Podchaser Logo
Home
Inhalable mRNA with Ethris' Carsten Rudolph, Ph.D.

Inhalable mRNA with Ethris' Carsten Rudolph, Ph.D.

Released Monday, 5th February 2024
Good episode? Give it some love!
Inhalable mRNA with Ethris' Carsten Rudolph, Ph.D.

Inhalable mRNA with Ethris' Carsten Rudolph, Ph.D.

Inhalable mRNA with Ethris' Carsten Rudolph, Ph.D.

Inhalable mRNA with Ethris' Carsten Rudolph, Ph.D.

Monday, 5th February 2024
Good episode? Give it some love!
Rate Episode

Episode Transcript

Transcripts are displayed as originally observed. Some content, including advertisements may have changed.

Use Ctrl + F to search

0:00

The business of biotech is produced by

0:02

LifeScienceConnect and its community

0:04

of learning , solving and sourcing

0:06

resources for biopharma decision

0:08

makers . If you're working on biologics

0:11

process development and manufacturing challenges

0:13

, you need to swing by bioprocessonlinecom

0:16

. If you're trying to stay ahead

0:18

of the Cell or Gene Therapy curve , visit

0:20

cellenginecom . When it's time

0:22

to map out your clinical course , let

0:25

clinicalleadercom help , and

0:27

if optimizing outsourcing decisions

0:29

is what you're after , check out OutsourcePharmacom

0:32

. We're LifeScienceConnect and we're

0:34

here to help . Considering

0:40

that about a month ago the CDC reported

0:43

that just 8% of US children

0:45

and 21% of US adults were

0:47

up to date with the latest COVID-19

0:49

vaccine , which at the time had

0:51

been available for a few months , it's

0:53

no surprise I brought the virus home

0:56

from the JPMorgan Healthcare Conference

0:58

. Between COVID fatigue

1:01

and vaccine hesitancy , getting into

1:03

the COVID vaccine game right now might , at

1:05

face value , seem like a bad business

1:07

move . That is if you're

1:09

coming at the COVID vaccine game from the same

1:11

perspective Moderna and Pfizer BioNTech

1:14

did back in 2020 . But

1:17

maybe if you bring a new vaccine

1:19

paradigm to the population , you'd find that COVID

1:21

fatigue and vaccine hesitancy are actually

1:24

the driving forces behind

1:26

commercial acceptance of your product . I'm

1:28

Matt Pillar , this is the Business of Biotech and

1:30

I'll never knock the incredible result

1:33

of the public-private partnership that we saw

1:35

during Operation Warp Speed . But

1:37

to cut to the chase , there's room for improvement on

1:39

many fronts . We all know the first

1:41

iteration of new technology is never

1:44

the best iteration of a new technology

1:46

. I won't put words in his mouth

1:48

, but Dr Carsten Rudolph , CEO

1:50

at Ethris , agrees with at least

1:52

some of this sentiment . Otherwise he

1:54

wouldn't be taking his company down the path

1:56

of developing intranasally administered

1:59

and inhaled vaccines and therapeutics

2:01

for a number of respiratory indications

2:04

, COVID among them . I

2:06

caught up with Dr Rudolph in San Francisco

2:08

to tape today's episode . Let's

2:10

give it a listen . That's where I want to start . I

2:12

want to start with a little bit of background

2:15

on where you came from and how

2:17

you got into this space . Obviously

2:19

, I did a little bit of research . I

2:21

trolled your crowd up on your

2:23

LinkedIn profile a bit and I learned

2:26

that you have a pharmaceutical degree

2:28

and have , quickly

2:30

on the heels of that

2:33

, you dove into the mRNA space

2:35

, at least it appears that way .

2:37

Yeah , yeah , what does it mean ? Quickly

2:39

, I did my PhD

2:42

exactly at the Department of Epidemiology

2:44

, at Pediatrics , basically in Munich

2:46

, yes , and at the Ludwig-Marsimius

2:49

University . My

2:51

PhD , I was mostly focusing

2:53

on creating

2:56

, developing a gene

2:59

therapy approach for the

3:01

treatment of cystic fibrosis , because

3:04

cystic fibrosis , of course , is in our specialty

3:07

or university

3:09

pediatric hospital . It's

3:11

a quite prominent disease

3:13

. This was always my idea to

3:15

do research

3:19

and providing a new potential

3:22

cure for these patients by delivering

3:24

genes , of course , that replace the missing function

3:26

in the body of these patients . But

3:29

I was mostly working on

3:31

non-biology and delivery . That means

3:33

just using classmates

3:36

and we all know . I

3:38

think you're also a biologist , aren't you ?

3:40

No , no , no , we should have set that record

3:42

straight from

3:46

the outset .

3:47

But then I'll give you a brief biology

3:49

tutorial . You've got to toll

3:51

the line here because I am not

3:54

.

3:54

Listeners of the podcast know I've said

3:56

it many times I dropped out of advanced placement

3:58

biology my senior year of high school

4:00

.

