon as you begin to have to scale up and so on, cost really increases dramatically and you don’t want to waste those money on betting on the wrong product.

So, so in that sense. It’s fine to pivot initially around it, but, but you need at one time to settle on something and then stick to it and then hope that that is the right thing. 

Philip Hemme: What, what specific, like, can you give, like, specific examples of, like, exactly at what stage you can still pivot, what’s the timeline looks like, or kind of, is there something a bit ideal, like, that 

Jens Nielsen: As soon as you begin to, you know, and make the real MVPs because then in, in biotech when you need to have an MVP, you need to have production in certain scale.

How many liters? So then we are talking about at least 500 liters typically, because otherwise you cannot get enough material to provide to, to potential future customers for testing and so on. And then. Of course, you can adjust a little bit, but, but it, it, that will really more be smaller adjustments along the way.

So, so I, it is at that, that, that stage that it’s more, becomes more and more difficult. 

Philip Hemme: Yeah. But 500 liters, I mean, takes a bit of time and capital. 

Jens Nielsen: Exactly. Yeah. You can, you can go to, to contract manufacturing organizations and, and, and have them to of course help you and so on, but still it is costly.

Philip Hemme: Yeah. So we’re talking already. I mean, a big seed round or 

Jens Nielsen: yes, yes, you would normally ask, I mean, that, that, that you will not really get far with less than 5 million euros as, as, as, as that would be a big seed round. Yeah. 

Philip Hemme: But so maybe, yeah, that’s, I mean, yeah, but that maybe you can reflect on the, the stage before, before, like basically on from, let’s say.

If we, if we still take this like pivot example from, let’s say someone in the PhD postdoc has found something, maybe patent application, thinking about spending out to this 5 million, like, then specifically, like how many pivot can he do or he or she? 

Jens Nielsen: That’s, that’s a, that’s a, you know, not too many because then, then it becomes time consuming.

But, you know, this is exactly what we are doing at BII. We are helping. These cases at an early stage. So, so we you know, we often, when we evaluate applicants coming to us we, we are looking very much at, you know, what, what is the, what is the scientific basis for this? Is there something to build on?

We look at what are the the founder’s first bet on a potential market, and, and then we also, of course, look at the team do is this team willing to also maybe pivot if it becomes necessary? So do they have the right agility and flexibility? So these are the three kind of criteria we’re looking at, but then in our first program where we, we give the companies about half a million euros, so it’s not a lot of money, but that’s really the key objective is to make the business plan so that is to validate the These hypotheses that the founders may initially have or validate some part of the exactly and, and, and, and, and, and, you know, see that and hopefully then based on this build a sufficient case to, to get to the seed round that we just talked about.

And we do see in, we do see in those 12 months very often that the companies pivot in, but, but normally, you know, it’s, it’s smaller changes. We have had cases where we have seen a complete pivot even happening at the bootcamp when they come in and, and so it’s a completely different company comes out.

But, but, but. That’s, these are rare cases, but, but there are significant adjustments in their business plan in that period of time. 

Philip Hemme: I find that’s a good point. I think that’s, I mean, it comes also a bit with experience, but it’s, I think that’s, I mean, you, we, we nailed right away, but I think that’s one of the hardest thing to do as, as an entrepreneur.

Yeah. Because, I mean, as you said, you have to be super focused, also not change direction too often, but at the same time being flexible and you need to be extremely convinced by what you’re building, but at the same time being open. I find this like, it’s a very fine line and it’s very individual depending on the market, the technology, even the team synergies, the whatever, even the location you are at or what access to capital you have.

I mean, it’s Very tricky. 

Jens Nielsen: Exactly. And all these factors are in play along the way. And that’s also why it’s, it’s important to you know, it’s important to take in advice, but you will have to be able to synthesize based on this advice because sometimes that advice will be. Orthogonal you know, and of course it’s great if you begin to get advice that are all aligning and you can, you know, follow that.

But sometimes you will have to make decisions and, and stick to them. And that is tough. That is tough. That’s also why companies sometimes fail because you make the wrong decision. But, but then you learn from that. You learn in that process and, and you, you can hopefully go back and reflect on, you know, what, what, what was that decision I made and, and based on what assumptions was it.

[00:19:34] Listen to your gut

Philip Hemme: In the learning process. Yeah. It reminds me that we added on episode three at the founder of Kurovac, Ingmar, and we talked really from, from how we started. As a PhD in the lab in the garage to building a basically was at some point was 20 billion market cap company pretty crazy At the beginning and I think one thing he said which one of the takeaway which volume was extremely difficult in this kind of decision as well, especially coming from a scientific or engineering mindset is like a lot of these decisions are basically gut feeling 

you cannot 

Put the table plus minus and it’s not obvious.

Oh there is like and And somehow, I mean, he took a lot of decisions, basically gut feeling of, okay, this is probably the right decision. And there’s also probably, okay, I just feel it. I’m convinced, but that makes it even harder decisions. 

Jens Nielsen: Yes, but that is true.

Philip Hemme: I think as a scientist, that’s very important.

Yeah. Just first to be aware of this, and then at some point taking the decision, sticking to it, evaluating. Whatever, six, nine months later, learning from it, iterating. I mean, we didn’t talk about iteration with Arno. 

Jens Nielsen: No, but, but let’s not forget that, you know, what we talk about gut feelings is that there is it is, you know, as, as the expression says, you know, it’s, it’s, it’s a gut feeling as such.

But let’s, let’s not forget that there’s a very close communication between the brain and the gut. And and, and what. mainly gut feelings are based on is actually a lot of information that we have obtained, and that’s what I said, you know, ask people and, and we, based on that, we synthesize that, but maybe why we call it gut feeling is that it’s synthesized to some extent unconsciously, by us, but it is based on a lot of data and a lot of information that we have collected.

And I think that’s important. So I just want to emphasize that because I think it’s important when you are an entrepreneur at an early stage, that you’re very open minded and ask people, talk to a lot of people and get that information in, because I think it influences you more than sometimes you actually know it does.

Philip Hemme: But yeah, I think, but yeah, I don’t know if I see the flip side also is like, you can spend a lot of time listening, listening, listening at some point, also need to make a decision. 

Jens Nielsen: Absolutely. Of course. Of course. Yes. Yeah. 

Philip Hemme: But also really, especially at the beginning. Yeah. Even if I reflect on the really beginning of, of lab biotech, which was basically 10 years ago.

We had no idea what we were doing. You know, at 23 years old. And yeah. You hear a lot of things and sometimes there are things contradicting, but at some point you also need to take a decision. No, I agree. I agree. Be laser focused. Yeah. Okay, so that’s, that’s, that’s good. 

[00:22:38] The importance of network

Philip Hemme: If we, what else did you see, what, what works or what didn’t work from a, maybe even more from a, let’s say, interaction between the different partners.

Because I think at the end, you’re also really. I mean, good. And there’s a big focus on this, of connecting with these academics with potential, let’s say, business partners or teams with VCs and kind of let everyone talk together and have an overlap. But what did you see there? Like, what kind of, 

what works?

What doesn’t? 

Jens Nielsen: So, so net, network is extremely important, to, to use in that, in that process. I, I think, you know, this is, this is the biggest challenge that you, that, that I would say that I faced as you know, being in academia is that it’s relatively still isolated. You know, you, you, you may, begin to have contacts to some investors that you talk to and, and that, that opens more doors and so on.

Or maybe you talk to people in industry and so on, but, but it is in many places, unless of course you are in, in a very, very strong ecosystem like Boston that is extremely well connected and so on. But that I think is the biggest challenge. And that’s what we are trying to overcome at BII to be that place.

And it’s, it’s, it’s harder than it sounds, but but, but to be that place where we can connect people, because it is important that you, you, you, before you get the right, yes, that you get some nose and you will get a lot of nose, as, as all entrepreneurs have experienced, but as I said, you, As a scientist, scientists are not, you know, we’re used to get rejections of our papers and so on, and grant proposals.

So, so the no is not necessarily the biggest problem. The biggest problem is that it’s something new. And, and and as we talked a little bit about that, being able to take in the advice in new areas, but actually make decisions and, and move on. So, but that’s, that’s what I think we, you know, we’re good at at BII that is to bring people into an environment where, where there is this connection.

And of course my staff is extremely helpful also for, for the individual companies on these travels. It, you know, it, it’s, it’s hard to give to, to, to, to kind of create, I think a winning formula to be quite honest, because I think it is to a large, large extent the people’s game. You know, you need to find people that can really support these cases and, and have a large network and can actually activate that network to the benefit of, of those who are helping.

And, and, and that’s You know, as I said, you know, this sounds good, but if I was going to write down a recipe for how to do that, then I think my, my, my only key recommendation will be to find the right people to help you to work with this. 

Philip Hemme: Yeah, you know, it’s a, it’s a good feeling to, to people. I mean, deep down, I think that’s also maybe a parallel with tech, but you know, in tech we say like, ideas are commodity, execution is everything.

Say people and execution is everything. I feel like in science is always, of course, you have something a bit more palpable with patterns or results, data, but still people are, I mean, she’s going to start about at least 50 percent of whatever the success factors are, but that’s also, people are also very hard to, let’s say change or adapt or changing a mindset of academic lab to make it more entrepreneurial.

I mean, this takes also. I mean, yeah, 

Jens Nielsen: but yeah, and I, you know, this is a, this is a good point, you know, at, at coming back and talking a little bit about culture in my research group, because this is also what I really tried to install in the group, you know running a very large group with a lot of ambitious people that are each having, you know, individual career aspiration.

It’s, it’s almost. A given, you know, design for disaster in terms of conflicts among people, right? Because they’re all but, but, but if you create a culture where, you know, everyone feels that they can safely share things, you know, I think I managed to ensure, first of all, collaboration, but, but that also taught me that this really brings in much more innovation and I’m talking innovation both in terms of new scientific projects, but also about addressing things.

And so, so, you know, and I, and, and that’s what I would like. You know, I, I think it’s important also for entrepreneurship, again, this, yeah, realizing that it’s not, it’s, you know, it’s a team effort and you need to be open for listening to, to the team you, as we talked about several times, you need to make decisions every now and then.

But, but this about you know, Creating a, a, a culture where everyone feels that they are on the same side and that they’re actually working and, and as I said, that’s tougher in a research group, but if you manage to do that in a research group, you actually also create an environment that I think is very open for innovation, going forward.

[00:28:05] Entrepreneurial mindset recipe

Philip Hemme: I think that’s, that’s, I wanted to touch this on like really those entrepreneurial mindsets inside academia or research that I think, I mean, that’s. One thing, what you describe, and I heard it from, I mean, from my best friend was, was a PhD at ULAB, so I kind of heard it also from the inside of the different thing, but I think it works, or at least it seemed to have worked, works really well, and it’s, I can see it very similar to some of the biggest bio entrepreneur labs, let’s say, Church lab, Langer lab at MIT, or some Boston or San Francisco based labs.

And I think this, I mean, this seems to, I don’t know what’s the recipe, but it seems to work that there is a certain kind of cultures that you can set up and it will lead to more innovation and yeah, entrepreneur. But I guess it’s very related to, I guess the people and especially to the PI. This culture has to be.

Yes, but I guess you can. I mean, I can imagine if you’re, I mean, also I’m trying to, in fact, but if you’re a pi, I, I was in some labs where the pi was, was very fundamental. Mm-Hmm. , I think it would be impossible for a pi like this to change and become more, or like, like become more like your lab or very entrepr.

I don’t think there’s really a, a real recipe there, like, what do you, you know, do you think let’s say PIs can become more, like more entrepreneur? 

Jens Nielsen: I think, what, what is of course important for creating an entrepreneurial culture in a lab is, is the open mindedness, right? Because as I said, often a scientific discovery, you know may.

you know, may find a completely different application than you, than you originally thought of. And if your eyes are suddenly open for that, you should, of course, be, be open minded, as I said, to kind of pursue that. And, and, and let’s just face it that most of the big discoveries in terms of, you know, startups and so on in life science and biotech have actually come out of basic research.

So it’s not. Then, you know, that, that I think, the, the, the researchers that are kind of really looking deep down to unravel some specific mechanisms in, in biology cannot lead to startups. But I think what distinguishes a lab, let’s have two parallel labs one from the other where innovation will come out and startups will come out is, is the open mind is to look at applications for that science.

And, and, and that’s more difficult because you know, how do we do that and how do we, and I think. It is by through role models and, and illustration of, of this by having more role models. And that I think is what actually eventually drove development of the Boston ecosystem. 

[00:31:08] Role models

Philip Hemme: Talking about role models, for you, what were, what were your role models, I mean?

I mean, you’re quite close to Bob Lange, it’s pretty well as a reference in this space. 

Jens Nielsen: Yes you know, I, I lucky wise had had many role models in my career early on. You know, various professors, including my own mentor and advisor was, was a, you know, very strong professor and, and did excellent research and so on.

So, but, and I think that’s the point also that you develop different role models, but what was important for me in my career was that I spent a sabbatical early on in my career in Boston and, and, and this, first of all, being more ambitious reaching, you know, higher levels, you could say in what you can aspire to, to do as a researcher was one thing I learned, but also this, that if, you know, That if you want it to find application, you should not necessarily expect anyone else to do it.

It’s not just publishing papers and then hope something will happen. You actually have to drive this yourself also. So this is what it taught me. And, and and so that I think is important to, to, to get that. 

Philip Hemme: I like that. I like this, like, lesson of what works on, I mean, if we sent. If I summarize what we just discussed on like focusing on the people, entrepreneurial mindsets, and really, yeah, being, being open, like driving the innovation.

[00:32:38] Interaction with industry

Philip Hemme: I think that’s, yeah, as a scientist driving it, not just doing the research. I mean, one point that I had here, which I think is also super important when you look from a, let’s say from a, when you talk to a biotech or life science industry, and when you look at all the products that are on the market, most of them.

came somehow through, through academia. And so most of them, I mean, in pharma, maybe some internal pharma, even though I mean, over the last 20 years, it’s going down and down. So it’s much more academia that translate to biotech. 

So I think, I mean, it’s key, 

it’s definitely key there. And I think definitely that’s one thing that, I mean, at least what I’ve seen what BIA is addressing is that the translation of science, let’s say, especially here, Copenhagen, Denmark, there’s much more translation that can be done to industry.

What, where, where, like, where is this interaction with industry? Like, I mean, except what we, everything we discussed, do you see anything else there where, where something can be more done? 

Jens Nielsen: Like, Yeah, this is, this is a very interesting discussion and something that I’m quite passionate about also, you know, and, and I think, let me wind back and, and say very early on in my career, I was fortunate to work very closely with industry.

on a research project. And, and, and the, the interesting thing was that they really asked, me and my group to, you know, to do some more fundamental characterization of a process that they were very interested in. And, and so, so, and because they needed to have this deeper knowledge and so on. So it was a perfect, you could say, synergy.

We could do research and get, you know, and public publish the results and so on. But they got more knowledge that they felt that they could use to improve their process and so on. And so what did. What this taught me was well, first of all, sometimes, you know, this idea that industry will just ask you to optimize their process.

This is absolutely not the case. Normally, industry is ask asking in, in research collaboration actually to do some more fundamental work because they need to have that covered in order to, you know, have a better understanding of their product. Or testing new ideas. Or testing new ideas or whatever.

They wouldn’t do anything. Exactly. So, so. So, so it, it showed me clearly from, from earlier on in my career that there can be a perfect synergy between, you know, doing research. But what it also taught me was that through this collaboration, I, you know, a number of other interesting research questions appeared that were, you know, that were kind of really interesting to pursue and more fundamental.

They of course now had relevance for the industry. Okay, so that became a perfect symbiotic so I could make even better research proposal to get funding for this because, you know, I actually knew that this was a real problem and this was interesting for industry. Secondly, it allowed me to, to bring many of these cases into education.

And so when I was teaching in the classroom, I could give examples. And of course this was at, at the Technical University of Denmark, so it is an engineering school and most of the engineering students are interested in, in real life applications and so on. But it resulted in that, you know, many of the best students came.

to me to do their, their, their, their project, their research projects later on, also as PhD students and so on. So I was fortunate at an early stage that I wrote, got really good students coming to me because they were interested in this. So I think this was, you know, and as I often say, and, and, and, and this will not, this is one example when, but, but I think It, it, it at least showcases that there is not a conflict between doing something that has applications and value for the society, and in this case industry, and doing good education and research on the other side.

I, I sometimes also use another, you know, way of explaining this, you know, as a researcher, you, you, you know, you can do, you know, basically anything, but, but maybe. We could begin to do research in areas that potentially would have some application. And I do understand that there’s certain research questions, but I, but actually I think it’s very, very small, even, you know, when you’re studying evolution, you learn some things that can, can, you know, and that would many people would characterize as very basic research, but, but you will learn some, maybe some techniques and stuff that will come out of this that you could find application for.

So I. I actually, I’m convinced that any branch of science, any activity of science lead to some new knowledge that could find application. But it’s this openness, as we talked about before, builds on everything else. 

Philip Hemme: Yeah, yeah. That’s why it’s so hard to make. I like your point of the synergies because it was something I’m always wondering and I think it gets down rapidly to that of what’s the role of academia and why, I mean.

What’s the use of public money in academia, whatever, if you spend 2 3 percent of your GDP. How much should be really, I’d say, I mean, if you try to split how much should be fundamental, how much should serve the industry, but I like in your case that if you look at it more synergetically, you see that it’s not one versus the other, it’s together and that’s 3 percent of the GDP will contribute in both ways and that the more you do on both sides, I mean that everything fits together.

I like that a lot. Yeah, 

Jens Nielsen: I, you know, I, I, I, I’m still, especially in biology and life sciences. Exactly. I’m still a strong believer in the academic freedom of course and, and, and, and, but, but let’s, you know, if, if I’m interesting also in the historical development of universities, I mean, you had the, the old classical universities that the, the old schools, they, you know, they was in theology and medicine, then engineering came and so on.

But, but really a transformative university was the Humboldt University. That was the first to say, let’s do research so we can do research based education, right? Because the training of people was the original foundation of a university. So suddenly then research and education became the two pillars of universities.

It was late 1800s. That was the late 1800s, exactly. And that was then picked up by Harvard, Yale, and many of the other, Princeton and so on in the U. S. They looked at Humboldt as a role model actually for doing this. Today, of course, exactly as you were saying, the, the, you know, so many taxpayers money are going into, into research at the universities that, that, that not just for education but also for research, that they, they are beginning to ask, what do we get for this, right?

And so that’s why universities are beginning to, to build this third leg that you can call innovation, where you do expect that, that some of it you know, comes back. Again, of course. Let’s not forget a big part does come back because many of the candidates that are coming out for, you know, graduates, PhDs and so on are taking jobs in industry.

So there is a link there, but can we do even more? And, and, I am certain we can in, in Europe, it’s, you know, we, now we talk a little bit about Denmark as a yes, we could do many more spin outs in Denmark. I’m pretty sure, but it’s, it’s a European difference compared with the U. S. 

Philip Hemme: I was about to say, I mean, I like the perspective, I mean, I like the looking back at the perspective at also what works now, I mean, what leads me to wonder, I mean, in Europe, if it’s so clear that there is synergies.

And that, let’s say, I mean, when you look at Boston, it’s crazy how well it works. Like let’s say, why is it not happening more in Europe? I mean, some in Europe are better than others. I mean, I think even you mentioned Cambridge, Oxford, Oxbridge, or Imperial or London. I mean, they’re super good. I think in Belgium, actually, VIB is very good as well.

Denmark, I would say, is actually leading. Some countries are probably less good, but why, why is it not more if it’s, if it’s so good? Thanks. Yeah, if it’s, let’s say if it’s so clear, why is it not more? I mean, I have some, some ideas of what’s, what’s the, 

Jens Nielsen: you know, I, I think it. It did become a little bit of a tradition, right, that you had segmented, you still talk about professors sitting in their ivory towers and so, you know, that, that kind of mental thing is still there and, and where let’s not forget you know, becoming a successful academics is, is extremely hard work and, and it requires a lot of time and effort and so on.

And so as I often say, you know most academics are very, you know, incentive driven. And so let’s not forget what incentives we have built that’s publications as a key incentive driver, right? And then many professors are hired this university say, yeah, we should also be able to teach, but but, but normally that’s harder to evaluate and so on.

So, but, so we have built incentives. For the, for professors to focus on that. If we, if we started to build in other incentives, I’m pretty sure we would, we would get for what how we decided. 

Philip Hemme: Do you see that changing? I mean, let’s say of money ways I begin to see that change? 

Jens Nielsen: Yes, absolutely. For sure. So, so I, I think, and also.

I see many more young academics being also interested in, in their research having impact and, and, and, and having impact as I said, it’s not just meaning a high impact publication, but actually having impact for society. So, yeah. 

[00:43:07] Results of BII

Philip Hemme: That’s good. Yeah, I was thinking back also from your answer, back to what you said about people, and I think also, I mean, for things to change, people to change, sometimes takes a bit more time or some generation thing as well, to, to get there, even having him.

Mm hmm. Even if you have the willingness, even if you design the system a bit differently, still, I mean, it’s heavy stuff and it takes whatever, I mean, it’s always in decades that you really see the change, I guess, you know, yeah, that’s good. It’s good. It’s I think, I mean, we, we covered, I think, pretty, yeah, pretty broadly there.

On, on the, I mean, on re translation, maybe if we go to, to BII a bit more specifically, I mean, you talked a bit about it as well, I, I think, I mean, I really like the, the model and the problem you’re trying to solve of really, like, enhancing this translation and being there, supporting. Maybe, I mean, I will, I will present it in an intro, but maybe you can talk maybe on, on the results.

I mean, it’s been now a bit more than five years. What’s the results so far, like compared to the initial hypothesis? Yeah. 

Jens Nielsen: Oh okay. I think that there we, you know, superseded expectations but, you know, it’s also hard to have expectations and, and set the right expectations when you start something that is so new.

So, but when you, when you build something as massively as BII, which is, you know, we are supported by almost 500 billion euros from the Nobel Nautics Foundation over a 10 year period. So that’s a massive, you know, investment you could say in building a new ecosystem, but still, you know, what we are, you know, what I’m impressed about what we have achieved, you know, we’ve, we’ve taken in now, we’ll take in a new cohort of companies here on March 1st, but then we will pass a hundred companies that we have supported so far.

Many of these companies are still in our program. So that means that they. You know, they’re still in fundraising mode, but still our companies and we have supported these companies with about 50 million euros in, in, in, in, you know, convertible loans and, and grants and so on. But they have raised about 500 million euros in, in investments and, and, and grants and so on.

So, you know we have really managed to, to, to gear the activities quite significantly. Okay. 

Philip Hemme: That’s fine. Yeah. I, yeah, you have almost 10, let’s say 10 X. Yes. Yeah. Multiple. Yeah. And then the, can you talk a bit specifically on the program of how, like basically what the companies get and what’s, yeah, let’s say the program, maybe a short summary of that.

Jens Nielsen: Yes. Of course. Yeah. So, so the, so, so we. If we go back to the, a little bit, what we talked about, the fundamental challenge you have as you’re sitting as a, as a young academic, and you have an idea and you, you need to build that business plan. And that’s really what we designed it out from because we, the, the, the reason why we do not see so many startups here compared with Boston is because we don’t have enough entrepreneurs that understand and can kind of go around at the universities and pull out.

Cases and make a business case and put proposals to investors. So. We see it a little bit as a long term objective of really training and educating more entrepreneurs in the space. And so therefore we designed this program that we call VentureLab, where exactly we take young academics, we take them into this program, we let them drive the process because we want them to be educated and trained in doing a business case.

And then they go through a 12 months, we give them half a million euro in capital. And then they get 12 months to, to go through, you know, how to build a business plan. Who are your customers? What are the regulatory constraints we talked about? You know, what are all the other, how do you secure IP and stuff like that?

All elements 

Philip Hemme: doing it and doing it, 

Jens Nielsen: not just talking about it, but actually doing it on your case, each individual case, then maybe also important is that we take in. Companies in, in cohorts, so, so, you know, they, they, they, there’s both some peer learning, also a little bit of peer competition, of course, but, but, but that works fine.

And, and, and, and so a cohort of companies go through this 12 months, it’s a little bit like going to class, but, but you’re really working on your own case that you’re maturing. 

Philip Hemme: And they have access to, you have labs here. We have labs. 3, 500 square meters. 

Jens Nielsen: Yes, exactly. We have labs, fully functioning labs.

Philip Hemme: So they’re all moving in office and lab and they are zero space. 

Jens Nielsen: Exactly. And we have created large investor network. We talked about the relevance of networks also, industry networks, you know, so, so we can facilitate that they can talk to people. Yeah. 

Philip Hemme: That’s great. 

I think. It’s, it’s one, it’s one thing where you are really, where there’s really the, the bridge of this like pre seed money that you invest, I mean.

To be honest, Devin, two years ago, three years ago, I was talking, looking for building an X Venture, talking with some, some scientists from DTU, we were talking, I was talking with BII actually, with some people from your team, I think this like 500, 000 kind of precede as in, okay, you have ID patent and then you can work for Torment.

I mean, that’s, you can really, like, it really can bridge between the academic lab to them being ready to raise a seed. Yes. In whatever level of seed or series they’re even directly, but you can really cross that bridge. And sometimes that’s, especially when you come for a committee, especially on deep science and reading, kind of, let’s say, advance on the MVPs, if that’s, What you need, I don’t know if you need 500k, I mean, if I look at more like YC or Entrepreneur First, which I would guess are in a similar stage as well, it’s more around 100k, you get 4th, but at the same time, usually they don’t really get 2 MVPs, they need additional cash there, so, but I think this is really crucial for getting them started, because if you don’t get This step, 

then 

Jens Nielsen: exactly, exactly.

Philip Hemme: Nothing happens, but yeah, and, and, 

Jens Nielsen: you know, it serves, let me just come back to, you call it investment. And let me come back to that, but I mean, it serves the purpose of course, of building companies. But I think it’s important to emphasize this, that I mentioned. That we see it really as an objective also of training people.

And we are of course very happy that the companies actually succeed, raise money, move on and hopefully, you know, bring products to the market that address unmet needs and all these kind of things. But. But, but we, and the, the reason why I’m emphasizing this about training people is that we’ve strongly believe in the idea of it’s possible to take academics and train them to become entrepreneurs.

And I, and for you coming from tech, you say, yes, what’s the problem with this? But I can tell you that many investors in life science in Europe. Do not believe in this this idea there. The sentiment for a long time has been lit as soon as we invest in the companies, let’s get rid of the founders. And, and, and we look at this very differently.

And that brings me also to the way we support them. We, we don’t see it as investment. Well, first of all, because we. Well, we can, we can take that, we can take this luxurious position because we get the money to, to support us as charitable financing from the No One Hurts Foundation, so we don’t have to give the money back but we we give it as convertible loans.

at very founder friendly condition. So therefore the founders feel that we are standing on their side also on the travel along the way. And I think that’s an important element also in, in, in, in this for they, they really trust us when they, when they have to do that jump.

Philip Hemme: I like that. I mean, and one thing that is, that I’m pretty impressed by from a BIA point of view is really the, also the team, internal team you were able to attract. Yourself, but also. I mean, I know about the CBO, Christian, and the Symbioteam and some others, I mean, which I think is one of the things that I always see in Europe, which is really liking all these, like, bio incubators and programs and, and then the people react.

It’s not like, it’s not people who have really, really know what they’re doing versus we had, we had Johannes from LabCentral BioLabs on the, on the, on the, on the podcast. I know he was also in discussions with Novo, but I mean, I think there is some parallels there where someone really experienced, has built companies, has VC experience.

And at the end, I mean, what we also discussed, people, everything, and yes, I mean, in innovation especially, it’s all about people and here, I mean, when the people come, it’s people, people, building companies together, and I think that’s, yeah, I mean, and you take YCEOEF, they have amazing people being able to try, which I think is a, is a key, I mean, yeah, 

I think it’s key.

Jens Nielsen: Yeah. No, I agree. I agree. And you know, I, I think also that’s of course an interesting observation over the years because I think in the beginning we were perceived a little bit as, you know, we would help some of these young startups and so on and, and that’s fine and by the, you know, traditional biotech environment in Denmark, you know but, but now we have grown to a situation where everyone is taking us very serious.

So we have all the experienced biotech people, you know, coming in and helping our companies and so on. And so in that sense. We have also really become that center for, for this. And, and that, that of course, I’m very proud of and happy to see that we have succeeded in. So it doesn’t become, you know, something parallel to, to, to what was there already before.

[00:54:14] How to be cost-effective in helping startups

Philip Hemme: Yeah. I know having quite a few of your entrepreneur residents or like mentors, which is. Basically all the best, let’s say, especially in Copenhagen or Danish, like, really, really impressive. I mean, one, one thing that I, I mean, it sounds like you’re really doing it. Yeah. Seems to me of results and works.

I think if I play a bit devil’s advocate, what you mentioned of like, we have the luxury of the novel. I mean, it’s a big luxury, but it’s also. I guess it makes it easier, like, if you have, you know, whatever, 500 over 10 years, that’s 50 million a year that you can invest slash burn, but invest and spend, I mean, that’s, makes everything easier, but I mean, still need to spend it well and et cetera, but.

Thinking of like, yeah, I mean, but it makes sense, like, 

but it’s. 

Jens Nielsen: I think a very, you know, what you may be alluding to here, you know, how could we replicate this without spending that much money somewhere else? And that I think would be key learnings, of course, from, from here, because, you know, let’s say we that that’s fantastic if we can build a strong ecosystem here in Copenhagen.

And that’s of course good for Copenhagen and Denmark and so on. But how do we help you know Berlin you know Heidelberg, Paris, other places in Europe where, where this also Would work, but, but, but do it more cost effectively because the most other countries don’t have a charitable organization like the Nova Nordisk Foundation to, to, to help them.

And, and this is something we are looking into. How can we, and, and maybe the models would be slightly different but still with the same goal in mind. And how can we then unleash, you know, maybe not fully commercial kind of investment type, but, but, but, you know, strategic kind of investment type to, to drive again as to to begin to do this, of course.

So, so this, but, but it’s, it’s difficult, you know, as I sometimes say it’s, it’s interesting when we have international visitors, you know, from France, Germany, England, and other places, you know, they come, they get super excited and they all say, Oh, we would like to have a BII until I tell them what it costs.

Then then, then, then the the interest drops a little bit because but But I think, you know, we will be able to figure this out and this is something we are working on right 

now. 

Philip Hemme: I guess that’s, maybe that’s what, actually, I always thought there was something puzzling with that thing that’s probably the biggest limiting factor as in access to capital, capital available, slash, let’s say, the return on capital compared to other options.

Because, I mean, I mean, but, but deep down, I mean, everything we discussed before as well, if you don’t have. enough capital, then you don’t have the right people. Yes. You cannot invest enough to seed the companies, et cetera. And then everything breaks. I mean, because at the end I mean, the formula you need to execute and find the right people and find the companies, everything, but still, if you don’t have enough capital to execute everything, like in any startup, if you don’t raise it enough, you can execute and everything.

[00:57:56] Novo Nordisk

Philip Hemme: Yeah. That’s a, that’s a good, yeah. I was looking, if we zoom out on the Novo Nordisk Foundation, because I think that’s one thing that, that, that strikes me here in, let’s say, in Denmark. I mean, I was in this building actually, it was like seven years ago, and we did like a documentary about biotech in Denmark.

And of course I knew Novo Nordisk, I knew Neuroscience, but then when I arrived here, I was like, okay, actually, everyone basically, or almost everyone, receives money from Novo somehow. Even like the research institutes are basically Granted or built even by, I mean, they have the Novo name here is basically one, one of the institutes.

And then the Novo, let’s say Novo holding as an investment vehicle, which is I think even in one of the biggest, let’s say, evergreen, such private equity fund in life sense, even globally. Yeah. Which is crazy. And people don’t, even in the industry, don’t know how, how big and how large it is. Which I find quite fascinating with the no foundation is that there’s not that many like.

drawbacks or like flip sides. If I looked into a critics, I mean, there’s a few, but it sounds like the ideal thing of super successful company, the word artist, which also, I mean, there’s not that many scandals behind in, let’s say, in a pharma standard. I’m like doing, I think, an amazing products. I mean, right now, especially it’s going crazy with, with, with JV, but amazing companies with a foundation that controls the company was.

Dividends and profits coming in, reinvested. I mean, sounds like the ideal thing. I was obviously looking into like, okay, what’s, what’s like, sounds too good to be is that, is that so? Actually, what’s your like 

opinion? 

Jens Nielsen: Yeah, you know, yeah, it is almost too good to be true and to some extent, but, but, you know, of course there is but that’s also why the organization, I think it, and I think that same culture was, you know, instilled in, in BII that we need to have a certain humbleness, we need to be aware of the, You know, the big impact that we actually have and therefore the way we, we do things and set up things has to be in line with this.

But of course, when, when you become so large and dominant, you know it, it does have consequences. I mean, if you look at the Danish, research environment and, and, and really performed a deep dive, I’m pretty sure that you will find that it, it, it’s, you know, to some extent skewed on what Nobel Nordisk Foundation is supporting.

Philip Hemme: One of the big critique is that most of the life science in Denmark is very oriented towards Beginning to, you know, all this 

Jens Nielsen: Lucky wise, you know, they still give a lot of money, you know, for open ended research and so on. But you could, you know, you could argue is, is that so problematic? I mean, after all, we are a small country, so do we need to be strong in all areas of science?

Is it maybe fine that we begin to stick up really in certain areas of, of life science? And I think You know, if we it’s kind of interesting if we look at our portfolio of companies in, and let’s take therapeutics because this is what most people can relate to in biotech. You know, of course we have a lot of oncology companies not surprisingly, any accelerator around the world will have that.

We have some of them that are strong. Some of them have raised a lot of money. There’s also a lot of VC investments in that space. So, you know, this is fine. Do, do any of them stick out compared with, you know, what you have in Boston and other places? I, I don’t know, I don’t know enough about it to, to be quite honest, but I, I doubt they would.

But they’re, they’re good companies and I’m sure they will be successful and, and hopefully many of them will develop new cancer medicines. What is interesting is that we, we probably is one of the accelerators slash incubators in the world that has the most companies in metabolic diseases, compared with our portfolio and even addressing metabolic diseases with peptide based pharmaceuticals.

You know, and, and, and, and this is, this is not by us selecting for that because we always select the best companies in every cohort we come in. But this is the outcome of that, you know, you can say over here, select, and then you suddenly, and then you look at that, and then You know, I say, of course, because this is where we have a very strong research base in Denmark.

And so, I think that is a reflection of, of what we just talked about, that, that Novo Nordisk Foundation has, and probably also Novo Nordisk themselves, sponsored and supported research activities in that areas over, you know, tens of years. And You know, there we are building some very, very strong and interesting companies that I think are more unique compared with what you find many other places.

And, and is that a strength? Yeah, I think it is. So, you know, one can, of course, be, be raised criticism around it. But, but as, as long as the foundation is, you know, thoughtful and careful about its supportive activities, I think it is a strength. 

Philip Hemme: Yeah. I don’t, 

I don’t, I really don’t see that many setbacks.

And even, I mean, what we discussed before, I mean, the synergies you have when you specialize a bit more in one space from the academia to even be a, to industry and the synergies you can create when people work in a similar space also, I mean, you’ve got a lot of great advantages and even, and the people working in similar space can transition from one space to one, one company to another.

I mean, yeah. Yeah. Now that’s, that’s, that’s remarkable. I mean, one, one thing that really I find remarkable and you mentioned the humbleness is, is really this that I see, I saw here, especially it’s really struck me, especially in Denmark and Copenhagen where basically the money comes from basically one organization, most of the money comes from one organization, everyone goes to pretty much the same university to DTU or business school or one program, but still like everyone is very humble.

I mean, it comes to the root culture, I think in the Danish culture. And I think everyone kind of works together. You compare to other where people are always fighting. I mean, Tim Harris, people are always fighting and one against another and like not working together. And working in a smart way, I don’t know, I, I feel like it’s always quite, it’s quite remarkable that it functions, it functions so long, and especially when you add so much cash.

Because usually the more cash you put on the, any balance, the more you have jealousy and fighting and stuff. And I guess it’s not ideal, and there are some of this, but still, I find it remarkable. And at the end, if you look from a long term view, I mean, the results is also, yeah, amazing. 

Jens Nielsen: Yeah. Yeah. It’s good reflections.

You know, there is a certain element, of course, in, in, in Danish culture that, that buys into this. But I think it’s also to some extent from, from the. The Noble Nordisk company culture. So, so if, if one reads about the early stage and the early foundation Noble Nordisk as a company were among the first to get, you know.

to, to, for example, begin to give maternity rights and maternity payments for, for women. You know, we’re talking about, you know, very early stage, right? So they were always very supportive of their employees. It was, it was voted for many years, the best place to work in Denmark. And, and, and building as a bigger and bigger company, of course, that becomes a benchmark.

And I think many other companies. So I, I think the impact is more than just the money. It’s really also was a culture that was established and that was pioneering of the founders of the company to, to actually. To think around this, but that’s, you know, we talked about it several time today. It’s more obvious that, that people isn’t, is the key asset you have.

But if you go back to that time to see that I think was visionary and, and I, I, I think that that is a part to play. But Danish culture is, is also, you know, we have this, you know. Something that I’m not a great fan of that they call it yendolo. I mean, it’s hard to explain that translate directly, but you know, it’s basically just saying that you, you, you know, you should not stick your neck out too far, you know, you should not, you should not break and so on.

Exactly. And that is so. And, and the reason why I, I, I am not to, you know, I don’t think that’s a very strong trait because it does prevent you know, people to take leadership sometimes and then, you know, and, and set ambitious goals and so on, but it, of course, because it is there and unspoken off, it also means that you as a leader need to make sure that, you know, you kind of embrace your team and make sure that everybody you know, follows you.

And, and so, yeah. 

Philip Hemme: No, that’s great. 

I think, yeah, I mean, if I, yeah, I like that. If I, if I look also, one thing even for our audience to understand as well, I mean, Denmark, think about a country, but it’s basically not even 6 million people. So it’s like half of Paris. Yeah. So it’s like, also, I mean, the, the scale is pretty, it’s smaller, but even compared to a city, I mean, it makes different dynamics, but still, it’s still, I mean, 6 million starts to be.

Quite sizable as well. And I mean, I think the second also, let’s say, you know,

thing to put in perspective is also there’s some, let’s say, luck, especially from the Nordic side of, I mean, diabetes, it was impossible to predict that it would grow that big. I mean, even though, of course, they, they, I mean, they executed brilliantly, I mean, and keeping a market leadership and like at that scale is insane.

Yeah. But still, there was some, like, let’s say luck of, no, it’s a market that’s exploded and when your market explodes, of course, your company explodes with it. But I mean, that’s, as everything in entrepreneurship, there’s a 

big luck component as well. 

Jens Nielsen: Of course, there was a, you know, being a company that is, that has as a main business focus on, you know, diabetes.

And yes, of course, because of obesity epidemics, you know, diabetes has also increased as a disease. So, so the market has increased, but, but suddenly being able to go from there to begin to address the maybe more underlying problem, obesity and then that you have drugs that can actually do that, that is, of course, a little element of luck in, in, in that.

And, but, but being able, you know, as always luck is, is good. You know, we have a, an expression it’s an old expression from a Danish entrepreneur, you know, that, that, that luck favors the prepared one, right. And and, you know, if you were not able to, to use it. Then you know, then, then you can be lucky.

But if you were not prepared for using that luck, then yeah. 

Philip Hemme: Yeah. I mean, I think luck is not, I mean, it’s, it’s part of every entrepreneur journey. I mean, I was listening to one entrepreneur, you were saying that basically 50, oh, more than 50% of the success was luck. Yeah. And it doesn’t mean that it’s necessarily like negative, it’s just more accepting that it’s part of it.

But then. Even if an average is 50 percent sometimes, can be even more, I mean, yeah, even that is, I think, was, what was the market, I looked, was like, 10x over, what is it, 20 or 30 years, I mean, this is, for a, market growth is insane, yeah, yeah, so, just from a, being in the right space, and being already there, and then the market explodes, it’s, yeah, I mean, there’s a big luck component.

It doesn’t, it still doesn’t, doesn’t like withdraw the merit of being able to capture that luck and execute on it. Comes down to it. That’s great. 

[01:11:07] Closing the lab

Philip Hemme: Maybe I want to finish more on the, like a bit more on, on yourself. I mean, you talked a bit about the mentor and everything we discussed, there’s definitely a lot of your personal lessons.

I want to finish on one thing that quite struck me when you joined BII, which was, okay, well, you joined BII as CEO and you’re still keeping professorship, but your lab, basically, you were kind of continuing what was ongoing with, I think now is pretty much closed, I’d say closed down or not running, which I found.

Like, I’d say quite surprising, slash, I don’t have many other examples of someone, I mean, your library, you had basically 75 people at some point, and then, whatever, each index, 150 or something, and like, really, I mean, building, as you said, so difficult as well to BPI at that level, and then, such a big transition, I, I, I mean, I was quite like, wow, like, surprised, and I don’t know many, but can you?

Reflect on it, maybe now, five years later, of like, how you took this decision, how it went, like, Well, 

Jens Nielsen: let me, let me first start by saying I have not regretted it. But but it, you know, it was a big decision. Of course it was. You know, I, as, as I often say, you know, I, I could happily have been professor until my retirement.

And, and, you know, I thought it was fantastic to to engage you know, and, and, and train young people. Doing research and work with them. That’s something I get a lot of energy out of, you know, I found it super exciting also to take some of the research and make spin out companies and so on. So all these things, you know.

But on the other hand, when this opportunity arose, you know, they say, okay, you could take leadership of BII and, and build this. And then as I know, the ambition level from the New Religious Foundation can be high, you know, that, that was a golden opportunity to, to focus a hundred percent on something that I’m passionate about.

And that is, you could say. Translating science from universities into, you know, startups that are viable and eventually, you know, address our many needs and so on. And yes, I could continue doing that in my research group, but here I could do it on a bigger scale and help many others to do that. And, you know, and it, it, it’s, as I said, yeah, it was a big decision, but, but maybe also published more than 800 papers at that time and educated, you know now my, my last students are graduating, you know, more than a hundred BSD students and so on.

And, and, and of course, if I could, I could do more of that, but, but, you know, I, I kind of had fulfilled, you know, some, some of the objectives you could, you could aim for, you know, probably for a lifetime. So, and if I was going to do it, do something else, it would be something like this. So, all this went into my mind, you know, and then I said, okay, let the Let, let me do the jump, but I can tell you many of my colleagues were shocked also, you know, they said, what?

I mean, but, but some of them also, you know thought that it was, was extremely brave to dare to do some, try to do something else, you know, and so, but anyway, yeah, 

Philip Hemme: I liked 

it. I mean. I have to say from all our discussions so far, I, and now what you have achieved, I can see much more, the, the much more connections synergies and how it fits together than when you did the job.

Okay. I mean, yeah, I mean, yeah, now it, from our discussion, it makes even more, yeah, even more sense, which I quite like. I mean, I think it’s also, and what we said also, I mean. This kind of jump is also more of a typical, like, let’s say, more U. S. style, let’s say, Boston style of, okay, you’re doing more dramatic changes, which, but it works also.

Jens Nielsen: Yes, 

yeah. And, you know, and let’s also not forget that, that, you know, one of the things I mentioned, you know, that I get a lot of energy of, you know, working with young people and training them and so on. But there is still that element here and that’s what I, you know that, that is the same. Both young people in my own staff but also, you know, all the startup companies that I meet and, you know, helping them and, and, and, and, you know, instigating hope for the energy and, and, and, and drive into them.

So yeah. 

Philip Hemme: Yeah. 

That’s, yeah, that’s where I can see a lot of overlap. It reminds me the, the the episode 11 that is coming out will be come out when, when it will be released, which is a Sander Van Deventer was at the Amsterdam and founder of Unicure was a professor and then transition, which become like many founding a gene therapy company and really running it.

And then now VC. Made also this big jump, but when we discussed is like what struck me is how everything is quite connected. Yeah, we had separate jobs, but deep down what you did, I mean, what you started our conversation was, was like, you have this engineering mindset and trying to apply and translate science and that’s like the pillar and then you can be more PI, you can be Incubator.

I mean, probably you can be a very good VC as well. I can imagine. And you are half, half investor there. So at the end it’s a bit different hats, but they are so connected. Yes. I like that. I mean, and I think the whole discussion here is what I like, how like also we had always realized how, how everything is quite connected and I think, I mean, if I summarize to my initial It’s a question of like what works, but what works is to realize that everything is connected and that instead of opposing whatever one academic or fundamental from other, like when you start seeing that everything is very connected and embracing this versus rejecting it.

Jens Nielsen: Yeah, exactly. 

It is a conclusion. Yeah, no, I mean, I agree completely. No, I agree completely. This is a very good song. 

Philip Hemme: Great. 

Thanks a lot. 

Jens Nielsen: Thank you so much, Philip. It was great. I think it’s one of the best roundup where everything fits together perfectly. 

Philip Hemme: Thanks a lot. Thank you. Thank you very much. 

[01:17:46] Thanks for listening

Philip Hemme: Thanks for listening to the end.

I love the episode especially how Jens is passionate and humble and how everything is connected and all the synergies when you’re trying to translate science into products. If you enjoyed the, this episode, please hit the like or follow button. And if you are on Spotify or Apple podcasts, you can even leave a review.

Any of these actions would help the show a lot. And help us get better speakers, improve the production, and help you get better episodes. I would also be curious to know what you think, so please feel free to leave a comment if you’re on YouTube or Spotify. And you can also write me directly an email to philip at flut.

bio Alright, see you in the next episode.

Sander van Deventer, VectorY & Forbion | Pioneering gene therapy | E11

We’re in Amsterdam with Sander van Deventer, the CEO of VectorY. We will talk about VectorY and what’s missing in gene therapy.

Sander has an impressive track record, being a professor, operational biotech founder and a VC. In 1998, he co-founded uniQure, which brought the first gene therapy to the market. He also co-founded the leading VC Forbion as well as Dezima. He is now building a new venture VectorY, again in gene therapy. I interviewed him in 2017 back at Labiotech so it’s great to catch up with him again.


Transcript

Sander van Deventer: How large? And I said, well, probably a thousand square meters. They said, how many are you? I said, we are two. I’m pretty sure it will grow fast. 

Philip Hemme: It sounds amazing having the vector and then the antibody produced locally. How well does it work though? 

Sander van Deventer: You come from somebody in a wheelchair to somebody who can do top sports, and that is in one career.

Philip Hemme: How do you manage to just still keep going and then going again? Sander van Deventer: I’m the luckiest guy in the world.

[00:00:28] Welcome

Philip Hemme: Hi, I’m your host Philippe, and welcome to a new episode of the Flot.bio Show. Where I interview the best Europeans in biotech to help you grow. And today I’m in a very rainy, you know, Amsterdam. I had to talk with Sander van Deventer. And Sander has a really impressive track record, both as a professor, as an operational biotech founder, and also as a VC.

In 1998, he actually co founded what is now Unicure, and which actually brought the first gene therapy product. drug on the market. Then he went on Forbion, leading VC, and also a big success story, Dezima. And now he’s back at it again, founding a company, Vector E or Vector Y, again in the gene therapy space with some new technology.

We will talk about Vector E and also about the gene therapy field in general, especially what’s, what’s missing still to make it big. So let’s head upstairs and talk with Sanne. 

All

right. Welcome to the show, Sanne. 

Sander van Deventer: Well, nice to be here. 

Philip Hemme: It’s great that it worked out. I mean, I mentioned in the, in, I made a post about the Series A and I mentioned I will try to have you on the show and it’s great that it worked and we managed to get it just by the end of the year. Thanks for taking the time.

I mean, it’s a busy time before Christmas and all.

[00:01:54] The largest Series A ever

Philip Hemme: Yeah, I want to start with, with actually the Series A. I mean, 129 million euro, I think, let’s say 130. I mean, it’s, I think the largest series A this year. One of the largest ever, I mean, European standards as we been. But I’ve heard also, it took a bit longer to close than expected.

Can you maybe share what, what happened? 

Sander van Deventer: Yeah, so basically, I think there were two factors here. So the company started as a, a gene therapy company, advertising, basically it being a gene therapy company and. As we developed the technology it turned out that most of the value actually is in the ability to, to address difficult issues in proteostasis in neurons.

So, so actually from the delivery side of things, like we still like the delivery being a gene therapy because it’s a one off therapy that will work for a very long period of time, which is really nice when you do that. In the CNS, the focus shifted more and more to, to the business side of things, you know, what exactly we are expressing.

And so, you know if you look at the efforts here in the company by, by far the most effort actually is in the binary technology and the ability to distinguish between misfolded and non misfolded native proteins, natively folded proteins. And so we actually found in the TDP 43 ALS program that there were some unique features here.

That is, that at the same time you can address a gain of tox, problem, whereas you also have an important loss of function problem. And that put us on a really nice place. And so, yeah, some of the investors who looked at the company early looked at it like, there we have another gene therapy company doing a sort of orphan ALS approach or something like that.

to the newer, investors who came in later or looked at it later you know, deciding, wow, this is a neat technology and they can really do it. And if they can do it in ALS, they can do it in other CNS diseases. So that is basically the, the shift of perception that we got at Factory. 

Philip Hemme: And I guess with that shift also, means also the potential is 

Sander van Deventer: Yeah, because, you know, just to give you an example in ALS, of course you have you know, really good data for SOD1 now, and there’s academic data on FOS, but those are the genetic forms of ALS.

And together, all, you know, all, all the genetic forms of ALS together are maybe 7 percent of ALS. And the technology that we developed is for all sporadic, so sporadic. It seems like the more rare form, it actually is more than 95 percent of, 93 or 95 percent of ALS. So the market is enormously larger also for, for, for our product than it is for, for product targeting.

So one. 

[00:05:01] What’s coming for ALS

Philip Hemme: Actually, that’s good. I mean, I had some questions on ALS because I think, I mean, it’s a huge unmet medical need. I don’t think there’s anything really on the market right now. And I think that there were some failures, let’s say, or some things were blocked. I think, I don’t remember the exact name, but somewhere were blocked.

So can you expand a bit there of like, how, how will you position yourself compared to what’s in development at the moment? Or, yeah. 

Sander van Deventer: Yeah. Yeah, sure. So, first of all, the market has, so ALS people think about it as a sort of rare disease. Yeah. But the the, the odds of somebody in the Western world developing ALS are about one in 340.

Yeah. So I always tell people, you know, if you board a regular train, then somebody in that train will get ALS. So it’s not that uncommon, actually. So the market is pretty large so that’s one thing, but the other thing is that in the last probably five years or so, five, six years it has become apparent why actually motor neurons die in sporadic ALS.

And it turns out that it’s a, it’s a protein which is very much involved in the splicing of mRNAs in motor neurons. Now neurons splice RNAs in many more transcripts than other cells to to give you an example, we have done some of these analysis. And so of the 20,000 genes that you obviously have in, in a, in any cell, but certainly also in a motor room, you have like 150,000 transcripts.

And so they need to be correctly spliced and TD four three is, is, is, is the key component there that is in the splicers arm, the key protein. that binds to these RNAs and directs correct splicing. So we found that one third of that complete transcriptome, so we’re talking about 50, 000 or whatever transcripts are dysregulated when Tdp43 goes wrong.

So, so that gives you also the answer why it’s so difficult to develop something in, in ALS. Because we do know many of the downstream 

Philip Hemme: you’re hitting. Everything. 

Sander van Deventer: Right. So companies have tried to do like STATMIN2 or, or UNG13A or all these other genes that that are definitely dysregulated and definitely associated with pathology and ALS, but it’s only one of so many.

So we, we actually correct about 80 percent of that complete misplaced transcriptome just restoring nuclear TDP43. 

[00:07:50] Vectorized Antibodies

Philip Hemme: Wow.

And just to go on the technology, I mean, You say about vectorized, I mean, that’s where the name comes from, so vectorized antibodies. I’m curious on, on really how, how and how well it works. I mean, it sounds amazing, like having the vector and then the antibody produced locally or in the cell. I mean, that’s how, yeah, how well, how well does it work 

though, Eddie, how, on a.

Sander van Deventer: Yeah, so some companies that are also under the umbrella of factorized antibodies that they make, antibodies that are secreted and they work outside of the cell. So, 

Philip Hemme: Voyager and Yeah. Most of it, they are Exactly. Probably one of the leading ones, almost. 

Sander van Deventer: Yeah. They are in that business, basically.

We are developing antibodies, and actually some academic groups have done that, and Voyager has done that in the past also. that stay inside the cell. And so you can put them in different formats. So we have used single chain IVs. We have also worked with VHH, binars. And and, and, and those can be nicely expressed long term in, in animals.

I think, you know, the longest is a couple of months that we have now, but I expect these to express years after a single dose. And so that works nicely. Now, it comes to the part where what you bind and, and, and how you actually then get rid of it. So, in ALS, you have aggregates of Td43.

Normally, 80 percent of the Td43 is within the nucleus, and that’s where it mainly works, because the splicing aspect and, and so on and so forth is, it does more than that, but let’s focus on that. And, and then only 20 percent is in cytoplasm, and there’s a recirculation. Of TD four three from the cytoplasm to the nucleus.

So we know that process quite well. But if you have aggregation formation in the cytoplasm and you get mis, placed, so to say T four three, then you lose it in the nucleus. And also that circulation is then disturbed because the aggregates actually aggregate more and more. TD four three is like a gluey lump of TD four three that stays there.

And so that interferes with that recirculation. So, the, the, the, the, the binaries that we make need to get rid of those ag gats. In some, we, so we, we actually de, developed a whole lot of binaries targeting the complete protein. So there was a lot of work has been done there. And so we first, 

Philip Hemme: you said 

Sander van Deventer: that’s why most of the people work on the binaries.

Right. 

And so we, we, we first defined where actually to target that that protein. So then we came up with a binder that did it very well, and we actually found out why. Because it binds to a part of the protein that is not accessible in the normal forms. But it is accessible when you have aggregation.

And so, so that’s one part of it. And then, of course, binding alone is not enough. You need to get it somewhere. And so those large aggregates actually then need to get, degraded by autophagy. And if you have smaller misfolded proteins, it’s mainly through the proteasome. And then also one other pathway you can think about is the lysosome.

So at the same time we made what we made dagrons, so those are motifs that we can link. with our binders. And the dagrons actually instruct the whole complex to go somewhere. So it can be, you know, to autophagy, it can be to the proteasome. And we actually have binders now that we engineered to have a dual function.

Basically, it says if you bind to an aggregate autophagy, but if you bind to a misfolded protein, it needs to go to the proteasome. So all that work also was done in parallel. So we call the binders vectabs and these dagrons we call dagrons. And so the ones that are engineered with a dagron we call a vectron.

So it’s a little bit because otherwise we have to always explain. And that works amazingly well. So the, the binaries actually are expressed at the, the through levels, the trough levels or the, you know, the, the, the, the levels that we have in those cells are really low. They’re so low that you sometimes even have to go to technologies like, LC MS to, to basically even to show that they are there.

But they function like very, very well. So they almost completely or completely normalized the phenotype of ALS motor neurons. So, if you look at, you know electrophysiology, or you look at dendrite length, or you look at signaling, or you look at you know all kinds of aspects, survival, and all kinds of aspects of health, they’re completely too normal.

Philip Hemme: What’s the matter? 

Are you? Yeah, okay, so. If for instance, you have the, you have your, your Your vector, your, your gene cycle, your vector, you will insert the gene in a certain cells, which then express and make the, the, I mean, express some of your binders, intra, intracellular. And then you can, target the aggregates, bind them together, and then lead to the autophagy and, and restore the phenotype.

It’s crazy. Yeah, 

Sander van Deventer: but it’s not only autophagy, I think. All of it. Or, or we can run it to the proteasome or proteasomally. Degrade them. You can actually, we, we have a bin that’s neutral, and so if you put a different deagon on it, it’ll start doing different things. So if it’s neutral, so it binds through, an aggregate does nothing.

But if we engineer it with an autophagy, Deon, then it will instruct autophagy or proteasome more degradation. It will instruc that as well 

[00:14:03] Developing binars

Philip Hemme: and can really engineers the binders. I’m curious what, so you mentioned VH or what’s specific in the binders? I mean. Just coming from, I worked a bit with molecular partners with some, all right, alternative proteins and I know one of the big challenge with antibodies, it’s just usually too large, also a bit harder to, to re engineer precisely, especially when you talk intracellular, so how do you Yeah.

What, what are you actually using? 

Sander van Deventer: Well, there are a couple of things here. So there, the whole development of these binaries is very different from, from antibodies. I’ve worked with antibodies for a long time. And so to, to start with, how do you select binaries that specifically recognize misfolded proteins?

Because the moment you start making these proteins as an antigen, for example, They’re always misfolded and they’re in the, you know, they’re never in the correct configuration. So you really have to do this on a high throughput cell systems. And so here in the lab, we actually have a high throughput screening system in, in for example, U2R cells, but also in IPS striped human cells.

It could be motor neurons or, or immediate spiny striatal neurons or cortical neurons. We have all that here. And also glial cells, like we have astrocytes here and all that sort of, we have everything here. So muscle, we can do all that. So 

Philip Hemme: This floor, and 

the other floor, and the other 

building. 

Sander van Deventer: Right, right, right.

So it’s all there. So, so the screening is very different. But also the pharmacodynamics and pharmacokinetics totally different because, you know in, in, in, in the periphery, you would like to have you know, a sort of, you know, at least a significant trough level. But here you know, the turnover actually is pretty fast for these small binars.

Talk about hours or something like that, but within the cell it doesn’t matter. Actually, they’re extremely active. You can hardly measure them and they’re highly, highly potent. So the whole pharmacokinetic model is also totally different from, from what it is outside cells. 

Philip Hemme: And I guess they 

are produced constantly.

That’s the point. You don’t need a massive quantity because it’s over time. 

Sander van Deventer: Exactly. And then of course, how 

Philip Hemme: you tell me what, what, what specifically are those binders like? 

Sander van Deventer: Well, we do have them in different formats. So we do have a, for our TD43 program, it’s a single chain FV. And for our Huntington program, it’s a VHH engineered with a dagger on.

and all that sort of stuff. So it’s, it depends. There are some arguments why, why to choose one over the other for a certain program. But you know, there are also aspects you know, in what compartment do we want that binder to show up? So if it, if it, if we don’t want, for example, the binder to show up in the nucleus.

And clearly, for TD43, that would be, you know, not something that we want. Then then we, a single chain FE format, you know, doesn’t go to the nucleus. But the VIHH might. And so we do have some aggregates that are nuclear, and for that program, we would use a different binder. So these are just some considerations. 

Philip Hemme: Yeah, that’s good. And I’m curious just from also a bit comparing it to, to different technology. I mean When I, when I heard about it and looked over, I thought right away about mRNA delivery as well. I mean, you could deliver instruction to produce the other binder as well. But I guess then the window. to produce is way less long or less over time. So for ALS, you will not have this like ongoing production of binders, which will fix it really long term with a single injection, or you will need like several injections. That’s, yeah, 

yeah.

[00:17:57] AV technology

Sander van Deventer: So that’s why actually coming to the gene therapy aspect here. So we are open to any non viral delivery or even use the, the VAC taps as VAC taps, you know, engineer them in any way or format.

Right now for the brain programs, there are two things. First of all, in ALS, we really, really specifically need to target motor neurons. So we have a whole vector strategy to do that and and I’m pretty confident that with the vector we have now here in house we, we achieve that goal. And then it’s a single administration and there will be very long term expression based on what we have seen, for example, in Huntington Project that we developed at UniQR in years.

So that’s a good thing. With siRNAs, of course the specificity of the transduction will be, you know, more, you know, common. I’ve seen really good distribution data, by the way, with silencing RNAs now in the brain. But it would be a repeated dosing. And the problem with repeated dosing is that you know, a lot of the troubles that you now see in the Roche program, or with Spinraza, or other ACE whole programs, is really, you know, increases in NFL, ventricles and all that sort of stuff.

Which probably is related to, you know, it might be related to the repeated dosing of the, of the ASOS. So yeah, we really don’t want to get in those troubles. So for now, for these programs in the brain, we use AV technology but we are open to any other delivery technology 

Philip Hemme: because I, I mean, there’s a lot of also downsides to AV.

Yeah. It’s, I mean, in general, it is one of the bottlenecks. So yeah. Okay. 

Sander van Deventer: Yeah. But you know, to, to come back to that when we started this company, I’ve been producing AAV since 2004 and, you know, we did two commercial products, Clibira and Hemogenics, and so I’ve been scaling it up and and I’ve been, you know, involved quite a lot with the technology itself.

So here in this company, when we started, we said, well, we want to actually even the next gen. So we want to get to 90 percent full particles. We want to have the particles filled with the correct and intact genomes and all that. And then we want to produce this all the way to a 2, 000 liter scale. So that the cost of goods come down and that we can do hundreds and hundreds of patients from a single run.

And that’s, that’s the proc, the process we have. Yeah. So. 

Philip Hemme: I, I saw on the manufacturing you, you, you already claim on the 2, 000 liters already? 

Sander van Deventer: No, we haven’t done it but we are doing it now. Okay. So we are at the, at the process now to, to, here in, in, in the company we only go to 50 liters. Yeah. And so then we transfer it to CDMO.

Yeah. With, with exactly the same equipment. It uses our materials in our process, and then our people are there, and that’s how we’re scaling up now. Okay. 

Philip Hemme: And I was on the, what you mentioned also on the, on the AAV, I mean, I think it rapidly gets pretty technical as well, but what I’ve seen is one of your poster or paper on, you really can engineer the capside and have some like tissue specific peptides on the, on the side that you can.

basically targets certain type of tissue, I guess, in the brain. I guess, for us, this is super important. And at the same time, you also showed that, that the capsaicin can cross the blood, blood brain barrier. So you don’t even need to inject directly in the brain. 

Sander van Deventer: Yeah, there’s, there’s several companies that have shown that, so, in, and also in primates.

So we only showed it at Xvivo. So we don’t, don’t even have primate data. We are generating those data, but 

Philip Hemme: Wasn’t that on PICS, the data you poster you showed? 

Sander van Deventer: Yeah, but vivo, right? That was in a model. But you know, Capsogen and Dyno and Voyager and Dev, all these capsids that can do that. Okay.

So we are not unique there. But the one thing I think we’re unique at is the structural engineering part, right? So we basically choose the target, and then we make a peptide ligand, and then we screen that ligand in different formats. And that ligand we engineer in in the outer capsid directly in such a conformation that it retains that binding capacity.

So you can actually, if you work that way, you can work much more targeted than if you do a million or a billion, library with random mutations. And then you have to select back. And also the optimization is more straightforward. So, yeah, we’ve done that. And we’re, we have now. library that actually is being tested also with novel binding.

[00:22:55] The pipeline of VectorY

Philip Hemme: Okay. That’s nice. Yeah, I like that. On the, on the, you mentioned on the pipeline, so you have ALS, Huntington’s, Parkinson’s, but they’re all in preclinical slash in, in cells at the moment, so. Yeah. So where you found, where is it true, true then A and D and, and even which first human? 

Sander van Deventer: Well, for ALS, I mean, we’ve done the TDP43 mouse model and we’ve done multiple.

Models going from mouse to pig to, to primate. And we’re now at the stage that we are in, we’ll shortly discuss pre IND have a pre IND meeting with the FDA, and the plan is to to complete our primate top study this next year, end of next year, and to have an IND meeting and to start clinical trial in 2025, in Q1 and with ALS, of course, the point is that you know, fairly rapidly get to an end point, because the disease is rapidly progressive, and so we expect, and then there’s a whole biomarker strategy which is very much linked to the exact TD PFORT3 pathology, because the consequences of this misplacing as a consequence of loss of function of TD PFORT3.

Are that you also get peptides that shouldn’t be there, that are misplaced, actually the result of misplacing. And those peptides you can measure, you can measure them in CSF, you can measure them in patients. So if you then, if you restore splicing, you should get rid of those faulty peptides. So all that work is ongoing.

So I actually expect that if it all works well we will And, we do have a whole panel of those and, and then of course there are usual biomarkers like NFL. And, of course, there’s a motor score also that we are following that is rapidly progressive. And so 

Philip Hemme: I start to understand also what’s first, what’s behind the platform and also where is the potential and also where.

I mean, it sounds like you have pretty solid already knowledge slash data really on the cap side, on the, on the binders, on the, on the disease. on exactly where you, where you target, where the trial is going. You already have solid preclinical data. Because, I mean, my, my question, I mean, basically what’s, to, to explain the, the, the money raised, basically, at the preclinical stage, I mean, it’s, of course, I mean, you’re, you’re involved and everything.

You can, you can pull out a bit more, but still, there’s limits to a preclinical, let’s say, evaluation, or how much you can really raise. But now I start to really understand that seems like a lot of the pillars are basically very solid from the foundations and then it’s about executing on the clinical trial.

Sander van Deventer: Well, the other thing is, you know, we said, listen, I mean where are we going to finance towards? And the discussion we had with our investors and in the end, everybody was very much on the same page. You know, it only, you know, value you create here with clinical results. So we wanted this financing for sure to get to meaningful clinical results for our lead program, which is VTX2 is cheap for 3 in ALS.

But we were also bringing the Huntington program to IND. And two other programs in the same proteostasis paradigm towards DC. So, you know, if, if, if you, if you do have a product that succeeds in ALS, of course the upside is, you know, much larger than the investment now. And but also if the technology really functions like a platform technology because we have excellent data in Huntington models also.

So if that also translate into political effigy in Huntington’s, then, you know, and the program. It’s a multiplier. It’s a multiplier. 

Philip Hemme: Especially I mean, CNS still today. There’s still, I mean, there are lots of, let’s say, failures or big failures and, but the need is, is so, so high that, yeah, if you have something that works, I guess.

[00:27:22] Where the money is moving to

Sander van Deventer: But you know, if you, if you look at the, where investments are going now, then there’s a sharp drop in oncology and there’s a sharp rise in well, metabolic obesity, of course, but also in CNS. So it’s really where the money is going now. 

Philip Hemme: It’s interesting. 

And I saw LSP invested through what is one of the dementia fund, which they started maybe earlier, earlier than the trend, but there are there are some other VCs who have also dementia or dementia discovery fund in the UK, which I guess, I mean, I was watching it, I think, I don’t know when they started exactly, maybe four or five years ago, where was still at least to me, maybe a bit early or was not clear.

Okay. Is that now, but maybe now it’s, it’s the timing. And I mean, I guess it’s the goal of a VC to be a bit earlier than, than too late. 

Sander van Deventer: Yeah. They did very well, I think. And and also the ALS investment fund put quite some money in in this company as well. And I know them almost from the start because we were looking at ALS investments and at Unicure, we had a GDNF program that at some point in time was considered to be.

Potentially something we should try in a LS and so I in already in those days I was talking about that.

[00:28:50] Meet the team

Philip Hemme: I think that’s the, the last part I want talk about is more on the, on the team as well. I mean, obviously you, you have co-founded Unique and then you even went back in 2017 as acting CSO and I think then the, you now CSO is, was the head of m and DI don’t remember the exact title at, at also, I guess.

I mean, the team is definitely very solid, but I’m curious also how it’s like, what’s the story behind some of the team joining or what’s like, yeah, or what’s the, the relationship with, with 

Uniqueur? 

Sander van Deventer: Yeah. Yeah. So, yeah, I mean, in general, of course, there are things like non compete and, I don’t think Unicure would like all their people to come here.

Actually they’re laying off people now like, like 130 or something like that. And I have a good relationship because actually they they’re actively helping people find new jobs, including here. So I have some people actually that worked at Unicure that work now here as of shortly.

The other thing is that yeah, I mean, listen, I mean, we started this company with two people, me and another and we, yeah, I, I quit Unicure and then I had a temporary job in China, as you probably know, when it was all about building an AAV manufacturing facility in Suzhou. And that all went very well was, you know, I wrote a little bit of a blog about it on LinkedIn being you know, in quarantine and what have you.

And then I came back and crazy. And we knocked here on the door where, where we are now to say, well, guy guys, do you have some lab for rent? And they said, well, yeah, how large? And I said, well, probably a thousand square meters or something like that. I said, how many are you? I said, we are two, but I’m pretty sure it will grow more fast.

So we didn’t get 1000 square meters, but, I think we started with maybe 300 or so. And but yeah, I mean, people just, we’re just like, and we were, we were like a magnet. People were calling us up spontaneously or. People really wanted to join. People left really good paying jobs to be in this company and and people actually, the founders invested their own money in the company and all that.

So that’s how it started.

Philip Hemme: I’m telling you, I guess you also, you also invested. 

Sander van Deventer: Yeah, of course. Yeah. And you know, the first thing actually I did is I went to the hardware store to to get some wrenches and stuff, you know, to, to put the the tables together and and and the wifi and what have you.

And so that’s literally how we started, but people really came here because there were so many people that, that we worked with in the past who knew us. They really want, the people like to work here. We have 15 nationalities here and we have about I think 65 FTEs or so, and the median age is 32 and we have 60 percent women.

It’s a, it’s a joyful group here, you know, with all these people coming from all over the place to actually do their best job here and that, that’s what, that’s how it works. 

Philip Hemme: I guess 

you’re hiring also actively, at least I’ve seen on your 

Sander van Deventer: Yeah, 

yeah, we just basically put out our, our vacancies, of course. 

Philip Hemme: I mean, people are listening to us.

[00:32:29] The Netherlands as a biotech cluster

Philip Hemme: But also, I mean, from what you said also, what’s pretty amazing also, Amsterdam or Netherlands in general, I mean, there’s a quite, I mean, quite a deep expertise in, in gene therapy in general. We are unique here, but also a lot of, of CDMO, manufacturers in, in the, in the space. Can you, can you expand a bit there, like on, yeah, on like how you, how you see, or like how you see that, or how it compares maybe to other clusters, let’s say, in Europe or?

Sander van Deventer: Yeah. Yeah. I think, you know, you have fantastic clusters all over the place. I mean, you, you talk about, Genepol, if you talk about a gene therapy but you know, all over the place and particularly also in, in UK, of course, enormous clusters of, of knowledge and know how. I think, you know, in the Benelux, actually, we’re, getting our act together, of course, with everything in Belgium going on, in Ghent and Leuven and what have you.

And, you know, Argenics, a company that we started with when there were five people at Forbion, we invested in it. And what is it now? It’s you know, now it’s 20, 20, 20 billion, market cap. So these are, these, these have become companies that are much larger than KLM Air France or Air France KLM more.

where companies like that, people just don’t know it, but you know, the cluster in Leiden is I was in the board there because I was a professor at Leiden also, I was in the, in the, in the board of the, of that cluster, and there’s more than a hundred companies there BMS is building a huge, cell therapy Cartisol factory there, Kite, of course here, so, yeah, there’s quite of that quite a lot of that and, but here in Amsterdam it started later, and, And it’s smaller, but we’re, we’re getting there.

I mean, we work with you know some companies here on, on site when it comes to promoter technology, for example and it’s growing fast. So yeah, I’m, I’m pretty optimistic about the, the environment. 

Philip Hemme: And I mean, also when you look at the map, Leiden, I mean. 

It’s nothing. 

I mean, far as you know, Paul, to the center of 

20 minutes here, 

I landed in Chippewa yesterday.

It’s crazy how close it is and the train, everything connected inside. That’s nice. Yeah, I was always surprised about this, like especially in the Netherlands. I mean, you had a few companies like unique here, but that’s a lot of like bigger pharma had their gene and cell therapy. here from a manufacturing or from development as always pretty like established trust me, 

Sander van Deventer: I was, well, J& J, Crusale, and of course we have GenMap and then, you know as I said, I mean, BMS, but big time now, Hostelos also in in in Leiden.

So yeah, there’s quite a lot. Yeah. 

[00:35:31] CRISPR approval and gene therapy

Philip Hemme: On the, maybe just on the, on the gene therapy field, if we, if we zoom out a bit, I’m curious what you think about the CRISPR approval, recently. 

Sander van Deventer: Yeah. So gene therapy I, I came to the conclusion that you know, the, the, you know, the, the promise of gene therapy. So people present you a slide deck and the first deck says, you know, there are 5, 000. genetic diseases and we can target them with with LALALALA virus you know, that doesn’t work, huh? So we already know that it’s difficult and you need to know what you’re doing. And so we are not doing gene replacement, for example, any longer. We’re just not doing that. So that’s not because it’s too complicated.

Yeah, it’s often very complex and really, you know, not so easy to do, eh? So, yeah, at Unicure we started already a long time ago with silencing, and so there was actually a press release this morning from Unicure that had some positive news in the Huntington program. But I think, so gene therapy can be very useful for all kinds of things, and, and particularly now with the antibody.

technology that we have, we can tackle things that are very hard to tackle with small molecules or, or, or with sinusine technology. So, but yeah, I mean, the next new thing, of course, is gene editing or things like that. So yeah, with CRISPR technology, I mean, the, the, the hurdles are simple. So it’s delivery for many of the diseases that they’re targeting.

You need to have an even spread of the, of the repair. For example, if you look at cardiac indications, you cannot just repair one in 10 cardiomyocytes when it, when it goes to conductors because you will induce arrhythmias actually if you do that. So, delivery is a big one. And then of course with CRISPR, Gas based technologies, I mean the construct is very large.

So at at at, at, at Forbian, we’ve been looking at space quite extensively and we passed on many of those. But you know, with recombinases, you know, if this company’s seamless, with relatively small constructs that have no, guide DNAs or whatever. You can actually do the same. So, so we became interested in that.

So, I really believe that those technologies at some point in time will will, will you know, of course they work ex vivo, so we, that, that’s a big one, but in vivo that they will start to work in vivo. 

Philip Hemme: Nice. I mean, we had Roger Novak on, on the show and. One thing that, that struck me is how he said, CRISPR, of course, to put the, the, the potential and whatever you can do is basically a dream technology and you could do a lot, a lot of things, but they really focused on ex vivo, only ex vivo and something a bit more easier to, to do and to execute and to make trial versus some other CRISPR companies were tried in vivo wide away.

And it’s just way too complex. Yeah. So I, I like, I mean, I think it, it goes into what you are saying as well. 

Sander van Deventer: Yeah, no, there are proposals like we’re going to CRISPR all the HIV out of people and you know, this is crazy. 

Philip Hemme: Maybe in 20, 30, 50 years, I don’t know. And I’m curious what’s your view on the, let’s say from a commercial success, from a market point of view, from a, because gene therapy, I mean, so far.

There’s clinical trials, you have approvals, but then when it goes to commercial, I mean, in the U. S., maybe it’s a bit better. In Europe, I mean, I think it’s limited, but what 

Sander van Deventer: Yeah, yeah. We need to think about that better. So of course you know I think hemogenics, if you do a pharmacoeconomic model, it’s quite clear that if you treat somebody with hemophilia B early on, and it works like for 10 years, then even if you have a crazy price for the product, it’s You know, you, you, you win, so the, the, the healthcare system does win, because you know, these Yeah, 

Philip Hemme: but why is the healthcare system not buying it more than 

Sander van Deventer: Because people just are not thinking about it.

So you know, if I, if I say here in the Netherlands a gene therapy product will cost one or two million or anything like that, everybody says, that’s crazy. And, you know, the health care system cannot afford that. But if I just tell people that and, and what about some patients with hemophilia that cost 300, 000 a year just in product, and then I’m not even calculating.

That these people you know, cannot work and that that they’re, that they need surgery and all extra costs. And so, you know, 300, 000 every year, that you know, that is a lot of money over 10 years, it’s 3 million, right? So people just don’t do those basically calculations and since our healthcare system is not you know basically yeah, maybe in France now with the thalassemias, they actually calculated how much the thalassemia patient cost to the healthcare system in France.

There are quite a lot of them, of course, in France because of the Mediterranean migration. And so actually they came to the conclusion that it would be much, much, much better for them to go for gene therapy for that disorder than to pay for. All the, all the, you know, all the extra costs that these patients bring.

So we need to have healthcare systems and leaders that actually think about this in a much more relaxed manner and also in a much more informed manner. 

Philip Hemme: But I, yeah, but from what you say, I mean, the, and I heard that many times, I mean, the case is pretty clear as in like, I mean, 10 times 300, 000 versus one or two million, it’s not like super complicated math or whatever, like, but, so I’m curious, I mean, as a payer, they can also, I mean, it’s their job to understand it or like to, to do it.

So why, I don’t get where it’s like, really blocking and because I hear the same mess already, let’s say, five years ago, was already the same, let’s say, argument, but I don’t see necessarily payers, like, paying more, acting more. Yeah. Or is it, is it me? 

Sander van Deventer: No, yeah, you’re correct. I mean, the most frustrating part of it is the the lack of any vision.

And, you know, I’ve been here for a long time now. We did the first Remicade patient. It was infused in MCEM in 1991. So that’s a long time ago. And there we had exactly the same discussion. So people were saying, yeah, you know, you can treat Crohn’s disease with steroids and with azathioprine. And why are we now going for a treatment that at that point in time would cost like 35, 000 a year?

I mean, it’s now much cheaper, of course. But then you know, everybody started realizing that actually you know, we, we, with the old treatment, steroids and azathioprine, we never had complete remissions, actually. Never, ever. And so that’s a, a complete different, therapy had, had hit and, you know, everybody now is, is, of course treated with those kinds of drugs, the same holds true for, for, for when we started working on natalizumab or now with ocrelizumab.

If you have MS and you’re treated with the regular stuff, it’s basically waiting until you hit the wheelchair, that’s what you get. With ocrelizumab and ofatimumab, those new drugs. It’s, it’s, there’s zero relapse almost, almost nothing. And so, you know, these are completely different. Approaches.

And by the way, I can give you a little bit of an anecdote there. And, I, I was closely involved with Natalie’s map and I knew about OV2MU map because I was my friend is Winkel, who is basically first developed at, at GenMap and of course went to Novartis. But, my my wife basically got MS like six years ago.

Suddenly, and we are actually mountain biking in the Alps and stuff like that, you know, very fit and bang, you know, she went out actually and and had severe problems, couldn’t see and all that sort of stuff. And then we went here and then it’s like, yeah, you know, you can do this. You can do that. I said, no, no, no, no.

We’re just doing what I think, you know, works. And we started immediately with Natalism app because that was available then. And and she, you know, she, she, she completely, almost completely normalized. And then when Oculism app came, I said, well, Oculism app clearly is better than Natalism app. You need to take that.

So that’s about, it’s about I think the cost is about 35, 000 a year or so for Oculism app here in the Netherlands. And I, I do this 

Philip Hemme: what’s the, the company’s behind? 

Sander van Deventer: It’s Roche. Roche. Yeah. 

Philip Hemme: And the other one? 

Sander van Deventer: Overture Memes, now partitioned. Yeah, of course, they’re finding their, they’re fighting their little Swiss battle, but, but they’re both good.

So probably Overture Memes might be a little better even than Opalism, but they’re both very, very good. And so I, I do these lectures and I had a meeting at health, the, the economy of the Ministry of Health here in in the Netherlands. I just showed them this. My, my wife is a professor of pediatric infectious diseases.

She has a big lab. She makes money. She pays tax and all that sort of stuff. So I did the, the, the commercial case of my wife. And I could, could actually calculate that it is much better for the Dutch. Taxpayer or for the Dutch government or whatever to have treated my wife for 35, 000. Then not to have treated her over because in six years in her case, she would have been in a wheelchair.

So I mean But, but it, you know, people just come to me and they say, wow, wow, you know, that’s really interesting what you said and blah, blah, blah. But nobody’s taking any action. So that’s really what worries me. 

Philip Hemme: Crazy, crazy story. Makes me think also about how beautiful and how life changing some of the biotech products developing are.

I mean, it’s The example of yours is crazy. 

Sander van Deventer: Philip, i, I, I became an internist in 1980, in 1987. And so I do have some slides, actually, that I made in those days about rheumatoid arthritis. There was a young woman, 30 years, in a wheelchair, completely, with all these deformations. I, you know, I have a slide about HIV, the HIV that we’re seeing.

I’ve just horrible. I have a slide of a patient with hemophilia in those days, with enormous swollen joints and whatever because of the bleeds. And if you, you know, I, I’m a, I’m a cyclist, I, I, I like to cycle and so a couple of years I was in, ago I was in Tuscany and there was a lady cycling with me really competitively.

Actually she was also, she had been in in the, in the, in the British squad I think actually for the coming out nationally. And she was on Remicade because of rheumatoid arthritis. And she was basically cycling like crazy. And then you come from somebody in a wheelchair to somebody who can do top sports, right?

And, and that’s in my, in my, it’s in one career. And so people just don’t realize how much, you know, things have improved. And then I’m thinking back, why have these things improved? Is it because we have better doctors? Is it because we have nicer hospitals or better ministries of health or ministers of health?

No, no, no. It’s a hundred percent because we have better medicines. A hundred percent. So, people need just to realize that. 

[00:48:15] Where Sander came from

Philip Hemme: Amazing. Yeah. Yeah. I like that. And it it’s, it’s great. It’s a perfect shift also to more, to yourself, to the personal story. I remember I asked you in 2017 that I think I, I wanna Get a bit into this again, and I think it connects exactly to what you’re saying as in, I mean, you’re, you’re really having these different hats as in like professor biotech founder, but not just scientific founder or, or professor, the founder, but also we executive and at the same time VC, I, can you, like, I guess from what you explained as well, you, you, you’ve seen I guess a lot of things that works in the lab, but it gets never translated and never really reaches the patient.

So I can imagine, I think that’s also a bit what you told me, that it’s pretty natural, it was pretty natural for you to push and go towards there. And even at some point you, you were raising money, but you wanted to see the other side. And also when you have the control of, or you have the bite of money and you can deploy it, it also makes you act differently.

Can you kind of maybe reflect on, on all of that? 

Sander van Deventer: Yeah. Yeah. So, yeah, to come back, you know, I’m the luckiest guy in the world, so I’ve done fundamental science. I was a scientist at Rockefeller University. And then I was trained as a, I’ve done a lot of virology in already in the early 80s. Basically doing molecular biology in the very early days that you have to do everything here.

You had to do everything yourself. You had to make your restriction enzymes yourself in the cold room and stuff like that. So, then I was trained as a doctor, internist, gastroenterologist and all that. And then I got involved in drug development and You know seen quite some success there in, on the antibody side the antisense oligonucleus nucleotide side, the gene therapy side, and then, you know, got the opportunity to become one of the founders of, And actually before that actually already founded of course Unicure and then afterwards some other companies.

So I’m, I’m, I really am very lucky because I’ve seen it from all sides. But in the end, you know, you have to think about, you know, what drives you? Why do you, why do you do the stuff you do? And so, I’ve seen, I’ve, for 20 years I’ve seen patients, okay, so patients that are very, very sick and for which we could do nothing particularly the worst probably was HIV because we had a lot of that in Amsterdam and I was in the smack in the middle of it.

And we could do absolutely nothing for these patients. And then later, you know rumored Crohn’s disease. You know 100 percent of patients almost with Crohn’s disease got surgery. And 25 percent of them almost, and always ended with a stoma. With, you know, with, you know, that’s really bad if you’re a young girl stoma.

That, that, I’ve seen that a lot. And then you know ALS and, and MS and all these diseases. So. I was seeing patients in the outpatient clinic and often couldn’t do a lot. And so that’s why in 2004 actually I made the decision to switch completely because until 2004 I was also in the clinic.

And I was thinking, you know, where can I contribute more to the health and, and, and the benefit of people, of patients being sitting here doing, you know, 20 patients in a shift in the outpatient department or developing something. And I thought, well, probably with developing stuff, it would be better.

So. And then, you know, we did Clibira, we did Amgenix, we did this program, of course, in Huntington’s and and of course, New Amsterdam Pharma with CTP inhibitors and you know, many different things that we did, I think will benefit patients quite a lot. And if you think back and you think, you know, how many patients now have been treated with MTT?

TNF in one way or the other, it’s a lot, right? More than I ever could have seen in the outpatient clinic. So, so that is the good thing about, I think, where I am now. 

Philip Hemme: Yeah. And you, I was about to ask you when you, if you look back, if you do the math, it’s always also I guess you. Yeah, I mean, you have impact, but when you are treating a patient, you have also, you know, a direct impact and you need also people to treat patients and you need doctors as well to deliver the drugs and to do all the following, but I guess for you, it’s really clear that you had more impact.

Developing the drugs didn’t, but 

Sander van Deventer: I think so. Yeah. 

Philip Hemme: At the same time, you also got, I guess, I mean, there’s always some luck involved in, in biotech, as in like, I mean, the failure rate is just so high in, in, in your case, it’s, it’s not only luck of course, but No, no, we , yeah. 

Involved. And it’s amazing that you managed to.

be involved in two or three, four approved drugs within not even 20 years. It’s, it’s very rare, actually. I can’t. 

Sander van Deventer: Yeah. And, you know, there, you know, you always think back and you think, you know, what actually was really, a career you know, changing or, you know, what were the big things. So one was actually that I was lucky to run into, you know, a promoter you know, for my PhD project.

who was really a visionary. So he was, for all his PhDs actually, there was a like, you know, you really have to go here or there and that usually would be the United States or whatever or, or other places. But you know, he had more than a hundred PhDs and he, he has died. But I think his offspring is like, I think we had a sort of, gathering reunion or, and there were like, 30 or 40 professors who came out of this one man.

And so for, he said you need to go to the United States and you know, I, I did my PhD on bacterial endotoxins and it was when, cytokines became you know, and discovered. Ah, and so Charles Dinorello and and and Sarammi and Bruce Boyler and whatever. And so I, I became there and then, then I came in the United States and I was actually accustomed to the European sort of thing like you know, you said, well, it would be really neat if we would do this or that.