4:00

However , our audience will

4:03

appreciate a little bit of flavor

4:05

, but one real problem for using that's plasma DNA then

4:12

, of course , is how to

4:14

get the plasmid

4:16

into the nucleus , Because the gene

4:19

, of course , is active in the nucleus , sarah , and

4:22

the lung tissue is a quite

4:24

post-metonic tissue , so it's not

4:26

fastly

4:30

proliferating when the

4:32

nucleus membrane breaks down , basically

4:35

, so it's really a limitation

4:37

. And then we thought

4:39

, well , and we knew in those

4:41

days basically that our delivery

4:44

systems and model particles that we

4:46

used , we could pretty well shuttle

4:48

the messenger RNA into the

4:50

cytoplasm . Yeah , so that worked

4:52

quite well . And then we thought

4:54

is there a nucleic acid , basically that

4:57

you could make use for the turgis that

4:59

is active in the cytoplasm

5:02

? And of course there's

5:05

the messenger RNA that can be translated

5:08

at the ribosome to produce

5:10

the missing protein . Then this wasn't the

5:12

idea . Well , why shouldn't we use messenger

5:15

RNA ? In those days no one was really doing

5:17

a lot of work in this space here and we

5:19

stuck it on this and we saw

5:21

, well , that works really well . They're

5:23

much , much more efficient than using

5:26

plasmids . But we saw at the

5:28

same time that just

5:30

the standard messenger RNA molecule

5:32

and we know this all now from the pandemic

5:35

vaccine development as well is quite emogenic

5:38

Because we have a lot of sensors

5:40

in our innate immune system that

5:42

is made for detecting these

5:44

foreign RNA molecules , because this could

5:46

signal just invasion of the virus

5:49

, like the coronavirus , which is an RNA

5:51

virus . So we thought , well , it's

5:54

very efficient , but it's also

5:56

quite emogenic , basically . And then

5:59

we thought , how can we navigate around

6:01

this here ? And then we thought about

6:03

modifying the messenger

6:06

RNA and introducing modified

6:09

nucleotides , because we know from you

6:11

, from the SIRNA field and the

6:13

oligonucleotide field in those days , that

6:15

if you do so you can really reduce

6:18

the immune response and recognition

6:20

of the RNA molecules . And then we

6:22

played a little bit of ground and we found really a

6:25

scheme where we can change the pattern

6:27

of the RNA molecules

6:29

so that is not recognized

6:31

any more that well by

6:33

their immune system . And then

6:35

we thought , well , that works quite well , shouldn't

6:38

we exploit

6:40

this potential of such molecules

6:43

? And then we started to find the problem here

6:45

. So that was the idea behind it .

6:47

Yeah , that's why the question I asked

6:49

you is you were talking about this research work

6:51

and we like who was we at the time ? Like pre-athrists

6:54

, who have we ?

6:55

at this . This was my research team basically

6:57

at the Department of Pediatrics and my

6:59

colleagues over there , and I shared the lab

7:02

space with another professor and

7:04

we were running those SIR

7:06

experiments in these days here and then you

7:09

know , I was always because we all know . You

7:11

know that there RNA molecule itself

7:13

, a negative RNA molecule , is not

7:15

taken up by the sample SIR . It does not

7:17

penetrate through the cell membrane and

7:20

so you always have to package this , basically

7:23

or the nanoparticles , and then taken up to

7:25

release the mRNA payload

7:27

in the cytoplasm .

7:28

Yeah , and .

7:29

I was always also intensely collaborating

7:31

with now also

7:33

my micro-founding partner of the company

7:35

, christian Klang , who was running

7:38

and a research group as a professor

7:41

at the Technical University in Munich so

7:43

that's their second large

7:45

university in Munich and he is a

7:47

really expert , well-leading

7:50

expert in designing our

7:52

carrier systems for nucleic

7:54

acids . And then we thought basically

7:56

, yeah , we can put this together , the

7:58

expertise and turn it

8:01

from the company and

8:03

then after we got the first business angels

8:05

convinced that then we were really

8:07

brave .

8:08

Oh , no

8:11

, we have to do it .

8:12

I will always remember this moment , this

8:14

funny moment . So my plan was just

8:18

something I want to become a full-time professor

8:20

, basically , but now I became an entrepreneur

8:25

?

8:25

Yeah , you brought it up , so now I want to dig into it

8:28

. You made that decision . So

8:30

many questions there . What

8:33

was your biggest reservation ? So you thought destined

8:36

for professorship . And

8:38

then you did this work and you realized well

8:40

, there's the foundations of a biotech

8:43

here . What was your biggest reservation

8:45

about making that leap ?

8:48

I would not say reservation , it was more

8:50

a , basically because I

8:52

always thought that if you do research

8:54

here at the university here

8:56

, this also

8:59

should be something where

9:01

you can , because I mean , this is basically the

9:04

both tax money here at France that we

9:06

raised there . That would be something that you

9:08

have to give back also and make

9:10

something useful out of it . So that

9:12

was always my intention of

9:15

my research that I did at the university . So

9:17

we thought , no , we just have to try it now . And

9:21

of course I mean , if we had failed , then there

9:23

would be always the opportunity then go back

9:25

to academia . So . But we

9:28

thought , no , if we don't try it , then

9:30

who will pick up those ideas that we develop

9:33

? No one . So we thought no , we have to restart

9:36

and do it by all .

9:37

That's how we did it . That's great .

9:39

We now have the first program in the clinics . It took

9:41

us some time . It's

9:44

a complex thing to develop

9:46

for a pulmonary messenger RNA

9:48

therapeutic .

9:49

For sure , and we'll talk a little bit about that clinical

9:51

program in a little bit . But

9:54

from the business perspective it

9:56

almost sounds like a bad joke . So two scientists

9:58

are in a bar right Doing

10:02

mRNA research . When

10:05

you decided that you were going to launch

10:07

a company , you said you didn't really have

10:09

reservation because you knew that you wanted to make a contribution

10:12

with what you learned . But

10:14

building a company is a different thing from

10:16

being a research academic , or professor

10:18

, to that matter . So what

10:20

? I guess maybe not reservations

10:23

, but what were some of the early

10:25

steps that you took around learning

10:27

how to be at the helm

10:29

of ?

10:30

a biotech company and the first thing is to

10:32

write a business plan . Basically that

10:34

was the starting point and this was

10:36

basically , I have to say , that was

10:38

a trigger . Then , basically , you know , we had in Germany

10:41

also business plan competitions , basically

10:43

for young startups , and

10:46

then we thought , OK , we try to just

10:48

put down the paper , our

10:50

thoughts , basically , and then you get , of course

10:52

, structured thinking around yeah , so what

10:54

you all need and what kind of

10:56

value proposition , what kind of

10:58

drug , and to start

11:01

or to create a financial plan

11:03

, budget and all these things

11:05

. And there I think we self-trained

11:07

ourselves mostly , I would say . And

11:11

then we thought , yeah , we will try it now and

11:13

we will do it and

11:15

we will learn while we do it . So

11:18

that was the starting point .

11:19

Yeah , did you have support resources

11:22

? So like here in the States ? You know I'm not

11:24

sure what the biotech scene looks like in Munich

11:26

, but here in the States you know oftentimes

11:29

startups will take advantage of communities

11:31

, incubators . You know even parent

11:34

companies that spend biotechs on . So

11:37

I'm just curious , like , to what degree did you have

11:39

the opportunity to lean into

11:41

folks who had founded

11:43

biotech companies ?

11:45

Yeah , so there was definitely some exposure

11:47

, you know , because our first business angel who

11:50

helped us also to

11:52

move this forward and

11:55

look

11:57

into the right things that you have to

11:59

basically consider yeah , so that

12:01

was . And then there is , of course , I mean , also

12:04

in Munich . You know we have a kind

12:06

of small biotech hub there

12:08

with their kind of infrastructure and

12:10

this was also good when we got some

12:13

exposure here to this

12:15

sort of creating business .

12:16

Yeah , yeah , getting back to the

12:19

science that you founded the company

12:21

on , you said it started

12:23

out with research and cystic fibrosis

12:25

. Right Was the by the

12:27

time you realized that you had something

12:30

that was really building a biotech

12:32

around . What was sort of your

12:36

therapeutic intention at that

12:38

time , like in terms of indications

12:41

and targets , did you think , well , we can move

12:43

this well beyond where it ?

12:44

started . Yeah , it was always this

12:46

. Focusing to the pulmonary space , you know

12:48

, because that's where I have

12:50

my home , my scientific home , so to

12:52

speak , and where

12:55

I knew this field very good

12:57

, the biology and , of

12:59

course , the exposure to our

13:01

thinking of what you have to consider for

13:03

delivery , etc . So

13:06

and with I mean there are

13:08

really a few severe diseases

13:11

, therapeutic

13:13

diseases , where there's not

13:16

a good cure available . Yeah , so

13:18

we thought , no , that's a good place to

13:20

go . Basically , I mean , we did also some

13:22

work in those days in bone

13:24

regeneration , you know , to locally

13:28

translate our proteins

13:31

that are in grafted oncology and sponges

13:33

to induce

13:35

bone healing and non-critical

13:37

defects , you know where bones are

13:40

broken and they don't

13:42

know anymore that they've been all

13:44

together . So you have

13:46

the help there with biology that we

13:48

did also some work in this space . Basically

13:51

, but mostly we decided

13:53

to look and go into the lung space .

13:56

Yeah , okay , remind me what timeframe

13:58

this was . What was after this ?

14:01

So we started to look into using messenger

14:04

RNA . That was roughly around the

14:06

2006-2007 timeframe

14:08

, yeah , and we

14:10

followed the initial most important

14:13

patterns of our technology in 2009,

14:16

. Yeah , and then we started

14:18

the company towards

14:22

2011, . Yeah , basically

14:24

and really truly operational

14:26

we became in 2012, . Yeah , it's

14:28

pretty much 10 years from now , yeah

14:30

.

14:31

That's , I mean relative

14:33

to the recent , I

14:36

guess , advance in awareness and

14:40

even growth , just in terms of the industry and the number

14:42

of companies who are playing in this space . Those

14:44

were early days .

14:46

Yeah , those were the very early days and also

14:48

, you know , in the academic setting , basically

14:51

there was pretty I mean , I

14:53

think , pretty much a lot

14:55

of the birth of the messenger RNA

14:57

drug modality . I think

14:59

this was came really from Germany

15:01

, yeah , so we had the longest

15:04

there . I think that were our colleagues from QVAC

15:06

, basically , I think more , who were the first

15:08

one who tried to develop

15:10

messenger RNA for cancer vaccines . Then

15:13

we had Hugo Schahe in the same time

15:15

, the founder of BioNTech

15:17

, who you were thinking of , developing

15:20

messenger RNA for the cancer treatment

15:22

, and it was us basically , I

15:25

think , to develop our pulmonary therapeutics

15:28

. So this is all roughly in

15:30

that timeframe , basically , yeah .

15:32

Yeah , what's fascinating in

15:34

those early days . You mentioned your business

15:37

angel your angel investor From

15:41

there to sustain the company and

15:44

to grow the company . What was the

15:46

investment scene like

15:48

in this therapeutic modality

15:50

in those alley days ?