And everybody would say, yeah, but you know, you know, the risk or who says it will succeed or you cannot do that or whatever. And then I came in New York at Rockefeller and it was like. Okay so yeah, this sounds like a good, a nice idea, you know, we are, we, you know, new, and what can we do, you know, what do we need?

And that’s how it went there, you know, there were also struggles to get into the lab, you know, we had to fight for a bench, but on the other hand, you know, people were really supportive in, in, in, in basically saying, yeah, you know. Sounds like a really interesting idea. How, how, how could it be done? And so that changed my mind and I never forgot about that.

Philip Hemme: How could it work versus how can it not? 

Sander van Deventer: Exactly. Why it would not work. Yeah. Oh, 

Philip Hemme: that’s, that’s nice. And I guess you, you kept some of this mindset even though you returned to Europe. 

Sander van Deventer: Absolutely. Absolutely. 

Philip Hemme: Yeah. I like it. I can second this. And I mean, I went, I was one year in, in, in Boston to finish my masters and say my, Especially in Boston, especially like it was 2013, 2014 was an amazing energy, vibe, everything.

And I learned also a lot from there. 

[00:56:14] How to have the energy to be a serial entrepreneur

Philip Hemme: Maybe just from the last, last point, which also fascinates me as an entrepreneur as well, is that you still have like crazy amount of energy to go again and to build again a company. It’s, I mean, it’s so difficult in any kind of company, but in biotech it’s so complicated.

I guess from what you said, there is impact behind and there’s a lot of motivation, but can you explain a bit there, like how, yeah, how do you manage to just still keep going and then going again? 

Sander van Deventer: Well, I do not really have an answer for that, but you know, there’s a couple of things I have very rapidly get bored.

And you know, also here at the universities in the Netherlands, you would get a compulsory retirement. Yeah. So I was just basically. Yeah. Yeah. Very, in a very, very friendly manner, actually told to, to not to show up any longer. They were really kind. I mean they were really, you know, they were, but they have to do that sort of stuff.

But yeah, I mean, listen I, I, I, I had a good friend dying from ALS. Then two other people I knew also died from ALS. So at some point in time it came. Yeah, I, I realized that particularly also because this pathology became apparent in the last five years, I think, wow, if this is the case, then, then, then I, I think I know what, what to do about it.

And so then I thought, yeah, if I know what to do about it and we have the ability to do it and almost obliged to do that. And it’s great fun. I mean, it’s just great fun.

Philip Hemme: Amazing, Sander. We could continue, but I think you are pretty packed schedule, so it’s 4. 00. 

Sander van Deventer: Okay, well, great. 

Philip Hemme: Thanks a lot for joining again. Thank you.

[00:58:15] Thanks for listening

Philip Hemme: Wow. Wow. Crazy, crazy discussion. It’s amazing what he has achieved and what’s behind. I loved how he’s really super focused on the patients, but the clarity to develop it and really focus on impact. Really, really inspiring. I mean, we need. Well, Sanders for Europe and for humanity. Super inspiring. Thanks for staying until the end I hope you enjoyed it as well.

If you did, please hit the like Subscribe button if you’re on Apple podcast or Spotify You can also leave us a five star review Any of these actions would help us a lot and I would also be curious to hear what you think So if you’re on on YouTube or Spotify, you can comment directly or you can send me an email at philip at Alright, catch you in the next episode.

Thomas Clozel, Owkin | What does AI Biotech really mean | E10

In an online special, we’re interviewing Thomas Clozal, the co-founder and CEO of Owkin. We talk about scaling AI in biotech and how it feels to be the child of billionaire biotech parents.

Thomas founded Owkin in 2016 and I first met him in 2017. At that time, Owkin was still a baby and I wasn’t sure where it would go. Now it’s a unicorn following a $180M investment from Sanofi in 2021 and has over 300 employees.

🔗 Links mentioned in the interview:


Transcript

[00:00:00] Intro

Thomas Clozel: The thing today where the field is going is everybody agreed it needs to be more data driven, improve the variety of success of drugs, get more drugs out, is the better population, but you need to be able to take horizontal bets. It’s like finding a panda in a zoo. A baby panda in a zoo. Yeah, a baby panda in a zoo.

I mean venture capitalists ate way less bamboo, but you still really have to find them. It’s a rare species. 

Philip Hemme: How did it feel and how did it impact 

Thomas Clozel: you? Having a social impact in life gives you less pressure. Because you did your best. 

Philip Hemme: Hey, I’m your host, Philip, and welcome to the Flot.bio Show, where I interview the best Europeans in biotech to help you grow.

Today, I am online to talk with Thomas Clozel who is the co founder and the CEO of Owkin. Thomas founded the co founder company in 2016, and we talked just after, around 2017, for the first time. And it was just the beginning, and I have to say, I wasn’t sure where it’d go. But today it’s a different story.

It’s a billion dollar company following an investment from Sanofi in 2021. 118 million, one over billion valuation and has a team of over 300 employees. Amazing journey so far. And so we will talk about AI or scaling AI in biotech, as well as how it feels to be the son of two biotech billionaires. So let’s connect our line and talk with Thomas.

[00:01:26] You need a good pipeline

Philip Hemme: Welcome to us. Welcome to the show. 

Thomas Clozel: Hey Philippe, good to be here. 

Philip Hemme: Yeah, I mean, it’s good to have you as well. It’s good it worked out. We tried for, I think, four or five months. Pretty much. We made it before 2023. 

Thomas Clozel: It’s perfect. That was the goal. 

Philip Hemme: Alright, so, so, for today, I think, Before challenging you on like Orkin, like specifically, I would like to start with, let’s say, the bigger picture in AI.

I get this request a lot. I mean, it seems like they have a lot of things happening, obviously super hot topic, a lot of deals being announced. I think it’s one of the fields, maybe with obesity drugs in biotech, where you have like, outperformed everything else. But at the same time, I mean, I think it’s super complex to understand, both from a technological point of view.

Also kind of a bit of a fear of missing out, fear of being replaced, whatever, you hear this like big AGI and everyone’s job will be replaced. Can you maybe start on the, let’s say on the top level view, what’s above Orkin and from a field perspective of like, what’s Yeah, it’s a different category, especially as it is like today yeah, as we act and today, yeah.

Thomas Clozel: No, you know, so my, my vision is really like, you know, the, the pharma model has to be changed. The probability of success, as you know, is super low. The pharma has spent a lot of money to create great drugs. Most pipelines are really empty, you know many, many pharma depends on one drug, you know only.

And so there’s a lot of things to be done. And the thing that is even the, the, the, the most like, you know, probably. Doable as a, as a, as a challenge to, to, to have the pharma space is, is so many steps are based on intuition and that could be replaced by data. And you want to go from an intuition based model to a data driven model to improve the priority of success and the number of drugs that can be out.

I think that’s really where, where, where, where the field has, has to stand. Another question is how do you actually do it? And so for us, and for me, the answer is using really good data and artificial intelligence. I think that today where the field is going is to be able to replace the farmer. A lot of people are taking very vertical bets.

vertical on just doing drug discovery, or doing clinical trials, or doing diagnostics. My vision is things cross fertilize way more. You can find biomarkers that can help you find new targets, or find the right population, or the right indication, or help you for clinical trial, and even do diagnostics with the same biomarkers and the same biology.

And my vision is like, biology can help, you know, for the full life cycle of the drug. And therefore beds should be more horizontal than actually vertical. And it’s not only vertical by the, which part of the farm I want to help, it’s also vertical by modality. People are just doing, you know, AI for pathology, or AI for omics, or foundation model for proteins, but, you know only the multi modal aspect of it can capture the right biology and the right way to help.

I think today we are, the field is going is everybody agreed needs to be more data driven, improve the variety of success of drugs, get more drugs out with the better population and better, but, but you need to, to really like being able to take horizontal bets and never go to the vertical side of it.

I don’t know if that resonates with you, but. 

Philip Hemme: Yeah, definitely. I mean, and I think that’s one of the big selling points of, of Orkin. You should have written to Orkin. Yep. Where should we go with it? I think, but what you mentioned, what I, what I like also, maybe you can comment on there. It’s like, I mean, you mentioned.

optimizing drug discovery, optimizing clinical trials, optimizing, let’s say, I mean, in there you have patient certification, optimizing, looking for biomarkers, all of that. And then even then optimizing, let’s say, really in the healthcare and how other drugs are prescribed or given. And let’s say in a better setting, you can optimize a lot of things.

But maybe there to understand a bit more, because I mean, you guys, you are trying to go For the horizontal, yeah, I think if you take these different categories, I mean, how can you are going very horizontal and try to cross except wet lab? If I’m, if I understood, okay, so you’re really going full horizontal, but if you go into the categories, I mean, if I look, there is.

In the drug, yes, I’m really on drug discovery doing whatever, like virtual cells. It’s the first one that comes to my mind, or like really, like let’s say specific step of the drug discovery. You mentioned pathology, I think like path. ai is probably one of your biggest competitors, really on like data.

pathology, but it’s pretty big also, but on the, and then the bigger differentiation is, is the ones who have their own pipeline versus the one we do deals with biotech and pharma companies with enough pipelines. 

Thomas Clozel: So can I, can I comment on that? 

Philip Hemme: Yeah, yeah, yeah, go for it. I’m trying, I’m really trying for a long time.

I try to 

Thomas Clozel: be, we try to decipher the categories. So, I mean, like the question of the pipeline is a bit different. First, the big question is what is the problem you’re solving, okay? For Okin, we are trying to solve the precision medicine problem. Why precision medicine does not happen? I was a clinician, and you know, when you treat patients the same way, pretty much, it’s frustrating.

So for us, like, the view of precision medicine was, we don’t have the right diagnostic tools to be able to augment the care in predicting response to treatment or survival or whatever. And we just don’t have enough drugs. And when we have drugs, they’re not matched to the right population. So the matching of population to biology is a real problem for the pharma.

So we believe that the problem we wanted to solve was precision medicine and therefore we had to bring diagnostic tools, AI enabled New therapeutics. In the right subgroups. So this is really the ion. We are, we, we, we take, so this is, I mean, we are trying to solve this problem. The only solution is to rebuild a full end-to-end pharma.

That’s really the thing we do. So I think like, how to defer the field in general. I think it’s, it’s first it’s not about creating a pipeline. Not creating a pipeline. Everybody has intake bio to create a pipeline. The big question is our pipeline is diagnostics and therapeutics other people is therapeutics.

The question is. Is your pipeline really data driven? A lot of companies, you know, have built a really amazing platform. For example, Knowledge Graph platform, but then had taken experts to choose the pipeline. So first the question is, is your pipeline completely driven by your technology and not by a few experts?

Because if you do a great platform, great technology, but then you go with a few expert driven decision, you’re not really making a revolution in the model. That’s the first point. And the second point is, Is your pipeline better? I know that the real pharma, have you found things that the pharma would not have found itself?

Not only faster, but better. In indications that failed in the pharma, glioblastoma, mesothelioma. And, and if you want to really be there, you have to wait for clinical proof of concepts, which is a phase two B in oncology, for example. And you really have to show clinical proof points. I mean, so the question is, is your pipeline linked to the platform?

And is your pipeline proven to be better? And there is no really other option to show that that. And that’s the, and the third point is, then this is what the first, the second thing you can prove. So the first thing you can prove, but then if you want to prove like even bigger metrics, like probability of success, can you, for example, double probability of success?

What we want to do at Token, go from 10 to 20%, for example. You really have you need a lot of trials. You need a lot of clinical trials to be able to show it. I think the problem as well is nobody can do the claim today that they have been able to improve productivity success by better chemistry, by better clinical trials, by better targets because you need so many trials to show it that there is no big claims that can happen.

And this is the big problem. Acceleration has been shown. Augmentation still has to be proven. And we are all here in the same game. And to finish on that, the solutions part of it, the solutions, the services. You know, for me, the services is only a way to cross fund your own pipeline. You have to get your own pipeline, otherwise you have no IP.

You cannot be a big business being a solution provider. So for us, we are doing like, you know, a lot of big contract with Pharma to learn from them, but also to get enough cash to cross fund our diagnostic and therapeutic pipeline. For me, this is how I view it. But you have seen a lot, so you can tell you may have other.

[00:09:24] How Owkin is scaling AI in biotech

Philip Hemme: Yeah, no, it’s great.

Can you, and there’s a, at least a few questions I wanted to jump in, a few points I want to jump in, but Maybe, can you be a bit more specific or mention, let’s say, the other players to see how you, how they, how they compare to you? I mean, without like trash talking them, but just more like about 

Thomas Clozel: I think first, so we’re very different.

We AI for biology because we believe that biology is the background to discover new biomarkers that can feed the full life cycle. What we don’t do is chemistry, okay? Why are we not doing chemistry today? Because I think chemistry space has improved incredibly with AI the space of molecules, but there are still things that are unsolved, like protein to protein interaction and others.

And today, if you ask me, should I, should I use AI to help chemistry? Yes, absolutely. But I still need the real, the real good chemists in the lab. Can be from Germany, can be Switzerland, and I still believe that, you know, you really have still to get really good chemistry in the wet lab. And, and AI is a help, you know, so today this is why we choose like to, to not do chemistry ourselves for now.

The rest, finding new biomarkers, new biology, new targets, match it to the right population, use these biomarkers to find the right subgroup and indication, improve clinical trials, synthetic control arms, co write adjustments, and develop a dynasty tools to be able to deploy the subgroup is really where the rest, but, but what we are doing and, and I think nobody’s really doing that today.

So first we, I think the only ones that do the end to end pharma, we really want to recreate a mini Rush, if we can. Very very, very mini. Very small. But, you know, I think this is what we wanted. The best step. Other Yes, the best step. Other players are A lot of other players are in the chemistry space.

That’s the first one. Other players can be in the biology space as well. Try to discover new biology. If you take, you know, like in situ or Or, you know, recursion. They’re amazing, but they’re using cell lines. For me, as a clinician, you know, if you don’t have the macro environments even if you’re using salines or organoids, you still miss the microenvironment, the immunity.

And I, I don’t have a huge belief in finding new biology in mechanisms with such a translational gap. And for me, the only way to reduce translational gap is using patient data. You still have to validate in the lab, but you know, as my, my CEO is always saying like you know, cancer was treated in mice 30 years ago.

So we have to find a new model. And for me, the model is patient data as well. And biology has to be discovered in very large patient data sets, deeply phenotype using new modalities, such as partialized transcriptomics. It’s a very important one. We’re doing this big mosaic project. And you have to get with an angle of finding a biology that is not saline driven.

Philip Hemme: That’s Isn’t then the a bit challenging, but even today, I mean, you said you had a wet lab as well, but you don’t do the chemistry, isn’t It’s the optimal to combine, let’s say, in situ with you guys. So you have the really patient, stratification, micro environment, bigger like patient picture at the same time with the biology, cell line, really the chemistry, biology, and you can kind of combine and cross 

facilitize.

Thomas Clozel: So we, we doing it too. So we we, we created the Okinaw and in Okinaw, what we are doing is using organoids with Sun Bioscience in Los Angeles, they’re amazing persons. You should be interviewed like the, the, the founders. And so we, we’re using organoids in the lab as well to, to test our hypothesis, but first hypothesis are being formed in patients.

That’s the difference, you know? And then we, we use a really big lab in the loop approach where we are creating perturbation screens to improve our models. So I’m not saying like, you know, the lab is still a very central place, but I still believe the first type of biology has to come from the patients and we show it in the Mesothelioma paper we published in Nature Medicine four years ago, we could discover a new mechanisms using a prediction.

From patient’s digital pathology and reverse engineering the model to understand new cell biology or gene biology that could explain different phenotypes. And there was a new discovery in, in, in, in, in mesothelioma. But the first step was having a lot of patient data access, multi model, making a prediction and reverse engineering.

And then you go to the cell lines and the guideline model to validate. But for me, the first thing has to be having 10 times more data and data. And when I say data, it’s also private data in the best hospitals that are from the patients with cubioniders. I don’t believe in public datasets. I mean, you know, like if you build your company on public datasets and some have done that, you know, that’s, 

Philip Hemme: I mean.

The quality of the data influence massive. Is the quality of your models or like the, and it’s directly linked, so you need really high quality data 

Thomas Clozel: and diverse and Yeah. No, because, because dive no, but diver. The thing about diversity, everybody believe, everybody always remembers that patient diversity matters of course, but doctors, diversity matters too.

Patients are being treated very differently in Europe and in and and in, in the us even in France. You go to Paris and you go to Y, you have a different treatment probably. And for the same tumor, and so, you know, if you don’t capture the type of heterogeneity of, of biopsy, of treatment and everything, you have a model that is not robust.

And by definition, AI is not robust. And this is why you need to have European data, as we do at Okin, and not only US patients. This is why data from Fat Tyrant, for example, have no use to do research. And also just because the clinical data, they’re not multi modal, but also just US based. in community centers.

We don’t believe in community access so much. We believe in best academic centers where you have QOP non leaders that help you curating really high quality datasets to go to the single cell level, you know. 

Philip Hemme: Do you think Rush agrees with that? 

Thomas Clozel: I think Rush agreed. I mean, Rush, I mean, I mean, come on, like today, 

Philip Hemme: it was very big. 3 billion, the deal with Flatiron or something. 

Thomas Clozel: So the deal was huge. The deal was huge. And Rush wanted to, the story of Rush is simple. Rush wanted to make the first move with Flatiron. They really wanted to do, to be the first. Then about Flatiron, Flatiron was super useful for market access, useless for research, very, very little useful for synthetic control arms.

Synthetic control arms using just Flatiron data is not proved to be very efficient. Then what they did is about Foundation Medicine, which was great, they tried to reconciliate both. There was only like 30, 000 patients, you know, in common. What, what has, you know, and, and, and, and they just showed that it was very hard for them to get multi modal data this way to do really great research.

I think now with Aviv Regev, they’re really trying to rebuild. It’s. You know Spatial Transcriptomic Data Sets, as Okene has done before. You know, I’m trying to go to building a new data lakes, and I think they’re taking the right strategy. But I think that the Flatiron, Flatiron was great for market access, not good for research, not very useful in clinical trials, and the reconciliation was very hard.

Tempus is doing better in taking, you know, the multimodal data from scratch. What is different from us is we, we are embedded within hospitals, so we can continuously enrich the data sets. We can add new things, Spatial Transcriptomic, long reads, single cell. And so we can always like, you know, enrich this, and we don’t have a static view like, you know, this type of companies do have.

And I think like Rush, Rush has been a bit jealous of Tempest’s approach in the past, and I think now they’re really trying to take things from scratch on the Genentech side. 

Philip Hemme: Okay, I, I read the, I 

will link it below, but there’s a good article in Endpoints about the, the head of AI from Genentech slash Roche and what he, what he’s building.

And it’s, 

Thomas Clozel: it’s. I think it’s, I mean, yeah, I think it’s Regev I think Regev is trying to build something amazing. I mean, I think what they’re trying to build, but they started, I mean, they started from scratch and they do exactly like us, patient data, special asymptomatic. And I’m so happy that we have inspired Rush.

Maybe they won’t admit it, but we’re doing this for them. 

[00:16:46] Augmenting AI in biotech

Philip Hemme: Get this quote there. Now 

that’s, that’s, that’s, that’s a lesson. On the I want to know a bit on, on, you said we, you haven’t, our AI in Biotech hasn’t augmented, or hasn’t shown the augmentation yet. Can you go a bit into what’s the, I mean, differentiating basically the vision of, let’s say, what could be done in five years versus what has been done really today and let’s say in the last five years and where do we really stand?

Thomas Clozel: Yeah, I mean, I think, I think there’s lots of great companies around, you know, I can name them, Xantia, Insilico. You know Path AI, Page AI, Recursion, I mean, I think they’re all great, and we’re all doing quite different things, and we’re all a bit playing with the pharma. I think what happened with the pharma today, the pharma has first, first, the story of the pharma and AI.

At the beginning, they wanted to do a lot of internal innovation, build the things internally. Then they started to realize that the problem is they cannot do it for two reasons. They have a very hard time building their own data lakes, okay? Very hot, very hard time to regroup their own data. And second, they’re not hiring the best data scientists.

Why are not the best data scientists going to the pharma? When you’re really good data scientists, you want to work for Google when you’re young, not for, you know, rush on a valley. And it, it’s sad, but it’s true. I think, I hope it’s gonna change. I really believe the best in the future will work for the big pharma.

But today the, the truth is the young data scientists super brilliant. They wanna work for, you know, like probably a, a, you know, like a a, a metaphor or Google. I, I don’t know, but it not really, it’s not still not super attractive. And, and I hope it’s gonna change. And then they really understood they have to do more external innovation.

And they tried to play with a lot of different players around. And I think we, they all make proof of concepts and we all make proof of concepts with them. Sometime we did not bring pipeline value. Sometime we were bringing things that were incremental, but then the CEO was coming and saying, how my pipelines changed thanks to this company.

I want to see my pipeline changed. And so you had the height of discoveries that was amazing and tell you, Hey, we did that. But the CEO was, I want to see difference in my pipeline. And I think the, the, how you can bring direct pipeline value really mattered, you know, I think in the for the pharma. And I think all of us, maybe we went sometime to, to, to upstream to show pipeline value.

And this is why, you know, we did a lot of clinical trials improvements to really show things that were more direct than very early discovery or diagnostic tools to improve, you know, but I think, yeah, this was a problem for all of us. And I think today, the pharma. Wants to, to, to have less partners, a few partners, and there is, they will have one chemistry partner.

They have a lot of great companies around, you know, in Silico, Exentia, amazing. And they, they might have a one for biology, you know, us, and another. But I think in the future, they, they, they, they have less partners. Another question is why the pharma has not really acquired, but BioNTech was great team.

They were really amazing team. And they were, I think they made a really good move. I think today, you know, I think the pharma is still a bit scared of the cultural difference. They would be scared that if they acquire a company, people would leave. And I don’t think it’s true. I think, I think like people understand that it’s complimentary and I think people will, will stay.

But I still think this is maybe the reason why the big M& A has not really happened, like the Flatiron level. I mean, maybe, what do you think? What do you think? Why, why is this M& A not, you know? I mean, 

let’s say I mean, I can see that they are trying to wrap their head, their, their head around as well and probably still like going one foot at a time, like touching the ground, seeing, okay, and testing here and there.

And I think the culture thing is probably like, it seems like a really big shift. I mean, you guys, you call, I mean, you always have like biotech versus tech bio and we did this tech aspect. I can imagine, let’s say, data scientists or AI engineer, it’s not just the salary, it’s the package, but it’s also really from a culture point of view.

If you work for a big tech firm, or even for you guys, it will probably fit, or it will be much closer to that kind of culture that the person needs, or where the person will strive than, let’s say, a really big farmer. I can imagine, so. This, I mean, to your point, this, I can imagine, it’s a big shift, I mean, Even, I mean, for me personally coming from, let’s say, more tech, I mean, half tech, half biotech, but I can see in the industry there’s really a big mindset shift.

And I worked for, for some, like, a bit larger organization, biotech or pharma, and just the way they’re working and the culture, they’re great cultures, but it’s, I think, very hard to fit with someone who’s really like an AI or in tech. It’s really from a, just from an operation thing. I mean, I’m thinking about, okay, they’re still working, you know, on email and PowerPoints.

And they don’t have a project management software and some basic things, which for me was like, wow, it’s insane. And you’re a billion, you’re a billion dollar company, but you have no like project managers whatsoever, which I can imagine for 

some. Yeah. And I think things are a bit changing, you know, taking the part of Sanofi, like, you know, the head part of Sanofi looks more like a startup type of office.

And I think people are a bit less corporate. The mindset is a bit changing. You see that? And and but as soon as in the pharmatics, we, we start to have like people like CEOs or our head of BD global that has just had to be really like interested in the topic, like profoundly or even chairman. Take Sanofi, you know, like in Poland, really love AI and like the new CEO, the new chairman, sorry, for the good AI is really has a deep, you know, interest for it.

You know, take BMS as well. You know the team at BMS is really interested in, at the C level role. I mean, I think at the same time, it’s adding C levels that truly believe not, don’t only want to have an AI bread, but really want to understand with companies like. Like all of us, where, where there will be impacts.

And I think this is also, like, where we need to go. There is other company, the Pharma, that have way more classical CEO ship, 

Philip Hemme: Do they, do they really get it? Or are they still in the more curiosity, like

Thomas Clozel: I don’t know. I think some are really, I still, I still did diving in it and I think it’s not just only about being an AI company, having a brand, they really want to show things, but, but they want to show that we can bring pipeline value once again.

And this is where I think they want to say to the world, this is pipeline is AI driven and that, but, and, and this pipeline has shown success where we failed. This is where the hottest spot is for him. 

[00:23:05] How AI can work for small/medium biotech companies

Philip Hemme: I don’t know. So, I want to switch gears a bit from, I mean, we talked a lot about big pharma and pharmas, but, I mean, you have also all the, like, biotech companies, let’s say small and medium size who are, who are pushing a lot of, like, innovative, innovative drugs, and I’m curious in their case how, like, how they can really use it in biotech.

I mean, from what you said first was like, Oh, like basically Big Pharma or anyone has to be like AI first and have an AI pipeline, which means even the biotech company would have to internalize some stuff or should they like work with some partners for, let’s say, for, for discovery, for the clinical trials.

What’s your, or like, what’s your view there, really, for biotech companies, let’s say, or whatever, select this I mean like, clinical stage biotech company. 

Thomas Clozel: First, I think some are very involved in it, like, take names like GeneMab, I’d also have been very driven in AI and, and have a lot of different stuff.

Of course, the prime of, you know, I think like the, for, for AI companies like us, the, the opportunity is amazing to be able to, to get in licensing drugs from those, and, and maybe also just to, you know maybe have revenues, royalties. It’s very hard to get royalties with the pharma, you know, you get clinical milestones.

But royalties maybe, like, the biotech will be more open to it. Even if you prove your value. And I think so, this is it. The problem, of course, is biotech has less money to spend and doesn’t participate in the business as much. So, you know, for companies like, like the, the, like the TakeBio ones, the big pharma is a bit like, you know, can invest in the, in the equity rounds.

So it’s a bit more structural in a way. But I think they both are very important at different levels to work with. 

Philip Hemme: But not just like partner with you guys, but more how you perceive it. Basically, what should they do? I specifically, I got like, what do you think they should do? Because I got the question from, let’s say, VP Discovery at the biotech company, had an employee with billion having next gen biologics platform.

And she was like, Oh, what’s, what do you think about AI in biotech? Like, what’s basically kind of, what should I do? How can I use it in biotech and how can I use AI? Like, what do you, what do you, like, what would you think? 

Thomas Clozel: If you ask me for, for like a single asset biotech, you know, midsize, it depends on the fields.

I wouldn’t go for what brings the most direct value, which is for me finding the right subgroup. Everybody knows that finding the right subgroup for your drug can improve the probability of success. But matching biology to the right population, finding the right subgroup, or maybe the right indication, is not easy for a startup.

Most oncologists, they never know which indication they want to go to, and then they have to build very expensive basket trials and take a lot of risks. I would go, if I was a single asset, to take a company that can help me on identifying the right subgroup and the right indication for my drug. Before I go, help me positioning the drug really well.

I mean, of course I’m taking Akin use case, but you know, I, I would, I would start with this use case very simply. And , I can do that. 

Philip Hemme: And what about a non, a non OIN use case? 

Thomas Clozel: no, but about new case? No, I think, no, because, because the problem of a single asset is not having a better pipeline, a bigger pipeline is, is even more having the success of your first drug.

Right? In

Philip Hemme: this case was a, was a platform use case more, it was a platform company actually with, with research. So. 

Thomas Clozel: So in the platform Nobody, I would, I could also work within Silico, you know, or on, on, on the improve, on, on augmenting my pipeline with some of the new drugs, you know, why not, you know, I think it both are complimentary.

I mean, I think I would, I, at least I would choose one use case I don’t want to explore but I wouldn’t try different things at the parallel and I would really create a. A little common lab where people can meet. I mean, I love this idea of really having a common lab where people can exchange a lot.

Or, I would merge. Why not merging with a mid size? I think mid size biotech and AI scale apps should merge together in the future. Because this is where I probably could work as well. Why not, you know?

[00:27:18] Always looking for more data

Philip Hemme: It might happen actually. Let’s see, let’s see. No, that’s, that’s, that’s great. I mean, we, we touched the, we touched a lot of things already.

Let me just let me just check. Let’s talk a bit about, about, I mean, on the competition, I’m still like, one thing, I’m still wrapping a bit my head around is like, you, you said something, I think a podcast that you mentioned, you wanted to have 10 times more data, or let’s say 10 times more and better data than anybody else, like, where do.

Where like, this is a, a wish, but where do you stand today? Like compared to the biggest? 

Thomas Clozel: No, we are, I mean, we have, we, we look, look in a look in our papers. We publish a court in Otium on 3000 patients, you know to find some new biomarkers in new biology was in nature medicine. Today we have another thing called Mosaic Mosa is the largest special for systemic efforts.

We are getting towards 7,000 patients dataset with five sensors. This is the goal. And this is really to build, you know, a huge effect on the, I think it’s going to be huge for the, for the field, because we’re going to be able to show that we can have huge data sets. It’s a 15 million investment, 5 0, you know, and we are building the right pipeline to analyze it.

Spatial transcriptomics is the only way to see visually genes expression and, you know and and, tissue organization. It’s, it’s an amazing play for computer vision. And I think this is something, this is the type of modalities that can also capture the macro environments. It’s the only way to capture the microparameter effects.

And so, you know, we’re working with like, you know, Nanostream, TEDx, and we’re really building this and five amazing centers to build this thing. And we were first, and we were really happy to see some competitors climbing on our feet. Like, no HH, no formal alpha coming afterwards. Once again, happy that we inspired people.

You know, it’s, it’s, it’s we are really building something that that I think, but it has to be big, because if it’s not big, if you don’t have a size effect, you cannot validate things in the right way test it train things, test things, find the right hypothesis, and you’ll never know if it’s a data number thing or just a, you know, and, and you need to also capture the heterogeneity, so this is the type of efforts we are making, very big bets, gross products and we, we really aim to go super high.

Philip Hemme: Okay. Right. In the, I mean, that’s, sounds high, high enough, yeah, high enough, I mean, in the thousands of patients, it’s already high. And I guess what you said is, the quality of the last quote, 

Thomas Clozel: the court published. Yeah, I mean, everything is about the quality and the multi modality. And I just want to just re explain why multi modality matters.

So you understand special asymptotic matters to capture the microenvironment. But in general, when you work in cancer and in oncologies, right? So when you work in cancer, you never know which modality will give signal to the prediction. So if you do only pathology, like PATH AI, you know, PAGE, sometimes pathology in lymphoma does not bring a lot of predictive signal.

You really need other modalities. Even if you anchor your model of pathology, you need to add multi omics. Sometimes, you know, CD scan or MRI are, or proteomics, you need to add other modalities because you never know where the signal will coming from. And so the multimodal approach is, is really complicated.

But it’s the only way to capture how you can understand the different skills of biology. So I mean, for us it’s not only about being multimodal, it’s also being multiscale. How you go from a gene to a cell cell to a tissue a tissue to a disease capturing causal signal. And this is the very thing.

So we, we really want to go to, to, to try to understand causality more than correlation. And the only way to do that is being multi modal and multi scale. We’re not there, but it’s a big work. And this is really the goal. 

Philip Hemme: But that’s, that’s where also AI makes total sense, because you can process tons of data very rapidly and figure out.

the relationship, the causality, like, really well and much, much better than any human can do. 

Thomas Clozel: I mean, yeah, but we haven’t proved, I mean, nobody, even us, we are still trying to prove a good explanation. I mean, before causality, you have other, like, success levels, like good explanation of a disease or something like that.

We have to go there, too. But, you know yeah, I mean, and what’s also interesting about data is today, you have to get to foundational models on every data modality. So here you need like millions of patients, you know, if you want to build a foundation. And Okin is, is spinoffing his foundation model for biology.

The first one that is going to be multi scale on our dataset. But we are really, you know, being able to work in millions of patients on pathology, millions of proteomic patients. And so this is where you need like, and then to connect the scales together, you need like multi modal longitudinal datasets.

So you need different types of data and numbers when, for the tasks you want to optimize. But you really like, you know, we, we for you, the, the, the number of data annotated, you know, not annotated, sorry, unlabeled, because you don’t need that. It’s going to be big as well. 

Philip Hemme: I looked into one company, actually, Valo in, in Boston.

I know quite well, David Berry from, from Flagship for, for almost 10 years, actually. Yeah, me too. And, To my understanding, I mean, they’re going a bit in a similar space as in having a lot of human data and then building a pipeline on it. And from what I see or from their presentation, they are also in the thousands of patients scale.

So how does that, how does that work? 

Thomas Clozel: Yeah, so I, I, I, so I’ve never read, I, So first, like, I think like the difference between that, I mean, the, the, we, we have discovered like, you know, very reference ways to analyze pathologies that we publish, you know, people are using and the multi modal, I, I haven’t really read so many papers in the, in the literature, in nature medicine, like us, or about vital technologies.

So I don’t know much about it. Okay. I haven’t read like science, so I don’t know. And then the thing is like, we are building like, you know we are building like, but I think David is great. He’s probably have an amazing team. No. I mean, the idea of in licensing drugs is a bit similar.

We’re also building dynasty tools. And we are partnering with 41 of the largest hospitals in the world. I’m not sure which partner For us, it’s really a win win partnership with the hospitals. We try to publish, we try to work with them. We are generating data against access. We also try to build a federated ecosystem of hospitals.

180 of the best Filipino leaders. I, I don’t really know about the, the, the, the, the partnership and the quality of the data. I can say, you know, so I, I really don’t know much about it to comment. 

Philip Hemme: Okay. I don’t know exactly either, but I mean, they mentioned acquisition of data, so I guess, and I guess they must, I mean, the data has to come from somewhere and probably the leading hospitals in 

Thomas Clozel: when you acquired that, so.

[00:33:58] Anonymization of data and federated learning

Thomas Clozel: So the, the thing about tech first, I’m a doctor. I like data to stay in the hospitals where they’ve been created. I don’t like to acquire, I like to have a remote access. I don’t need them to acquire them. I don’t acquire, we don’t acquire data. We have remote access to it because it’s privacy preserving. I believe in privacy.

Yeah. If you, if you wonder, data is c is the new oil. Try to wonder for every company, what is their policy? What is their vision on data access? Is it a sustainable way to access data? Is this, is this good for the patients? I mean. For Okin, it’s easy to privacy preserving access, you know, federated, to break the research silos.

We really want to break research silos and competitive silos between centers. You know, I think that at least it’s clear. We were the first one, and we’re still number one in federated learning for clinical data. This is why Google Venture came in years ago. I think like today, today I mean the broking of data, it’s something that I believe will happen and everywhere, but it’s not a very amazing vision on the, on, on, on data to, I mean, as a doctor, it’s not mine.

Yeah. 

Philip Hemme: You, you switch to the next topic on federated learning. It sounds amazing, honestly, and, and you’ve proved that it’s worked, I mean, the, the Melody project was, was, I think it was 10 Pharma, you proved also that it worked with, with hospitals. Yeah. But the other model of acquiring the data also seems to quite work, especially in the US.

I think it’s more a question of also of price, I mean. If a hospital, I don’t know what kind of deal we’re talking about for this, but if it’s in the millions to acquire, I mean, I think a hospital could be interested and I, like, I guess there’s a question of price and then there’s probably maybe some workarounds around privacy.

Let’s say if you anonymize the data, then you cannot identify the patients. Then it’s, it’s, so 

Thomas Clozel: you cannot, that’s never true. Philip, that’s not true. No, it’s not true. If you take, I, I put, I put 40 hackers on any data set. I come back to the identity of anyone. Today, there is no anonymization of data that works out.

We have to be very clear. None. 

Philip Hemme: None? Why do, why do Apple claim that that anonymization of the 

Thomas Clozel: Well, I’m pretty sure to try to give us data and put like 40 hackers. Why? Take a, take a pathology slide, okay? Pathology slide, by definition, if you take the name out, whatever, you can anonymize, absolutely anonymize it.

However, at Token, we’re able to predict genes based on this slide. We can predict signatures of genes on the pathology slide. If you predict for the signature of genes, you’re coming back to the genome of people, and you can really get back, and you can cross with genealogy database, 23andMe, whatever.

Today, I, I, I, I personally don’t believe that there is 

Philip Hemme: You predict the gene on the top level, you don’t predict the sequence to then identify the patient, like 

Thomas Clozel: You sell predict, you still predict signatures of t-cell or B cells, which is quite, you know, identifying. However, I mean, I’m just telling like the, the, the roots that could, like, predict more of this is the tumor.

But you know, you can still, I, I I don’t believe that, I mean, we have to prove, the only way to prove that something is really, is to put a dozen hackers on the, and see if they cannot come back to it. I mean, for me, I mean, I mean I’m not saying that federated or aggregation and I, I’m still think that it’s fine to buy data sets.

But I think both have to happen. In Germany, very hard to do data brooding, right? So that works for the U. S., but in the global scale, correct. 

Philip Hemme: I know the German problem, but I am curious on this, like, where is the model because, I mean, when you talk, it sounds like, yeah, it’s good to hear you both, because when you talk, it sounds always like the only solution is federated learning, but that’s from the reality, I think, yeah, okay.

Thomas Clozel: Yeah, reality is between both. However, just can I say that, however, there have been deals in the past that were exclusive deals on data generation with hospitals and scale up, and this is not okay. We have to all be okay that we are treat, we are trying everybody of us everything by is trying to treat patients and help medicine.

And if you’re making exclusive deals that your data cannot be even used for internal research within the hospital. You know, of course there is no commercial, right? When you generate data, that’s okay, but you still want things. You wanna, you don’t wanna have a hold on data sets for anybody else.

And we’ve seen it in the past. I don’t wanna name people, but you know, we know. That there’s been deals where the people couldn’t do research, they couldn’t participate with anybody else on even the, the, the, the, the, the data sets that were not generated before, and I think this is not okay, I mean, we have to see that if you put a hold on data sets and a key lock that nobody is able to use, you’re not helping research.

And so we have to find, we have to promote companies that are really trying to, to, to help open this system. 

Philip Hemme: Well, I guess this would be a, I mean, this would be a very challenging It may be a very challenging problem because I mean, data is basically new gold or whatever. And your model also, I mean, quality of the data improves.

So if you increase the quality of the data or you decrease the quality of the competitor’s data, maybe your model can be better or whatever. I mean, basically what Google is doing with YouTube, they’re not really sharing all the YouTube data. They’re Gemini. I mean, 

Thomas Clozel: Of course they’re not. Of course they’re not.

No, no, but, but I mean, yeah, I think you agree. I mean, but, and no, no, no, they wouldn’t. But I mean, just take the example of telemetry. I mean, take nice example of telemetry. Melody was the first time people were collaborating and it was the largest database of small molecules virtually. But take the example of glioblastoma, very important to my heart, a brain tumor.

You know, glioblastoma is 35 clinical trials. There is only one positive. I think it’s Novocure, an amazing company really. And so if you take there is so many silos of negative trials. Happening in different pharma anywhere, and, and they will never share it. Sharing, nobody will, they will never share the negative Klingon trial.

If you do federated learning on, on that, you could really capture new, you could have enough data to find some, maybe some new biomarkers for glioblastoma. It doesn’t mean you will find a new molecule, because there’s still a long way to do, on the chemistry side. But however, it’s very frustrating to know that people have silos of negative data, not collaborating, and that there is a technical solution.

Why, why are we not doing it at Okin? They have customized a small market, so we will have very low support from the pharma. We won’t have enough money to do it, it costs a lot of money, and it’s a big headache to work with multiple pharma with federated learning. So, you know, we’re just not doing it because we don’t have a business case.

But it should happen, I mean, the data are here. 

Philip Hemme: I mean, yeah, yeah. Feels a pity. Yeah, let’s see. I want to jump on what you said about the, that it’s mostly a European problem. And I think that’s, you’re touching a point where I think it’s probably one of the biggest value propositions of Orkin as in, I mean, GDPR and let’s say Germany, even Switzerland are super strict on, on data privacy.

So I guess, and also even for you as a Orkin, as a business, I mean, I think that most of your employees are based in, let’s say Paris or Europe. And I think also most of your pharma partners are Europeans as well, even though you have some in the U. S. as well, so. No. Not true? 

Thomas Clozel: No, no, no, no, no. On this side of the, I mean, no.

First, I don’t think what is a pharma or pharma is that all global. But but no, no, no, I think today we have more U. S. partners than Pharma. In the Pharma space, we have more U. S. partners. 

Philip Hemme: Is the, are the U. S. one partnering with you for the European data or as a, from a global perspective?

Thomas Clozel: Everybody’s, everybody’s, people are partnering with us for two reasons. We have global data. We have as many hospitals in the U. S. even more than in Europe. But we can, we have both. The second is the fact that we can enrich the data. When you acquire data, you cannot enrich it. You cannot go to a new analysis, accessing the samples.

We can, theoretically, access samples from the patients. Of course, in a, in a very strict ecosystem, and with the but, you know, we work with the patients, we are really relation, we have a relationship with hospitals. And so people work with this, this, this federated access data worldwide. Then they work with us because we, we can bring the AI layer and the biomarker, and then we can implement this within the life cycle of the drug.

We are a one stop shop for the pharma in biology. This is why they work with us. We can do clinical trials, diagnostics new therapeutics, indication discoveries, so we’re discovering new targets. So I think today they would like to work with us for that. And and of course I work on the chemistry with great players in Silico, Exentia, whatever.

I think today this is where, where we stand but we don’t give access to our data anytime. So nobody has access to our data. We are in the middle. 

[00:42:18] Becoming a biotech company

Philip Hemme: Yeah, that’s good. Maybe last point on Oken is the, the transition to being let’s say full fledged or, or full biotech. I mean, you You were saying, I think you, I heard you you, you want to reinvent the pharma biotech full stack.

You mentioned Tesla building everything end to end. 

I mean, 

and I mean, you hired some, some, some big people. I mean, this is Andreas, the CFO of Merkur Partners who joined you and he built Merkur Partners, co founded Merkur Partners, co founded. That’s a biotech company. You had the global SVP BD from Sanofi.

a really like heavy, really biotech, biotech people, I would call it biotech or drug development people. I’m curious on, on where, how, how did, I mean, how are things going there on like building your own pipeline? I mean, I think it’s been, these people joined a bit more 12 to 18 months ago. So how, how did it go there on the Like on the pipe.

Thomas Clozel: Yeah. I mean, so, so first we, we build a tech. Yeah. Thank you for mentioning them. You know, we build a tech bio and it’s led by Jean Philippe Vert, who is one of the very top specialists in the world of AI for biology. You know, he used to lead a Google brand in Paris and, and professor at Berkeley and then Paris at Le Muin.