15:53

It was tough , basically because

15:55

there was not this awareness

15:58

of messenger RNA . Now everyone

16:00

knows what messenger RNA is . Of course , in

16:02

those days they were rather not

16:04

. There

16:07

was also a time where the SIR RNA

16:09

therapeutics there were waves . It goes

16:12

up and down . There

16:15

were sometimes some into the SM

16:17

and then I remember

16:19

a time when Roche stepped

16:21

out of their SIR RNA . Then

16:23

there was again less

16:25

appetite . Then it took a little bit

16:28

again to create the awareness

16:30

. But this was early . It was really

16:32

entirely new in those days .

16:36

So how did you , as an academic and founding

16:38

the company with an academic beyond that

16:40

angel investor , how

16:42

did you learn to navigate

16:45

the biotech fundraising

16:47

scene ? What did you do in those early days to maintain

16:49

Speak with a lot of ?

16:51

people yes , speak with a lot of people . Basically

16:53

they are going to

16:55

meetings , to conferences to

16:59

find investors . That's what

17:01

we then also found , basically our

17:03

first investors

17:05

. We then basically

17:07

invested with the company so that we could really

17:09

do the first steps .

17:12

Did you see a spike

17:17

in interest from the investment

17:19

community during COVID ?

17:22

Yes , I think so Because

17:26

of the awareness that

17:29

we got from the mRNA

17:31

space . I think it's very

17:33

important for the theater . In

17:35

the end of the COVID pandemic there was a

17:37

nightmare for the world , but

17:40

for mRNA it's

17:42

a great success . It

17:46

could be shown that the full potential of

17:48

messenger RNA that you can develop really

17:51

fast drug products

17:53

. Most important , we have

17:55

approved our mRNA

17:57

vaccines . It's also now clear

18:00

that this is a new drug

18:02

modality that can

18:06

achieve a proof of that . I think that's very

18:08

important for the entire field . Now we

18:11

have so many new great

18:13

areas where people

18:16

now

18:19

explore the potential of messenger

18:21

RNA . It's a new drug modality

18:23

that's fantastic to see .

18:27

Events like that obviously a

18:31

global pandemic is going to have an impact

18:33

on biotech

18:35

, not

18:38

just in terms of the repercussions

18:40

that we saw accessibility of

18:42

raw materials and

18:45

outsourced capacity , that kind of thing . But it can also

18:47

alter the course

18:49

of the company's

18:52

pipeline ambitions . We saw it in COVID

18:54

. There were hundreds

18:56

of biotechs that didn't have

18:58

COVID therapeutics .

19:02

Suddenly we had a little bit of the same policy

19:04

. We embarked

19:06

on one program at the beginning of the pandemic

19:09

. We're with a partner

19:11

, a company in Switzerland , nurengun

19:13

, which are incredibly

19:17

knowledgeable in identifying

19:20

human antibodies . We

19:24

teamed up , basically , and then

19:26

, from the first patients that were accessible

19:29

to them , they isolated and

19:31

identified a really super potent

19:34

human , a patient-derived

19:36

antibodies . The idea

19:38

was that we translate

19:41

the protein code into an mRNA

19:44

code which can then

19:46

be inhaled and you produce

19:48

the antibody in the lung directly

19:50

at the site of virus infections

19:53

, so that you have , to speak , a protective

19:55

level , a level

19:57

of layer of antibodies covering

19:59

the lung tissue . We

20:03

could nicely in

20:05

those days hamsters were used

20:08

as a model and we could nicely

20:10

reduce and avoid this

20:13

weight loss that

20:15

you experience after infection with SARS-CoV-2

20:17

. We moved this

20:20

pretty far . But then what happened

20:22

? All in the sun , and that's what we also know

20:24

, this virus changes

20:26

its face quite rapidly

20:28

and then

20:30

, when we reached the

20:33

Omicron level

20:35

, basically , of mutation , we then unfortunately

20:38

saw that the antibody

20:40

was not neutralizing

20:43

the virus anymore . So

20:45

we unfortunately had to terminate

20:48

that program . So

20:50

that's for one , and the second one

20:52

is basically , you know , the program

20:54

that is now moving in

20:56

. The ball is in the clinics now . We just

20:59

started the first phase one trial

21:01

just before Christmas . This

21:03

also a little bit goes back basically to

21:05

the pandemic time because you know that's

21:07

the concept , eth 47

21:09

, where we have

21:12

a method , rna that encodes

21:14

for a type III interferometer , and

21:17

these type III interferons in the

21:19

lung are called , yeah , it is capable

21:21

of programming the epithelium

21:23

in a very broad antiviral state

21:26

which interferes with

21:28

replication basically of any respiratory

21:31

virus . That's a very broad activity

21:34

, this molecule . And this basically

21:36

goes also back to the pandemic

21:38

time because we've got a

21:40

big

21:43

funding for this program from the Bavarian

21:45

state , yeah , so who were looking

21:47

in those days not only to put

21:50

the bet on only vaccines

21:52

, because no one knew in those days will

21:54

the vaccine make it , but we also

21:56

need therapeutics basically . So

21:59

in the end the pandemic

22:02

had an influence on what we did .

22:04

Yeah , it sounds like a win and a loss . Maybe

22:06

A

22:08

bigger win than loss . Yeah , absolutely

22:10

, biger win , definitely . What was

22:12

your pipeline structure before

22:15

those two opportunities presented ?

22:18

Yeah , just mostly centered only on

22:20

rare pulmonary diseases

22:22

. So that's still a focus

22:24

of our company , because for

22:26

a program here , what I just mentioned , eth

22:28

47 , this is

22:30

a program where you need really

22:33

a partner with the Global Footprint

22:35

app to develop this drug basically

22:38

, but it's a small biotech company

22:40

. We really want to become

22:43

a fully integrated biotech company

22:45

that develops manufacturers

22:48

and sells its own drugs , and

22:50

that , of course , is something you

22:52

can achieve in a space where

22:57

you have a very decidable

22:59

patient population that

23:01

you can serve by yourself , the

23:03

biotech company , and that's , of course , the

23:06

rare disease space . So this

23:08

was always our goal and that's why we've

23:11

looked at our major focus

23:13

is on rare

23:15

genetic pulmonary diseases .

23:21

So it's interesting . It seems the exception to the rule

23:23

of late . Like most of

23:25

the companies that I've talked , most of the CEOs that

23:27

I've talked to are very clear about their intentions

23:30

, and it's decidedly not to build their own

23:32

manufacturing facilities and become an integrated

23:34

biopharmaceutical company .

23:36

Okay , if you , in the end , fully manufacture

23:38

everything in-house , that's a different question

23:40

. You can use , of course , wonderful CDMLs

23:43

that's what we do right now but we

23:45

established all the manufacturing processes

23:48

in-house . And that's also important

23:50

, you know , because we have

23:52

for pulmonary drugs you have rather

23:55

higher doses than for vaccines

23:57

. So we always looked very

23:59

early on in creating

24:02

and developing our easily scalable

24:05

manufacturing process . So

24:07

we have our narrow process developed

24:09

that is not

24:12

based on HPLC , so

24:14

it's just built on tangential flow filtration

24:17

, which is easily scalable Now , and

24:19

also the advantage that it's purely

24:21

APS , no solvents involved

24:23

, and

24:26

this is what we transferred to CDML . We worked

24:28

with a fantastic CDML there together manufacturers

24:31

, our drug product

24:33

.

24:33

Yeah , I'm interested

24:36

in your perspective on the

24:38

explosion of the RNA

24:40

, anything RNA space . We'll stick with

24:42

mRNA , but I mean you can put any letter you want

24:44

in front of it , right ? Yeah , yeah

24:46

, in a COVID and post-COVID

24:49

world , and I

24:52

mean , I think about it like you've been working

24:54

on this . You know science and this technology

24:57

since you know the mid-2000s

25:00

and before , and

25:02

for years and years . I'm

25:05

not going to say it's going unnoticed , but it's not

25:07

necessarily , you

25:09

know , in the media cycle

25:11

, and then all of a sudden this pandemic happens

25:14

. And then , on the heels of the pandemic , it's

25:16

like being the only person in the world who thinks

25:19

the color blue is cool , or even knows

25:21

about the color blue , and then , all of a sudden

25:23

, overnight , like everyone's- like blue is awesome . We

25:25

love blue , right I

25:27

? mean there had to be . That had to be kind of a

25:29

you had to be aware of

25:31

it , right , and like the explosion

25:34

of interest and explosion

25:36

of companies . I mean , there's so many companies in

25:38

this space right now , so

25:40

what's your take on that Like .

25:43

It's great for the field . I

25:46

mean , it's fantastic . So the

25:48

more approaches

25:51

we see , the better we get

25:53

here . And there are so many diseases

25:55

that can be potentially

25:58

treated with messenger in A . This

26:00

can't be done by only one party

26:03

basically . So that's great

26:05

that we have so many different approaches here

26:07

, not only for the vaccines . So

26:10

I think we will see

26:12

some new

26:14

potential approvals rather shortly

26:16

, also for additional vaccines or in

26:18

infectious disease space . But

26:21

also look into the cancer

26:23

space here . I think we see really very

26:25

nice good signals in this space where

26:27

people use messenger RNA as

26:30

a drug fidelity . But

26:32

then look into genome

26:35

editing . There is a great potential

26:37

for messenger RNA to

26:39

be used here . And I mean we have

26:42

the first success now for CRISPR

26:45

being approved , crispr

26:47

technology . So that's a huge

26:49

step basically and I think

26:52

a huge stimulus

26:56

also for moving this

26:58

forward with using messenger RNA

27:00

. Basically . And then , of course , there is

27:02

this field of rare genetic diseases

27:05

where we see , I

27:07

think , pretty good programs

27:10

also for liver genetic

27:13

diseases and of course we want

27:15

to achieve the thing with our pulmonary programs

27:17

there and to really offer

27:20

hopefully as soon as possible

27:22

some help for

27:25

the patients suffering from these diseases

27:27

. So I think there's not

27:29

really moving forward right

27:31

now .

27:31

Yeah , in

27:34

the context of your pulmonary

27:36

focus , what do you

27:38

see as the

27:41

biggest potential risk to progress

27:44

? But whether

27:46

it's a technical risk or a market risk

27:48

or a scientific risk , what's

27:50

the rate limiting factor or big risk that

27:53

could stand in the way of progress ?