So on the, you know, we have an amazing data science lab and people that we also hire like a really great medical team, you know, Vassily Sumilis, who is a professor of immunology and, and immunology. To have a really medical expertise because we believe it’s important. On the pharma side, what’s, what’s hard is to find people from the pharma that are platform minds, you know, that have this that are, they have the sense of urgency and really want to accelerate things.

It’s not that easy. And I think, yeah, Albon de Sablier, our Chief Operating Officer, came from Santa Fe, and he has this sense of urgency. And Andreas brings this, like, you know, four IPOs. So I think we are building up. We’re hiring SVP to discovery. You know, we are hiring people in the labs that have been through you know, so I think this is the hardest part, for sure.

Finding the right people that can bring this this value. Still, understanding the transactional side of the tech is very hard. But I mean, there are people in the pharma space that do that. In the venture capital space, You know, venture capital is another story. Very few people get both, you know, understanding what representation learning means, how you find the right data and understanding, you know, the, the, the, what is a really good pipeline or the diagnostic.

I mean, it’s pretty much a very rare species. It’s like finding a panda in a zoo. You maybe have one every a hundred zoo. I think like a mix of baby pandas around. Yeah, baby panda in a zoo. And, and and I think like they eat. I mean, venture capitalists ate way less bamboo, but you know, we, we, we still really have to find them.

It’s a rare species. 

Philip Hemme: Yeah, that’s, no, that’s, and so specifically, where do you stand now in the, in the pipe? Can you, do you, can you share anything? 

Thomas Clozel: Yeah, so we, we’re going to have, yeah, I cannot share with anyone else, but we have two dynasty tools approved, you know, the first one is being deployed. It’s called MSI Intuit.

The second one is gonna come very soon. We have the first phase one coming in. And we really, you know, are working with the pharma. We are on 20 years of chemistry. A really, like, best in class drug. Everything we built in diagnostics is first in class. Everything we do in diagnostics, in therapeutic, is best in class.

But once again, we are really trying to show, we, the big thing for us is try to show the world that it’s not just another pipeline. Because a pipeline is a pipeline, you know, and you can have a hundred molecules if it doesn’t show Really great success. And I think for me, for everybody in Silico, we all have the same problem, you know, it’s showing that our pipeline or value of health, showing the pipeline is different.

Philip Hemme: Everyone wants the hard one. 

And 

I’m curious on the type of drug, is it like small molecules only or are you agnostic of the type of technology? 

Thomas Clozel: No, we’re, we’re agnostic today. We, we, you know, I think we, we, we, we did this thing today. We, we’re going to announce a big, a big partnership with a very famous CRO on small molecules.

But we are open, you know, because we, we want to discover biology first. If you find a protein at the surface, they can be only targeted by, you know, by biology. And there is good companies doing AI and, and good CRO on the, on the, on the, you know, the biology side. Biologics is also important. We, we, we couldn’t restrain us because we want to discover new biology first.

So today we open. We don’t have any internal expertise on one or the other. We externalize this expertise, working with the best CRO there. It costs a lot of money, of course, but you know, we have to get there. 

Philip Hemme: I think, I mean, from my view, it’s always something that’s not puzzled me, but I was a bit cautious about that.

Most of the AI biotech, especially the ones doing their own drug development and having, they’re mostly doing small molecules. Which I think it is great and there’s still need for some amazing small molecules. But if you look at the pipes, most of the pharma, like 90 percent of the pipe is biological, biologicals or advanced therapeutics.

And I haven’t seen that many biotech. We have like breakthrough biologicals thing. And the first that came to mind, where I was like, oh, wow, it was exciting to, I think, mid of this year, they announced that they have, Now they, they can, they can generate antibodies, I think, or biologicals. And I was like, okay, now, 

[00:47:29] Small molecules

Philip Hemme: now it starts to be like really, I mean, probably really interesting.

Thomas Clozel: Very smart companies. Yeah, yeah. No, I, I, and there’s so many mechanisms, especially in the microenvironment, small molecules is not the solution. You can’t. First, it’s biological first, but we don’t, you know, once again, it’s working with CROs, but the problem of biologics is, of course, like, in a way, small molecules are a bit, a bit simpler, probably because you have less optimization, you have less stuff to do, but the patent is shorter, there’s a patent problem as well creating a bit less value.

I agree with you. 

Philip Hemme: Do you think there is a, I mean, or at least from my view, was a bit like, maybe the, the biological complexity? At the moment, it was just too complex for Gen. AI or AI in general to really, to, to, like, manage. Is that one of 

Thomas Clozel: the I think so, but also, also, also Yeah, probably, probably the, the, and this is, this is the first part.

I think the second part is accessing privacy data sets. It was the idea of Melody, accessing small molecules, libraries from the farmer, not public ones. And, you know, I don’t think there’s enough like access to really high quality private data of really high quality with once again, the idea of continuously, I really insist on the idea of being able to continuously enrich your data sets.

What happens again, we can, we can add new analysis if needed for the same patients. And this is really what matters, because there is so many new technologies that are coming in, you want to be able to do that. And I think this is what’s missing in the chemistry space. A lot of people are using, like, public data sets to work on the chemistry side.

And, you know, there will be a limitation too. But I think, I mean, come on, Insilico, Exemshar, they’re all doing a great job. They’re doing great stuff. It’s crazy. I, well, 

[00:49:10] Limitations

Philip Hemme: I, really, I’m playing the devil’s advocate here and trying to challenge you. I’m trying to understand, really, like Where are the limitations?

What’s hype? What’s reality? Where is it standing? So I think, but I think you did a really good job of presenting, 

Thomas Clozel: yeah. Thank you. Thank you. First limitation is data. Technology is the second, but data is always the biggest limitation. First. Always. And then I think, yeah. But I think, and then the other limitation is time to show proof points.

Time to show that, you know And this is the terrible thing about this field. And once again, the terrible thing is waiting four, five years. to get your face to be showing that you, you should prove something. Of course, you can accelerate and whatever. How? This is so frustrating. I mean, it’s, it’s, but it’s true for every biotech, right?

Philip Hemme: I mean, health, yeah. Drug development. I mean, you can bend time or bend a bit time, but once you know a bit the complexity of it, it’s, you cannot cut too many corners. I liked what, what Mark, the guy that actually, we, we talked about it, that you watched the episode, but he said that. Yeah. It’s very hard.

Great episode, congratulations. Thanks. Thanks. It’s very hard to find actually the balance between speed and quality in a clinical trial because you’re, and you have so much pressure to get the faster in terms of capital, capital limitation and you want to help patients, but at the same time, if you compromise too much on the quality of the trial, It can backfire like pretty, pretty big times.

So I like that. I mean, it’s, it’s, it’s not, it’s not like a, I mean, in tech you can cut a bit more corners and probably you can scale maybe faster. But in healthcare and with patients, I mean, we saw Theranos, we saw even 23andMe. I mean, when you cut too many corners. Yeah, but it’s 

Thomas Clozel: a great, I mean, but I think 23andMe is better than Theranos.

But still, 

Philip Hemme: there are some No, 

Thomas Clozel: no, no, it’s discussable. Yeah, but I like it because, yeah, but I like the fact that 23andMe takes patient data and finds molecules and stuff, and I think they’re doing a better job. I mean, one thing that is also very limiting is the Phase 1. Phase 1 is so in oncology. It’s so limiting.

They all look the same. You never know. I think phase 1 is a big topic. We are trying to improve phase 1. You know, trying to, not finding evidence, you can’t, but insights, with counterfactual outcome, building like, trying to compare the response, the actual response to a predicted response. Sorry. But I feel like phase 1 in oncology especially is a big limitation because you never know what to think about it.

Yeah. And we try to work on it. That’s a good point. 

[00:51:51] Growing up with billionaire biotech parents

Philip Hemme: I want to switch a bit topic more to the personal side and, and talking about more your background and, and kind of your mindset. And the first thing that comes to mind is, is talking more about your, like, let’s say your parents and where, where you came from.

I mean, I mean, of course, I mean, your parents built, I tell you, I mean, super successful. One of the few, let’s say billionaires in biotech, at least in Europe. And must be, how did it like, feel and how did it like, impact you, growing up that way? 

Thomas Clozel: Thank you. So first my parents, my parents are, first my parents are doctors.

You know, they’re not like lawyers, like some of the CEOs are doctors, they really think about their patients. And they’ve been like putting up drugs that are amazing. Like for example, take the example of Q Vivic, it’s their last drug and it’s building up. It’s an amazing drug. It’s revolutionizing sleep. I take it all the time.

You dream more. You sleep so much better. And of course, it’s not the success of, of, of of WigoV today. However, I believe that sleep and obesity are both very important for your life. And I hope it will come. And I think my patients are just like, extremely like, idealistic also very, sometimes maybe too much.

Doctors, I think that my, my, my parents give me the first, the, the, the patient first thing because they’re doctors. And I’m a doctor too. And, and, and they teach me that patients matter first, and when you bring patients value, there is a business. At the beginning of Actelion, they were building something for, you know, like, for, for for permanent hypertension.

People were telling them the market is too small, whatever, whatever. They were like, this is the first one. And, and the second thing is, I think they also, like, show me that you need to, to trust in what you’re doing because don’t, don’t listen too much about people, you know? I mean and, and my, my father is very, like, go forward.

But I think that I listen to, to trust in my, my instincts as well. Listening to advice, but I’m And the last thing is not being afraid to fail. If you do something good in health, you know, really, and I really promise, I don’t feel the pressure to fail. And I think they haven’t either. It will give you some strength because when you’re a doctor, you see people having very severe things, diseases, you know, that you can cure.

And it makes you very humble about life and really help you to, to say that if you fail what you’re doing, there is one thing in life, which is your health. Health matters more than anything else. So my parents have been very cool about failing, even when I was a kid at school. If I was failing anything, they were always super cool about it.

Not really pressurizing me anything and giving me the right values, and I think it really helped me. And even when I stopped being a doctor to to to they were like super supportive, and and some parents could have said, you should stay with your job. You know but but however, they’re not really helping me day to day, you know?

I’m I’m they haven’t helped me on deals. Ha ha ha, still. Because they want me to succeed, and that’s helping me. I mean, the American way, really, that’s, and, and this gives me, this gives me the, I wanna make my own money. I wanna make my own job. I wanna make my own success, and I wanna do better than them.

That’s kind of what driving me and the show. I’m that doing, I’m not working. I never work with Act Docia. I’m making my own stuff. And and yeah, I, and I my father always made the joke saying, I’m the, I’m the, I’m the father of, of ma Arts. This is how he present me. Just for the joke. And I hope, I hope one day will mean something.

Today, not that much. Today, like, okay, who is this guy? Well, you know, I don’t know. But it’s, you know, but like, my parents always take huge bets.

Philip Hemme: I like a lot the, I like a lot the, the, inspiration lessons part, but also very hands off and like, giving you the, the freedom to explore, to fail, to, To build yourself and to build yourself differently.

I think it’s very good. A good mix. 

Thomas Clozel: Very. And being single-minded. My, my fa my father, my, my mother, they refused all the boards. They, there are not in any boards. They’re only in my father, only in I board, and my mother say no to hundreds of boards. You know, honestly, just because they’re very, they also teach me to be single-minded, single about one thing you wanna do in life well and just do it.

Don’t, you know, there’s so many people that love to be bored people, being in the X, they don’t care. You wouldn’t, I mean, they’re really out of the radar at every point of view. 

Philip Hemme: Yeah, I can tell you, your dad is very focused also on PR, or at least he, at 

Thomas Clozel: least I’ve never managed to do it. Yeah, he never do anything, he doesn’t care.

Yeah, he never do anything. He doesn’t, he doesn’t invest, he doesn’t do anything else, you know. 

Philip Hemme: It’s pretty impressive, I mean, I love it’s pretty impressive actually, I have to say. 

Thomas Clozel: It works, I can’t lie. No, no, I mean, but, but they are doctors. Always remember, they’re just doctors. They really don’t see, they really believe that all the patients giving their drugs is their own patients.

They really are. 

[00:56:37] Becoming unfocused and having fun

Philip Hemme: On, on on focus, it’s, it’s funny because, I mean, I mean, she, that is very focused and you apply this, but at the same time I heard you in an interview on, on Fast Company about that you want to be more unfocused and Being open to every opportunity versus being ultra focused and that lot of VCs challenged you on that, of where is the focus 14?

Thomas Clozel: Yeah. No, I think it’s, it’s exactly, so at the beginning I had so many board members, a few members say, you have to do one thing, take one modality pathology, just do one use case. And I was like, I, I want to discover, I wanna build a net of technology that will I help to understand where the right use case.

Because once again, this is new technologies for the healthcare, we don’t really know. What we’re really discovering and how much it’s going to impact patients tomorrow. I want to be way more open and a very more unsupervised mindset. And, and they were all, I know I need to be focused. And I, I’ve never agreed to be focused in Farrokhine.

I’ve always agreed to go large and to be ambitious and try to cross fertilize things. Because it does cross fertilize. Of course, like I got one board member that fire me once and you know and 

Philip Hemme: I see, I guess you, you learn, you learn from your dad there. No, he had some lessons to share.

Thomas Clozel: The lessons from my dad, which is like Was an activist 

Philip Hemme: investor. I heard about this. Chloé was like, whoa. 

Thomas Clozel: Yeah, yeah, yeah, yeah, yeah. Oh, yeah, yeah, yeah. Oh, yeah. No, but you, yeah. I mean, I mean, I mean, the board, I mean, the board is a big topic, you know, for a company like us. Because, you know, it’s very hard to get board members that get to the right level of understanding of what you do.

We have some very, very but it’s not an easy topic. So, no, no, no. And, yeah, I mean, I, I’ve always fight for what I, what I believe. And if this company is all of us, we are moonshot companies. It’s in situ in Silico Exentia. I mean, we, we want to try to change big stuff and at the same time, it’s, but you know, as, as of course, all the big tech people have we’re not like an operation.

Hey, I mean, we have to be in some ways and we, most of us will fail. Maybe me too, but I think it’s, it’s fine. We, we all have to, to be in this mindset and, and. Out of focus is the, is the way to go. Try to 

Philip Hemme: I like, I like that choosing the case for when to be ultra focused and when to be less focused.

And I can see for me personally, it’s something that I learned recently and I try to to be much more like Malleable or how you say it in English, more like flexible rather than, I was like very rigid, like always ultra focused, always deep work and like very rigid. And I find that in some cases, whether it’s with a company or whether it’s even personal or whatever, or, or in the time of project, sometimes like choosing the right setting of focus works actually better.

Thomas Clozel: I, I like it exactly. That’s good. And I think also it’s the same when you, when you, when you go out for a walk and when you are sitting at your desk. When you’re sitting at your desk, you’re in a mode, it’s a concentration mode. You know, you really focus. When you go out to walk, you’re more attentive. It’s called attention versus concentration.

You’re more, you know, ideas coming and flowing and everything. And I think to be successful, you need both. You need to walk out and be sitting at your desk. You need to focus on the mission and you need to have very precise QP key objective and key results. You need to have. You need to know what you’re solving.

Sometimes people don’t know the problems they’re solving. You need to have a clear vision on data. You need to know the technology you wanna be number one on. You need to be always number one. We are number one on multi scale AI for biology. But then you need to be also attentive to what the world, to, to do ideas and come in, you know, need to be both.

I think to do a bit of both. And, and, and usually people try to, to make you only go for the focus side. Yeah, that’s, 

Philip Hemme: that’s, that’s, I like that. On the, maybe a bit on the, on the, on the other personal topic is on, on fun. I like a lot, like that, at least it seems, but even from our discussion that you’re having fun in what you do.

And yeah, I remember you, you were like whatever doing interview and wearing like a pink elephant sweater and I messaged you about this and then you just posted out the JPM blue bike. It’s, it’s small things, but first it’s about having fun a day to day and even making a bit more public and, and, and have it sharing that fun.

Can you like, first, I mean, I like that a lot. I can connect to it and relate to it, but can you elaborate a bit more on like, maybe what’s, what’s behind like it? 

Thomas Clozel: First, yeah, thank you. At first, I think like, once again, I, I know what’s important. I mean the, the, being a doctor and oncologist for, for not that many years, but enough to understand what that health is, the only thing that matters, your health, your family, your friends, the rest is a, is a coup d’etat.

You know being a billionaire you’re still going to die. Not going to absolutely be more happy. I mean, I know that I, I feel, I think I understood my own way to things. It’s like health really matters. My family, my friends, the rest is a great top on and, and it’s just an adventure. That’s the first one.

Then I, I think, you know my way, you know, I think I love to have challenges in life. It makes you, makes everybody happy, makes me happy. And there are doable challenges. And, and Okin is an amazing challenge. Very hard company. You know I need two aspirants after every day, or each board four aspirants probably, maybe 20 and, and and but it’s also super doable, maybe doable, maybe we can achieve it, and it’s just so amazing.

And the last thing is, I’m surrounded by so cool people we, we keep the culture, I’m talking very childish culture. And that is, you know about about about being, you know, together we are playing games with the Pictionary yesterday, you know, together. And I think I love the people and it makes me so happy to have people that I actually really do love.

I think we are 320 people. There’s nobody I could not have a lunch with. There is none. And that makes me so fulfilling, you know, in a way. But, but, but the most side of it is, you know, if it fails, it fails. I promise I won’t be, you know, because life is bigger than that. And my, my, my, my human adventure with my colleagues is the best thing I’ve created in the company.

And I really mean it. And I love so many of them, like from the bottom of my heart. So I think it’s something that makes me just giving a bit lighter and lighter hearts because, you know, and, and this is where social doing something like, you know, we are all in the same space, right? Having a social impact in life gives, give you less pressure.

Because you did your best, you know, like, you’re trying to help, and people that just work in the big tech but don’t understand what the impact they want to create socially, I think they’re missing something at least it gives them more pressure. And if I was building an NFT for, if I was building an NFT company for whatever, I would feel way more pressure.

I haven’t succeeded, but I absorbed. NFT 

Philip Hemme: for what? Well, NFT for, 

Thomas Clozel: I don’t know, for us. For 

Philip Hemme: biotech, I don’t know. 

Thomas Clozel: I don’t know. I mean, there’s great companies around that as well. It’s just not my stuff. I like that. But you invited, I hope you’re going to, I hope you’re going to come on our, please, at J. P. Morgan, we have to do our tuk tuk tour.

We have a tuk tuk at J. P. Morgan. It’s going to be, it’s going to be, because I want to be outside. I hate to be inside. So we’re going to have hot chocolates. We’re going to be outside. We’re going to go by the water, by the Embarcadero, come back to Union Square. So I’m, I really hope we can come and everybody can enjoy our tuk tuk 

Philip Hemme: meetings.

I will show the, I will, I will show the picture post it. It looks, it looks really cool. I like, yeah, it’s, it’s, I like this, this, I like this, this mindset. Because on, on pressure, I mean, I like your view that because you are doing something with high impact, this relieves pressure. My feeling also is that quite a lot of people, let’s say, in the industry, because it’s such a serious topic and high pressure, I mean, you’re talking about people with, like, cancer or, like, really heavy stuff, sometimes they become, I feel like, very serious about it and, like, a bit, let’s say, not boring, but it’s a bit judgment, but a bit, like, too serious, too stiff.

Which, I quite like your view of it, because basically it’s the same, like, whatever, the same cause, but the interpretation, how you go about it is completely different, and I feel like, 

Thomas Clozel: yeah, I like that. I mean, I think both, both make sense. Don’t get me wrong. Both make sense. I mean, I would, I’m, I’m, I mean, we also need people that feel the pressure.

I mean, it’s not because you have no, you feel less pressure that you don’t feel the sense of urgency. It’s different, right? I do feel the sense of urgency for the patients. I mean, it’s, it’s unbelievable that we still have cancer with this level of understanding. Take, take cholangiocarcinoma. It’s increasing.

We don’t get it. Where is it coming from? It did not exist 20 years ago. What’s the food effects on it? You know, whatever, we, we are so humble You have to be super humble about what you, what you’re building Because we, the, the, the The biology makes you humble But I feel like we also need people that are stressed out.

I mean, it’s just not my way to go. And I’m not saying I’m never stressed. I am sometimes, but, you know, at the end, it’s so hard. Biologists really are, I mean, and I’m so, I just want to, I’m so grateful for people that work there as well. Everybody in the tech, in the biotech, in the big pharma, in the labs, in academics, even more in academics where people don’t make so much money.

I’m so grateful because you can see so much money around like the new large language model, the, the, the, the, the, the new me too of open AI raising a lot of money. Cool. But it’s amazing. But I mean, I mean, and, and, and having like small biotech startup having so much hard time to raise money. It gives me like you know, sometimes a little bit and I’m a bit angry, but it’s like, I mean and you have to be, you know, so I’m really grateful for, for the people that work in health, wherever.

Philip Hemme: I like that. Still on the fun topic what, what I, what I question. What I like is that I think even if it’s serious, if you need the urgency, I think having fun at work or having fun with your colleague is just something so like fundamental from a human level, or at least for some personality of people, it is so fundamental that I think if you don’t have it, you cannot do like really good work.

Thomas Clozel: Do you see that as well? I, so, yeah. I mean, if you don’t come, if you don’t like to come to the office in the morning, you know you know, it’s, it’s, and it’s hard because, you know, I think also the work environment is the more you grow, the more you have to be closer to, to some type of work principles.

Work is being like, tried to create new, new, mid. And I think we resisted quite well for now. It’s not easy, you know. But you, yeah, you have to keep fresh, you have to, to keep and I think, like, also being in your, being more European company, it’s easier, you know, a little bit. Why, 

Philip Hemme: why is it, why is it easier?

You’re less exposed to the, how do you call it, the woke virus in the, in 

Thomas Clozel: the U. S., so. Yes. Yeah. I don’t want to be political, but No, no, no, no. But we, I mean, we’re still very careful about these values and American has also brought really important values, and we are very respectful of diversity and everything.

I, I just say like we, it’s a bit like, it’s easy. It’s harder to talk about religion or, or things like this, for example, or politics in, in the US you have to be a bit more careful and I think it, I mean, we also have a big US company, but. Our French spirits, we quite free, we very we have really revolutionary company.

I mean, we, our company is like, we know, we very like my, my, my co founder was probably the smartest people I’ve ever met in my life. And, and one of the coolest, he’s so French, you know, he was like, when, when we had Google Venture coming in and the company was like, we have to say no. We have to be the first company, no, saying no to Google Venture.

And he was like, and he really meant it, I mean, he meant it. And Gilles has always been saying revolutionary. And, and very revolutionary. It’s the coolest guy around. But you 

Philip Hemme: still said yes at the end. 

Thomas Clozel: Yeah, I believe. So we got a bit of American pragmatism. American pragmatism coming again. 

[01:09:02] Getting the best investors

Philip Hemme: When you saw the money on the table as well, I mean, the money and the value, I guess.

How is it actually to have Google Ventures as investors? Because they invested quite a lot. I mean, I think they’re in past AI as well. And then obviously with Google background, I mean, they’re crazy view on, on AI in general, and they have also their own, let’s say, bio efforts. So how, 

Thomas Clozel: like, how was it? Yeah, no, I have nothing specific to say.

I mean they’ve been supportive and nice. I mean, I, I mean, we were the first, I think, in Europe when they invested at the time after they made more investments. So we were the first deal. I think what happened at Google is, it’s not really about Google Venture. It’s, it’s not about the funds that invest, it’s about the partner.

And at the time we had somebody called Adam Gobara that has created top Harvest Capital. Which is one of the coolest people I met in my, in my life in my, in my work life. He’s, he’s been always super fond friendly, who was supporting us with a real vision for the company. You know, and he knew why he was investing.

He, I mean, you want people that are not in what we call momentum investing, which is a lot for a lot of VCs investing because other people are investing. You want people that invest because they have a very, very precise theory about what you want to do. And, and and you know, you really want, and I, I also took momentum money and I probably shouldn’t have, you know because the people are coming because all the people are here and not the people by earning your value. You want people that we had the same with Gwendolya from First Capital that invested at the beginning he, because he believed in us, in the vision, you know. It’s more about the partners and the funds, really.

Okay. Can 

Philip Hemme: you, can you share a bit more about this momentum money and like slash the fit with investors? 

Thomas Clozel: Yeah, I mean, for example, at the beginning, you know, we had like people like Nicole Druckerman, she’s a German amazing investor. You know, you’re more of like first Google. I think they are Adam Gobera.

This is really people more than funds. That Jacky Abitbol from Quetta is that really believe in us. You know, they were really like believing in the stories. They didn’t want, they didn’t care if an American fund was coming or whatever. Or another fund was here. A lot of funds are just waiting for leads to come, for the diligence to be done.

And when they see good names, they’re just coming in. And it’s, it’s part of the problem of venture capital. Is they really work by FOMO, right? We all know that. Some of us, but, but there is amazing stuff. I, you know, take the example of Sofinova. Sofinova has been an amazing biotech fund, as you know.

And now they’re creating this great digital fund. And I think like Ed and Simon, the two partners, have really, really strong view on things. And, and they understand the, the, the intersection. That, that we start to have in this field, people that have, you know, that have balls and that have ideas. But, but you also have people that just want to invest because whatever, blah, blah, blah, blah.

Philip Hemme: Mm. I was about to mention Sofinova. It’s, it’s, it’s, it’s great. But I think. Talk about this maybe from your perspective on like, because I mean, there is always a level of FOMO that is, let’s say, healthy or that, that, that, that works. I mean, especially when you talk about venture rounds. I mean, I don’t know if you’re talking about angels coming at the beginning.

I mean, angels, there is a bit more believing and really the fit was a person and really long term investing. But let’s say from a VC, I mean, you need to be in this like sweet spot of, okay, it’s not too early. It’s not too late. And what will be my return kind of. having a decent return possibility. And this is also very dependent on, on let’s say, the markets, other investors, pharma partners who follow, they quite like momentum and follow also a lot of others.

And the public market, which is very momentum based as well. So How do you, like, wrap all of this around and then you take the decision of which investor to take, like, you build an AI model for this? 

Thomas Clozel: Yeah, no, no, I think it’s a personal thing as well. You want to, I think it’s like buddies. You want to take people that you want to work with.

I mean, I, I took people that I did not want to work with in the past and, and in I’m, I’m proving my, today, I’m really choosing, it’s the people, it’s not the name, it’s not the real thing because people make the difference. So I think, you know, you have to take people and you have to understand what part of momentum investing or a part of like real theory, you have to ask them, why are you here?

Why do you want to work with me? Well, you know, well, what’s, and, and, you know, but, but the problem is what is the actual value in our company, in TechBio in general? I mean, there is people that bring you great insights, but I have to say the truth, money is money. Because nobody has done this before, right?

Ha, ha, ha, ha, ha. So it’s very rare that somebody’s going to bring you, I mean, at the earliest stage, nobody’s going to bring you something dramatic. Then, when you have to structure, when you need to understand better, when you have to position the dra Then the, the, the, they’re going to come and help a lot.

At the beginning, you know, I think they’re more functional than just exponential, you know, in what they can bring to you. Mm, mm, I like that. And actually, I’m going to be loved by the VC community. Oh yeah, it’s good, I mean. You will have it. They’re not going to like me, but I don’t care. 

Philip Hemme: Yeah, you work with Sophie Noble already as Venture Partner or something, so it’s okay.

Yeah. No, 

Thomas Clozel: yeah, I have to mention, true. But, but because I really believe in Ed and Simon and they proved me right. I mean, it’s a great company. They’re very fun guys. They’re fun guys. So it’s fun as well. But I think just something about investing. I, I, something as, sorry, just about that. I think it was like, we, we, we have taken as both observers people from Salafi from Nestle and I mean Venkat from BMS.

And I think like Venkat leading, like, you know, a big part of data science from the discovery. They’ve been amazing to help us, you know, and I would really, I really recommend for people to take also people from the industry in as board observers or board directors, you know, I just like to help you build things because I have an industry view that is also super interesting and for me, they’ve been extremely helpful.

Yeah, 

Philip Hemme: especially as you said, on certain aspects of your business or your case where it is kind of industry, where there is some industry part and industry knowledge needed. I mean, I wanna ask you one, one question also on the investor side, because, and act, I think you, you’ve billed it more as a, as a tech company as well as in like raising money, but with not too much dilution.

And even, I mean, when Sanofi invested, I think was a 180 million, but at a 1 billion valuation, I think they got 10, 15% of equity, which is, quite low and a very big multiple, much more tech multiple than a biotech multiple, where probably they would have whatever, have 30 to 50% of the company, and I guess what, what translate behind as well is that you guys as founders instead have much more, say, control or things to say on the actual company versus the typical model in biotech where a VC fund will come in and, and take a lot of control.

Sorry. 

Thomas Clozel: Can you hear me? Yeah. Yeah. No, no, no. So I think we, we, yeah, yeah. So 

Philip Hemme: can you, can you, yeah, the question is more like is that the case? And can you elaborate a bit on it? 

Thomas Clozel: Yeah. No, I mean, we, we, we took, we, we, we, we were not very delusion sensitive with my founder, with my co founder, because I always prefer to have a very small part of a huge company, which is not huge today.

So I hope Okin will become huge. So we were not really, we, we know we took the money when we had a really great fund that came in and we took the money where it’s coming from. And so we took maybe too many rounds. So we got delusion. However, we don’t control the company, my co founder and I. But we’re careful, of course, you know, we’re careful to, to still try to get the right dilution and everything.

But, you know, we are not in the, in the, in the position where we have, we control the company. Which maybe is healthy as well, you know because it’s such a complex thing, you want more people to control it. But I think, you know, in the, you know, the more you grow, it’s my first company, right? So you know, I learned that you know, of course, like dilution matters for the control of it a lot.

So, you know, 

Philip Hemme: But, I mean, you, you don’t I thought, control maybe You don’t control more than 50 percent with your co founder, I guess, but you still can’t, you still have, you still have more shares than in a typical biotech setup was a founder CEO ends up with whatever before percent or two, three, or one, two, 3 percent or something.

Thomas Clozel: Maybe a, yeah, for now. For now. More than that. More than that. But you know, not 50 percent either. Yeah, yeah, yeah. But you know, I think like, you know, we, we, we, when there is good money coming in and because we have to do, we have to build. You know, technology, we have to find data, we have to build molecules, so much to do.

It’s a costly project. So, you know, we’re taking the money and you can’t really stay, you cannot really like do like bootstrap it and the money and program your own thing. It doesn’t work out because in biology, all the weight lab thing also works a lot. This is the problem of biology. You always need funding.

Philip Hemme: You need a lot of it. You need a lot of cash. And as we discussed on the whole field as well, I mean, the field is moving so fast and you have quite some competitors as well. And like, Yeah. You need to move also fast to be relevant and to still be the, the best while keeping, while 

Thomas Clozel: keeping, yeah. While keeping always years of cash ahead of you, you know?

Mm-Hmm. Talking, we have at least three years of cash ahead of us. It power you because you, you have to be careful with that. That’s great. 

Philip Hemme: It’s great. Thomas. I think we are, we are about about time. Was was a great discussion. I hope the audio I think worked. I think it worked. Normally it records offline.

I think it worked. Our least. This will be, we, we will edit up. 

Thomas Clozel: Yeah, I think you were us and them. A little pixel ice. And and I love the hats. What is the, is your hat for this, this wing, this weekend or this winter? You gotta take it for skiing. 

Philip Hemme: It’s, it’s my Christmas hat. 

Thomas Clozel: It’s so cool. You have Christmas hats.

I love a Christmas hat. Actually. See you soon. 

[01:18:51] Thanks for listening

Philip Hemme: Wait, wait, wait. I forgot. We need to take a picture together so I can put the hat on. Can you just say, wave, say hi? Great. Thanks for listening to the end. I really enjoyed the conversation. I think this topic is, is really complex and there’s a lot of, like, hype and everyone talks of I’m doing AI and biotech, but I think Thomas really tried to challenge him and I think he’s really good at it.

Really good speaker, good at answering, but I think he nailed it pretty well, explaining also what’s the difference, the key differences between really native tech bio, AI biotechs, what Oaken is doing. I really like the approach of starting with the patient data and really like a huge amount of data and crazy data and then going backwards towards assets instead of having a pipeline versus a lot of other AI tools where It’s harder to capture value in biotech.

I hope you enjoyed as well. If you did, please hit the like, subscribe, review button, especially if you’re on Spotify or Apple. And I would be also curious to hear what you think. So please, if you’re on YouTube or Spotify, you can comment directly or you can also send me an email to philip at 

Thomas Clozel: float dot.

Philip Hemme: All right, catch you in the next one.

Marc de Garidel, Abivax | How to make two Billion-dollar exits (“smoothly”) | E09

Today, I’m in Paris to chat with Marc de Garidel, the new CEO of Abivax. We talk about the challenges behind the exits and why Abivax could be a third big success. Marc is a biopharma veteran with an impressive track record. He has made two billion-dollar exits, was CEO of Ipsen, and has recently made the biggest French IPO ever with Abivax. He has also raised $60M series B for Massachusetts-based Corvidia Therapeutics before being acquired for $2.1B by Novo Nordisk. He also was the CEO of CinCor Pharma which sold for $1.8B to AstraZeneca in 2023. I’ve never met Marc but I heard many great things about him.


Transcript

[00:00:00] Intro

Philip Hemme: It’s great to finally meet. I heard great things about you. 

Marc de Garidel: Well, I don’t know if I deserve them, but I think at 11. 30 p. m. I got a second offer from a competitor to Novo, and actually the terms were not very different. A few years later, Novo is one of the dominant market cap in the world. It’s been a great deal for us.

Philip Hemme: Was it already your plan to go in? Like exit pretty early. 

Marc de Garidel: Maybe I’m not smart enough to do the other way around.

Philip Hemme: Hi, I’m your host Serap and welcome to a new episode of the Flot.bio Show where I interview the best Europeans in biotech to help you grow. Today I’m back in. I was quite sunny and nice to talk with Marc de Garidel, who is the new CEO of, of Abivax. Mark is a, one of these biopharma veteran with an impressive track record.

He made two billion dollar exits. He was a CEO of a French biopharma Epson and now made the biggest IPO ever for a French biotech on the NASDAQ with, with Abilax. Actually, I don’t know him personally, but I’ve heard great things about him both on the track record side, but also on the personal side.

So I’m very excited to, to meet with him. And we will talk about, actually, what were the challenges behind the exit, as well as why Abilvax could be your next big success. So let’s head to the office and meet with them.

[00:01:32] Two big exits

Philip Hemme: Welcome to the show, Marc. 

Marc de Garidel: Thank you very much for having me. Great. 

Philip Hemme: It’s great to finally meet. I mean, I heard great things about you. 

Marc de Garidel: I don’t know if I deserve them, but it’s good to be here. 

Philip Hemme: Let’s, let’s see. Actually for today I want to start with the with the two exits you did prior to, to Abyvax. I mean, both were in the billions, billion, biobucks, such billions.

I mean, Corbidia, I think it was 7, 75 million upfront dollars. Synco was 1. 8 billion. It was in cash, which is impressive. Actually, I don’t know many people who did. So it exists like this but I’ve also heard that it was not a, a smooth ride so could you like elaborate a bit more on what happened and what didn’t make it a, a smooth ride?

Marc de Garidel: Well, as you know, you know, biotech is always a bit of a lively experience. So I think in the case of Corvidia, the you know, the challenge was that we are developing an, actually an anti inflammatory treatment for arteriosclerosis. And we had been actually in discussion with one of the big pharma, fairly early on when I joined.

But what they wanted to see was actually the phase two completion before possibly doing doing doing a deal. The challenge also of the program was related to the fact that Phase two was relatively simple to execute. It was a 250 patient trial, but the phase three would be more like 3000 to 5,000 patients with, you know, costs already ranging from 300 to $400 million for the phase three.

So it was clear from a covid standpoint that the step from phase two to phase three would be a a, a difficult process. And we were preparing actually for an IPO in that context. So we actually ran. Sort of a, a dual approach. One was indeed, you know, execution of of, of the phase two and seeing whether a partnership could be possible and still in parallel trying to raise money.

Yeah. For, for, for, for the phase three in case a partnership would not happen. So I think what was very interesting is the, we got actually a first offer from from, a big pharma company. Again, maybe 

Philip Hemme: not from Novo, not, 

Marc de Garidel: yeah, from another one who who came about and the first proposal was very, we thought was very low.

So we told them, you know, we cannot accept, we keep going with with our phase two. But as the phase two started to get toward the end, then We had a very productive discussion, actually starting at Jeffrey’s, which was in November preceding, basically, the deal. And Novo started to show real interest into possibly looking at our face to data and possibly doing a partnership.

So We at J. P. Morgan organized a more thorough meeting with basically all the research team and R& D team actually of Novo and ourselves to try to come to a potential agreement on conditions for a deal. So, Novo followed that procedure and essentially gave us a term sheet in March of that year. It was, yeah, but this was upon the condition of success in a phase two, which, you know, again, could be always argumented and and channeled.

So we you know, so we are on one hand very happy, but forget still to wait for the result of the phase two. And then when the phase two results were uncovered. We are obviously very happy with the results and we communicated that to to to, to Novo, but that in the meantime, so I gave a call to the other party saying, you know, now we have some great results, let’s say, are you still, are you still interested or would you consider you know, doing a partnership with us?

Because we you know, we have already a term sheet from from a great a great, partner. And you know, I remember vividly because I was actually, you know, in my house, this was, you know, COVID time, you know, so this was when everything was Was virtual. I was you know, in my home in, in near Boston, and I, and I got this call from the head of r and d from this big pharma.

I knew actually from, from history, it’s told me not. Oh. Mark, we are very interested in possibly, doing a deal with you, but give us a bit of time because we need to analyze all your phase two and our confidentiality, but we’ll come back by Friday. Midnight of that week. Okay. So, you know, Which month are we talking about?

We are still, we are, we are probably at the end of we are in June. June. June of, yeah, June of 2020. So then, that Friday, at maybe four o’clock in the afternoon, I call the head of BD, that company, and I say, you know, I haven’t heard anything from you. Are you going to submit anything or because otherwise, you know, the other party is ready to sign.

So you know, they, this this this business officer says, Yeah, yeah, we are, we are preparing something, but give us a bit of time. We know midnight is is is the is the deadline. So I think at 1130 have a PM Eastern Time or your time? Time. I got the you know, the second the second bid from from a second offer from, from a competitor to know and actually the terms.

We are not very different. So, I sent an immediate message to the board. I had given them, obviously, the heads up that things were cooking. But I was not sure. And then I told them we need to to probably organize a board meeting tomorrow to review these these two bids. And, and and see what you want to do.

But I think I would propose we take a bank to help us in through the process. So, to cut a long story short. We put in place a bidding system, the bank organized, so by the following Tuesday, we would have essentially competing bids and the best offer would win. And what was very interesting is a big pharma who thought they would win, actually lost to Novo, who actually, you know, have a bid, an increase, you know, within two or three days.

The terms changed su substantially. It was wonderful for , for shareholders, social shareholders was wonderful for us. And I think ultimately, I now Novo is developing I think they, I believe they are doing three phase threes and for, for the drug. And hopefully in the next couple of years we’re going to know whether this re is going to be bigger.

a big you know drug for reducing inflammation and arteriosclerosis in patients. 

Philip Hemme: Yeah. I mean, I, I heard also, I think it was one of the biggest deals of Novo around that period, or even like, not ever, but in the, in, in these years around, they didn’t make that many big deals. 

Marc de Garidel: That’s right. Yeah. It was the first, actually, I think it was in 

Philip Hemme: I think Ablynx had lost it to Sanofi.

Marc de Garidel: Yeah, exactly. So, so for Lenovo, it was a big step forward. And I remember the discussion with Lars, you know, the CEO of Lenovo, who’s a great, great CEO. And I, you know, he told me, yes, this would be already a big step for them. So we’ll see. You know, a few years later, Novo is you know, one of the dominant actually market cap in the world, without a drag, but yeah, it’s, it’s, it’s been a great job for us.

That’s nice. 

Philip Hemme: On the, on this outbidding, it reminds me another story, which I forgot the name of the biotech, but it was, I think, last year where they was like outbidding two, three times, and it went up to Like billion dollars when the first offer was like five or six times lower. Yeah. 

Marc de Garidel: I mean, in our case, we said, you know, we said, you know, on Tuesday.

Yeah. You need to have your bid by, by noon. We have a board meeting at one and two o’clock. We’ll let you know who has win and who has lost. That’s pretty quick. 

Philip Hemme: That’s a quick, that’s a quick time frame. But I’m wondering more on the lessons learned and for you. I mean, as running a biotech, I don’t really understand why you.

Wouldn’t do such a process, I’ll try to get, I guess. What’s the limiting factors if you have not enough interest from pharma? Or what would be the 

Marc de Garidel: No, I think, well, I think part of the challenge was, you know, in order to finance the phase three, it is going to cost, you know, three to four hundred, five, five hundred million.

You know, there are not so many so, so many pharmas that at the time would have been interested. And by the way, cardiovascular, you remember, was kind of ignored starting in the mid 2010, period. Because of exactly this this, this studies that are very big and you need to do an outcome study.

So you need to, to demonstrate that you are already literally saving lives. And, and a number of farmers like Roche and Novartis just to name a few, you know, had, had, had spent up to $700 million in, in studies and then everything yeah, that passed because of the result were not good. So I think.

Already, Corvidia was one of the first, I mean, certainly one of the most surprising DR at the time. One, it was because it was COVID, everything was kind of dead. And two, it was in a, in a, in a, in a targeted area where no one would have expected a DR. Yes. 

Philip Hemme: Wow, that’s, that’s amazing. And I guess from what you just said also on the running phase three, there’s also good chance that the milestones would be paid because I think we have 75 up front, but then there was one point, 

Marc de Garidel: there is another 1.

4 billion in in in my drama, 700 million under related to basically registration of the drug. And then there are milestones on sales. Yeah. Yeah. We’ll see. I mean, I 

Philip Hemme: think it’s trusting that, and I mean, I would, the dinosaur, I mean, cardiovascular already was, I mean, as you said, from big pharma dropped it all less interest.

But especially as a biotech, very few biotechs going there. Also from the size of the trial and the money you need to raise. But you are basically coming with Syncor, but also in the cardiovascular space. Yes. And it worked also pretty well. 

Marc de Garidel: Yeah, because I think, you know, at the end of the day, almost regardless of the therapeutic areas, what you have to try to find are drugs, which can, you know, bring a therapeutic value to patients.

And because, a lot of companies were in oncology you know, everyone tend to flock to the same areas. Actually, in cardiovascular, there was not so many assets, but there were still a few good assets. And and and you know, synchroasset was actually a drug that was initially at Roche. Roche decided to get out of cardiovascular.

They had done all the really, very good quality experiments in you know, in being preclinical setting. And what happened is you know, an incubator basically negotiated with the app of fin Investments and Fin our Partners the extraction of of the drug out, out of of Rush to, to start developing it you know, in, in in human clinical trial.

But all the foundation of the work was done, it was first class work for, for me when I was contacted again by Sofinova to, to to be part of this new adventure, I was very reassured that the foundation was very solid, you know, the early preclinical work was first class, and also the positioning of the drug.

was going to be very differentiated from anything that had been you know, inexistent to treat those hard, hard to core, hard to treat resistant hypertension patients. 

Philip Hemme: And what, and then, so when, what does the, so then you joined, and then what, can you go through the, what happened? I mean, this was even faster than Corbidia.

Marc de Garidel: It was faster, but you know, it was also a fairly lively experience because A few months into the job, first we got a lawsuit from, the incubator who was in Cincinnati, because we had transferred some of the employees in the incubator into into Syncor, which by the, by the way, was part of the program everybody had agreed to.

But there was some question about compensation and things like that stock options. So we, we already ran into a lawsuit from with the incubator, was also part, part of our shareholder basis. So. In the meantime, you need to raise, you know, funding, you know, to accelerate and to conduct four phase twos pretty much in parallel.

So the good news is we were able to, to raise you know, summer of 20, 21, about, 190 million in in a series B. That was the first, the first big basis. And then we wanted to do an IPO. By the end of the year or, or early in 2022. And then, as you know, 21 was the year of where I, so things started to shake up, especially in the second half of of 21 where it became unclear as to whether we could do an IPO and then we had this lawsuit.

So we eventually was a good story that we were able to, to resolve the lawsuit. And then we were able to be out, do the first IPO of of 2022. We are the first one listed on the, on the NASDAQ. And actually we did actually very well because I think everybody was excited about our program and I’m going to raise another 200 million.

Yeah, through that through that through that process. You know, a few months pass, we have the first results of our study in this R2 TRIT. 

Philip Hemme: It was already the first, was the first two results. 

Marc de Garidel: Well, yeah, a few months later. And on that basis, it was really successful. And as a result, we were able to do another follow on, raise another, you know, 200 plus.

So we basically raised 600 million within a year, which was, again, one of the highest you know, in the world in that period. And, while we were doing this, AstraZeneca was actually watching us because they thought, again, this mechanism of action, so it’s basically, inhibiting a very key hormone that plays a role in hypertension, and they were very interested in this mechanism of action, possibly combine it with their SGLT2 for CKD patients.

Okay. So, you know, that discussion with AstraZeneca took almost a year to complete before they made they made an offer. But then You know, life in the biotech, as I said, is never easy. The second phase two study that we were doing in a less severe population, actually, it was a disaster. Okay. So everything, you know, basically the results were poor.

Not because the drug didn’t work, but essentially, the patient we enrolled and the centers that were used, even though they were the same center in the R2 T3 population, the, there was a lack of compliance in, yeah, in, in, in, in the study. So basically, the, the criteria for inclusion were not very well respected by, by centers.

And we had a huge placebo effect. So after years, it was, I think, One of the worst studies ever seen in with a placebo effect of 16 millimeter for mercury. We’ve never seen that before in any study. So it was an entire disaster. So, so crazy how, okay. But the good news was because we, we, we, we had taken, you know, blood samples of of the patient at baseline and at the end of the study, we were able to demonstrate, you know, over time that, again, it was, it had nothing to do with the drug.

It was really poor execution of of the trial by the, by the, you know, by the centers. And that actually nothing went wrong, by the way. We shared it with the FDA, the FDA, the phase two meeting. did recognize the same thing and AstraZeneca was able to, to, to complete the transaction. 

Philip Hemme: It must be really hard also to frame the story or even explain.

I mean, usually the phase two results are kind of the headlines or like, oh yeah, it’s what it is. And then it’s like arguing and every biotech when there’s a like, say, or like a low result, they try to argue. But at least from my experience, it’s never that clear that it’s like. 

Marc de Garidel: Oh, sure. I mean, when we announced the result, the stock went down by nearly 50%.

But again, we, you know, because we, I have a very good scientific team who are also by great opinion leaders. We should, because we had such amazing results in this hard to treat population. Everybody knew. It was not making sense, it would not work in a less severe population, right? Because if it works for a very young population, it can’t.

And it had something to do with, you know, the design of the study or the way it was conducted. So anyway, just to say, yeah, we went from, you know, going up, then we crashed, and then we came back. And then, you know, Araz, you know, boss and in the context of competition, because there was another company that was, that would’ve been interested probably to, to, to get bought.

But you know, in the end 

Philip Hemme: did you do also out bidding process there? 

Marc de Garidel: So we had some discussion with a few a few, companies. But I think some of them were a bit, you know shaken by the you know, the early population results. So they decided to pass. Okay. 

[00:20:48] Lessons on compliance analysis

Philip Hemme: But I’m curious about this, on the, on the compliance analysis from the clinical trial.

What were your lessons there? What would you Let’s say, do differently. 

Marc de Garidel: Yeah, well, I think what you, you learn is, is that which by the way applies to, to Abilax I said to the team here. is, the discipline of conducting a trial is extremely important. You have to, there is a balance between quality and speed, right?

And the danger that we all have in this industry is because of money reasons and, you know, the time it takes to develop drugs is to go very fast. And when you want to go very fast, you go to centers. Breaks. Well. You know, they are not they are not cheating on purpose, you know, that’s not what I’m saying.

But their standards may not be as high as you want, or what the doctors do is that they don’t, you know, pay the same level of attention when they, you know, recruit the patients to some of the criteria. And you know, so the lesson for, for us, for RabiVax is, we have to work on the placebo effect in these studies, and part of it, He’s having MSRs to, to, you know, to help, train the the centers so that they target the right type of patients.

And to do, you know, sometimes audits of the centers extensively so that again, those sensors are not enrolled if they don’t if their discipline is not as high as as we would like to. 

Philip Hemme: And you will do, you do that or even the audits internally or then it’s also, I mean, I guess you’re working with a CRO.

Yeah. So 

Marc de Garidel: it usually is the CRO who does that for, for, for, for you or you can independently depending upon, you know, some, some companies. Do it independently, but the CRO can do that. I guess also you, you 

Philip Hemme: are very also careful in choosing your CRO, even like supervising 

Marc de Garidel: and where I am. Yeah, yeah, yeah, sure.

And probably to me, the second lesson which we have applied actually here at at Amivax is we had a limited number of oversight, at Syncore. So we are fairly, you know, relying on, on the CRO. It was a good quality CRO, but nevertheless, We, we probably, you know, trip out too much. Yeah, yeah. And I 

Philip Hemme: guess with, with any kind of service providers, I mean, the, the, the way you manage the service provider makes also a huge difference.

Marc de Garidel: Yeah, but yes, I mean, but that I think we, we had in the relationship with the steering committee, we, we, we, we reused a number of things, but, I think still the key to me is you have to hire, you have to, you need to, when you are a small biotech company, you know, again, when you have been used to an outsourced model, you still need to make sure you have core competencies and, and some people you hire who have skin in the game who are on your side.

Yeah. You know, to review what the CRO is doing and, and the, and the centers makes sense. 

[00:24:09] What to do when the market is against you

Philip Hemme: Let’s, for the, so taking a step back to, to what you mentioned on doing IPO on Synco, at the period where you were one, the first or first only one of year, and now you did again with, with switching to AB bva. Yes.

Especially one of the only one this year, at least in this size, and I think was. It’s the biggest one of the French biotech ever on the NASDAQ. Even I compared data, I think in the last five years, at least must be the top five in Europe, even which is really impressive as well. How, how did you manage this when it’s like, how did you manage it when the, in both cases, the market was just not favorable and you still managed to push through it.

Marc de Garidel: I mean, Well, obviously, I mean, there are a number of factors at play. I think part of the success was already ingrained in the fact that in February of this year, the company had done a crossover round, attracting some top notch crossover fans. So I think there was a foundation of investors, deep pocket investors, U.

S. investors who knew well, you know, the story and were going to help back the, IPO story. But I think part of the reason we were successful is that, you know, we brought also a team on top of the existing team. We did a great job for this too. We brought much more us experience in the management team quickly.

You know, we did a strategic review within the two months. We laid out a plan, which is not, you know, six months plan, but we The seven year plan in terms of how we could exploit the potential of bifidimod, which is, you know, a very, very attractive drug, potentially, because it’s an oral treatment. It’s an immunomodulator, so it’s not like blocking one pathway for, for, for, for, for you know, for specific reasons.

So it helps probably reduce the risk of you know, safety, adverse events, issues, which we have seen, by the way, in phase two, where so far the profile has been, has been very good. So that, strategy with a great drug, great potential drug like this was, I think, you know, became a bit clearer to the, the, the, the, the financial community.

And then we were helped also by the fact that we released two year data on the drug. That was, oh, I think, you know, again, it’s not head to head the study, but this is the best ever seen in in ulcerative colitis. So I think the conjunction of, you know, having already some key investors upfront to have a management team that I think, you know, add the credibility in terms of the strategy, but also execution with some of the arina people who came in.

Plus the phase two data to your data, which was really best kind of best in class. combination, you know, the key elements were there for a successful IPO. Then, you know, the market, obviously, was, was not, was not good and actually got worse and worse because obviously you know, not, not only inflation, not only the Kremlin war, and then, you know, what happened in Gaza.

And in Israel, so we are, you know, certainly lucky to go through this this IPO, but, but, you know, you know, the conditions here were not. Not the best. And, and the price, you know, was, was not initially you know, we, we had a, a reaction, negatively reaction after the listing. There’s a listing, but now we’re essentially back where to the price of the IP.

So again, you go through this ation, but fundamentally what was important is to get the money to execute the phase three program in ulcerative colitis. To do the phase two, the in Crohns, in Crohns disease. and potentially develop a follow on product you know for the long term. Yeah, 

Philip Hemme: that’s, that’s because I want to go back to, to the, to the pivot was, with Sophie Nova coming in, you coming in, because In the IPO, I think you raised 200, over 200 million and, but before you raised, what was it from?

The stuff you never invested, I think was 150 and then debt on top in total was 500 million, if I’m correct on the numbers. 

Yeah, 

Marc de Garidel: so what we raised this year is 130 million euros. In, in, in February, we did, bank sorry, debt facility. In August 450, and then we raised you know, about 200 million euros net in the IPO.

So we raised, yeah, all together about 500 million, which, by the way, is in the top five, you know, in the world, you know, of biotech this year. Yes. And certainly, you know, the, the largest amount, not only in France, but also in Europe. Yeah. That’s impressive. Within a year, yeah. Yeah. 

Philip Hemme: Can you go back to the, the story, I think?

I think I heard that, so know, played really a pivot or, yes, in course the, I don’t know if you call it a wheel launch, a we boots or let’s say a pivot. A pivot. Can you go a bit like specifically of what happened, how they contacted you or Sure. Everything fitted. 

Marc de Garidel: Sure. So again, you should go back a, a year, A year ago company again was planning to be sold after the phase two.

Unfortunately, it didn’t get get solved, and, as a result It had still now that it was moving to phase three with you know, a lot of potential commitment of you know, already 200 to 300 million euros. So company got into a severe situation where, you know, it had a good drug, but it was not well financed.

So Sofinoa. Partners played a role, 

Philip Hemme: they were a bit stuck, I guess they couldn’t run start the phase three the same time. Exactly. Hard to Exactly. 

Marc de Garidel: So they delayed a bit, the phase three as a result because they thought, you know, this is a good trial, so someone will show up and, you know, in the end nobody showed up.

So, so part of, of you know, the key role that, so partners played. Already a year ago was they said, okay, we need to reshape the way we operate we need to, to change the chairman. I mean, to have a chairman who has probably more experience in the phase three development program. And also because of the sheer scale up again, the complexities, we need to change the CEO to, to to address, you know, the complexity of a large large program.

And someone who can also help, you know, raise a large amount of money. So that’s where Sophien Auvard came in, in, in in essentially September out. And the rise would be for, well, yeah, yeah. So my story was was a bit of funny because when I got out of Sync Core, I should, you know, I announced the the deal at at GPM, and I said I was I’m dead.

I’m finished. I want to retire. So, I want to go back to Europe, because I was based in Boston before. I want to retire, you know. But, Sophie Nozad, part there, she came back and said, Oh, Mark, you know, can we do a bit of an excerpt? One more. No, no, not one more. I said, OK, you know, he’s Could you be a chairman of one of our companies?

So I said, well, being a chairman is obviously a different story. So that may be we have obviously a great story together. Why not? So I asked, you know, Antoine. Can you tell me, you know, what are your two top picks in your portfolio? Because I don’t want to do the, the, the work. But, please, please tell me what are the best companies in your portfolio where you are looking for a challenge.

So, Abivax was one of the two. And as I got into, you know, then the discussion with Kidam, who was on the board of Abivax, I was more and more attracted by, you know, the science. By what had been done by, you know, this sort of turnaround that needed to be, to be, to be done. And my conclusion, which I expressed to Kylian, was at the end, I think, you know, you’d be better off having me as CEO than the chairman.

Because, you know, there is very little time for this company for a turnaround, given the fact that, you know, the fact it’s not well financed. Field 3 has started, but it’s not really going, you know, it’s fast enough. So, you need someone to execute, to, to, to execute, and I’m willing to do that. Okay. So I changed my mind.

Philip Hemme: And so you, you sold yourself to do it again. 

Marc de Garidel: Exactly. Exactly. But I have no regret. So so, so it’s you know, it’s a great, it’s a great undertaking. You know, we can possibly change, you know, the fate of of patients in the long term in this, setting. You know, this is very, very unique.

Now, there are always risk when you, clinical development. And, but you know, it’s a wonderful opportunity. 

Philip Hemme: Yeah. That’s a, that’s a main story. And I’m quite impressed by how quickly you executed also since joining, I mean, raising this kind of rounds of even changing or hiring more, most like some more us based C levels.

I mean, it seems like you went very fast. Yes. What’s like, what’s like, how did you manage to go like that fast? 

Marc de Garidel: Oh, I think it’s when you have experience, you know, the tempo, I think you’re going to, if you look back at Syncore, Syncore was already you know, managed in a very I think at least, you know, overall, yeah, efficient way.

So, you know, once you’ve done the analysis of what is the challenge of the company, the biotech, then it’s more, you know, are you convinced the board on one hand to get the resources you need? And, and two, to put the process in place so that you, you know, you get the money you, you need to execute, which was which was easy, the IPO and then put in place a team that will be ultimately responsible.

And I think where also I got some help is that Sofinova and the board attracted in in in the spring, so in, in March Sheldon Sloan, who was you know, the person who ran the arena studies. He’s a. The UC study at so at Arena, which ultimately got bought by, by, by Pfizer. So Charlotte had a great understanding of how to run, you know, a large phase three illustrative grade program.

So, and we both 

Philip Hemme: independently, well, 

Marc de Garidel: independently, yeah, he came before me. Even though I actually, I was part of the process indirectly of because I, I was, you know, being considered to as a chairman. So they asked me to, to speak with him, even though he didn’t know that I was going to be ultimately the CEO, he thought I was going to be the chairman, but yeah, so it was part of this process.

But the fact that we, we had people we knew and then we tracked it, you know. bunch of people very quickly. Yeah. So we didn’t need to go through, you know, headhunters and, you know, all the gyration of normal recording because we knew people in the field could you know, be able to, to execute such a complex track.

That’s good. 

Philip Hemme: I mean, I guess what also, it helps too, as you said, to have gone through it and I mean, I guess massive network as well. Yes, exactly. From the . 

Marc de Garidel: Yeah. 

[00:36:08] Phase II compared to standard of care

Philip Hemme: That’s great. The, for the, for the clinical results I’m curious on if we can go a bit more into the, the science and slash the clinics.

Because, I mean, in, in UC or even in IBD, I mean, there’s still, at least in IBD, there’s quite a lot of drugs also, like, around, I mean, Can you go a bit on how, like, how good how good were the Phase II, how it compares to, to the standard of care? I was like, sure, what’s in development, like, and sure. 

Marc de Garidel: So I think, again, first you know, to help a bit understand the, the, the setting.

So when you have ulcerative colitis, you know, the first, the symptoms you have as a patient, you have, rectal bleeding. You go very frequently, you know, you need to go to the bathroom very often including at night. And then, gastroenterologist who treat you ask you to do you know, sort of a coloscopy to check.

You, you know, the, the lining of your the tro of your intestine is being altered by you know, many ulcers or not. So from that sort of, conjunction of looking at those three elements, there is a score that is being, you know, sort of used by, by the gastroenterologist to, to assess the severity of your, of your, of your disease.

So, and this is a population, again, for mostly people are diagnosed between 20 and 40 years old, so it’s a very, it affects a really young population. So what happens is, for the treatment, usually what happens, you get a five ASA, so that’s sort of an old you know, generic compound that is, that start to stabilize your disease for a little while.

But very often, after a few months, or maybe a year or two, you know, disease continues to, to, to progress. The, the doctors start to put you on corticosteroids. Corticosteroids. They are, again, good to move to against a flare up. So when you have a peak of inflammation, however, if you prescribe corticosteroids in, in the long term, it can affect, you know, the really the.

And then massive side effects, systemic side effects. Yeah, systemic, like diabetes and and, you know, increases the risk of death and so on. So doctors really want an alternative. So before we talk about Obesosimod, until now, what happened was mostly you had what they call, you know, advanced treatments or biotherapies, which were, you know, anti TNF.

Yeah, yeah, yeah. The anti the IL 23, I mean, the, RIVOX, the Jack ones. All those drugs add, add their utilities, but they face a few challenges. One is some of them have black box safety warnings. Because they have some serious, you know, adverse events in terms of either, affecting the human system and therefore, being more prone to infections or causes cardiac problems.

Some of them have, more recently, who are, who have arrived, like the S1P, the Ozanimod from BMS, which is the first real potential oral treatment before RINVOC has pre initiation requirements so you need to ocular exam because you have the risk of loss of vision up to 30%. So Essentially, you have a lot of drugs out there, some are highly effective for a while, but then over time, either their efficacy starts to wane, or two, the side effects, the adverse events start to show up and they are really problematic, which forces doctors to change treatment for patients.

So Obifazimod. Even the phase 2 resource we have seen is ideally placed now as what we call a first line or advanced therapy agent. Why? Because we think we could be well placed after again 5 assays and corticosteroids, but before all these advanced therapies would generate their own set of challenges.

And, and the profile of a Bephazine Morality is such that when you look at the Phase II result, is that first, after eight weeks, it’s, it has a competitive response for patients. It’s, it’s, it’s, you know, it’s, it’s really in the landscape of all, all the other drugs. But then over time, after one year, two years, you see this lasting efficacy that we don’t see with many of the other.

The three years data. The three years data, yeah. You know, you have, you know, If you take the, the, the, the, the patients who were enrolled at, at the beginning of the study, two years later, regardless of the profile of those patients, you know, 50 percent of them are still controlled. If you take now the patients who responded after eight weeks, So two thirds of these patients are controlled after two years.

I mean, I mean, no one, you know, again, it’s not, it’s not, we cannot claim superiority because we don’t have head to head, but in, in absolute terms of any published studies, we have the best response. And this is what was presented at the recent Congress at UEGW, which is basically the gastro week for Europe in Copenhagen, where we were, you know, in the second, in the, in the, Session new kids on the block, so basically new product and our data was judged.

as, you know, the best abstract, because simply, you know, it is, it is the best that are out there you know, so that’s why I think Obifazimod has a very important role to play and to finish just on a safety front, you know, because doctors are, you know, this is a chronic condition, doctors are obviously very worried about it.

About the safety of that patient. So what we have seen is we have treated more than a thousand patients now, and we have, we haven’t seen any opportunistic infections. We have any, we have not seen cancer, which could be you know certainly a risk when you you know, depresses you the immune system.

Nothing of this nature so far and that’s more than a thousand patients. So again, we thought it could be, so, so, so we think again, this drug could be a very important for the doctors. 

[00:42:51] IBD and the microbiome

Philip Hemme: I’m curious about, you see an IBD, I mean, it’s very tight to the microbiome. Yes. It’s down. So how does it connect, let’s say, to microbiome or even to microbiome drugs, or even lifestyle biotherapeutics?

I mean. More from a alternative way to treat it like, yeah. 

Marc de Garidel: So it’s a new mechanism of action. So what it does, it upregulates. So model in itself upregulates a key molecule, which is called mere 1 24. So micro, yeah, yeah. 1 24. And it’s very sim specific to immune cells. So what microRNA 1 24 does is essentially, you know, we’ve proven that in, you know, in the in preclinical experiments.

Is if you take a a mice, that with immunity is okay, so with no IBD you’ll see you, you, you administer od you see no impacts. On, on, on the, on the on the on the cells of, of, of those those m mice. When you take a a, a mice, who has the ulcerative colitis, so it’s called a DSS model.

What you see when you administer a, of a FuzzyMod, you see the production of of microRNA you know, ac accelerating, and basically regulating, you know, different pathways so that essentially the inflammation that is caused by psoriatic colitis, this is tampered by by you know, microRNA. So microRNA 124.

He’s essentially a regulator of of of you know, the information process. And what’s wonderful, it doesn’t block, you know, everything. It’s just you know, controls it. And it’s, you know, pleiotropic in the sense. That means it’s a physiological, you know reduction of inflammation throughout, for example, you know, the IL 17 pathway, the IL 23, you know and the macrophages, for example.

So it’s a, it’s a more subtle, I would say, way to, to, to, to reduce inflammation than, boom, you know, you just attack one, one pathway, which is great in the short term, but because of the, you know, the, the, the body, your, your redundance. You know, then ultimately the body finds a way to, to, to to, to, to do the information as one of the pathway.

Philip Hemme: And I guess that’s, that explains, I mean, the small subtle way and yes, explains also why it lasts longer in the safety profile. Exactly. Exactly. But still on this, on the cause, because at the end. The cause is still more micro microbiome. We are, we are not, we are not fixing the cause, we’re fixing the symptoms.

Marc de Garidel: We are fixed. We are fixed. We are, yeah, we’re controlling the disease. We’re, we are not, we’re not addressing, you know, fundamentally the the origin. Which, by the way, it 

Philip Hemme: makes sense to address, I mean, not for you specifically, but let’s say in the current development to actually address it directly. 

Marc de Garidel: Yeah, I mean, there are, well, there’s been a lot of companies who have tried, it’s extremely difficult because, you know, the understanding of IBD, especially, let’s say, if you take a user centric crisis, it’s based upon polyconjunction of things.

One, it could be genetics, so it could be some mutation, possibly. Two, it could be hormonal, too. If a disorder in hormones could also accelerate. And three, it’s also the lifestyle. When you eat, you know, things that are preconditioned, with a lot of you know, sort of bad things, you know, that ultimately accelerates the inflammation of your, of your of your body and your intestine, and then creates this this this, I mean.

So. The, the, the, the, you know, the science behind, IBD sources is still underway. There is a lot of, of work to be done in order to really fully address what what you are. Okay. 

Philip Hemme: Because I think, at least from my understanding, a lot of microbiome companies are going in IBD. Sure. But have not really managed.

Marc de Garidel: Well, microbiome is extremely complex, right? Because you have, you know, billions of of bugs that are as their utilities, which one are the ones that you want to, to, to, you know, are fighting and stuff. Yeah. And it’s a very recent, it’s a very recent field. And unfortunately, until now, until now, it has not been really highly successful.

[00:47:27] Microbiome therapeutics

Philip Hemme: I like this. Okay. I like the, the, the complimentary of, okay, this. It’s a microbiome therapeutics that can cause, I mean, can fix the cause, but it’s like, it’s a longer term horizon. But at the same time, we need also even more small molecules in the first line that can be taken easily 

Marc de Garidel: or a long time. Yeah.

Philip Hemme: That’s like, because that’s one thing I was also wondering, even if we zoom out a bit, Corvidia Zinco and Abivax are all small molecules. I was 

wondering what’s your feeling there if you just look from a, let’s say, small molecules versus biologicals, or like more complex cells, gene, cell, what do you How do you see it?

Like, what’s your, 

Marc de Garidel: I mean, different modalities have their own values. So it depends upon the disease and what you want to do. So I don’t think one should say it’s one versus the other. I think it depends on the a bit of the circumstances of the disease and what you’re, what you’re trying to address.

But one thing we forgot to mention is, for example, for the future of IBD is a combination therapy. So a bit like in oncology where You know, when you basically combine different mechanism of action to essentially to to find a better solution for our patients. So we, we believe based also on the feedback of our key opinion leaders around the world, they told us Obifazimod with this you know, sort of unique mechanism of action could lend itself.

With other products that are out there and they can complement each other, maybe by, you know, doing some of these highly effective drugs. Maybe that those could be reduced. You could add, which is on top or you could start with one of those these drugs and they put In maintenance. So there could be, you know, the variety of combination that could be very helpful to to to, to patients.

So this is one thing that we’re start going to start actually preclinical experiments right now. And hopefully we’ll have results of those combination in the course of 2020, four by the end of 24, so that in 25, once we have the induction data. Hopefully, you know, very positive data, we can start doing combo work as well.

Philip Hemme: Okay. Okay. Now, that’s, I mean, I like your approach of not necessarily opposing small molecules versus modality because I feel, I mean, quite a lot of biotechs, I guess maybe because they come from the science, they’re very focused on the platform, on their like modality and will compare it to the rest.

When, I think, at the end of the day, what tends to matter more is, okay, what are your, yeah, what are your data, yeah, 

Marc de Garidel: and what are you serving for, yeah, sure, yeah, I think that the challenge, for example, with cell therapy and gene therapy is, cell therapy is more the production, this process of manufacturing is extremely difficult to do, and the time it takes to, to, to, to do that for those patients in the context of some acute disease, and then gene therapy, I think the next challenge is, as you know, It’s been, you know, what is, you know, everybody thought, you know, you do one injection, you’re done, you’re, you’re fixed for the rest of your life.

Well, turns out that it’s not that simple, unfortunately. Seems also to have a waning effect over time, that’s one. And two, you have the, the safety side. of which, which, what is the support of you know, which you know, virus or what, which, what is the platform of your gene therapy? There are different platforms.

Yeah. So that you know, when it is administered to patients, you don’t also create some, some adverse events that are not not welcome. So, yeah, I mean, again, science, it’s is always in evolution. So you need to learn and need to be open about different avenues. 

[00:51:20] Why is 70% of the market in the US?

Philip Hemme: Yeah, that’s it. On the Just a question about on the, more on the market of where’s the markets very connected to Avivax, but also more a bit more of a general question.

I think if I’m wrong, that one of the press mentioned that 70 percent of the markets for Avivax is in the U. S. Yes. 

And I feel us, I mean, is, is it, is it because there’s more patient there from it’s, or it’s more just from a marketer policy from a reimbursement policy. Yes. 

Marc de Garidel: Yeah, so in the U. S., for example, I’ll give you some numbers.

Yeah, in the U. S., there are about 800, 000 patients who have ulcerative colitis. When we look at the population we can address, we think we can address about 25 percent of those, because they would not be, they would be Either, you know, already, once they have finished their first conventional therapy treatment, or we could get switched from current, you know, advanced therapy treatment where they fail, or they have too much side effects.

So that’s about 200, 000. In the U. S., just to give you an idea, the price of treatment, the net price, after rebate to the PBMs, is about 50, 000 per year per patient. So it’s, it’s a, so if you multiply what we think, you know, is a potential, the addressable population for us out of the 800, 000, we think we could have about 25%, patients.

Times 50 times 50, 000, that’s a 10 billion market opportunity. So, so, you know, that’s why we, we, we think, again, provided we can show again in phase 3 what we’ve seen in phase 2. You know, this is a 10 billion market opportunity, so we are not going to get to see all the market. But, you know. 10%. It’s already been.

And this is only a s ulcerative colitis. The, the market in Europe, in terms of patient is actually slightly higher. But as you know, that the challenge in Europe, our prices have been going down and down and down, and government are all reluctant to pay, you know, SD for, for, for big innovation. So, yeah, the US is 70% of the, of the market.

Philip Hemme: So. So it’s, if it’s, if it’s 10 billion, so what is it in, in Europe would be two or 3 billion potential 

Marc de Garidel: market. Yeah. The, the Europe is about 20% of, of the, yeah. 

Philip Hemme: And what do you think about this from the, if you reflected, I mean you’ve mentioned already that’s, it’s, it’s hard in, in Europe or for, let’s say for biotechs to get the innovation finance.

How do you reflect on this? I mean, I think for, for me, what I see is, I mean that’s, that’s kind of a fact. But I think it’s very, makes it very challenging for European biotechs to actually, let’s say access the market or forces them to go after the US market, which is not necessarily that easy of a talent location access to capital.

Yes. So what’s your, like, yeah, what do you, do you think about this situation? 

Marc de Garidel: Well, I think the, the, so if you are a biotech in Europe and you have a good drug, you know, you can start indeed the experiments in, in, in Europe, but, but quickly you need to take a look at the U S market. And because this is where, you know, in general, most of the opportunity is going to be.

And also this is where most of the sophisticated investors and deep pocket investors are. So. It means that for, for you know, the management of biotech, the need to have CEOs and management team with Europe slash US experience is becoming more and more more and more important. Otherwise, what would happen is if you can do that and what you have to do as a biotech in Europe, you have to partner quickly with, with you know, a big pharma company.

It is going to do the job for you in the US and we’ll do it obviously very well. But you know, obviously the value creation is very different if you do a partnership very early as a small biotech versus later when you get the benefits of having, you know, produced you know, good results and and having partly address again, the challenge of the of the US market.

Philip Hemme: I’m thinking maybe also from, from a, from a policy point of view, I mean. Talking Europe or even in France about financing innovation a lot and more on the financing side a lot in the early stage side. But there’s a market that’s not there. 

Marc de Garidel: Yeah, no, the irony in the end. I mean, I was recently, doing a speech at the Ministry of Finance here in France, I think, three weeks ago.

You know, part of exactly the irony is that Europe helps, you know, the foundation of the small biotech. At the early stage, I think, there is a really good process, good, good help, and they are, you know, good investors. The challenge is as soon as you go into more intense, you know, clinical trials, funding requirements, and so on.

Then, unfortunately, because there is less money available in, in, in Europe, you need to go to the U. S. And, and and then, because of the cost of, of doing those trials, the return that I expected by investors are such that today, and mostly it is you know, it is in the U. S., it’s the U. S. system that rewards innovation.

So, you know, many of the CEOs of, large pharma companies, including, you know, European, like You know, Pascal Souriau, the last at Nomadis have said, you know, the European system needs really to, to, to rethink itself in order to adopt, you know, innovation in the long term because the risk If we don’t do that, you know, you’re going to have the haves and the have nots.

Philip Hemme: Yeah, and I mean, if the, if the market rewards less innovation as well, means, in a way, less returns, means less investors. 

Marc de Garidel: Exactly. I guess it’s a society choice, I mean. Yeah, yeah, these are, again, they are complicated, they are complicated issues, but, you know, our role, at least as a leader in the industry, is just to give the heads up that, you know, there are consequences to what, So what you know, public, you know, politicians do and we again, you know, are, we, we are here also to try to help find solutions, but there is, you know, they need to, to, to reward the people who take risks, right?

Because it’s a very hard industry. It’s very hard to develop drugs. You know, a lot of them fail. And you know, so there’s a few that are making it to the finish line, you know, should should be a reward. 

Philip Hemme: Yeah. I mean, it’s, I guess it’s a very complex decision, even on the political level. But I, 

I feel like it’s not really the same proving, at least I’ve seen in France or even in Germany, that a lot of even innovative drugs are even withdrawn from certain markets in Europe, even though they’re already approved for, for more for pricing and market access and while they’re already in US.

Yeah. 

But let’s see, let’s see how it evolves. 

[00:59:07] The best way to sell a company is to make a great product

Philip Hemme: Maybe more on the, on, on your, more on the personal, your story, like, so, I’m, I’m curious if, on both Corvidia, Syncor, even Abivax, is, was it already your plan to go in and, let’s say, not flip, let’s say, but like, exit pretty early, or was it more like, opportunity based depending on what, what happened?

No, I, 

Marc de Garidel: you know, I think the, the fundamental of, What we do is develop good drugs. So, I, I am actually absolutely, you know, I’m trying to preach that the best way actually to, to, to sell a company, like a great product, is to make a great product and to have, and to believe it is a great product. You don’t control, you know, a CEO.

You don’t control what a big pharma is going to do. People will think it’s easy to, to do a dr You know, this, this is one of the, the hardest thing to do. So the only thing you have tool you control is, is to develop a good drug and, and think long term. Think about, you know, what this drug could be in three in six years.

And then you need to to, to rally everyone. Including the investors to support the program so that the drug needs to be well differentiated where I, you know, part of my criteria when I have looked at, at drugs is differentiation. You, you can’t bring, you know, drugs that are me toos of me toos and me toos you know, the system cannot serve.

I still am convinced that bringing something that makes a difference to patients and that doctors would, would like to prescribe, that’s why we are in this business. So fundamentally, you know, I’m sort of a, like, a bit like we in buffet type of, of person in the sense that you, you need to create things you know, that are tangible that are going to help patients.

And then whether you do it with GI or not. This is, to some extent, secondary because it’s a, it’s a mean to the consequence, it’s a consequence. And I refute, and I think, you know, my judgment, the best CEOs are this way. People who want to make money quickly on, on technology, okay. If you’re lucky, yes, you can make it and make maybe a huge amount of money.

But this is really more the exception than the rule and probably it is not good because what you do when you’re too short term oriented You know, you try to hide things or you don’t give up the drug as well. Yeah, and And ultimately that’s not good for patients. So I think fundamentally what we are here for develop good drugs Companies I know are bought and are not sold No one, no one can tell, so I didn’t come to say, okay, I’m going to sell, you know, in one year.

I had no idea. The only thing I know, you need to develop well drag. You need to do it with tempo and quality. And this is you know, the sort of the combination of the two, which is the hardest thing to do. And you can, you could see Zincore on one hand, we did wonderful. We still, you know, blew up one study.

At the end of the day, we had a great outcome for, for, you know. 

Philip Hemme: I like that. I like to, I like it a lot actually. Actually it’s a great transition to, when I talk to Bomi, we actually from, from Sphinx, she said the first time she met you, you gave her, she said, he gave me faith in humanity. So I thought it was very, like, very nice compliment.

I was waiting to meet you. But from what we discussed, I think. I can see where it’s coming. I like this, like, in some very caring and focusing really on the patient and the problem to solve. Yes. And then the rest. 

Marc de Garidel: Yes, with Marvolo, and exactly. And then probably because maybe I’m not smart enough to the other way around.

No, but I think fundamentally that’s why we are, we are four. And that’s, you know, when you when you are, you know, building a company. You know, you’re attracting people who want to have a, a, a, a mission. Yes, money is obviously important. Everyone wants to make money for sure. At the end of the day You know, if you don’t do anything meaningful when you come every day to the office, you know, what is your you know, what, what, what, what’s your role?

What’s your, you know? So I think this is where, you know, the, the, you know, it’s a kind of a passion and you need to do like that and some people do, some don’t, 

[01:03:48] Find people who inspire you

Philip Hemme: but. I mean, I don’t think, yeah, I mean, what, I don’t think it’s, I mean, a lot of people in the industry do, especially in, I would say, in the biotech industry, maybe more than in some big pharma where it tends to be, maybe.

Bit more diluted at least from my experience, but not all big pharma are like this. There are a lot of people who care also on this pharma. But I’m curious on your side, maybe if you can zoom out even, let’s say where, where did it like originated? Is it like something that you had from very young or you cultivated or some experience you were confronted to that?

Marc de Garidel: I think it’s probably hard to say. I think it’s partly you see when you are, you know, I was raised on a farm and I was raised, you know, in a sort of in an environment where things were, you know, sort of difficult. So I think what you learn is, yeah, life is not simple and everything you do, You know, it takes time to, to, to to do things.

But I think the, the, the, the, probably the generosity comes more from, I think, from my mother who was a very kind person and and tried to help actually poor people. So maybe I have a bit of, a few genes from, from that. But I think the, honestly, it’s a bit of the luck of, you know, if you ask me whether I would be in this position 40 years ago, I would have said never, even 20 years ago.

So. You know, life takes turns, you know, I think the importance also of meeting key people, people who are, you know, who will help you grow, in your career. You know, that’s also what I say when you’re looking for a new job. Think about, you know, the manager, your, your to me, the company is one thing.

Yeah. There is a job is a manager. You have, because the manager can help you grow or can destroy you or can make you indifferent. But, so you, you do love this kind of skills because you get inspired. You see some people who are, who are very, very you know, they’re charismatic or doing things. And by the way, you know, this principle of do the right thing and then money will come.

This was, you know, the first time I heard that was one of was the CEO of American Express. He was in Paris. He attended his, his conference and Amex became, you know, one of the most successful company at the time. You know, again, 40 years ago. And he said, you know, always. You know, don’t think about money, think as an executive, think about building something tangible.

Philip Hemme: It’s funny for a financial company, right? Yeah, yeah, 

Marc de Garidel: yeah, but well, but he did a remark, it’s 

Philip Hemme: even more important. He did, 

Marc de Garidel: yeah. Yeah, but, you know, look at some people who have done a remark, it’d be well, it’s gonna get Warren Buffett, I think. You know, you need to have a mission, you need to have something in which you believe.

And then, you know, the luck of life is that sometimes you meet some people who are going to encourage you and, and give you this this, this this thing, so, 

Philip Hemme: and, I mean, I like that as well, that’s, that’s a precondition from your family from your origins, and then also connecting to the right people.

But I guess this, this precondition also led you to admire or to work. with the people I’m going through. I mean, you went through, I’d say, bigger Pharma or bigger I think at Amgen and then Ipsen, and you still have this like deep care and, and mission, which is, yeah, it’s amazing. Maybe as a, as a last question still on the personal, actually, I know your son after you actually, I didn’t, I didn’t mention when he was running his co founding a biotech here in Boston and he’s all the co founder.

So we, we talked once, but I’m curious on, with such like intense career and high, high level position in between two continents, how did you like balance, let’s say, more personal life and how you, how do you approach it? 

Marc de Garidel: I think it’s also part of luck. He’s he’s been lucky to, to, to meet a wonderful wife who’s been very understanding with you know, with he has the pace of my career.

But also what I think you can only be successful in your pri in your professional career, I think, if you have also some balance, you know, outside. So some people, you know, take that in different ways and do sports, they do, you know, the thing. But I think if for, at least in the long term, I think, you know, personally having a family, a strong foundation of having kids, always again brings you back to, to reality.

Because when, when you come home, Who cares, you know, whether you’re a CEO or not. You still have to do certain things. You still have to cook sometimes. You still have to, to, to, to, And that gives you, again, this, this, this balance that I think is, is, is, is, is essential. So, you know, yes, I’ve been, again, lucky at home.

I had a good a good person with me and who has a very understanding, who helped me also, you know, tells me also sometimes where, what I do wrong and what I should do more. But yeah, you, you need the balance. You need to, to do things, you know, to relax as well. Yeah. The intensity of there’s a big danger, especially, you know, when you are on CEO job is important versus urgency.

You always get, you know, the guarding files every day when you come in the office. There always when you, and you know, something pops up. But, but as CEO, you are paid not only to resolve the, the, the s but really fundamentally to shake. You know, the future and to alter the course of the company you’re running.

And this can only be done if, at times, you step back, you get out of this crazy world. And you think, you know, you know, what do I need to, to make with, you know, with the team you know, to, to change the course and to make a real difference in, in what what you know, what you do as a, as a company.

So it requires a bit of discipline, but it’s important, you know, like on the weekend to relax and try to avoid to, to watch, you know. All the time, you know, I, you know, I see your iPhone, because the pressure is, is, is constant, but stepping back. I like that. 

Philip Hemme: I share it as well for running a company, I can second this as well.

Yeah. It was great. I think, yeah, it was great talking to you. I think it was actually the, the first time you, you’re on the English podcast, or at least in the recent years, at least talking with Avi Vax, which I think is great as well for the audience. I’ve seen you on French TV or plenty, but I think for, for the audience, at least in Europe and the US, who don’t speak French, I think it’s great as well.

Thanks a lot for joining again, and good luck for everything. 

Marc de Garidel: Thank you, Kai.

[01:10:45] Thanks for listening

Philip Hemme: Thanks for listening to the end. I really enjoyed the conversation with Mark. It was super inspiring, especially how his mindset behind all these like exits, IPOs, how he focused on the core product, on the long term, and how everything else follows. If you Enjoy the conversation, please hit the like, follow button or even leave a review if you’re on podcast.

Any of these actions would, would help us a lot. And I would be also curious to hear what you think. So please leave it in the comments below of this post or send me an email to philip at flot. bio. Alright, that’s it for now. Catch you in the next episode.

Mike Ward | The Best and the Worst since the 80s in EU Biotech | E08

Today we’re in Munich at BIO-Europe to chat with one of the best commentators in European biotech, Mike Ward. We talk about the best and the worst of the European biotech industry since the 80s.

Mike has been in biotech content since the birth of the industry in the 80s. He has worked at BioCentury, Informa/Scrip and now Clarivate. I’ve known Mike for many years and I like how clearly he sees things while being no-bullshit and fun.


Transcript

Mike Ward: It’s just like wow, I’m not normally the subject. I’m normally the interrogator. The creation of humanized antibodies and what eventually became the best selling drug ever, which is Humira. That was the reason why people didn’t like me. I know more people who’ve gotten Nobel prizes than probably watched Love Island or whatever.

Philip Hemme: Hey, I’m your host Philip and welcome to a new episode of the Flot.bio Show. Where I interview the best Europeans in biotech to help you grow. Today, I’m in Munich at BioEurope. So you can see a bit from behind. To chat with Mike Ward, who is one of the best commentators in the industry. Mike has been covering this place actually since, what is it, the beginnings, in the late 80s.

Has worked with, with Biosentry, with… Script, and format, and now is with Clarivate. I’ve known Mike for almost a decade, actually, and I like how sharp and clearly he sees things without at the same time being no BS bullshit, and also being just a fun person. I thought I’d even call him the, the rock star, but it’s like, we will talk about the European industry really from the birth to now, and from the best things to the worst. Stories. So, let’s head and meet with Mike.

[00:01:32] The best and worst of biotech

Philip Hemme: Alright, welcome Mike, welcome to the show. 

Mike Ward: Well, thanks for having me, Philippe. 

Philip Hemme: As we were talking, now you’re on the other side. 

Mike Ward: Not exactly, it’s an interview. It’s just like, wow, I’m not normally the subject. I’m normally the interrogator, so, so this is, this is gonna be an interesting experience. Not too stressful.

It’s okay. It’s okay. I’ll just do what I usually do. Tell the truth. It’s easy. It’s easy to remember. 

Philip Hemme: That’s great. So let’s start with the truth. I think I want to start with, as you have covered this space since really the birth of the industry from, from the birth to now. So what’s kind of some of the best and some of the worst you have seen?

Mike Ward: So you’re right, it’s almost 40 years. So, so I mean, covering the sector, so, Europe was, there was hardly anything here in Europe. There was maybe about 10 companies. So we’ve seen a lot. The best and the worst, I, actually, I think there’s one thing that it’s almost two sides of the same coin. Yeah. And that is around, The creation of humanized antibodies and what eventually became the best selling drug ever, which is Amira.