27:55

I mean , in the end it's

27:58

always the basic concept

28:01

of developing a drug and

28:04

this all centers around right

28:07

biology you have to really

28:09

understand your biology then

28:12

right exposure

28:14

, so that you have the right

28:17

PK , you know that your drug gets

28:19

there where it needs to be for

28:22

the right time basically . Then

28:25

, of course , pretty good , the right safety . That's

28:28

all the basis of a

28:30

successful drug in the end . And

28:32

then and that's what you mentioned is

28:34

there the right patient ? You have to

28:36

understand your patient , basically and

28:39

then the right commercial . So I think it's

28:41

a kind of umbrella , basically

28:43

, that you have to look at and

28:46

go into very detail for each of

28:48

these elements basically and

28:51

we did this for PCD , for

28:53

primary sedatives , canisia we

28:56

believe that we

28:58

hit those boxes

29:01

basically . So we tackle

29:03

very interesting biology because

29:06

in those patients who suffer from the

29:08

disease , they

29:11

are miscellaneous proteins

29:13

that drive the motility of the tiny

29:15

airs and our lungs and our airways

29:18

that are responsible for moving

29:20

the mucus out of the lung

29:22

. Yes , that's called mucosilir

29:24

escalator and it's made

29:26

to remove any dust that we inhaled

29:29

from the lungs so that we have a fully

29:31

particle-free surface of the lung

29:33

. But what is important to understand

29:36

here is that these CDR

29:38

, they are just made by

29:41

the airway cells , once

29:43

in the cell life and

29:45

in the half-life of the airway

29:48

. You said something

29:50

like four to eight weeks and that

29:52

means you can imagine if

29:54

you're delivering a messenger RNA

29:57

during the cediation process

29:59

, quite persistent effect

30:01

. So if you translate

30:03

a quite short lift

30:06

drug itself the mRNA is gone

30:09

in the lung after two and a half

30:11

days , something like this , into a quite

30:13

persistent effect . And

30:16

so we believe that's a

30:18

good starting point from the biology

30:21

and that's why we

30:23

, for instance , picked the PCD

30:25

the patients . It's a very

30:27

clearly defined because it's a genetic

30:30

mutation . So you take this

30:32

risk out of it , basically of the explosion

30:34

. We know that we

30:36

need to potentially

30:39

restore the CDR

30:41

function in about 20%

30:43

of the CD8 cells . We believe that's

30:45

doable on what we see in our

30:47

preclinical studies and

30:51

there is a commercial potential because

30:54

it's a life-limiting

30:56

disease basically , and it's

30:59

a high unmet need because there are no drugs on

31:01

the market . So that's why

31:03

we believe that's

31:06

a good spot for messenger

31:08

RNA computing .

31:11

I mean I could go down a rabbit hole here . Am

31:14

I hearing you correctly ? You talk about this cellular

31:17

regeneration like

31:20

a cycle and you're saying

31:23

that there's

31:25

an efficacy opportunity

31:28

at a specific point in that cycle

31:30

.

31:31

No , you just have to restore the function

31:33

. Basically once the CDR

31:36

are restored , you have a quite persistent

31:38

effect Because the cell

31:40

does not change the CDIA , so it's once

31:42

built and then they

31:44

work until the cell

31:47

is removed

31:49

from the body and replaced by a new cell . For

31:53

the lifetime of the cell Gotcha

31:55

.

31:55

OK , see , these are the questions I should have been asking

31:58

in AP biology before I

32:00

dropped the class .

32:01

My senior year .

32:03

Not long ago I had

32:05

your senior director of formulation and

32:07

aerosol research done on

32:09

a live event that I hosted

32:11

for Bio Process Online , christian Domen

32:13

.

32:16

Great guy , by the way . He is fantastic

32:18

.

32:18

I tell you , yeah , absolutely yeah

32:21

he was super valuable , just

32:24

brought a whole lot of wealth and wealth

32:27

of information to that conversation . But one of

32:29

the things that we talked about quite a bit on

32:31

that during that conversation was LNP

32:34

stability and some of the challenges around there . So

32:37

is that , do you see that as potentially

32:39

a rate limiting factor ?