And yeah, how this works is that one, obviously brilliant science, right? Greg Winter and his, his lab. Nobel Prize. They got the Nobel Prize for it and, and, and quite rightly so. But, and, and this was a time when nobody believed in antibodies. They’d been… They were used as diagnostics, but that was it. But what we actually had is a Greg Winter had come up with this, this science.

It was backed by the MR, the, the medical research council. So obviously that that’s the best, the best of things. 

Philip Hemme: It was in Cambridge,

Mike Ward: in Cambridge, London, 90s, golden triangle, golden triangle. But what actually had was, is that science, it will also actually, it didn’t just. Yeah, wasn’t just around the creation of of Cambridge Anybody technology.

It was also morphosis Yeah in Munich. Yeah, and it was also a an Australian company called pep tech So that the the IP had kind of been spread around and that was part of the problem. Yeah, because Actually, there was a sort of a dispute between those two teams What you also had though was at that time in Europe funding Was, you know, if people think it’s bad now, it was non existent.

I mean, but the amount of money, the whole of the European biotech sector would raise, you know, these days you can see companies. Yeah. Could raise in a one series. Hey, and that was the totality. So money was really hard to come by and the public markets, for example, the London stock exchange, they had recently changed their rules to allow biotech companies to float, but even though you didn’t, they didn’t have a trading history, but they had to sort of demonstrate that they had assets that.

Were interesting and one of the ways you did that was doing a deal So what we had is we had Cambridge anybody’s technology at the time. They had a molecule called d2 e7. They’re a great memory It’s just we’ve just called which is which became Humira and they and they licensed it to canal which was an the pharmaceutical arm of Of, of BASF.

Yeah. And eventually that got acquired by AB Abbott, which became, became AbbVie. But we had this company that was struggling to get finance. Yeah. And, and more, FOIS was also in a, sort of a similar, in a similar space. Yeah. These guys are working on human antibodies. Okay. In the States, at the same time, we had Apgenix, and we had Medirex I think, and both of those companies eventually got acquired.

But at the sort of the, the turn of the 1999 or there abouts, these two companies were able to go to the to the Nasdaq capital market and raise a billion dollars or, or in that kind of order. Whereas Cambridge University Technology and Morphosis really, really struggled to raise a lot. 

Philip Hemme: Was it in the hundreds of millions that worked? They raised. 

Mike Ward: It was about that. I mean, that makes difference. But, and what, what it meant was that, you know, for example, for Cambridge antibody technology, they just basically ran outta steam. Yeah. They, they’d managed to do a deal with AstraZeneca. So AstraZeneca owned a big, big chuck. Yeah. And, but the company was eventually sold.

The, the price that AstraZeneca paid was about $400 million because they already owned a big, big chunk. Because AstraZeneca wanted access to antibodies. That’s it. Now, the, the, again, it was still early days for, for the development of Humira. And AstraZeneca sold that asset to AbbVie, or their share of that asset, for 400 million, which is about the equivalent of one week’s revenues peak for, for, for Humira.

But what we, what we had, we had a situation where… We had a little biotech company in, in the UK that created the best selling drug ever, which was probably the original, you know, pipeline in a drug because of all those different indications. And it struggled. So I remember once interviewing David Chiswell.

So I was saying, yeah, how does it feel to be the CEO of a company that created the world’s best selling drug? But he did it all. Yeah. And that, and that was the situation. So that’s where, what did he say? So it was the best of times and the worst of times. What did he say? He said, that was the reason why people didn’t like me, that kind of question.

I mean, he was only joshing, he was, he was in front of an audience, but, you know, the audience did go, Oh, whatever. And then he realized that it was, yeah, I think I sort of made, made a comment like, so I guess I’m off your Christmas card list, but it, it, it, it was all, it was all fine. But, but that’s just interestingly, you know, when we sort of look at the sort of the European biotech scene, I’m certainly brilliant at the science.

And potentially translating it, but it just, it just doesn’t seem to work because of, you know, the capital markets aren’t here. And, you know, there have been attempts, you know, to sort of see it, but, you know, what we’ve seen in in, in the, you know, for, for a lot of European companies to have been successful, they had to go to the States now.

And it’s worth remembering all that original science that in fact spawned the U. S. biotech industry. A lot of it actually originated here in Europe anyway. So, you know, for example, the sort of the monoclonal antibodies. That was, that was MRC. Genentech’s first products. Is it? There was tissue plasminogen activator.

Came out of Belgium. The human growth hormone stuff came out of Sweden. You know, it’s like, it’s… A lot. And then, if you’re like, 

Philip Hemme: buy anything… M& A, lots of M& A come from Germany, or from Europe, 

Mike Ward: yeah. Yeah, yeah, that’s, so, so, it’s, it’s, it’s, Europe’s always been great at the, at the fundamental science. It’s just always been, sort of, being able to, sort of, like, translate that into commercial success.

And, again, sort of, thinking about, you know, the best and the worst, that now and then, I, I would be asked by… Venture capitalists or, or, or, or CEOs of companies to sort of talk to CEOs who are pitching to other VCs and they show me their, their presentation because I’ve seen lots and lots of presentations and often it’s the first advice I have to, to the European CEO is you have to channel your inner American because you know, the number of times they’re, they’re giving the presentation and they’re already A. community. Telling you what the ends are with it and well, there’s this challenge and that challenge It’s not like oh, no this, you know, we’re this is gonna be the biggest change in Britain Which is why when you have people like, you know Abgenix and medirex who are able to raise a lot of money based on chimeric chimeric, you know mouse antibodies not human antibodies they’re able to do all that and yes and and it’s and it wasn’t because The, the, the, the CEOs that we had, you know, the sort of, the management talent wasn’t that they, they weren’t any good.

’cause, you know, Simon Maroni, who was running Morphosis, you know, a giant of European biotech, you know, David Chisel has been very, very successful in helping your mentor and grow other, you know, European businesses. It’s so, yeah. So, yeah, it’s a long answer to the 

[00:11:08] The beginnings of the EU biotech scene

Philip Hemme: No, it’s great. It’s great. To start, actually, what, what strikes me is that. Some of the challenges that existed in the 80s, 90s are still here today.

Mike Ward: No, absolutely. So, so sitting in panels, and especially now that, oh, it will, everything’s really, really hard. You know, so I find that thing is whatever. And there are some sort of like VCs who were around in the 90s sort of thinking.

Well, you guys haven’t got a clue. You know, they said, you know, what happened between 2019 and 2022 in terms of, you know, the amount of money that’s gone into, into the, into the industry? That’s the anomaly. All we’ve actually gone back to is the pre pandemic norms. Because in, well, in the 90s, there was, there was hardly any money whatsoever.

I did a, a report for Arthur Anderson, right? When it was famous. I can’t think for right. Yeah Nothing to do with everyone but But we’d we’d written I’d helped them write a report on the state of the UK biotech industry and this was about 1992 and We’d come up with a projection that for the Europe for the UK biotech sector over the next three years it needed to raise a billion pounds and Okay.

I remember a member of the CEO who was a member of the BIA, the Bioindustry Association board came over to me and sort of says, well, you have single handedly killed off any potential for the UK biotech industry because That was kind of unheard of, right? The idea of to raise a billion pounds, and it was only over three years.

And that was the hot, everybody I don’t think we had, at that time, we had people like Celltech, right? And other, you know, sort of like, you know, companies that were You know, top of the, top of the class in, in, in, in European terms. And, you know, that was, that was the sort of situation. Oh, by the way, in that three years, they actually raised 996 million.

And I said, we’re only four, four million. You round it up, you’re at the… But it was because there was, there was already, we were beginning to sort of see the fruits. We saw the pharmaceutical industry at this stage, around 92. You know, this was, you know, Jan Leshley, Ed Smith Klein, et cetera, who began to bet on things like human genome sciences.

Kind of recognized that the future for the pharmaceutical industry was biological. Before that, it had been the synthetic chemists. Because, you know, most of those pharmaceutical companies were, in fact, the drug arms of big chemical companies. You just name any of them. It was the sort, they were, and they were…

The sort of sort of release. 

Philip Hemme: I know at least from, I mean, Sanofi, all the history comes from, is from the What, Rogue Black? Health, which is basically, basically total. And then BASF is still the chemical company. They spun out, and Bayer as well, and Yeah, yeah, Rundpulak, and, well, so they had Rogue Black, 

Mike Ward: and they had Hertz.

So Hertz went into, into, I mean, Sanofi was a whole bunch of people. Novartis? Yeah. Yeah, that was Sieber, and Geigy, and Sandoz. You know, they, they all, it’s, you know, to create their life science. AstraZeneca was your ICI pharmaceuticals. Yeah. And then they merged with the with this Yeah. ASRA for, from, from Sweden.

So you could sort of see all these, the, the, these, these, these these pharmaceuticals. But at that time it was, it was all, they had a chemical heritage. Yeah. And I remember sort of like in 19 88, 87, 88, you’re talking to. pharmaceutical executives and their view was that this biotechnology thing, while it’s okay for making you know, you know, some proteins that either had originally been derived from dead bodies or they were chemically modified from from pigs, i. e. sort of insulin and hemoglobin, but yeah, it’s, it’s, it, they’re never going to make a business out of it. And the early shots at trying to make therapeutic antibodies actually Really withered on the vine. Yeah. It wasn’t until 97 98 that you started to sort of see sort of change, but some of the early players though, were the likes of of of Smith Klein Beem.

Yeah. Which eventually merged to, with, with black. So to create GSK.

Philip Hemme: Well, you talk about, it’s super interesting to see the, the parallel, well, you talked about this biotechnology thing, and I think, I mean, in the. Late 70s, 80s. This, the term was not even really invented. Like, was it, or was it, or how, how was it like really described?

Was it corn? I mean, I think Genentech with IPO really kind of started that. Yeah. 

Mike Ward: Well, so Genentech’s IPO must be, what, about 79? Yeah. The, what the the, I think the, the, probably the first biotech company was a company called Cetus. Yeah. Which is like, you know, well, became part of Chiron, which then eventually became part of novartis. But at that time, yeah, so the phrase was being coined because I, I actually, my first job was it had been advertised by a, a, a magazine called Chemistry in Industry, which is published by the Society of Chemical Industry. And the job advert said that they wanted a chemist biochemist who could write about this, you know, new thing called biotechnology.

because we believe it’s going to be kept consequential for the chemical industry. So, so in 84, it was there. But the whole point was is that, you know, for, for the pharmaceutical guys, it was an intellectual curiosity. For the ag, agrochemical guys, they kind of realized that, you know, the opportunity of modifying seeds, therefore, you know, people like, you know, Monsanto, or, or in fact, quite a lot of the…

Again, this is a perfect example that… Plant, bio biotech, agricultural biotech. It was, it was Rolan plant genetic systems in, in, in Belgium, Europe was way ahead of the game. Yeah. But of course it was the the, the sort of the, the antagonism towards this from the sort of the, the public, the regulation, the pol the, the, the, yeah.

The regulations said, which just killed it off.

Philip Hemme: I like, I like the, if we and. I, I have several things I want to jump into or even go from the past to today. One thing is this, this biotech term, like they’re calling the technology and the company a biotech and how you define it. I found it always like it works, but it’s pretty far from perfectly.

And now, and now you get like. Biotech means even a small re company, even something not really innovating. What do you think about 

Mike Ward: this? Right. Okay. Yeah. Yeah. So, so it it, it it is. So yeah. Sometimes there is that sort of the enabling technology is Yeah, biotechnology, okay. Which covers everything. Right.

A whole, a whole raft of, of, of, of things. And then of course, which might be one of the problems that it’s had. And then, of course, then you have, you know, sort of the biotech company and, you know, what is a biotech? And I used to define biotechs as basically drug, R& D drug companies that weren’t making any money and actually had no, no chance of making money in the foreseeable future.

Philip Hemme: Not too bad, actually. Huh? It’s not too bad. I like it. 

Mike Ward: Yeah, but it was, they were, they were emerging. Now, of course. 

Philip Hemme: It’s kind of, let’s say in the startup world where a startup is anything that doesn’t have a, that is not profitable. As in the, at the end in the farmer world, you take the same parallel. I mean, giving you this definition, basically a biotech is a pharma startup.

Mike Ward: Yeah. Yeah. I mean, it’s not profitable. Whereas now what you have is you have people that they are biopharma companies or we sort of see, Oh, well now they’ll call themselves a gene therapy company or so, but they’re still, it’s still kind of like. Descriptives. Yeah. And they could all still be lumped as biotech and yet they’re doing pretty different things.

They could be a cell therapy company or they could be you know, developing some sort of gene therapy for for hearing. It’s all, all, all over the shop. So it’s just a sort of generic term. And yeah, unfortunately I think that that again has been a problem because. Often, and, and certainly in the early days, a lot of these companies only had like, you know, one or two assets.

Yeah. So it was a completely binary situation, and, and, and you’d see someone that everybody was really, really excited about, because there’s all, there’s a huge, I mean, people sort of think there’s height and bubbles these days. In the past, it would be, people would get excited about hardly anything. A little, a little bit more, don’t they?

And it’s all… Yeah, sky high and then they get the result then and the company goes you just think that’s the basic Yeah, and of course what’s actually happened is by the time it’s it’s got all that hype and everybody’s really really excited by it You’ve got all the what they call the tourist investors or the generalist or they all reading real check retail investors I’ve come into the space and then they’ve got They lost their shirt and, and they say, well, and it take a few years for, I’m never, I’m never gonna do that again.

So it’s one of these things where 

Philip Hemme: until they come back again in the try, I think in the cycles, you basically see, 

Mike Ward: well, it’s a, yeah. Yeah. 

[00:21:38] It’s all in cycles and echos

Mike Ward: So talk about cycles. So oh, crikey, what was his name? There was a, a, a, a famous Goldman Sachs banker who basically sort of described how it looked like the biotech sector was.

On a sort of, in terms of finance, it was on a sort of seven year cycle from boom to bust or whatever. And, and he, he had this theory that actually it was not much to do with the sort of science and the technology. It was more to do with the life cycle of, of analysts. So when the analysts come in, they’re all, they’re all really, really excited.

And, and, and sort of like you’re breathless about the sort of the science, et cetera. But eventually they kind like get jaded and they, then they decide they don’t want to do it anymore. They burned up for six, seven years and, and then, and then they’re gone. And they might go and join you sort of companies or, or, or, yeah.

Yeah. Do, do something. It’s, it’s not too hard on them. And then you get the new, the next crowd coming in and, you know, it, it’s, it may be apocryphal, it might be, it’s, it’s, it’s a fun story to tell. Yeah. But there’s probably 

Philip Hemme: one, I mean, there’s probably one thing there as well of cycles, but not even just the analyst, even in VCs, even in the people, some people, I mean, even go in seven years, usually you have time to forget what happened or like.

Mike Ward: So, so of course I do. As you pointed out though, you know, I’ve been doing this for a long time, therefore he is just like, oh yeah, this reminds me of your X, Y, and Z. You know, so that’s the, or sometimes you get the CEO’s recycled or whatever, and you go, didn’t you used to do such and such with? But Oh yes.

So so, so you, so you, so you do see, see these cycles and, and then sort of the echo. So again, sort of coming back to the point of, you know, where we are now. Yeah, we, in 2000, 2001 when you know, a, after the, sort of the geno well, the, the sort of the in internet bubble, right? That, that burst. But also at the same time we had the sort of, the first little draft of the human genome program where Bill Clinton, who was the US president at the time, and Tony Blair, who was the UK Prime Minister, basically go, well, this is gonna be, this, this, and this.

And, and, and there was a sort of a sense of, they kind of like killed the potential because they, this was. Oh, the promising it, but it was also, Oh, nobody should be making money out of this. There was that kind of like that view of this has got like to democratize healthcare and, but with also with the, the internet bubble bursting, there was a nuclear winter for four years.

So it took four years to recover in 2001. And then in 2008, you know, we saw all of those back, I mean, you know, they were You know, big backers, you know, Lehman Brothers was, was you know, a, a strong supporter of, of your life sciences, et cetera. And, and I remember, you know, I was, you know by a century at, at the time.

And you, we had the meeting, you know, when, when Lehman’s went bust, it was, it was this a sort of, you know, an existential crisis for, for, for, for the sector. Because. At that time, still, we didn’t have companies, you know, making much money. I mean, there were some that were making money, but the sector still required, you know, tons and tons of capital.

And the idea is, is that, you know, if it wasn’t there, you know, there would be challenge. I mean, we did a, you know, and particularly in Europe, it was even worse. I mean, that’s why, you know, the sort of, when we talked about, you know, the sort of the tip, the tip of the iceberg, where we sort of did an analysis of The, the financial requirements, because when you look at the clinical trials that they’re doing, you can sort of say, well, okay, you might have raised this much money, but actually you need to raise an awful lot more to be able to push all these things through the clinics.

So, and again, if you look at, if you look at the sort of financing, it took about three or four years for it to recover from the 2008 crisis. So now when we look at, you know, where we are now. And with all the, sort of, the, the macro, you know, political externalities, you know, sort of, war in Ukraine, you know, war in the Middle East, you know, disputes with, you know, mainland China, and, and, and then other places, it’s, you know, you know, you see people, sort of, like, talking about, oh, well, you know, when the IPO market, you know, reopens, you know, it might be next year or whatever, you know, I’m thinking, wow, it’s, it’s, Yeah, it could be a bit longer and that’s the reason why you’ll see in the VCs who have raised tons and tons of money They’re raised crazy.

They’re able to raise the money. I’ve been worse raising a new fund But what they’re what they’re what they’re saying to their companies is You have to have financial discipline Yeah so we’re seeing them cutting back on their programs etc because they don’t know when They’re going to be able to start raising proper money again.

So therefore these, these VCs are having to deploy more or put more capital to back their existing portfolios. Which means that we will see, no doubt, a drop off in new company creation. And there are, there are companies, you know, we’re at, in the the BioEURA meeting, where there are lots and lots of companies running around.

You’re hoping to, you know, raise funds or, or at least increase their profile so that they can sort of, you know, raise funds and it’s, and it’s tough and, and of course they’re, they’re, they’re being told, Oh, there’s plenty of money, big farmer sitting on, you know, hundreds of millions of dry powder and, and the VCs, well, you know, billions of, of, of been raised, but the problem is it’s, it’s dry powder.

You’ve got to do something with it. Yeah. Yeah. 

[00:28:14] Biotech of today

Philip Hemme: That’s good. Lot of things I want to jump on, but what do you think about the current, like, I mean since 20, what? 2020? It’s been three years that it’s been like really slow and wintery and to my feeling is also like longer than the, the previous ones, than some of the previous cycles.

I mean, you mentioned the 2001 that seems around 2, 3, 4 years, but let’s say 2007 or even the fixed from 1314, it didn’t last that long. And, and like, oh, do you agree? 

Mike Ward: Well, so I, I, I mean, the, the way I, the, the way I see it, it, it’s, again, it’s one of those, like, is your, is, is your glass half full or half empty?

Yeah. Because I think that, you know what happened during the pandemic where the life sciences sector basically saved the world and therefore, and also it was like one of the few places that people got actually a, that was working, but, but actually people could put their money to work and or whatever. I think that what we, so everybody’s really, really excited and, and that kind of, it did, you know, create a bubble and, and, and now you have all these, these, and, and some of them who were saying this were actually responsible for this happening.

They’re sort of saying, oh, companies went out, they were, they were too early, they shouldn’t have done IPOs, they they, they raised at high levels and now people are having to take haircuts, et cetera, and I, and I’m sat there thinking, You were the guys who did that. So, it’s almost like, oh, you know, I, I, Nothing, nothing to see here.

It’s not my fault. So I think that where we are example, you know, I’m not gonna, I’m not gonna name names because I’m still, I’m still where I am by actually, you know, not upsetting scary people. So can upset some nice people. Yeah, well that, that, that, that, that, that, that, that, that’s okay. You want to have fun, you know?

So, so, so watch it, Philip. All right. So So, so where we, where we sort of

Philip Hemme: just basically talking about some, some VCs, I guess some of the leading VCs were pushing companies to IPO to take the money when they was there and at the same time and spend more and at the same time, yeah, now they’re the same people saying, okay, watch out, the market is called

Mike Ward: if you just look at sort of, you know, the, sort of the, the, the companies that have gone out and floated, it’s, it’s about 85 percent of them are underwater and they’re not, and when you consider what inflation is, They’re really, yeah, probably, they’re, they’re probably moms almost all underwater.

So, the, the, there’s, there wasn’t so much discipline there, but of course, when people now try to float, you’ve got an awful lot of investors who went, well, last time I touched that, I lost my shirt. And, you know, and some of them have lost huge amounts of money. I mean, it’s just like, well, it’s, it’s incredible.

And, and of course, in hindsight, everyone goes, well, what did you expect? It was like pre clinical, you know? So Joe, that’s, you know, that, that’s sort of your part of the problem. 

Philip Hemme: But at the same time, I mean, on that as well, I mean, it’s whether dancing chairs or like, I mean, when there’s money available, everyone wants to pick it and not be too late.

I’m not being left out. I mean, it’s also normal during COVID when, if there’s so many generous investors or people wanting to put money to go and raise it, I mean. 

Mike Ward: Yeah. I mean, you know, I, would you, if there was money on the table, would you go, Oh no, no, I don’t, I don’t think it’s right. No, of course not. I mean, that’s, so the mantra always is take it , you know, never leave money on the table.

Yeah. Right. It’s like, yeah, 

Philip Hemme: take, but your point is very don’t put, don’t take too much or don’t oversell. And then 

Mike Ward: Yeah, well it came No, you have to, you have to deliver. Yeah. And, and I think that, that, that is the issue. So, so now I think that the, what we’re gonna sort of see is we’re gonna see some discipline within the market.

So companies will probably be able to, to, to. to do IPOs. Not, not in any way the sort of the volume that we saw for, for, for a few years. People are going to have to be exceptional, right? Not, they’re not good sides, exceptional sides. And, you know, people, and there’s got to be a clear route to, you know, what does, you know, how are they going to get regulatory approval for this product?

And are they going to get reimbursed? 

Philip Hemme: I think it, at the end, it raises the bar, I mean. When there’s higher competition or lack of money, higher competition raises the bar. I remember from Holger from Forbian was saying basically the same thing as in so many new VCs were created and now, and there’s still some, a lot of LPs putting money, but still you also see the competition and at the end you see also the strongest surviving and you’ll probably see that with companies.

I mean, today you said that people being companies being underwater, but it’s crazy. I mean, when I looked at some Fort markets, most of the European companies are trading at cash 

Mike Ward: or even below cash. Yo, yeah, no, no, no, no, what, what, what, what is it? Something like 40 percent of companies, public biotechs, are trading below cash.

So what, what they’ve got in the bank. That’s great. Yeah, I mean, have you seen that? Well, look, I mean, now, now and then, I mean, not, not in the same, not in the same volume. I mean, in the past you would sort of see it where you would look at companies and sort of say, Oh, they’re, they’re trading, you know, they’ve got more cash in the bank than, than, than their market cap.

Or I’ve actually seen companies that had spun out a a business where they still owned 20 percent of the stock. And in fact, the, the, the subsidiary they spun out was actually worth more than the, than the parent company in, in, in, in the first place. Yeah. I, I seem to recall a Oxford Molecular spun out a company called, I think, Cambridge Combinatorial, which was eventually bought by Millennium.

And when it first started the, the CEO of Oxford Molecular was a guy called Tony Marchington. Brilliant. I mean, as, as, as a journalist, he was the best. He sang like a canary . Okay. I would then sometimes he told me things for, I, you know, even I realized I don’t think I’m meant to write about this. Yeah.

And I would, I would actually phone up. Right. The, you know, the, the, the CFO of the company go, okay, he’s told me he’s gonna do this, this and that, . Sure. And, and I, and, and this is a public company, I said. Yeah, this is not in the, I just checked it, this is not in the public domain, is it? I mean, it, it was, it was like insider information or whatever, and he’s like, oh, no.

So, you know, so, I had to have like discipline in terms of, which is why again, sort of the longevity, you know, I never ever, like, basically screwed anyone over, you know, sometimes, but you know, sort of Tony was, you know, one of these guys, and and, and Cambridge Combinatorial was in fact a a business. I, it was called Cambridge Corporator set up by his brother.

Yeah. Allard Marching, who is now a, a vc, et cetera. And, and that company actually grew and and then was bought out by Millennium, because I remember the, the launch bot, Takeda Takeda, which came, yeah, it was now owned, owned by Takeda, but they. I remember the launch of, of, of Cambridge Combinatorial.

Yeah, so I was asked by the, the PR, Oh, are you going to write about it? And I says, you know, what do you mean? You know, so, but, but, it’s, it’s, it’s a combinatorial chemistry company. You know, it’s like, you know, it’s, it’s, okay. You know, I mean, the, the guys I think were all to be an ex fighter, etc. But it was, you know, it was interesting, but, and, and also, well, you know, well, Yeah, Oxford Molecular’s put a load of money into it.

And Tony Marlington was a The guy who’s the CEO of Oxford Molecular was a big, big fan of steam engines. And he, he bought the Flying Scotsman, which is one of these iconic British steam engines. And, and I remember saying to the PR person, What are you talking about? Tony spent more on his train set than he spent on his brother’s company.

Now, that was obviously one where I actually got it wrong, But Cambridge Conventorial, that was big. You know, went on to, to, to, to do well, but you know, initially it’s one of these things where, yeah, and you’ve been there where you have the PR, Yeah, and they’re trying to sell you something, and you’re like, oh, come on, you know.

Philip Hemme: I think we’ll talk about journalism in we can get there later. I think, I mean, yeah, there’s a lot of things we can cover there. If we go back to the cycle, I think if I summarize what you’re saying as well at the current cycle, you have this like, Hi from COVID. Lot of money coming in and at the same time kind of the seven year cycle and market instability and these two combining together makes it probably harsher slash lasting longer.

Yeah, if 

Mike Ward: I summarize that’s yes and and and part of the true of the driver for that is the if those You know those externalities are not nothing to do with Yeah, the brilliance of the science or or the technical achievement for the market And that, so that’s the issue, that the, and, yeah, it, are we gonna see resolution in those?

Well, nobody, nobody knows, and that’s the reason why, because this, it’s completely out of control, flight, flight for risk, I mean, no, nobody wants to do, do anything, and, and even when you sort of see, you know, some chinks of light, somebody then manages to slam that window shut, it, it’s, so, you know, I, you know, At, at this, at, at, you know, this, this conference, there are, there are a whole bunch of panels and it’s interesting that there, there’s this kind of underlying optimism as like, you know, hey, yeah, everything’s going to be alright because there is an ecosystem, we’ve got, we’ve got VCs, we’ve got lots of money, we’ve got pharmaceutical companies, we’ve got lots of money, and we’ve got all this, this, this brilliant science, but when you actually talk to these execs on a one to one basis, it’s like, oh, Oh, it’s, I’ve never known.

It’s so bad. It’s actually how, and it’s only because they’ve forgotten that it actually, it has been worse. It’s better.

Philip Hemme: I mean, I’ve never seen something as bad luxury. I like it. I mean, 

Mike Ward: are you a young boy? I, 

Philip Hemme: I’m young, but I mean in, in, in almost 10 years of at least covering and looking at it. But it’s definitely way worse.

Mike Ward: Yeah. But yeah, in the last decade. In the last decade, you’re absolutely right.

Philip Hemme: And like by far, like it’s crazy. Yeah. And talking about Europe, it’s funny, like I remember last year in Leipzig, it was one of the big topics and people were like, okay, it’s winter, how long would it last? And people, some were like, okay, it’s coming out soon, like six months.

But most, especially the farmer were like, don’t expect anything by 2024, end of 2024, mid 2025. And now one year later, it’s basically the same words. And people are still like,

like, you know, it will take some time. It’s curious to me to see that even over 12 months, the situation was basically pretty much the same. And the uncertainty is pretty much the same as about the outlook, which is also a bit worrying if you think about all these companies were in the same stage, like almost one year ago.

Yeah. And, and we’re already trading below cash. 

Mike Ward: And, and, and, and that’s the issue because of course, so one of the advantages, for example, the U S companies had in the past. Compared with their European counterparts is the fact that because they were able to access cash, big boluses of cash, much more than the Europeans could do, is it meant that they could actually really accelerate their efforts and And they could also try things out and okay, it wasn’t a disaster if, if something had to get sunsetted or whatever, but they were able to accelerate.

But what we actually now have is, A, we had the pandemic when it was very difficult if you wanted to start a clinical trial, unless you were doing something in COVID. And all these COVID trials. Yeah, but that was, but that was it. But you, you, so we already saw a delay there. And now you’ve, you’ve got the situation where.

You know, it’s at least 20 million to run a, you know, start doing, you know, first in human trials, et cetera. And if you haven’t got access to that kind of cash, then you’re held back. And the clock is ticking on these. You know, not only in terms of sort of the intellectual property associated with it, but also the science is moving so quickly.

You know, there’s this sort of, you know, there’s your innovation convergence. You know, that actually. The next wave, the next generation, is already upon us. I mean, you just sort of think about, you know, in the Hep C space, in 2020, 2010, there was nothing, really. It was like, you know, you got interferon and ribavirin or whatever.

And then also we saw the… Yeah, so the, the, the protease inhibitors coming along, it’s like, you know, Cefaldi and Harvoni or whatever, and they shot up nearly everyone. Because they were like, you know, nearby queues, completely killed the addressable market off in about five or six years. But you saw just the sort of the waves and waves of innovation.

It was and we’re going to see that again and sort of like, you know, look at CRISPR. Well, not CRISPR, but look at gene editing. Yeah.

It’s, what, 2012, Doudna and Achapentia, you know, published their paper, and then Zhang, you know, a little bit later you know, from the Broad Institute. That’s ten years, right? So it’s They’re almost, almost on their 90th month. And, and, and, December the 8th, right, we’re all gonna look out and see what happens.

Now, that’s for the CRISPR Cas9. Of course there is, there are issues around ip, around, you know, Chris, depending on whether you are based in Europe or whether you’re based in the US So small issues. So it’s just like, oh, I’ve seen this before, somewhere. You know, it’s that, that sort of, oh yeah, guys, resolve it.

Don’t put money in the pockets of the lawyers. Just play nicely in the sand pit, resolve it. Cross license. And everybody’s happy, right? Which is, you know, potentially your one solution. So, because there’s enough of a pie, but what actually is going to happen is that CRISPR Cas9 is affecting the first generation of this stuff.

And a bit like, if you look at the antibodies, you know, it started off where it was… Yes, straightforward. Mouse antibodies, then you had the chimeric, then you had what were called humanized antibodies, and then you had the human antibodies. Yeah. And it was every, every two or three years, you saw these breakthroughs coming through.

Yeah. And the results of that were more and more useful, to the point when, until recently, all the best selling drugs were all biologic, and were all effected in monoclonal antibodies. That human monoclonal antibody. Yeah, most now. 

Philip Hemme: Most of us. Some enzymes are placed, I think, still, but most 

Mike Ward: of them, yeah. But 20 years ago, zero, I think.

[00:45:12] Can biotechs make money?

Philip Hemme: You mentioned, yeah while we’re still on the public market, what do you think about Abivax IPO? I mean, Abivax managed to raise 200 plus million. 

Mike Ward: Yeah, so, so, again, I think, I think that if you’ve got a story that, that resonates and, and again, you know, people understand now cause of COVID, et cetera, people got a much better understanding of, of vaccination and the, and the value of, of vaccination, et cetera.

So I think that five years ago, they might, it would be more of a struggle that it is, that it is now because of people go, Oh yeah, yeah. Oh, vaccines. Effective vaccine is good. So, I think that But I don’t, I don’t think that that’s the, oh That’s the, that’s the The window is open I think that that’s What 

Philip Hemme: we’re going to see It’s quite surprising to see it in that timing I mean, it was surprising to me at least to see that they They managed to raise that much Right now 

Mike Ward: Yeah, I don’t know how many insiders were involved.

Yeah, because that’s one of the things that you would, if I was now a journalist and I was sort of analyzing that deal, I would look and sort of see like, okay, so, A, how much was floated? Did it go out at the price or was it at a discount? And what… How many insiders actually participated in the round? It was a big party.

So there’s a whole, there’s a whole bunch of, so I, I saw it and I, but I, I, I didn’t necessarily you know, look into it in any, in any great detail.

Philip Hemme: I think, I mean, so Finola is behind and put quite some money in, but there’s some, some some U. S. crossover fund who also put, but also the market, and I think, After trading, they were at minus 15%.

Yeah, just after the first day. Which is a sign also the market corrected as well. But, I just thought about this. If we go back to the definition of biotech as well, I mean, we’re taking a bit of a loop, but, towards also what’s happening today, now you have biotechs who make money. And we still define them as big biotech or biotech, let’s say, whatever, I mean the biointech, in Europe you take the biointech, and…

I mean, the, the Argenics et cetera, also Moderna in the U. S., but it’s funny also how do you, just, even just from the term, how do you 

Mike Ward: define that? So, so I wouldn’t call them biotechs anymore. We, so we call it biopharma. Okay. Right. Which also, you know, some of the pharmaceutical, well, in fact, now most of the pharmaceutical companies have a huge molecular biology, biology.

Most 

Philip Hemme: of the pipelines also. 80 plus 

Mike Ward: percent is that biotech, but you know, even, even some 15 years ago, there were though. Yeah. I mentioned, you know, people like Smith Klein were, were clearly on, on that route where others were still very much firmly wedded to the small molecule and weren’t necessarily involved in the sort of the, the, the molecular biology and the biology.

Whereas now they all deserve to be. 

Philip Hemme: But you get to the biofarmer word, then you have to do, I mean, then it’s basically conflicting with the mid cap farmer, who all call themselves biofarmer. Yeah, 

Mike Ward: so, so, so there’s, let’s 

Philip Hemme: say, Epson, Servier, which for me is very different from a, a genmap is very different from a Epson, for example.

Yeah, yeah, yeah. 

Mike Ward: And, and, and therefore that, so, it’s, it’s, it’s kind of. Because obviously we always had to sort of like, you know, find some sort of like jam jar label that you could give these companies because you’re right, there are, there’s sort of, there’s almost like, you know, sort of what’s it, you’re mid to scratch or whatever there’s a, where there’s sort of mid tier of, you know, people like, you know, Ibsen, Servier, Almaral, and they tend to be, they’ve either, you know, come from a sort of like a private foundation, you know, Lundberg might also fall into this category.

Right. And have been located at, you know in a region and they’ve, they’ve supported that region, but never really globalized. So why Ibsen now is, has got global, you know, ambitions. A lot of the Japanese companies were Japanese and now you’ve got people like Takeda, Estella. Yeah, these guys have now sort of, you know, taken a much more sort of, you know, global view.

So those, you know, again, it it’s one of these sort, sort of their, you know, sort of mid-size pharma Yeah. Is, is, you know, how we would necessarily describe or, you know, sort a reach, re a re a regional player, right? So like Spanish pharma or a, you know, Italian pharmaceutical company or whatever. Then we have, you know, and I’ve heard for example you know, bio, the, the Biotechnology Innovation Organization, it came up with.

Almost like how it was split in the market for what we would call biotech in terms of R& D stage companies and clinical stage companies to gain sort of like, you know, sort of differentiate that revenue, 

Philip Hemme: revenue stage are profitable. 

Mike Ward: Yeah. Yeah. Yeah. So, well, I mean, there were no basic commercial stage.

Commercial. Yeah. Yeah. And, and that’s the, and it’s, you know, part of the evolution. So it’s a way of all this stratifying. Yeah. So, you know, it’s like we’re, we’re all, we’re all humans. So we’re all biotechs, but in fact, you’re what kind of, you know, you know, when, when we actually deep down and we start stratifying it.

So, oh, well, yeah, but you, you got, you know I like me to, you know, sort of be, you know, sort of end up with Alzheimer’s or whatever. You can sort of stratify it that way. So, so I think that, actually, what’s in an egg? A rose would smell as sweet by any other name. I was about 

Philip Hemme: to say, but I was just curious to hear your definition and from seeing also these, these, Terms evolve over time.

Yeah, and where you are stuck. So at the end of the day, I agree with you I mean, it’s just a word it tries to encompass something and to help when you hear word, but it’s not a hundred percent If it’s such a 

Mike Ward: so it’s a good going back. It’s like, you know when I was it I was a technical science of technology editor at European chemical news and I was writing about biotech so right about cell tech right which is a I’d originally been backed by the MRC.

It was an MRC chief. All the science had come from the MRC. They had a sweetheart deal with the Medical Research Council of the UK. And I was working on European California News, which was basically focused on, I mean, on petrochemicals. And ICI was the bellwether of the UK economy. It was the biggest company on, in, in, on, on, on the, on the FTSE 100, etc.

I remember one of my colleagues said, Why do you write about this thing called Cel tech? Like, who cares? And I, I sort of then said, So one day, Cel tech will be bigger than ICI. Now, what happened is in 92, ICI then sort of split into its, like, chemicals business, and then it split off its life sciences into what was Zeneca, which they’ve now asked to Zeneca.

And, over time, I think it was the next 5, 6 years, Chemicals became, you know, less and less important. So because ICI actually became smaller, smaller, smaller, smaller. And then one day, the Sultec actually got into the FUZIO 100. This was before it was acquired by UCB. Got into the, the FUZIO 100, and I boned up my former Just Talking about, man, you owe me a beer

That’s what I, so I said, I told you it would happen . So yeah. Let’s about, and you owe me beer. Yeah, that exactly. Yeah, I bet. Good. That’s 

Philip Hemme: the good one. And stay on this like big biotech or biotech farm biopharma. I mean, I think that’s a really interesting development that, I mean, it never happened within Europe, where you have like biotechs who are still like standalone companies, let’s say.

I mean, Jen, Jen Jen expiring tech or worse north of 10, I mean, more than 10 billion and making actual, I mean, billion new revenues and are profitable and are still like, as a standalone and this, I mean, basically never happened. Well, I mean,

yeah, I mean, in the last, let’s say the last decade, this happened before never really happened. No, 

Mike Ward: no, no, no, no. And. 

Philip Hemme: So, so, so, so, so let’s put Italian as well in there, which was already profitable before being acquired by G& J. But basically, I mean, it’s like a handful of companies who made it commercial at the billion level in dollar or euro level 

Mike Ward: and being profitable.

Also, what, what, what do they all have in common? When? 

Philip Hemme: Biologicals. Most of them. They’re also European. Yeah. I mean, I’m focusing on the Europeans, but 

Mike Ward: no, no, so yeah, I know because we’re. Yeah, we’re not going to go down there. But anyway, that route, but so, so, so the UK actually has always struggled to create businesses of critical mass because they get to a certain level and then all of a sudden, they just don’t get acquired.

And, you know, and that’s a shame. And I think that, you know, you look at, you know, and you mentioned softening over. Softening over were a big, big backer of… Yeah. Kelle and, yeah, and and Act When it was first created, the first Anto, think Antoine was called it on the board. Yeah, he was. He was.

Yeah. That, and you look at some of those other you Argen X and you know, a links. Yeah. But they got that growth Genmab, you know, these companies, they were backed by what you call clever man. Yeah. And actually by VCs who they are patient. I think, 

Philip Hemme: I think Fabian was big back. 

Mike Ward: Yeah. Yeah. And, and you can sort of see, oh, and who are the guys who are now able to raise you?

Significant. It’s the same guys. Yeah. You know, it’s, you know, the, the sort of the guys at Fabian and the guys and, and it’s, it, it’s, so when we always look at sort like Europe, so Yeah. The UK biotech sector and, and I’m now gonna get just a lot of people. So I go, what are you, what, why you sound like you have a bad mouth in the UK?

Philip Hemme: But if you just look at the facts, 

[00:56:31] Funding big vs small

Mike Ward: but, but, but if you look, so, okay, while the UK takes a disproportionate share of in the funding, right? So it’s, you know, somewhere around maybe 33 percent Yeah Yeah, could be up to 40 percent and it always tends to be either number one or number two in, in the sort of the totality of the money raised

What you see elsewhere in, in Europe is that the investors actually, they put big bets on potential winners. So, you know, you, you might sort of say, Oh, Switzerland, Switzerland in this past quarter raised 250 million, you know dollars, right? In sort of VC financing. And then you look at it, and actually 240 went into one company.

And then, and, and that’s, so, so, so interestingly, the sort of the… These, these European VCs are actually operating the way that American VCs in the past operated. And, you know, I don’t sort of, I don’t sort of see the same, you know, big boluses of cash. Now, it might be because actually a lot of the interesting UK companies have already upped stakes and gone to the US.

So, they may still be headquartered in the UK. What actually, you know, what is the CEO, the CFO hangout? Yeah, 

Philip Hemme: where is it? Let’s tell, yeah, I didn’t, I didn’t mean to really compare and I would, I mean, it’s an interesting point you make between, between the UK and the rest of Europe. But I was, I was thinking also really more from the like, today you have these like, I mean, whatever, three, four, five large biotech companies.

And this, I mean, this is a really interesting evolution at the end. Edit. As you said, a lot of this is made good returns on it, and also full of people. I mean, the people I’ve experienced in commercializing and bringing these drugs to markets and I mean, it’s, it’s, yeah. Super beneficial. Yeah. I mean, 

Mike Ward: but it kind of makes sense though because if you sort of think about it Yeah.

Some of, some of the, the, the, the most successful pharmaceuticals of all taught companies of all time and certainly. In terms of innovation, I’ve been European. I mean, I mean, you look at Basel. Switzerland’s a tiny country. That’s crazy. And you, you look at, it’s, it’s capable. You look at Sweden. Again, a tiny country.

Or Denmark. These are, I mean, we’ll see, I know we’re Nordics now. Like the, the, the, the largest European pharmaceutical company. Markitcap. Yeah, by 

Philip Hemme: Markitcap. Oh, you’re the largest Markitcap company in Europe. All right. 

Mike Ward: Yeah, but it’s, it’s, you know, wow. That’s. Yeah, that’s good. You know, it, it, it, but it, but it, but it’s, and you can sort of see that there has to be, you know, cross fertilization where, you know, people sort of like moving around and, and I, I think that, you know, we, we, we’ve seen the sort of some of the recycling of of, of, of executives you know, et cetera.

And, you know, I think that. I think that’s interesting. I think that when we sort of think about Europe, I think we should sort of, yeah, sort of compare it with Massachusetts, right? The whole of Europe with Massachusetts or the whole of Europe with, with California, partly because, you know, the way that the VCs look at things is that yeah, I’ll go to things that I can sort of visit in an hour, an hour and a half.

And in Europe, you, you know, if you were sort of like in Munich, are they, we, we, we saw the sort of the strategic, if you’re in Munich. You can get to everywhere within two hours and yeah, if we saw a Western Europe as a cluster Then all of a sudden it’s like well, well, yeah, just no punching above its weight.

Philip Hemme: No, it’s good I mean my my point of it was also like I mean, maybe you you could think it would be a natural Evolution that some companies become billionaires But I think if I look back at it in 2010 or even 2015 was not obvious at all And you had the same discussion as it are In Europe, a bit like in the UK, all the assets get acquired much earlier because they don’t have access to, I mean, we, we know all the problems, not access to massive rounds, et cetera, but at the end, it’s not really what happened.

And you had, you have these like four or five companies who made it really big and prove that, that it’s possible. I mean, it’s more like a fact and it’s interesting to look back. 

Mike Ward: And, but, but, but in most cases, it’s, it’s kind of almost, and it never happened. It’s by luck rather than design. I mean, it happened 

Philip Hemme: five times.

I mean, by intake, I would guess there’s some reluctance from the pandemic that’s unpredictable. Okay. Okay. Okay. Okay. Okay. 

Mike Ward: Okay. Okay. Okay. Gemma, right? Gemma is a really interesting situation. So it actually originally spun out of medirex. Yeah. Okay. And I actually remember being at the first ever pitch that Lisa Drayton, who was the CEO of Gemma back in 1998.

Yeah. Well, the first day of pitch, because in She was at an investor conference. She wasn’t meant to pitch. And somebody dropped out. And somebody said to her, Well, do you have a pitch? And she, she didn’t. But she came over to me, she said, Did I have, you know, access to the internet? And I, you know, was, I did.

So I had my, I was able to. You know, connect and we downloaded her presentation from the states, so she was able to give her a presentation, which basically meant I always got the red carpet when I visited Gen Map. Right. Because I, I was there at the beginning. But their, their product Darzalex? Yeah. Okay.

It originally was partnered with GSK and then there was, there was, there was some issue. GSK handed it back. Okay. Now you, when, when you were reported on a sort of day-to-day basis, and you saw. a company, a pharma company, hand back an asset, 80 

Philip Hemme: percent of the time, it’s, it’s a bad 

Mike Ward: thing. It’s, it’s, it’s not good news, right?

Oh, we’ve got all the work that they did on it. We’ve given us all the sort of the, the, the material files, et cetera. Oh, it’s like, and, and we got that money as well. Yeah. But what we’re, we’re, and the vehicle was really, really brilliant, right? With the fact that he then managed. To license it to Johnson Johnson.

Now, Darzalex is one of the top 10 best selling devices in the world. Now, the whole point is, is that the technology platform that was associated with Darzalex, yeah, it’s a platform company at Genmat, right? And obviously they’ve been able to actually sort of, you know, develop their own products. But it’s a really sort of…

It was one of those things that what looked like a, probably sort of an existential crisis for the company. Actually, it came out the other side, you know, all, all, all seeing all dogs then. So so Genmab is one of those companies that, you know, it’s, I’ve, I’ve really, really, like, like Biontech, I’ve really enjoyed watching their progress because I saw them at the start and how it’s grown.

And. You know, I’ve I, you know, one has one’s faith, and you sort of, you want them to do well, right? Because you like the scientist behind it, or you like the CEO, or, or whatever, and you just sort of, yes, go for it. I mean, 

Philip Hemme: yeah, it’s a good, positive, I think, involvement, and it comes back to what you said at the very beginning, and then, it’s the same, I mean, you’re in the phase of the coin.

Sometimes there are great news and bad news, but the bad news becomes the best news ever for a company like Genmab, which is amazing. And it’s, I think, a great kind of closing on this, like, let’s say, biotech sector.

[01:05:06] Biotech and journalism

Philip Hemme: And I want to go a bit more on the personal side, and I think we mentioned journalism, and you mentioned here as well as like, okay, there’s a bad news, and how is it twisted, or how a lot of, you get pitched a lot, and a lot of stories, and a lot of stories that are really interesting a lot, so I thought I’d…

And I think, I mean, journalism and biotech, I mean, I would say from talking to people in the space, it’s not that well understood how it is in media. I mean, media first is not that many media, but also it’s not that well understood how things work in the background also. So maybe we can talk about this stuff, like I, yeah, what was your experience like, it’s probably a very general question, but like, what’s, should to explain to someone, let’s say like a, you know, someone like average, Managers or average attendee here by Europe, like what is really happening behind the scenes and, and how, how’s the media slash journalism actually works and in biotech specifically.

Mike Ward: Yeah. Okay. You’ve got one hour. Oh, okay. So, so, so the process would work, you know, something like, like, like this, because there are different, like, again, there are different, like, almost like tiers of yeah, probably. So there are those who. are effectively just telling you what happened, and that’s it. So, get the press release, and this is what happened, and don’t actually challenge anything, okay?

And you see them, and in fact, you know, sometimes, they even still keep the, you know, the signature around the press release, and contact, blah, blah, blah, at the end. You know, and I’ve seen that, and I’m thinking… Oh, come on, you could have at least edited that off. So, so, so, so, so you have that. But those, those, there’s no value in them.

20 years ago, so, so, so again, what’s actually happened in the sort of the time I’ve, I’ve, I’ve been doing it. When I first started, a fax machine was, wow. This, this was, this was like, you know, at Houston, landing people on the moon. I mean, having a fax machine. The closest we had to sort of fancy technology before that was the Telex machine.

Okay? But most, most of the… 

Philip Hemme: You’re getting out of my… Yeah. memory. 

Mike Ward: Well, no, that’s the point. But what would happen is that most of the stuff that would come in would be papaya mail, like snail mail, run like you’re fast. Most of it was snail mail, and it would be… And you get a press release. But of course, nobody had that, you know, the only way that people would ever actually know what was happening was what they read in the newspaper or what they read in the trade press.

That was how, you know, they, they, and, and, and, so that, that was the first thing. And of course, how people differentiated themselves was getting on the phone and talking to people. And, you know, asking the right questions. Okay, so, so that, that’s, you know, I have a necklace. It kind of got a bit quicker and a bit quicker, but, you know, even, you know, in the late 90s, it was, you know, the press releases would come out on Factorship, you know, very, very few people were, were using the internet as a resource, and there wasn’t much there on the internet anyway, and if you did actually have access to the internet, it’s, you know, pages come down the screen, it was like, it was agonizing, and of course, most people didn’t actually have sort of web pages, What we’ve seen since the sort of, you know, in the last two decades, or certainly in the last decade, is also, everybody’s now got access to all this information.

Therefore, it’s become, information has become commodified, and, and therefore, what has actually been required is for people to add intelligence in some way or another. And you can do that either by Actually looking at sort of the fundamental data that you, you’ve collected, et cetera. Yeah. And therefore putting something into context or actually getting out there and actually talking to people.

So if somebody was to sort of say to me, well, so you know, how, how did you do what you did? I said, I spent a lot of time talking to people, but I spent more time listening. To people. So, you know, I would say to my journalists, you know, you’ve got, you’ve got one mouth and two ears, use them proportionately.

Yeah, yeah, I like that. And, and the, and the, and the other sort of, you know, key was, you know, make sure that you understand what is being said to you. Yeah. Okay? And don’t be frightened. Unless this is like the guidance I gave to the journalists. Do not be frightened to sort of say, look, yeah, I, I don’t get that, I don’t understand.

Or, you. Repeat back what you think you’ve heard, and they’ll correct you. Now, I said, you know, okay, the person that you’re interviewing might think, this person’s clueless, right? They don’t know what they’re doing. I said, but at least, right, if they then explained in words of one syllable, what they’re trying to say, when you write it up, everybody else is not going to realize that you’re clueless.

Everybody’s going to say, oh. You seem to know what you’re doing. I’ve been getting away with it for decades. That’s sick. It’s, well, it’s, it’s, it’s, it’s again, it’s, it’s about, about having the, sort of like, an open an open relationship. And, you know, one of the other things I would say to, like, for example, company CEOs, et cetera, about, you know, when dealing with the media, is you have to understand what is the motivation of the person who’s interviewing you, okay?

What, what are they, for example, on their own career path? Are they looking for the shock horror probe that then gets them, you know, a, a position on a much more high ranking sort of like daily newspaper where they can now write about, you know, Kanye West or, you know, and that kind of stuff? Or are they actually sort of serious?

Because, you know, one of the… One of the sort of the, the, the, the important things, sort of the relationship that I had with the, and this is partly ’cause I kind of grew up with them. I mean, you know, a lot of the people who are CEOs and VCs or whatever, I met them when they were, you know, brightly minted PhD students or brightly minted postdocs.

It’s and now they’re, you know and people sort say, oh, you must be so smart. I way, if I was that smart, I’d now be, you know, living on an you an island in the Mediterranean. So. Now all I’ve done is I’ve just been there, so I’m going That’s interesting. Oh, that’s interesting. So, 

Philip Hemme: but also you have been there for such a long time as well, that you have a compounded value of like every year you stay analyzing, you add contact, and you see the same people again and again.

And even last, I mean, I don’t know how many times we’ve met, but 10 plus time and Okay. You get a feeling for the person and you know. What has worked or not? Same for the CEOs when you already know them. I mean, you said Genmab, but some CEOs or some VCs, you know them from other deals, from running other companies and you see it.

I mean, there is definitely something there as well, but 

Mike Ward: Yeah, no, no, no. And it, it, it helps. But there are two things that are important because it’s, it’s about us having that, that, that, that relationship. So somebody will sort of said to me about. So do the right and I said, yeah, sometimes it’s what I haven’t written is more, it’s more important than what I have written.

Yeah. And they go, what do you mean? I said, well, well sometimes, you know, again, when the PR have been very impressed PR companies have been very, very successful at pushing a story. And, and I haven’t believed in it. And you know, I just thought they, wow, they’ve all, they’ve all been drinking the Kool Aid, right.

They’ve all taken this on board. And you know, what actually happens in that, in a situation is that. Yeah, if I haven’t written about it, it’s because I have made a conscious decision not to write about it. And, and when I had the team, right, when I was running teams of journalists, there had to be, when we were looking at sort of the information that was coming through the door, or through the window, or through the, through the internet, when we had that information, they couldn’t just sort of say, oh, not interested.

They had to have a reason why. I’m not going to spill any ink on their story. So there had to be a sort of conscious decision what we were going to write about or what we’re not going to write about. What we wrote about is, did we have something to say that was interesting that would move the, the conversation on and develop the conversation because there’s no point us wasting our time just repeating what everybody else who’s, some of the stuff that is free.

So therefore it was And, and, and then the next question would be, okay, this looks really interesting. What else do we need to do to make the story even more interesting? And the way again is by having the sort of the network, one can phone people and sort of saying, did you see that news? What did you think?

And that’s, you know, a sort of, you know, a sense of. Or getting a sanity, I call it getting a sanity check. But yeah, sometimes also, you know, even with the PR companies, you know, one eventually builds a relationship where, you know, they send you and I sort of say, right, you’re not going to try to sell me a part because you know that I will never talk to you again.

And, and they believe me, right? So, I mean, I would ask them and they would, I sort of say, look, am I going to look an idiot if I actually ignore this? Yeah. And I have a relationship with, with some of them that, that go, yeah, it’d be all right. Yeah. Don’t, yeah, yeah. I’m not, I’m not going to push it, right? I’m not going to say, oh, you must do it, you must do it.

Philip Hemme: I think that’s an interesting point as well. I mean, you, if you take for the PR agency. So, I mean, for those who don’t know in really the communication agencies work, trying to get the message out and obviously talking to media, trying to convince usually that the story is interesting and sometimes adding background as well.

[01:16:15] The EU biotech world is very small

Philip Hemme: But, and if you expand it also, I mean, all the, like, let’s say executives or all the network, it’s a really small world, the biotech world, especially if you think European biotech is extremely small. And you want to be, have good relationship and not screw up people, you want to be friendly, but at the same time you want to remain, like, to your values or like, stick to, to what’s most interesting and not bend this.

And I think this is a very tricky bit. 

Mike Ward: So, yeah, so, no, you’re absolutely right. Because I remember once, there was one CEO where his company had some really bad results. And in fact, it was an existential crisis for that company. Eventually, right, yeah. It managed to, like, limp for about the next two years, but eventually it died.

But at the time, the CEO sort of said, Oh, I’ve seen your story. And he goes, wow, you really stuck the boot in. And he said, but, You’re right. Yeah, it was fair. Why? It, well, I, I didn’t, I didn’t overreg it. It, it, the, the, and, and also, they had the opportunity. So, if ever, you know, I saw something, I always gave the CEO, you know, The CEO didn’t see it for the first time in the publication.

I gave them an opportunity. This is what I am writing. Yeah. Okay. You have an opportunity to respond to it. Yeah. Was, was, was part of that. The second thing is also is that the, the, I mean, there were times when I would, I would come across some information or I would triangulate Yeah. And the, the CEO or, or the investor would go, this is.

Because you know, none of the stuff that we were writing, it was like, you know, we are going to break the world or whatever, but it was, you know, they would sort of say, it’s, it’s a really, really, yeah, you’re, you’re absolutely right. This is what’s happening, but it’s a really, really delicate situation.

Could you hold off? We’ll give you the exclusive when it’s, you know, ready, ready for pride time. And, you know, that’s what I did. Or sometimes when I’m sort of like pursuing a story, the CEO would give it. Give me some information off the record, confidential for my information to help me understand. I couldn’t write about it, but what again, it at least enabled me to write my story.

Right? That, you know, it, it, it was on, it was on solid ground. It, it, it, it was sound And coming back to this thing about that, 

Philip Hemme: that’s where it’s tricky that you’re like, it’s like. We’re helping you, but at the same time, you can help back, but not help too much, it’s, 

Mike Ward: it’s, it’s… No, well, you, you, you… I find it a very tricky thing.

You’re part of the ecosystem, but the, but the whole point is, as you sort of said, everybody, you know, it, it took, it, it takes 40 years, right, in my case, to build up a reputation. Yeah. And, you know, what, stitching one person up would be… It would disappear in a moment because people say, yeah, don’t talk, don’t talk to them.

You can’t trust them, et cetera, et cetera. And I know that that’s how it to people, you know, because I’ve always been interesting in sort of seeing, right, who’s, who else is in the space. 

Philip Hemme: I like that a lot. Like you’re, you’re really, I didn’t perceive that part of you that much. I mean, I, I enjoy, and I enjoyed how you dance and how you stay like the bss, but I, I didn’t see the behind the part of like, where it’s quite conscious and really long term.

You, you’re in the long-term game and you’re playing that for, for quite a while and it helps your benefits. That’s very interesting to see actually, 

Mike Ward: look, I mean, 

Philip Hemme: and that was, and now it seems very in hindsight, but 

Mike Ward: it’s, yeah, that was my, yeah. My view that I, I am always going to, you know. I always sort of saw myself in this space.

Philip Hemme: I like this part also, that’s the, between someone who’s serious, someone who, who like, will jump to something else. Same for me, I mean, I can see myself in the biotech space for a very long term. And then, you make your decision differently. And definitely also, it’s also a good filter for, you want to work with a person or not, or like, what’s happening.

Mike Ward: I like that. I mean, it’s an easy, it’s an easy thing to do though, I mean, it’s, it’s, it’s like, wow. You know, Yep. I love what I do. Yeah. Okay. I still, I still love what I do because I get to, to talk to people who are super smart, who eventually go to win Nobel Prizes. Yeah. Okay. So, you know, I sort of say to people, you know, you do know that I’m actually connected and, you know, to a whole bunch of people who’ve got Nobel Prize, that I got more, I know more people who got Nobel Prizes than probably watched Love Island or whatever.

Right. In my, in my friendship crew. It’s, it’s stimulating. No, no, no. The great thing is, is that it, and it’s evolving, and you’ve got new people coming through all the time, and it’s, it’s fun. Yeah. 

[01:21:45] The changing media landscape

Philip Hemme: What I like also from what you said, and maybe I can add a bit from the, the media landscape, I mean, you had a particular view also coming from, let’s say, Scrip, or even Biosensory, which is always premium journal, so paid for publication, so.

Really, the audience pays for premium content, pays the journalist, and so its publication will tend to be more high quality, more high analysis, versus some free publication, which now tend to be, at least, probably the most read now, let’s say, I mean, oh yeah, if you take the, the, from the older one, from the fierce, fierce biotech, which is still around and still very popular, to, to even the Endpoints, to La Biotech, to, still La Biotech today, to some others it’s more the free, but I mean, there’s still some analysis done, but typically it will be a bit, let’s say, lower, lower, lower analysis, or more like the journalist had spent a bit less time on the story to really get all of it, more or less.

Yeah. And sometimes the model would be, it’s more to give context to the audience, so it would be free or there’s a freemium model, let’s say for endpoints. Yeah. And then you have really the free, free or more like press release directories. I’d say biospace in the U. S. still mostly do that. Some others, I think, those ones today are like, more like, less than whatever.

Mike Ward: You can have a Google alert and, and get as much context. And you have everything, yeah. 

Philip Hemme: Yeah, yeah. And at the same time, you have, I think, one interesting trend as well in the last several years, but really social media. Let’s say LinkedIn, especially TwitterX, also, but mostly LinkedIn, where You can get access to some stories, which is interesting is that you can get access sometimes to the personal backstory from, let’s say, the CEO sharing and like where they were, and sometimes it has some angle to it, which is not obviously not journalistic, but has some context, interesting context, so it’s more like to to give the whole like landscape.

Mike Ward: Interesting. So for example, yeah, a young journalist starting out today. That’s what they should be tracking. Yeah, yeah. Right? I mean, I would look at, obviously, where people are moving to, etc. In the old days, I mean, one of the things that, you know, how I started, trying to work out what companies were up to, is that they would advertise in the back of Nature, in the back of New Scientist, PhD studentships that they had.

And it would be in areas. And then I’d look and go, Okay, is that an area that they’re already in? Or is this a new area they’re exploring? And of course these companies, you know, then when I would go and interview them and I go, so, so yeah, well, what are you thinking about this space? Whatever. And all of a sudden you can see them going, how do you know that

And it’s because, you know, they, they, they, there’s, there’s a lot in the hiring. You, you advertised it. So I, I, I think that’s that. But, but coming back to the the sort of the various tiers, what was interesting is. You know, with, with Biosentry and, and also with, with, with Scrip and on all those publications.

Even though there was all this free content that was actually, you know, potentially competing. All it actually did was destroy the potential for display ads. Right, so yeah, Scrip, when it was, because it was, and it just forced us to go. You’re digital only and not even produce a hard copy because in the past the hard copy would be there and it’d have Display ads and classified ads in the back, etc People were still and they are still willing to pay for highly curated quality Contents the economist for example, you know, it’s its revenues have gone up It’s got more subscribers than than it used to have Okay.

Now it’s available, you know, on, on, on, on your, your iPad and, and, and whatever. So they, they kind of like digitized it as well, but it’s not cheap, but people are willing to pay for your quality analysis. And, and I, and I think that that is, so therefore there is still a, a, a, you know, a space for that. I, I, I, but you know, to come back to it.

So, you know, again, sort of how people deal with the media, you know, I think one of the things, and I think one of the hardest, so a lot, a lot of journalists. They kind of, what I call it, they retire, and they go into PR. And it’s because they sort of say, oh, that looks a lot easier than, than what I do.

Because they obviously would just be, you know, hanging around, having parties, etc. And they all sort of think, oh, and also they tend to get paid probably a bit better than, than most journalists. I sort of say, well, no, hold on, you don’t realise that you’ve got to get up probably at 6 o’clock in the morning to make sure that the press release went well.

That’s what I’ve been working on. You’ve got to chase around. You’ve got to deal with… Journalists and analysts, are they going to write the story, etc, etc? You’ve got to deal with the flak if they’ve actually misinterpreted what was in the press release. Because, if everything goes well, it’s because the CEO and the company are brilliant.

Anyway, so that would happen. If it goes wrong, it’s your fault. And also, if you go, you know, as a journalist, you know, if you come to a survey, I mean, people want to talk to you, right? Yeah, everybody’s, yeah, I’ll talk to you. Because they, they won’t. Like coverage, et cetera. I said, but if, if you are a PR guy and they see you, they say, well, I’m not your clients, therefore you are probably gonna pitch to me.

And therefore they’ll, they’ll walk at the other provider. Yeah. I said, so it’s a, it’s, it’s a sort of, it’s a, a tough job. So again, you know, it’s one of these things where, you know I have respect for, for, for the PR guys, but I did also sort of say, never ever just use that. Never tell me AP, right.

Because. 

Philip Hemme: But this year, you never stick. Yeah, I mean, it’s, it’s, I mean, I think deep down also, you two things we haven’t talked about in media is the value it provides to readers. And I tend to think that it’s, it’s very high, whether it’s a premium publication or not. I mean, there’s a lot of value in like people knowing what’s happening even for us at La Biotech.

I mean, we had 150, 000 readers a month and we had so many like deals and financing rounds and whatever that happened because They read and they know, oh, they discovered the company, or discovered the fund, or the startup got discovered, and all the people kind of connected. I think from an ecosystem point of view, this has a crazy amount of value.

Yeah. I remember Jørn Aldaik telling me from, from now he’s at Hoopipa, was telling me, I think the European ecosystem would have grown much faster with a European biocentury. And I was in like, in the U. S. Biosentry had a huge role and they were like super early and new role in build and like connecting the ecosystem.

We were saying, okay, like biotech, you had a bit this role, but like, I mean, we have that much and et cetera, but yeah, it was basically everyone was like, okay, media, even that has, has an impact on the ecosystem. So, so it’s very hard to quantify. Yeah. 

Mike Ward: So with the European, you know, Biosentry, well, I was, right.

So what happened with, with Biosentry was that. Around about 96, where we saw sort of like the creation of the Neue Markt, etc. Dave, Dave Flores and Karen Bernstein, they kind of recognized that Yeah, something was going on in Europe that was going to be potentially called Sprague Shop. And so therefore they had in their, on their board, they had a number of Europeans either from pharmaceutical company or from VCs, like, you know, David Lethers or whatever.

And what they said was we need a journalist, right? We need somebody in Europe to cover. Yeah, biotech, so what they did was and everybody said, Oh, well, well, you need to talk to Mike. But the problem is that Mike has got his own newsletter. So I had my own newsletter, which was called biobusiness, which was because I’d seen biocentury and I thought, well, I’m not going to bother competing with it.

But what I would do is I would just focus on European biotech. So I had conversations with, with Dave. And, and then Karen and, and they sort of said, tell you what, why don’t we acquire your, your newsletter? You work for us, we’ll, we’ll, we’ll pay you a salary. You just keep doing what you do. Everybody who was a subscriber to your newsletter will switch over immediately to, to, to buy a century.

And, and also I, I had a kind of a, a, a burnout deal on, in terms of not only the Yeah, the renewals, but also any new subscriptions. Okay. Okay. Now the whole point of this is that my, my newsletter cost 200 pounds and this was like 2000. Right. So it was a huge difference. So getting, getting a 10 percent royalty was actually, it was just the same anyway.

Philip Hemme: It paid a bit of set on the island. 

Mike Ward: But so, so I had a, I had a superb incentive to get in this many, many readers and also. That ain’t like hell for European biotech. I didn’t know this story. So what happened? What happened is somebody once sort of said to me Oh, do you, you know, sort of Yeah, so you, you’re, you’re the European correspondent.

I said, no, I’m, I’m the rest of the world correspondent for, for, for biocentury. And, and they go, oh, oh, well, yeah, what’s that? What’s that mean? I said, yeah, anything outside the Bay Area. Because in that, in those, because Massachusetts wasn’t even that big. Right, we’re talking about 97. Yeah. And, and it was, you know, Biocentury was absolutely fantastic.

Yeah. Why, it was great fun. Every day we would have a meeting, half past four European time. Yeah. Half past eight in the morning, where we would talk about what had happened that day, what we thought was important, what would we need to do. To move the story forward. How would we, how were we going to, to, to do this?

Who should be responsible? And that’s, and at the news room, we did that every day. That’s cool. Okay. And it was, and, and you know, the, the, you, you’ve, you’ve, you’ve, you’ve met them, right? Yeah. Those go all. absolutely brilliant place to work, where also we knew. So, so it’s interesting what Jorn said, because we knew we were making a difference because at that time, there wasn’t really much, much else out there.

Philip Hemme: It’s a great start. I think the, the, the second point of the media was, is actually also money. And I mean, even for me, just knowing the background at Labaretek or so it’s. It’s not easy to run a media company and journalists are not the highest salaries, highest paid, but still it’s, it’s a lot of money to run these stories and it’s, yeah, it’s, it’s actually tough business.

And if you look, even if you zoom out, I mean, journalism basically one of the first industry that got completely disrupted by internet media in general or at least disrupted as in like had to change big times. And, and also even from the, from the business point of view, I mean, most of the media companies even today are like struggling or like, 

Mike Ward: And, and, and, and it’s because it’s, it’s, it’s, it’s a question again, sort of, you know, sort of, you know, reinventing you know, oneself.

So yeah, by a century realized, you know, always had to add value that was done. And, and when I joined Informer and sort of was the already sort of script set. That was exactly, you know, we, instead of writing every story, right, printing all the news that was fit to print, we actually focused. On the most important.

And, and it was one of these things where, and because we had access to lots of other data sets, then that obviously could inform, you know, what we were doing. We, we had opportunities to talk to the analysts. So that’s what, that’s what in the informer group what we were doing. And now Clarivate, so coming back.

You know, the, the great thing about Caravate is what I’m doing is that I’m now able to write, you know, what I like and sort of, you know, write, you know, thought leadership pieces, you know, talking about sort of trends, et cetera. And I’m, I’m able to dip into all the data sets that we’ve got at Caravate and it, and it covers the whole waterfront.

It’s huge. It’s absolutely huge. And, and it’s, and it’s free. That’s the point. For, for, for, for people to download by the sort of the content that we produce and, and, and, and it’s because, you know, what we’re doing, it’s almost a sort of like the reverse model of the, you know, sort of the fierce or whatever, where what they actually really do, they were, they were harvesting email addresses, which then could then be used for marketing purposes, et cetera, et cetera, and sold on for, for, for, for those purposes, whereas for us, it’s, it’s a way of actually Getting people thinking about, you know, what, what does this company do?

Because most people don’t necessarily know the name, but actually then go, Oh, oh, they’re part of you. I, because it’s a sort of this archipelago and different sort of data sets. It’s all decision resources. 

Philip Hemme: It’s high quality content. It’s a form of content marketing. In a way, as in, you raise awareness, you raise people to come to you and, but it’s interesting because you’re doing, it’s content to serve a goal, as in sell intelligence products, versus let’s say messenger, which is content, selling content directly.

Yeah. So 

Mike Ward: it’s just interesting. So what it is, it’s, it’s a soft sell. So the idea is, you know, frankly, it’s a, you know, A, if they’re giving this stuff away for free, how good is the stuff that, We have to pay for it. That’s, that’s part of it, but it’s also just so that, that actually Clarivate is invited to be part of the conversation.

So, so, so, so my, my view in, in what I’m trying to do is I’m looking to bring together other thought leaders around, you know, topics that are keeping the executives awake at night. And the idea is, is that we don’t have all the answers, but at least we. You open up a conversation and, and it also then brings other people in and sort of say, Oh, well, we saw this.

And again, it’s, it’s like, you know, sort of like you’re moderating panels at these conferences, et cetera. It’s, it’s, it’s part of the same process. You know, I’m not there to just get, show these, you know, give these guys an opportunity to sort of say, my company’s doing this, my company’s doing that. We want to move.

The conversation or we want to trigger you know, thinking elsewhere, we want to create debates because, you know, even if people sort of says, I think you’ve got it completely wrong. I’m not offended by that. I say, okay, why explain why 

[01:38:35] The rockstar of biotech

Philip Hemme: it’s great. I think we’re coming out of time, but the last, the last question to, to wrap up is, I, I mentioned in the intro and I’ll stop it a bit, someone called you, you’re, you’re the, you’re a rock star of biotech.

Mike Ward: Ah. 

Philip Hemme: But as in, it’s one thing that I like a lot with you actually, you’re very like, very sharp and you see things very clearly, but at the same time, relaxed, no BS, fun, I mean, I don’t know if people can see your, your shirt, I mean, you always have something there. What, what do you think about it? What’s like?

Mike Ward: So, so again, it’s, it’s one of these things where. Actually, no, just be, just be honest, you know, it’s, just be yourself, okay? Some people, you know, might 

Philip Hemme: It’s easier to be yourself than to try to be someone else. 

Mike Ward: Well, that’s it, you know, and I, and I think that, you know, it’s somebody once said to me, so like, oh, you know, they came up to me and said, you know, what, at one of these conferences, you know, you know, why do you wear such wacky shirts and, and, and jackets?

And I says, Well, this is why and then what do you mean? So what you just call up to me? So that’s fucked away Okay, and that’s just and that’s the whole point. I said, you know if I was anonymous Well, I wore a suit or whatever. You wouldn’t come and talk to me. I guess you’re 

Philip Hemme: fun I mean, but it also expressed a bit deeper.

Yeah thing of like, okay, like let’s not be too boring I mean the industry is frankly quite conservative and quite boring So, like, also, like, let’s just be yourself and adding a bit of this twist. 

Mike Ward: And, and so, so, so I’ve had a sort of a couple of occasions where you know, sort of one you sort of in fact, it was the same boss that it was once where we were going to meet the new CEO of the company and, and therefore, you know, some board members.

And he said to me, he said, Mike. Any charge you’d wear a suit and a tie. And I said, I don’t have a tie. Well, I said, the only suit that I got is a, a, a dinner suit, . And, and I said, and I’ve got the tie to go with it. And he goes, oh, well, well, he said, no, you can, you can borrow one of my ties. I said, no, it’s okay.

I don’t need to do that. So anyway, we rock up around next morning and my, my two colleagues who are there, they’re, they, they’re dressed like funeral directors. Yeah. And, and I’m dressed as ico. And then in comes the CEO and his, his posse, right. Of other board members. And they’re always sort like, you know, polo shirts and, and, and, and Chinos and I, and I said, I turned to, to, I said, he feels a bit over dress.

And then there was another time we went into just sort of a, a marketing company. And again, it was, we we were, we were doing some. They did a lot of stuff for brand awareness, for like, serial gummies or whatever. And we went in, and everybody else in the office was dressed like me. And, yeah, I, my two, my two colleagues were dressed, yeah, like, again, funeral directors.

They stuck out like a sore thumb. So that’s part of it. And it’s just, again, it’s just to sort of say, right, okay, this, this is how it is. It does, it does get conversations going. Yeah. And, yeah, it’s. The, the, the challenge is, is that I can’t wear the same jacket two days running, so it’s a bit like that, that school, school uniform thing of like, well, if you’re wearing the school uniform, you don’t have to worry about what you’re wearing tomorrow because you’re going to be wearing a school uniform.

Think about it then. If all you do is wear a dark suit, white shirt, and a dark tie, or, you know, some kind of tie, you know, nobody notice. Yeah. But, you know, I I remember your shirt. Well, this one, yeah. This is good. This is one of my favorites. It’s good. It’s good. So, so it’s good. So, yeah, so, but again, the whole point is, is just also making it comfortable for people to come and talk.

That, that’s, that’s art. And of course, the best way of actually gathering information is, as I say, is through us. Cool. 

Philip Hemme: It’s a great conversation. Thanks, Mike. 

Mike Ward: These have been a lot of fun. I don’t know how this is going to go, but this is great. great. 

Philip Hemme: Thanks a lot. 

[01:43:18] Thanks for listening

Philip Hemme: Thanks for listening to the end. This episode was great fun, but also quite stressful as there was a bug in the audio and we almost basically lost the microphone audio, which would have been able to publish the episode.

Quite some luck there. But overall, I mean, I really enjoyed the discussion. Mike has really that, that clarity and has seen over 40 years, but still able to deliver it in a very, like, clear, fun fun way and and, and enthusiastic way, which is great. Yeah, if you, if you enjoyed the episode, please hit the like.

Subscribe, share, button, any any of these actions would, would help. You can also leave a review on non Spotify or Apple. And I would also be curious to hear what you, what you think. So please, if you can, comment on YouTube or Apple or Spotify or you can just send me an email to syllop at flot.

Alright, that’s it for now. See you in the next episode.

Agnete Fredriksen, Nykode | Being one of the top 50 women in 🇪🇺 bio | E07

We’re live from Oslo 🇳🇴 to catch up with Agnete Fredriksen, co-founder and CBO of Nykode (ex-Vaccibody). In this episode, we chat about being one of the leading women in bio in Europe, the Nordics, and immunotherapies.

Agnete founded the immunotherapy company Nykode in 2006 which is now worth around $500M. For her PhD she designed and developed the first Vaccibody vaccine molecules, improving the outlook for cancer patients. I’ve known Agnete for many years and I like her sharpness while having this Scandinavian humility.


Transcript

Agnete Fredriksen: My tips, my tips now is to, this is one of the things I think my learnings personally is to leave early.

Philip Hemme: I know it’s your October months. 

Agnete Fredriksen: Not only October, I think in general. In general. I even had to do like manufacturing in you know, a short period that I never necessarily liked or excelled at it. 

Philip Hemme: I think you announced from this week that you raised.

45 million dollars if I convert in dollars. It’s a private placement with new international international investors. 

Agnete Fredriksen: We are also so happy with the data we have on the DNA. So, so let’s see if that’s you know, maybe more likely that we would explore with a partner with the mRNA competence.

[00:00:52] Welcome

Philip Hemme: Hey, I’m your host Philip. Welcome to a new episode of the Flot.bio Show, where I interview the best Europeans in biotech to help you grow. Today I’m in the really cold and wintry Oslo to talk with Agnete Fredriksen from Nykode, ex MaxiBuddy. Agnete founded the company, this immunotherapy company, in 2006 and the company is now worth around 500 million dollars.

I’ve known Agnethe for several years and I like her sharpness while having this like Scandinavian We will talk about being one of the leading women in biotech in Europe about immunotherapy and about the Nordics So let’s go to her office somewhere in this building and talk to her. See ya

[00:01:36] Women in biotech

Philip Hemme: All right, so for today I want to start with talking about like women in biotech a general topic I mean, you’re actually the first woman guests on the show and I, I took the ranking, I would rank you I think in the top 50 women in biotech in Europe, according to myself. And I’m curious on what’s your experience like 

Agnete Fredriksen: being a woman, 

Philip Hemme: being a meeting woman in the field.

Agnete Fredriksen: Yeah, no, I don’t, I don’t necessarily think about it that much every day. But definitely there’s, there’s been episodes where you feel that you’re treated differently. So. I’ve started to think that it’s important to start to talk about the differences even though some of them are more general than person specific in order just to, to figure out how we can can make it more attractive for, for more women to enter into leadership positions in biotech.

I think it’s important not to just say it’s it’s no difference. It’s just, you just have to want to be a leader in biotech as a woman. I think you do experience differences. 

Philip Hemme: What happened? What made you change from, I just don’t want to talk about it, to I want to share more? Is there something that happened?

Agnete Fredriksen: No, I think, you know, you get to a certain level of position or confidence that I feel that I missed. To hear from other women that were in front of me that they actually talked about the differences. Cause I, I feel, I felt when I was younger that what I heard was that there is no difference.

Also from women in leadership positions, and then I felt that I experienced differences and that I actually felt was a bit unfair because I felt that I would be better prepared if someone actually talked about it in public or to me specifically. Yeah. So that I could also feel that it was not like only my, my experiences were unique.

And also that you can be more prepared to, to handle it. 

Philip Hemme: Must be also a bit confusing if you hear from eating woman, Oh, it’s the same. And if your experience is not the same, it must be like, Oh, is it me? 

Agnete Fredriksen: Exactly. Yeah. So I guess I, I just realized that I don’t want to have that burden put on the next generation of women coming after me.

So I’m happy to tell them, you know, in private or to some extent in public that there are differences and there are ways I’ve been, I’ve learned how to handle them. And there are things I cannot change myself. And there are things that… I think I know how to handle which makes it easier for the next generation.

Philip Hemme: That’s great. Can you share what you can, maybe some lessons or some things you want to share publicly of especially things that were in your control and that you could influence? 

Agnete Fredriksen: Yeah, I mean, so I, I think I don’t necessarily feel that I personally have experienced too much difficulties in the professional settings.

Also, I mean, obviously when you enter a room. There are people, if you’re not known to them, that normally can, can treat you as you’re not the, the founder and CBO or CSO or something. So, but that normally can, can change within a couple of minutes. And I think recent years, most of the people I’ve met already know who I am, if it’s in a professional setting.

So I haven’t experienced that too much personally, but I know it’s an issue. 

Philip Hemme: It’s interesting. How do you, in that example, how do you handle it? Like, like, do you behave in a certain way or how do you handle it? 

Agnete Fredriksen: No, I think there it just helps to be prepared so that you don’t have to be agitated in that setting and just, you know, hi and introduce yourself and.

And it also helps obviously to be with colleagues that, you know, you’re, treat you as who you are and support, support that aspect of it and maybe that, that is aware of it. So I think for me, it’s been important also to talk about these differences with my closest colleagues so that we are all prepared and can handle the situation in a way that, that I also feel is with some dignity in, in that setting.

But then I think that the more we move into something that is a bit more social and networking that’s maybe where I felt it was, has been much more challenging. And and I think that’s important also for, for other women to know about, particularly younger women in those settings. I think those social networking events are set up primarily by men and how they like to network.

And I think I’ve been very… It’s, you know, stubborn that I should not have less opportunities to be in those settings and also do. You know, get connections in, in this networking events, but it’s not easy. 

Philip Hemme: They’re very precious for business as well. I mean, that’s where networking of the parties at JPMorgan, by Europe, that’s where you make also really good connections, business connections.

Agnete Fredriksen: Exactly. Exactly. But they are not necessarily set up for, for doing that across the genders. And and you know, 

Philip Hemme: how did you, how, how, what’s your, 

Agnete Fredriksen: no, my tips, my tips now is to, this is one of the things I think my learnings personally is to leave early. So I don’t know how to, to change that immediately and also to be close with with colleagues and not, not be there alone.

Then I think. think to some extent it works, but I don’t think you have the opportunity the same extent to make new connections with people that you didn’t know before in those kinds of settings as a female which, you know, and then we could always joke about, you know, hunting or mountain biking and I both hunt and I do mountain biking, but I’m never invited to those kinds of things.

Networking events, that is not a party. Those are, in principle, I feel, who are males, connecting with other males. So I’m joking about, you know, should we set up a knitting cafe or, you know, should we have breakfast meetings with, you know, poetry over there? I mean, there are… I don’t know. But, but the, these settings, there’s a lot of these settings that are set up where male have the opportunity to connect and get new connections where we are a bit excluded.

So, so that part just feels that you have to focus on, on the more professional settings. Sometimes I think it’s unfair and limits the opportunities for us. but we’re slowly getting there. 

Philip Hemme: I think, yeah, I think, I mean, I think overall the situation is, I would say, improving, probably not fast enough, but still, I think improving.

I’m probably curious to hear on your side of, I mean, I remember from the, I think it was JP Morgan, I think it was the 2016, the scandal. Yeah, exactly. I mean, it was a woman strippers at the party, which is definitely not made for us. 

Agnete Fredriksen: It’s not made for us. You know, it’s, it’s really not made for us. 

Philip Hemme: Did you see any change since then?

I mean, it’s been over five years. Did you see something like changing? 

Agnete Fredriksen: Yeah, I think so. I mean, you know, from the early years when I started this 13 was outrageous. I think. Versus. I like that. Versus. I like that. I like that. Worse, yeah, but so I, I think it’s slowly changing, but you know, I mean, people are used to this.

There’s a lot of people that’s been in the industry for a long period of time and and they’re not necessarily there to talk business if there’s a woman in the room for good and bad or mostly I think for bad. So, so I mean, and it’s obviously not everyone but it just, you make, it makes you as a woman a bit more.

on the edge in the, in this settings. Cause you definitely just have one ambition and that is to, to also connect. I mean, we are as a special person. It’s a business setting. We also want to talk to people and we want to joke and have fun with with people we know partly and, and that we haven’t met before, that we would like to make a connection with and that can improve, please.

Philip Hemme: I think what you are raising as well, the like talking to your colleagues, male or female, but about, about the, the reaction and how to, or what’s kind of in the, I guess in the, what’s in the control of the group to, to improve. I think it’s super important, but I think also even now raising it up also for men listening to be more conscious.

I mean I can talk for myself, like I there are a lot of things that I don’t really notice. Something I would not notice. I’m not aware of it. No. But once I’m becoming aware of it, then it’s also easier to change. I mean, more from like some events, I would not be aware that it’s like made for men or that some women will feel uncomfortable.

But once I’m aware of it, then I can also change it.

Agnete Fredriksen: Yeah. And I think that that’s, you know, it’s not always fun to talk about these things I think, but I feel that we are a bit obligated to talk about the differences. I think we also talked about it about hiring. And realizing that, you know, I’m an ambitious woman. I like people to be surrounded by other people around me with the, with the passion to get things done.

And also, you know, in many settings, I do appreciate it if something happens urgently and we can discuss things at 9 PM in the, in the evening and realizing lately that with those kind of. Wishes and you give those kinds of wishes to a recruiter searching for someone that would be a new employer employee, sorry that would be more men that say I’m happy to be available.

And obviously I will always be there and work also at 9 PM if it’s if it’s something I know I’m so passionate. Well, a lot, you lose some women in the process just because they’re getting a bit concerned naturally. But then again, once they’re hired, you don’t necessarily experience the difference.

On the gender basis is in my experience. So it’s something about how we approach it when we recruit and how the recruiter approaches it. And trying to also see through the filters of how we naturally just behave potentially a bit different. I mean, obviously there are exceptions but between the genders on some of these differences between how we just respond to those kinds of questions.

Philip Hemme: But do you, yeah, I guess it’s a good example, I think. I guess you, you can mention that the, let’s say the potential employee can respond differently, but I guess the employer could also ask a bit differently. I mean, it can be on both. 

Agnete Fredriksen: Exactly. Yeah. And you have to, you have to both ask the question, not in a way that favors how men like to display themselves.

Yeah. not necessarily follow up. So that’s at least some of my experiences that it’s very tough for a woman to say, yes, I’ll do this. And I will always be there and I will perform day and night and 24 hours a day. If they know they cannot, you know, fill it, fill it up with with actions in the end, they are don’t necessarily like to overpromise.

And you, and you can experience that in the other aspect with, with some that come into an interview and say, I’m happy to overpromise. Maybe there is on a general gender basis, more. Men that overpromised than women, particularly women with families so they don’t necessarily feel it’s a good day if they’ve been in an interview, come home to their husband and say, I overpromised in this interview that I will be available every day, even though we all know that I need to pick up children in the kindergarten or whatever.

But maybe, maybe that’s not necessarily the same the other way. So then in the end, maybe the woman is at least as diligent and following up in, in my experience. So I think at least I’ve been more open to start talking about it. I know some women say. We shouldn’t talk about the differences because it’s all generalized and there are so many exceptions.

But I don’t think we change it if we don’t start to, to discuss it. 