32:41

I see that this limitation

32:43

in the field , because I mean just

32:46

look into the commercialized

32:49

vaccines , mrna

32:51

vaccines there . So first

32:54

we see that they need very

32:56

cold temperatures to be stored

32:58

and that was OK

33:00

during the pandemic basically . But

33:02

now we move into the post-pandemic

33:04

phase and we really want to have

33:07

temperatures that

33:09

the pharmaceutical

33:11

industry is used to use and also for

33:13

the patient . So that means

33:16

this overall cold chain needs

33:19

to be improved and need to be overcome

33:21

, and I think we have done a lot here with our

33:23

technology where we can contribute

33:26

to this and achieve this . But

33:28

with our technology we have , for instance

33:30

, the livalization process in place

33:32

where we see that we can store

33:35

the drug product already for

33:38

a year in the fridge , two to eight

33:40

degrees , and the studies are ongoing

33:43

, but we don't see any change here . And

33:45

even at room temperature you

33:47

see this already for more

33:50

than half a year . So that's , I

33:52

think , a huge step forward . And

33:56

then also you have to see and look into

33:59

the stability of the

34:02

drug product itself . Look

34:05

, the commercialized vaccines are

34:08

ready to use drug product so

34:10

it can be used for chemo-laws

34:12

basically . And we have with

34:14

our SNAPs what we call

34:16

our LNP . So we have already

34:19

achieved six days at

34:21

room temperature . That means doctors

34:23

can now really plan for

34:26

using the drug for an entire week . So

34:29

I think that's a large improvement . And

34:31

on top we have developed

34:34

our recall and stabilizing XEPian

34:36

technology here . Those

34:39

are a certain class of excipients from

34:41

the inactive ingredient list

34:44

from the FDA . That's what you can add

34:46

to the LMPs

34:49

and you make them mechanical

34:51

stable . And why do I

34:53

mention it ? Because on the label of

34:55

the commercial vaccines that's mentioned

34:57

don't check them because they are very

34:59

prone to aggregation , basically . So

35:02

and we can really now stabilize these

35:05

nanoparticles so that you

35:07

can even vortex them and shape

35:09

them at full speed and

35:11

we don't see any changes in

35:13

the particles . And of course this

35:15

gives you confidence to also

35:18

the doctors and pharmacists

35:20

to use the drug , because they don't

35:22

have any concerns anymore

35:24

that might potentially aggregate , and

35:27

also for manufacturing . You know

35:29

, when , in particular , the philipinibus

35:32

step , these nanoparticles

35:35

flow through little tubings

35:37

to basically go into

35:39

the bile , then

35:42

of course you have to avoid aggregation

35:44

, to avoid any batch

35:46

failures . So I think that's

35:49

really important , what we have achieved

35:51

here . And

35:54

then I think another I mean , when

35:56

we speak about limitations , I

35:59

think another limitation maybe and I think

36:01

that's a little bit unspoken also in the field is

36:03

that there are . You

36:06

know the pronunciations from the commercialized

36:08

vaccines they biotip

36:10

, distribute to the entire body . Yeah

36:14

, from the documents , from

36:16

their authorization

36:18

, you see that 30% goes

36:20

into the liver , it goes into

36:22

the heart even if you just locally

36:24

inject the muscle . Yeah , and

36:26

even 2 to 4% of

36:28

what's found in the plazas found

36:30

in the brain , basically , and

36:33

we think if you don't need it in the brain , one

36:36

should not deliver it to the brain , basically , and

36:38

you know our SNAPs that are Seems

36:40

logical . Yeah , okay , and

36:44

you know our SNAPs that

36:46

are made from catechonic

36:48

lipidoids , not

36:50

catechonic lipids . They are a little

36:53

bit more positively charged so

36:55

they can package the messenger RNA

36:57

more tightly , so you need just

37:00

half of the formulation but they

37:03

lead to a full retention

37:05

of the mRNA

37:08

drug at the site where you

37:10

deliver it . And that's of course so

37:12

you can avoid this biotip distribution . And

37:14

this is you know for us for our pulmonary

37:16

drugs that we develop . We want to

37:18

have the full activity of

37:21

the messenger RNA , of course at

37:24

the site where it's needed , that means in the

37:26

lung , and avoid any biotip

37:28

distribution . It's also safety aspect of

37:30

that . So that's why we

37:33

think really we have achieved pretty

37:36

much here to

37:38

move the field

37:40

in our products also really

37:42

forward with

37:45

the advantages and

37:48

to try to overcome those limitations

37:50

that we just spoke about before .

37:52

Yeah , what's been

37:54

your experience with the ? I

37:57

guess on the regulatory scene . So

37:59

I have conversations with my colleague

38:01

, anna Rose Welch , who's leading a

38:03

new project that's going

38:05

to be called Advancing RNA right now and

38:07

we often talk about , you know , some of the uncharted

38:10

regulatory waters in

38:12

this space , so what's been your perception

38:14

there ? I ?

38:14

mean , I think overall all the regulators

38:17

are really very

38:19

, very educated now from

38:21

the commercialized vaccines . I think that's

38:24

a great step , but you

38:26

know what I think for the field

38:28

it would

38:30

be very helpful , but that's ongoing

38:33

already . The other is to

38:35

consider a kind of platform approach

38:37

, so where you can more

38:39

easily , if you leave everything

38:41

the same and your drug formulation , the mRNA

38:44

formulation , but just then replace

38:46

this with another messenger RNA

38:49

, so that you get there certain

38:56

efficiencies . Exactly , exactly , so

38:59

this is for us also very important this approach

39:01

, because these primary

39:04

cellar dyskinesias they are

39:06

not caused by a lack

39:08

of acidic fibrosis , by only a

39:10

mutation in one gene , but

39:13

there are more than 50 genes

39:15

known , when mutated , that

39:18

can lead to primary cellar dyskinesia

39:20

, because you can imagine this is a very complex

39:22

proteinaceous apparatus

39:25

. To get this beating

39:27

ongoing , and

39:33

for us it would be very helpful

39:35

to swap out one

39:37

messenger RNA for another messenger

39:39

RNA to then serve

39:42

another genetic mutation that

39:44

causes the disease . So

39:46

those things that would be really

39:49

helpful , but I think that's a lot

39:51

of thinking into this direction already

39:53

.

39:53

Yeah , yeah

39:56

. When I look at the ATMP , space

39:58

, cell and gene therapies

40:02

, there's a lot of bad news

40:05

. I mean there's a

40:07

lot of consternation about

40:10

accessibility and cost

40:12

and

40:14

even commercialization . I mean there have been some not

40:17

so smooth approvals

40:19

in terms of commercial access how

40:22

does the mRNA field collectively

40:24

ensure ? I mean , obviously with the

40:27

COVID vaccine it was sort of a non-issue , right , we all

40:29

got our shots . But

40:32

how does the mRNA world , as it

40:34

addresses more complex

40:36

indications and perhaps charts

40:39

some complex manufacturing waters , how

40:41

do you make sure that you're not making some

40:44

of those same ? I don't even want to say

40:46

mistakes , but trying to maintain an

40:48

efficiency that

40:51

results in patient accessibility

40:53

at the end of the day , I

40:56

think you're referring to those gene therapies

40:58

that were upfront .