Philip Hemme: I mean, yeah, it is super complex, super complex topic. I like you in the example. And I think I saw what one study from a big consulting agency for Harvard Business Reviews about. That’s one major factor of the salary difference, I think, was for the U. S., was actually connected to overselling or being too humble or too, like, allowing oneself. And that this had a huge impact on salary negotiation. I was surprised, actually, I was in the U. S. setting, but I guess we’re not talking, you know, a cultural difference, but because of sharing the gender and culture, it becomes multi complex.

Agnete Fredriksen: I’m not an expert on this and statistics and everything. It’s just some of those things I’ve been starting to experience myself that, that I seem to also get a lot of. When I talk to a recruiter, the ones that are presented to me are dominantly men. And then starting to realize maybe that’s also due to the fact that I’m telling this recruiter how I want my new employee to, to behave.

And then slightly changing it. And then you see a difference. So. 

Philip Hemme: No, that’s interesting. Yeah. I mean, on the statistic, on this topic, I mean, it’s, it’s a very, I think, quite subjective topic and we have some stats, but the stat, it’s not like science. I mean, you cannot measure it and reproduce it. But still, I think having a bit of stats data or some studies is probably better than even though I’m, I’m sure the studies contradict themselves like a lot in social science studies, right?

Agnete Fredriksen: Absolutely. And you know, there’s like, there are as many different men as there are different women. So, so, you know, it shouldn’t be a general topic, but, but. Maybe there are something that we can do. 

Philip Hemme: What’s interesting is when you cut, when you have some statistic cut with your own personal experience, and I guess your personal experience is not only once, it’s like several times in recurring, then it starts to be a bit more like a trend and more solid than just, just a stat or just a single experience.

Yeah. And I’m curious about the, also more from where we are, Scandinavia, Norway, Scandinavia. I have the feeling that, and I think there are some studies also that, that gender diversity in Scandinavia is, is better than the rest of Europe or than the US. I mean, first thing that comes to mind is the prime ministers, Finland, Norway but I think, well, I think most of the Scandinavian country had a. Women Prime Minister, which is not the case of not a lot of other countries, France, France. Did this like, I guess, have an impact or how much did it have an impact on, on you? Well, do you agree? 

Agnete Fredriksen: Yeah. Yeah. I, you, I mean, I think it’s also a lot of, you know. Gender equality has come further, I think, in the European countries is everything from from the opportunity for, for maternity leave, paternity leave which is also sort of more or less mandatory in our countries.

It’s also about. The cost of kindergarten being much less, so it’s obviously stimulus that women do go back to work and everything. I don’t necessarily feel that I had an experience that there was a difference in my career opportunities until I started to travel out on Norway. So, so definitely there are, there are, I think Norway has come much, much farther than that.

And I think when I travel. 

Philip Hemme: So you mean in Norway slash Scandinavia, you didn’t feel it, but you felt it much more when you went outside? 

Agnete Fredriksen: Yeah, particularly this, you know, if you establish some some business relationships with people in France, for instance, and then after a year or something, you start to talk about, you know, I have three kids at home.

And you see the shock in their face which is, you know, I think it’s pretty normal here that you do go to work and you can also travel, et cetera. Even though you have kids, it’s a different community, it’s, it’s not supposed to be different here if you are male or female in the, in, you know, the normal settings.

But do you experience that, that’s a, that’s a shock for. more people in the international world.

Philip Hemme: And the shock is more like from a they’re surprised or also a shock and it changes the business relationship. 

Agnete Fredriksen: I don’t think, I don’t think I felt that it changes the business relationship. I just feel that it’s like. That it’s almost like they’re judging you as to, I would not allow my wife to travel when we have kids or something in that that respect.

And I think maybe also some curiosity. So who is taking care of the kids? You know, are the kids fine? Do they grow up? I don’t know if it feels, it feels like something like my kids are so beautiful and so smart and so happy. So I mean, obviously that makes it easier for me also to, to travel. But it’s differences.

And I think, so it’s interesting when you talk about prime minister and all these things where there is a lot of gender equality, as you say, in our countries. But it’s also interesting in what I’ve experienced lately, because we do have some contacts with politicians, the amount of support they are offered, as there are always someone there prepping them, helping them getting there to the next meeting, making sure everything is in place, et cetera.

Which I think is really important. Think to some extent is also a bit comforting or attractive for women that enter into this leadership positions. And you know, I’m starting to experience it more as you may have seen today compared to like five years ago when you have to do everything yourself.

And you have to order your flight tickets and you have to make sure you come to every single meeting in time and you, you know, you need to have the logistics, you, you have to pay the bills and approve and you have to do everything which, which is, you know, demanding. It’s as busy now. But, but I do also have people that support me through the day and make sure that all the important stuff’s are taken care of.

So that’s. 

Philip Hemme: But this, so you, you put, yeah, I mean, but. you think in, let’s say, in the political world, they have more support. 

Agnete Fredriksen: I think you experience them when they come in here. I mean, there is always someone sitting next to them in a taxi that has prepped them for the next meeting. And this is the topic, you should remember these things, this is the things you’re supposed to say here.

And then, you know, they come and say, now the time is 1054. And now we have to leave and I’ll guide you to the next, et cetera. So that they can spend their time doing. Exactly what they’re best at. And not also simultaneously having to do, do all the other stuff. And that’s not the case with the founder position.

Yeah. You don’t have any support in the beginning. And you have to fight. Yeah. Quite a lot. You have to stand up for your own opinions in front of, you know, investors, partners, employees. So that, that may make it a bit more challenging for women in that, that early phase where you, you just have to do everything yourself.

Philip Hemme: I’m curious if I like, because I experienced that myself as a founder as well, where we would like do a lot of things. At some point where the switch, especially at Laboratec, when the team was growing and we invested a bit more in support functions, especially like a team support who could handle like a lot of different things.

And this was really game changing, of course, as founders, but I mean, as male founders, but I think as women founders, there’s also this learning curve of like, okay, a lot of small tasks. And when you accumulate. Yeah. Accumulated, it takes a lot, a big chunk of time. It does, yeah. And also mental time. Yeah. And having someone who can handle this.Agnete Fredriksen: It’s also stressful, right? Because you have to think about all these additional topics while you are actually trying to drive the business to the next. You also may have to make sure you’ve paid all the bills and you actually have a flight ticket tomorrow or not. Yeah. And it’s it’s,

[00:25:35] Gender equality on the board

Philip Hemme: and the last question on, on, on that topic is on the let’s say board membership, the board of, of Mercure partners. Did you, like, and I think, I remember that I think on the NASDAQ, there are some rules now for gender equality. Like, and I think that actually, yeah, and what do you, what do you think about, like, having more, like, rules, it’s all, like, not laws, but more rules coming more from top down.

Like, what do you think about, about those? 

Agnete Fredriksen: No, I, I actually think we’ve experienced here in, in the Scandinavian countries that, that it’s needed to, to be an accelerator of the change. And I think as I mentioned earlier, I mean, whatever we’ve done in Norway as to paternity leave and cost for kindergarten and all this, these things has facilitated that.

Yeah. No, women do work. We don’t have a, a whole lot of women that just stay at home in this country. So that’s, that’s an important thing and it comes from above. It also paternity leaves, sort of gets the, the male fathers to connect with the kids and take care of them on a. you know, have to be alone with the small kids.

That makes a change, not just for those months, I think it makes change for, for for afterwards. And I think also with the, with the board positions and also, you know as you, you know, in the government and everything, there are this, these kind of sentiments that we should show off gender equality.

I. I think it has been important to normalize it. I think it’s also, to some extent, maybe necessary to get to the first steps. Then maybe it’s not necessary in 10 years or 15 years or 20 years. But, you know, we all want to be in a company where you hire someone with previous experience and per definition, as long as it’s not equal, then you will continue with the same bias if you want to have people with prior experience.

So someone needs to take the leap and be brave and hire someone that don’t have prior experience. Or a bit less. Or a bit less so that the next company then can hire them again as board members or whatever, because then they have had the experience. So I think, I think it is necessary and I think it’s a, it’s a good thing.

And you know, you can always fight about this. I don’t want to be on the board just because I’m a woman. But I don’t think it is like that. I think we need to stop and, you know, we shouldn’t start saying, Oh, 50 percent needs to be a woman. We can start with, with something and then slowly this will change, it will normalize, you will get more and more with experience, it will get more and more that enter a board where you’re not the only female.

And then. Maybe sometime from now it’s, it’s positive, obviously everything is based on that we do think it’s a positive thing, but I feel that most, most companies do feel it’s a positive thing to have the balance of the genders and so, so then I think we need to get there. Yeah. 

Philip Hemme: Because on the board, I remember I was talking with Patch and I know that it was much harder for them to find a woman that is fits in the, in the board with the same experience was actually really, really difficult.

Agnete Fredriksen: But it will. Yeah, but I hope so. At least I enjoy being on the board. Yes. Thanks. And for me, it’s a great experience as well. And I think, you know, there’s a diversity. I come with a different background on, on aspects than than the other board members that are currently all, all there are made. 

Philip Hemme: Is it only?

Agnete Fredriksen: On the board. Yeah. Yeah. Yeah. Yeah. But, but. I think, I think it should be positive, but I also think, you know, someone needs to take that decision and say, okay, we need at least one. And then we maybe want one more. And then I will have the experience. 

Philip Hemme: Much more than zero. Exactly. Statistically. Yeah. 

Agnete Fredriksen: And I will have that experience for the next board and et cetera.

So, so and then it’s easier for me to also help motivate other females to be on the board. So. It’s a long journey. I don’t think we fix it overnight. 

Philip Hemme: It will take time. I mean, but as, as any, let’s say society changes, I mean, there’s so many things to change on so many different levels that it will take.

I went to a museum last week. There was a photo exhibition about. What was it? 1968 or 1969 like strikes for women rights. And the signs were like, women are not good only at typewriting. And I mean, in most countries, women had no voting. And you think within, yeah, I mean, it’s been 50 or 60 years. Which can seem very long, but also can seem not that long.

It’s always hard to know, okay, is it changing fast enough or not fast enough? It’s tricky, but I think there’s definitely some changes. When I saw it, I was like, okay, this seems like 200 or 300 years ago. 

Agnete Fredriksen: Exactly, yeah. And sometimes it’s good to take, like, to look back 10 or 20 or 30 or 50 years to see that, you know, realize how much it has changed in the right direction.

But there are so many differences in different European countries and also in other countries. And I mean, we travel all the time. So I think that’s also for Norwegian women that come from a country where they don’t necessarily experience the gender differences. as much, then I think there can be, they can be not prepared for what they experienced abroad.

Yeah.

Philip Hemme: I mean, yeah, that’s, we know, yeah, I mean, I think you are traveling a lot in 12 months. I am. It’s October. It’s October. It’s October month. 

Agnete Fredriksen: Not only October, I think, Philippe, yeah, in general, yeah.

[00:32:22] CBO rather than CEO

Philip Hemme: That’s great. Maybe let’s, let’s change to… then even your role, it’s one thing that’s I think I already asked you, but I want to ask you again.

When you’re co-founder, you’re founded the company, but you are in the, it’s ACBO role, you don’t want to take the CEO role. Mm-Hmm. . What’s, what’s behind that? What, what’s, what’s behind like, 

Agnete Fredriksen: no, I think, I think, you know, my major driver’s always been the company and the company’s success. And I think starting a biotech company and coming from academia and believing that that makes you the perfect CEO with all that entails which does not have a lot to do with the science only I think is not good for the company normally.

As long as you are able to work very tightly with the CEO on the strategy and the science and you have a good collaboration, I don’t think it’s the optimal thing to move into a CEO role. There it is. I mean, you can always structure this the way you want, but I think it’s good to have someone in the CEO role that has done it before.

And I think that that’s what I still feel. I mean, I don’t have, I, I think I could. I strive being a CEO in a new company that is, you know, starting at an earlier phase than what we are now, because now I’ve already done it and I do work very tightly with the CEO on all aspects, but we do benefit a lot from, from that tight collaboration and coming from different angles.

So, so I think it’s good for the company and I don’t, you know, think it’s good to take over the CEO role moving into a phase that you’ve never been involved in before. I think that goes for, for many aspects. But then again, I have more, most likely compared to a lot of other founders that come from academia.

I’ve had a more interest in the strategies, the business development, the investor relations, what else is needed to make this company a success. And that has always been something that I felt was super important because it doesn’t help if you do a good experiment in the lab, if it doesn’t, you know, drive the company forward.

And I, I guess that’s the reason for the change from being the CSO to the CBO in the end. So I could focus across the entire company and the strategies and how to move it forward in the right direction. 

Philip Hemme: At the end also, I guess the, I mean, it’s a letter or a title, but what you mentioned, how you structure it internally.

It’s also makes a huge difference. I mean, I guess, I mean, you are from, for so from what you said, sounds like you are doing with a lot of things cross whole company and it’s almost, I mean, I, I guess almost co CEO, like then like, but I guess, I don’t know in the Nordics how it is, but then some.

Countries that don’t like the CO CEO thing. Like France is like completely, yeah, yeah. Completely unknown. Yeah. In Germany it’s very common. You always have two or three managing directors. Yeah, but I think, I mean, and it doesn’t really matter then I think what you said that

Agnete Fredriksen: I don’t think it matters what we call ourselves.

Yeah. Yeah. 

Philip Hemme: Bring the best value and that it benefits the company. 

Agnete Fredriksen: Yeah. Exactly. And I think we benefit a lot, this company benefits a lot from me and the CEO collaborating so tightly. So we do a lot of the business development things together. I mean, we were the ones that were negotiating the deals with Genentech and Regeneron, leading those discussions together.

That’s, that’s the two of us that’s been on the road for this capital race that we did this week, talking to all the investors together in, you know, we compliment each other in what we can answer there so that we cover everything in the company. So we don’t have to take a lot of the questions back in those meetings and say, Oh, I’ll, I’ll go and check with someone internally and then come back with an answer.

Most of the things. We are able to answer in the meeting at the, at the level that is needed there. And I think that drives us forward in the right direction. And we, we get all that feedback from the external world as well, that we can take home and discuss amongst the entire leadership team as to where should we go.

What is the focus and the interest? 

Philip Hemme: I liked it. And I think it’s also great to be as a, how you say, do it or like, yeah, whatever the word in English is to also at the top level, because it helps also a lot, it first, it feels, I mean, you always have the thing, it’s lonely on the top, but I think if you’re two on the top, well, it’s lonely and being together in the meetings also helps a lot to get feedback, share thoughts, discuss the same thing that has been lived.

Agnete Fredriksen: Yeah. Yeah. And I think that’s like all the dinners, all the airport lounges, et cetera. It is a constant discussion about the, you know, the last meetings, the sentiment, what should then be the focus and the topics and what should be. Which, what should we tell back to the organization and what should we tell the board and cetera.

So, so I always liked it. I also liked it when I was CSO because I, I was on the road quite a lot when I was CSO, obviously the company was smaller, but this opportunity to then be out there, talk to potential partners, talk to investors, you know, understand what is. of interest. And what is, what do they think is just boring?

It’s not always that you can have that same sense if you are just working in the lab when you see I get good results here and I get good results here, which show those experiments. are the ones we should pursue further in order to make this company do something that is of interest for patients and, and shareholders in the end then.

Philip Hemme: I mean, in a way your investors and your partners are your indirect customers. Exactly. And they’re the gateway to the patients. So even in any, usually even in tech companies, like talking to the customer helps them to. Exactly. Funnel it back into the product. 

Agnete Fredriksen: Yeah. Yeah. That’s where I thrive the most, when I’m allowed to be there, like in the middle, so we can, we can try to take the right decisions that we are proud of in a half a year or a year from now, because we made something that was differentiated and meaningful for the patients.

Philip Hemme: But it’s also. It’s extremely, it’s an extremely hard position, also hard to get this experience and get this like, I can talk super deep about the science, the clinics, plus also investibulation in New York, and the business strategy, making a deal, I mean, it’s, it’s a very Oh, yeah. large range of skills and experience sets.

Yeah. Which I mean, 

Agnete Fredriksen: but in principle you need it a bit early as a founder and you know, in the leadership of a very small company, there is normally at least here with the limited funding, you don’t have. the best CBO and the, and the, you know, you don’t, you don’t have, or you don’t have investor relations right in the beginning.

So you need to take care of, of everything. Obviously, maybe I was a bit more interested in, in, in that part than, than all founders. But I, I’m also, you know, maybe extremely curious, curious and I like to, to do new things. So new thing. I think in these companies always for me been for the best of the company.

So if someone comes in and covers, you know, like I even had to do like manufacturing in, you know, a short period that I never necessarily liked or excelled at it, you know, then someone comes in and. And can take that position, then I can quick, pretty quickly move out of the daily operations and the decisions in that aspect.

And then I can focus on something else where we do have a gap still. And that’s maybe something I like that there is always somewhere we have a gap and I love it every time someone comes in and can cover part of what I’m doing today. So I don’t have to take everything on board anymore. Obviously this company’s grown all the time.

So that’s part of the journey. And some of those processes have been more painful than others. Also for me and also for those that come in. Obviously. But, but I think that’s what I like. If there is a gap, how can I do to fill it?

Philip Hemme: I mean, you grow also, I mean, curiosity and personal growth and you grow also in this pain, as long as it’s kind of bearable and the challenge brings a lot of stimulation or mental stimulation.

We could talk further about 

Agnete Fredriksen: Not a routine person. Maybe.

[00:42:10] Raising $45 million

Philip Hemme: And you. I think you announced from this week that you raised, so it’s 45 million if I convert in dollars. It’s a private placement with new international international investors. I mean, coming, what you said, being in the meeting and closing it.

And I mean, at the, at the moment also, it’s not the easiest time necessarily to often close. So can you talk a bit about, about it or like what? 

Agnete Fredriksen: Yeah, no, no, it’s been very interesting. I think as a background information, obviously we are born in Oslo in Norway, and we’re not a VC as such funded company in the beginning, it was in Norway, it’s been some interest from local family offices, et cetera that are not necessarily health, health care specialists, but had an interest in, in biotech and those, those local.

Investors have been very loyal and been with us all the time, but we haven’t been in the market for. any capital raise since 2018, which is very rare for a biotech company. You had upfront payments. Because we got the upfront payments, like 200 million from from the genetic deal. And then subsequently 50 million from Regeneron and then some Milestone.

So we spent the time really to trying to build this company and, and execute, but simultaneously knowing that at some time at, at, at time in the future, future, this company will need specialist investors and also specialist investors with really big funds. In order to support the ambitions we have, we have a platform technology.

We can make multiple different products within oncology. We can multiple products within now autoimmunity if it works. And, and that gives us an ambition to build a big company in the future. If, if it succeeds. And then Norway is most likely not big enough for really supporting a company with this.

And also that understands what’s the difference with medium overall survival of this compared to that, et cetera. What’s the difference if it’s a single arm with these data versus randomized, you know, what’s openly, you know, these kinds of details we need. And to have more specialist investors on board.

And now the market has been horrible the last few years. And and it has made it challenging as to know when are we going to race because we still had like 174 million in the middle of the summer. So no immediate rush. But if we want to support the journey of this company, we need to slowly first get some high specialist investors on board.

They don’t buy the, you know, normal stock on on the stock exchange. They want to be part of, of a capital race. So we’ve been on the road for a long period of time trying to get everyone to know this company that was born in Oslo and familiarize them with the technology and now getting some of the really, really, really best funds to say, I’m interested and I’m interested to the extent that I say.

Let’s do it now. So we got particularly this one big fund. It’s not yet public, but I think it will be soon. I’m super proud of of being able to, to start a collaboration with with this fund starting with a 20 million U S dollar from, from, from those. And then obviously made it easy to get the interest from, from the rest.

I think it was You know, multiple times oversubscribed and in this market that’s, that’s not normal. Yeah. So and that, that means a lot for us. I think we are now, I feel that we are now at the second sort of platform, I think for me it was you know, you’re climbing and then there’s a platform and Genente came on board.

So okay, validating the technology, also a lot of funding. Now, it’s also been a lot of climbing, but now it’s, yeah, I feel it’s like, it’s hard to get us down from this level of validation that we have now as well. So so very important for us these days. 

Philip Hemme: And like, I guess also what you said, but I mean, also, I guess it’s also a pretty small, I mean, a smaller investor community.

They’ve been very helpful, but I guess you rapidly tap into European or even US investors and especially US investors for big money like Most of the European companies, they get bigger funding from,

Agnete Fredriksen: I think if you’re like a single asset company, you can do very well in Norway, but I think as a platform company in the future, if this is to be successful, we need we need as we also said that we are obviously exploring a potential listing at the NASDAQ in the future and in order to get there, you should not go there.

with primarily local investor base. So this is an important step also for a future successful Nasdaq listing for us.

[00:47:47] How the science works

Philip Hemme: That’s great. Going to maybe more to the science, I mean, the, and how the science works starting from the beginning. I’m curious. Yeah, maybe you can just on the, on the short, like how, how the science really works.

How was antigens we shown to the APCs like I was, I mean, there’s some questions there. 

Agnete Fredriksen: Yeah. Pardon my, my recent point. It is a complicated technology in in principle. It’s not that easy to explain in, in simple words no, but we were, so we focus on, on immunotherapies primarily vaccines in principle.

Meaning that we want the immune system to elicit an immune response to certain antigens we call them. And antigens can be like the covid spike protein, if it’s antigen people know about or it can be antigens that are present in tumor cells or something that the immune system react overreacts to in autoimmunity.

And every time you elicit an immune response towards something, it has to, the antigen has to be has to be processed and presented within an antigen presenting cells. So it’s a certain subtype of cells we have in our immune system that is super important. And that was the original idea, just to make sure that these antigens do.

get effectively delivered and loaded into this antigen presenting cells. We sort of combine the antigen with a molecule that binds to these cells specifically. So so that was the original idea, but then again, realizing that different targeting units, so what we use in order to, to to bind to the antigen presenting cells.

They will all bind to different subtypes of these antigen presenting cells. And when they bind, they will be sort of internalized with different kinetics. So how rapid, and they will also go into different compartments. And all of these things determine whether you get this immune response profile or this remote immune response profile.

towards the same antigen. So that again gives us the opportunity to sophisticated technology and make sure that for cancer, we know we want primarily CD8 killer T cells and towards the antigen. We want it to multiple of the antigens, et cetera. And there we identified a targeting unit that makes sure the antigens goes to a certain subtype of antigen presenting cells and.

It’s routed within those ELs in a way that gives us the broad and strong CDI T-cell response, and that is something we apply for all cancer vaccines. Then for autoimmunity that we cancer, 

Philip Hemme: sorry, I think I still follow . Yeah, just for the, for how you present the antigen to the antigen to the APCs. Like what’s like, so you can engineer to select.

Which kind of APCs will pick it up or like, and then how do you, what’s the delivery mechanism there like? 

Agnete Fredriksen: Yeah, so all APCs are different surface receptors. So for cancer, we actually use a chemokine, an inflammatory chemokine as the targeting unit. So this is sort of linked to the antigen through a dimerization unit.

So when this is expressed or you give it and deliver it, we do it intramuscularly. Then the chemokine part of our vaccine actually attracts primarily dendritic cells, certain types of dendritic cells. So that these ends up being in the muscle cell basically, you know, in the context of where the antigen is.

So it’s very effective because the antigen is taken up sort of by the neighboring cell. So that makes it very effective to load the antigens. But then again, when it binds to the kiwokine receptors on these particular APCs, it triggers internalization through a different compartments within that dendritic cell.

As if it didn’t have the chemokine. Okay. And if it has the chemokine, it goes into sort of endosomal lysosomes, et cetera, and makes the epitopes be more effectively loaded and presented on MHC class one molecules or HLA class one molecules, which gives us. When we look at all the data, we see a broader T cell response.

So if you have a large antigen, we can get a T cell response to multiple different regions. And these are primarily driven by CD8 T cells. And we see that if we change the targeting unit, It will go to different subsets of antigen presenting cells and maybe into different compartments. So, so we have identified the targeting unit for our oncology platform that gives us, you know, independent of which antigens we use.

a differentiated T cell response that is optimal for the immune responses. 

Philip Hemme: The platform is really more like the IP and the platform is really there of like being able to present different kind of antigen. And deliver it to the APCs and make sure that there will be, that there will be the correct response for a certain type of…

Yeah, 

Agnete Fredriksen: exactly. Okay. That’s super easy. Yeah, and that’s what we’re now using in a completely different direction. It looks very similar, but then we changed the targeting unit. We can make sure the antigens are delivered to tolerogenic antigen presenting cells and be internalized in a different manner and give a different signal to that cell so that the immune response we trigger is still antigen specific, but it will be regulatory CD4 T cells.

That’s what we mean. And that’s what we use in order to then make sure that we get a response that is applicable to treat autoimmune diseases. We can work on this for like other aspects that would be more optimal for treating certain bacterial diseases or viral diseases, et cetera, in the future. But so far, we now focus on oncology and autoimmunity.

Yeah, 

Philip Hemme: that’s, that’s, yeah, and I think I’ve seen from your last presentation that you will, I think, announce a candidate by the end of the year in autoimmune. 

Agnete Fredriksen: No, that’s in oncology. Yeah. 

Philip Hemme: Okay. Yeah. Yeah. But you also, you already have assets in autoimmunity in the. 

Agnete Fredriksen: No, autoimmunity is very new for us.

So that’s where we’re working on identifying the optimal platform. And now we back in September presented some very convincing and nice data in, in preclinical models. And it’s in a sort of MS like model and a type 1 diabetes model, but it’s smart that there are, you know, preclinical models where we can test the principle.

So we haven’t decided which disease we will move forward with. We will start that work, or we have started that work, to identify our… Our first candidates, but also in parallel, you know, really identify as I said with this particular chemokine, that’s the one we use for all oncology, for for our autoimmunity programs.

We still need to decide exactly which targeting unit. We have multiple that seem to work. We need to find the best one. And also should we give it as DNA or protein or… MRNA or, you know we have all these abilities to, to change. 

Philip Hemme: That was my, my next question. Yeah. On the, on the form you’re, you’re giving it to get, I mean, so for oncology, if I understand well, so antigen is already synthesized and combined with the chemokine, it was a.

Agnete Fredriksen: So in the clinic, we have, we so far worked with a simple DNA plasmid based vaccine. So we have the, the plasmid, the plasmid backbone that has then the target, the chemokinase, the targeting unit directly linked to the genes that encodes the dimerization unit directly linked to the genes encoding the antigen.

So it will, after we give it to give it intravascular, it starts to secrete this fusion protein that, so the fusion protein, it’s a pla 

Philip Hemme: when you, so when you inject, your plasmid will automatically translate into cells, muscle 

Agnete Fredriksen: cells express. Yeah. And the muscle cells secretes this new protein that is not just the antigen, but then the, at the protein level, the antigen is linked to the targeting unit through a ization unit.

Philip Hemme: Okay. Yeah. Yeah. But how, so I’m still curious how the, when you inject it, how it’s automatic, like it transfects automatically or goes into the cells to be expressed or what’s the? 

Agnete Fredriksen: Yeah. So it’s not very effective if you just like take a needle syringe with DNA. You need to either formulate it in, in in some sort of formulation that makes it cross the cell membrane or what we are using is a simple device, a jet injector.

That basically gets it with enough speed, it’s not a gene gun and such, but it’s, it’s just like me, it’s a tiny orifice and a spring. So it just pushes it instead of a needle. It just pushes it through the skin and into the muscle. At such high speed that and it meets enough resistance so it spreads and it goes, you know, across the cell membrane and and gets into the cells.

So that’s very patient friendly way of delivering it. In principle, this device is also being explored as an alternative to needle syringes also for those with needle phobia and, and other aspects of it. So it’s a very simple device. I mean, we came over that bit as a coincidence many years back and we are one of the first companies that tested this for DNA vaccines in the clinic.

Not the first, but one of the first. And so far it seems to, to allow protein expression and without, potentially also without too much of this adjuvant activity. That’s the things we’re also thinking now when we see the clinical data. It seems that we generate T cells. That, you know, come up and they are not, it’s a DNA, so the protein is being expressed over a long period of time compared to, for instance, mRNA, or if you give the protein, it’s sort of a very short period compared to the DNA.

But also. There are obviously so much of anticity, you can say with the chemokine, some CPG motifs in the DNA, but not as much as with an mRNA, for instance. So we don’t see any signs of exhaustion of the T cells. And we also see this very long term, long lasting T cell responses that even one year after the last dose in our patients and on the trials we looked at, we still see effective T cell responses.

That may be, you know, a good thing with the DNA and with the jet injection where we have the IP to deliver it as an mRNA based vaccine if we wanted to. We don’t. necessarily know all the differences in the kinetics and how that will affect whether we get more exhaustion of the T cells if we move forward with mRNA versus DNA or another something that we would potentially like to explore with a partner.

You’re looking into it. So we have preclinical data with the, with our vaccine formulated as an mRNA LNP vaccine. Those data look good, I mean, if we compare it with the standard mRNA based vaccine that only express the antigen, we see the same benefits when we add this chemokine, and we see it faster, we see it stronger, we see also very importantly, a broader benefit.

Meaning two more epitopes. And that that obviously is so interesting. So it, it seems in that aspect as an improvement of the current mRNA based vaccines, particularly then with this chemokine for oncology purposes. So, so it’s intriguing to, to also get that tested. But we are also so happy with the data we have on the DNA.

So, so let’s see if that’s you know, maybe more likely that we would explore with a partner with mRNA competence.

Philip Hemme: I guess I want to say that some mRNA companies must be also intrigued. 

Agnete Fredriksen: They could, could potentially be a bit intrigued. 

Philip Hemme: And they’re pretty deep pockets at the moment. 

[01:01:20] Explaining the clinical results

Philip Hemme: And what I was what was curious also is.

I forgot my question is but on the, on the, on the on the results, I mean, this also explains the positive results you had. I mean, I just looked up from, from this year, so positive phase two, April, 2023 was a combination with a PDA one from, from Bosch, the centric in advanced cervical cancer.

And also what’s. I mean, super nice results first, I mean, especially for phase two. You doubled the median survival rate, and also you had a lot of very long lasting, remission free response, which I guess also comes from what you just mentioned, that the DNA injection like, generates a very long lasting T cell response.

Can you expand a bit more on the… On the clinical, on the results. Mm-Hmm. On? 

Agnete Fredriksen: Yeah. Yeah. I can , I mean, principle, we tested this this particular asset we done 16 earlier in the preclinical or in, in a precancerous cervical lesion setting. Mm-Hmm. Where, so we sort of knew that it was safe could elicit very strong and long-lasting t-cell responses.

As well as we saw a very nice correlation with lesion size regression. This is obviously much earlier stages and it was monotherapy. So we had very strong belief in the asset as such and its opportunities. Then we quite rapidly designed this trial that’s called VBCO2 with testing this VP1016 in advanced cervical cancer.

And I think over the, the period, we as management, so we didn’t see the results, right, but getting more and more concerned about opening the book and looking into this data because of realizing that the patients coming into the trial is. It’s not only having one prior line of systemic anti cancer therapy, but two and three and four and five, you know, really late stage cancer patients and cervical cancer is per definition a high unmet medical need patient population from the start and, and getting really the sickest patients into the trial.

It’s, you know, challenging and risky from an opportunity to see that the vaccine has the ability to change it. 

Philip Hemme: Especially in the immune set up, I guess, immune system is already very… 

Agnete Fredriksen: Immune system is, is, yeah, yeah. And it’s, it’s a lot of inflammation in the blood and it’s not the optimal setting. We’re not set up for success.

And then in the meantime, as well, we saw other cancer vaccine companies moving yeah. Away from late stage in any indication, and while, while almost focusing on, on this early stage adjuvant setting. So we were quite concerned, I would say about whether those data would would give us anything that we could build up on for future development.

And I think we were so happy that the day the, the team had worked like on this for a few days before I got to, to know it, which is the worst period in my life. And then presenting the data with a lot of enthusiasm. This was first the interim data a year before. Where we saw, oh, really, that, that looks very promising and then a bit more optimistic when we should read, have the final readout.

But the overall survival data being as strong as they are I think I think that’s transforming for the company and obviously most likely also facilitates that we could do this capital race now with interest from specialist investors and, and really. you know, move this forward into now a potential registrational trial.

So, so for me, it was a super important days. 

Philip Hemme: I mean, that’s the phase two efficacy for other biotech companies. Yeah. It was huge. I mean, you talked about Yeah. Reaching plateaus. 

Agnete Fredriksen: Yeah. That’s a plateau. That’s a huge one. It’s, it is absolutely. Yeah. And I, I know, you know, our chief development officer is planning these days where he had like paper printouts all over his floor.

Every single patient, you know, double checking how long they survive and how long they actually progressed, et cetera, before coming to us and presenting the results. So. So, those data are, those days are extremely, extremely, and you have to be so sure that everything is, is correct before we go out and it, it feels much better afterwards.

It’s a single arm trial, but it’s a single arm trial and will, you know, what looks like be at least a doubling on the medium overall survival. Now they came, new data came out now with the skyscraper for trial testing again and it’s the same trick in more patients than in PD L1 positive patients, again, heavily supportive for us repeating that medium over, so I was less than 11 months.

Meaning that, that we get more and more confirmation that these VB1016s seem to, to help patients. And that’s what we’re here for. Yeah. 

[01:07:19] The immunotherapy field

Philip Hemme: Talking about the, if, if we zoom out a bit on the immunotherapy fields. I mean, so it’s massive breakthrough and massive benefits for patients, L1. At the same time, I heard that quite some, let’s say, some other checkpoint inhibitors or some other pathway didn’t work.

Hmm. As well. And I think a lot of combination trials also didn’t work that well. I mean, the one that works that comes to my mind is the one that Moderna did with Merck. Yep. I think that was very positive. Yep. Especially for other mRNA. Yes. Cancer vaccine. Yeah. Can you. Expand a bit there, like, kind of a zoom out view on, on where does the field stand?

Agnete Fredriksen: Oh, wow. Yeah. No, I think we definitely experienced also when the MERS Moderna data come out, came out that it was very much creating a boost in the interest of cancer vaccines in general. So I think these things are super important and it supports the entire field. Moderna’s data are with personalized or individualized cancer vaccines.

Thanks. which we are also heavily invested in. We have partnered our individualized or personalized cancer vaccine program with Genentech and are doing studies together with them so far in advanced setting, but obviously it’s Yeah. Of more and more interest also to, to move into earlier stages.

Also with our assets. And I think they, I mean, they are supportive as to cancer vaccines should have a role. I think in the future that cancer vaccines should be the first thing you’re offered. When you have a diagnosis, there are really, as far as we know, there are no safety issues with the cancer vaccine compared to all other cancer treatments out there.

And, and all cancer immunotherapies basically do work one way or another because they allow cancer specific t-cells to, to, to be functional within the patient. So adding a vaccine or just giving the vaccine first, which. Its primary role is to generate those cancer specific T cells, should also always be a basis in every single patient as long as there is a cancer vaccine available.

So I do believe that if we look at 

Philip Hemme: the cancer vaccine is also effect, I mean, shows, 

Agnete Fredriksen: yeah, and it should show most likely more efficacy the earlier you come in the treatment. So I think the entire field is moving in that direction to get cancer vaccines in as early as possible. I do, however, think that while some of the other cancer vaccine developers have stated that they’ve basically given up on advanced setting for cancer vaccines and think it’s too challenging, I think our data clearly indicates that with this differentiated immune response profile that we generate.

That we can help patients also that have failed other therapies. So we will not limit ourselves to only early stage adjuvant setting, but obviously also want to operate in that space. And I think the data we’ve seen, if you see is long lasting T cell responses, we really see very few patients progress if they first have either stabilized or had a resist response that tells us that in an adjuvant setting after surgery.

The likelihood of this vaccine really helping the patients to not get recurrence. is very high. So we obviously want to operate in that space. I think the fact that some of the other cancer vaccine developers have stated that they’ve given up after failures in advanced setting, they also see maybe some signs that it works much better in adjuvant setting if it’s even ctDNA negative versus ctDNA positive, meaning as long as there are as you know, tangible amount of tumor cells present.

It is more difficult for some of the other cancer vaccine technologies to really provide a meaningful clinical benefit. I believe our technology with the, with the differentiated CD8 dominant T cell response to multiple epitopes over time can be the right platform in advanced and in early stage. 

Philip Hemme: Yeah, I mean, that’s, and that’s actually the, the question I forgot that, I mean, what’s interesting is that you are, you know, you could be potentially agnostic of DNA or mRNA, so that’s whatever delivery, 3T, if you’re agnostic of advanced or early stage, I mean, this makes sense as a platform.

Yeah. Diagnostic of cancer and, then it starts to. 

Agnete Fredriksen: Exactly. That’s why the growth ambitions are pretty, pretty big. Opportunities are big, but we also, I mean, we do have partnerships. So, Regeneron is working with with three oncology programs and two infectious disease programs. I see with the mRNA data that we broaden the the scope of potential partners that would be interested.

So far. Since we have focused ourselves on the DNA, even though we said, I mean, you can use mRNA if you want to having more of these data available makes it also likely that we broaden the scope of potential partnerships in the future. So, and then let’s see what works best in the end. 

Philip Hemme: I mean, it’s also a bit chicken and the egg thing.

I mean, you have to start somewhere and what’s in your control is to generate the data. And I guess advanced patient is also a way to have a proof of cancer. Well, I mean, you’re showing that and then going up the, I guess I can imagine, at least from my knowledge, hearing from a lot of biotechs, it’s very, very hard to access cancer patients earlier.

More from the equipment and from, 

Agnete Fredriksen: Yeah, a bit dependent, I think, on, on, on the safety profile and things but, but yeah, I think it’s getting easier than it was 10 years ago to move into earlier stages. Also for us to do combinations with checkpoint inhibitors, because we see checkpoint inhibitors do move earlier and earlier.

So, so now we recently got strong indications that Ketruda or Pembrolizumab will get approval in. adjuvant setting in cervical, which is why we are now pursuing a combination with that in, in in earlier stages of cervical cancer and expecting potentially to see the same sort of movements in head and neck soon, which opens the space.

So I think, I think it’s easier and easier to, to get access to those patients, but those trials do take longer before you. You get to the proof of concept you need more patients because they are healthier per definition. So they take longer before you get the readout. That’s the good thing with advanced, if you look at it from a development perspective, but not from any other perspective.

So now I think we’re on the right path. So we started with late stage, can move to earlier stages. We started with DNA. We can broaden if if we want to through partnerships, et cetera. And now we focused on oncology expanded autoimmunity. We can do other diseases in the future if we want to, but let’s focus step by step.

Let’s do, let’s do it step by step 

Philip Hemme: from the, the combination. You. could also do it as a standalone or it just works better? 

Agnete Fredriksen: No, I mean, so interesting. I mean, as I mentioned before, we have done a monotherapy study in pre cancerous cervical lesions which looks good. So I think it depends on, you know, how That’s how sick the patients are potentially, but actually in this CO4 trial that we are about to start there we will have the first part of that trial will be testing VB1016 plus decentric versus VB1016 alone.

I believe that the combination in second line patients, as this will be will have a higher likelihood of of providing the optimal clinical efficacy, but we will see. 

Philip Hemme: So in the trial you will, you will test your OVB 016? Yeah. Alone? In combination with checkpoint inhibitor and then the checkpoint inhibitor alone as well?

Agnete Fredriksen: No, not the checkpoint inhibitor alone. Because these patients have just failed Ketruda or Pembrolizumab. So the, the, the investigators and the community does not believe in re challenging with another checkpoint inhibitor alone. So it doesn’t make sense. So probably very hard to recruit patients on that arm.

So in that particular setting, it does make have 

Philip Hemme: one in combination. One not in combination. 

Agnete Fredriksen: Exactly. And exactly. Yeah. So it also gives us some, you know, level of contribution of the components with which for, for FDA’s so importance.

[01:17:02] Biotech in Oslo

Philip Hemme: Nice. They, for just about to, to finish, to wrap up, but connect it to Oslo.

I mean we have an amazing view here. , the whole show a shot. So many people can see it in the intro. And how, how was it to start the company here? You said, you said that access to some investors or generous investors, how was it here? And also, I think you have an office in Copenhagen, Wadi, and maybe one in the U.

S. soon, I don’t know. So how, how was that from a, more from a location point of view, connected to your story since 2006? Yeah, 

Agnete Fredriksen: yeah. No, I mean, the headquarter is here, basically, because that’s where it all started with I did my PhD across the road in university in principle. So so that’s where we started.

It’s been good to, to, to found a company in Norway, but it’s, you know, honestly, it’s very few biotech companies and, and not all companies, biotech companies that were around are around anymore, which is logical. No, it’s fewer and fewer maybe, and then there are new things coming up, which we, we hope will succeed.

That has made it more challenging, I think, also from the, you know, the entire value chain with yeah, I mean, so there is not a VC, strong VC community here, maybe getting better. But compared to Denmark, for instance, it’s, it’s very different since there are no, you know, not a lot of other biotech companies, it has been more challenging.

Not necessarily the first part with having people working in lab, because I think there’s a very strong academic science community here, but to take those decisions of what am I doing with that science? How should we move it forward? The development part of this, the business development part of it, there is not a lot of prior competence that we can access.

So that’s one of. The reasons now that we’ve grown into a company with close to 180 employees is that we set up an office also in, in Copenhagen, where they do have a longer history of biotech and also biotech that has entered into the later phases so that you have, you can access competence. They have done it before, as I say, I still, you know, also here believe.

That we do break boundaries, but we don’t need to break boundaries without having people that have already prior experiences. So, so that’s the, the Danish office is around 30, 35 employees. And where we have more of the late stage regulatory development people, a lot of management at the moment while here we have the labs, all the preclinical, a lot of CMC.

Yeah. So that works as a good combination.

Philip Hemme: No, that’s amazing. I mean. I mean, it, and it is an amazing office and the whole science project looks really nice. And I think they’re also building up or building. 

Agnete Fredriksen: Yes, another life science building here. So they are trying to build build this industry. I think we are now a bit separated, right? So we have this very nice floor.

We have the lab in one end of the building and we’re a bit separated as a company, but there are also a lot of benefits. So. Or being in a science park where the infrastructure and also the opportunity to connect with companies. 

Philip Hemme: Oh, great. I think that’s a great final word. I mean, congrats on the whole journey.

Thank you. It’s, it’s, it’s amazing. It’s amazing for, for the ecosystem here, for, for patients, for everyone, I mean, it’s, it’s great. So, and thanks a lot for taking the time. 

Agnete Fredriksen: Yeah. Thank you for coming.

[01:21:08] Thanks for listening

Philip Hemme: I really enjoyed the conversation for having it open on such a sensitive topic and the company she’s built.

I hope you enjoyed it too. And thanks for listening to the end. If you did, please hit the like, subscribe button on any platform you are. You can also leave a review on Spotify or Apple Podcasts. Any of these actions will help us a lot. And I would also be curious to hear what you think. So you can please leave a comment on YouTube, Spotify or any platform.

We are reading all the comments or you can also shoot me an email at philip at flod dot bio. All right, now I have to get back to Berlin and see you next episode.