41:00

You paid a few millions basically

41:02

for a treatment . And

41:05

I mean the situation , I think

41:07

, with messenger RNA drugs is a little bit different

41:09

because it's more like a standard

41:11

drug that you repeatedly

41:14

administer to

41:16

the patient . So I mean

41:18

for our PCD programs

41:21

. Basically , we believe that's a little bit

41:23

into the direction that you

41:25

see for cystic fibrosis

41:27

, where you have already the

41:29

pricing then here on the market .

41:31

So you don't anticipate any indications

41:33

where that cost

41:36

control might become

41:38

a factor .

41:39

If you use messenger RNA or possible

41:41

genome editing , that means a one-time

41:43

treatment and then you are done . Basically

41:46

you face the same problems . But I think also

41:48

the people start to think creatively

41:50

how to solve this . What you mentioned , yeah

41:53

, how can we find financial models

41:55

basically that we

41:57

can also serve those

42:00

patients suffering from these

42:03

rare diseases ? Because

42:06

in the end , for one rare disease just

42:09

a few patients , but if we sum up

42:11

, all patients that

42:14

suffer from all the genetic , no

42:16

rare diseases that we have , those are

42:18

many patients basically , and

42:21

I agree with you . But I

42:23

believe that the field is looking into

42:25

creative reimbursement models

42:27

because we have

42:29

to find a solution there , because we want to provide

42:33

medicine to these patients as well

42:35

. But for the mRNA I

42:37

think it's a little bit different because it's not a one-time

42:41

treatment , or at least what we do , and

42:44

then you fix it , but it's those like

42:46

standard drug basically .

42:48

Yeah , very good . Just a couple

42:50

more questions that I will need to wrap up , but

42:52

I'd start with what

42:55

? Give us a clinical update , what the next big steps

42:57

for atherists are . What's

43:00

on your immediate ?

43:01

horizon ? Yeah , exactly so . For us important

43:03

and I mentioned this already for the first program

43:05

we just started our

43:07

first phase one

43:09

study just before Christmas

43:11

. So this

43:14

is our study

43:16

that has three arms basically

43:18

One arm is where we deliver it into the

43:20

nose and one arm is

43:22

where we deliver it into the lung and

43:25

then we will have , after defining

43:27

their highest dose , basically

43:30

a combined arm . And

43:32

we do , of course , safety . That's

43:34

their major clinical

43:37

endpoint . But we look into

43:40

also targeted engagement , which is very important

43:42

so that we can measure in nasal

43:44

swabs and in sputum from

43:47

these treated healthy volunteers , the

43:51

production of their recombinant

43:53

protein produced by the messenger RNA

43:55

. And , more important , you know that we

43:57

also see the downstream activation

43:59

of the genes that are induced by

44:02

the interferonamda , which gives us

44:04

the indication it works , it

44:06

is really doing what it is expected

44:09

to do . And this is the

44:11

study we will have completed

44:14

towards springtime

44:16

. So we think we will

44:18

plan an interim readout towards

44:22

early spring basically . So that's the

44:24

first very important news that

44:27

we expect then in

44:29

2024

44:32

. And then afterwards we

44:34

are also in the midst of putting

44:37

the preclinical package together

44:39

for finding

44:41

a CTA towards autumn

44:44

this year for

44:47

our first PCD candidate , and

44:50

so it's exciting time For sure , yeah

44:54

, very good .

44:55

And then , just to wrap things up , when you come

44:57

to an environment like a giant

45:00

investor conference at this stage

45:02

and atheroses sort of continual

45:05

non-clinical bath

45:07

, what's your MO here ? What's your

45:09

goal ? What are you looking to make happen ? Raise money

45:11

?

45:13

No , seriously . I mean now we enter into

45:15

the sort of clinical scene , basically , and

45:17

then we need many

45:20

more financial resources than before and

45:22

that's our goal here at JPMorgan and

45:24

also to fund partners , because we have

45:26

a really powerful we

45:29

believe best industry platform

45:31

technology and of course this

45:33

can also enable and help

45:36

other potential partners to develop

45:38

new drugs with our technology

45:40

. So it would be great to leverage on

45:43

top our technology also outside

45:45

our focus area .

45:47

Basically , yeah , excellent

45:49

. Well , I wish you well the rest of your

45:51

time here and I appreciate you coming all

45:54

the way from Munich solely

45:56

for this opportunity to be on the business , of course

45:58

, math yeah . Absolutely

46:01

my pleasure . I enjoyed conversing

46:03

with you and learning more about what Ethris is doing . Same

46:06

here , Matt .

46:07

Many thanks for having me . Thank you All , right

46:09

Very good .

46:11

I'm Matt Pillar and you just listened to the business

46:13

of biotech , the weekly podcast

46:15

dedicated to the builders of biotech

46:17

. We drop a new episode with a

46:19

new exec every Monday morning and

46:21

I'd like you to join our community of subscribers

46:24

at bioprocessonlinecom

46:26

Apple Podcasts , spotify , google

46:28

Player anywhere you get your podcasts . You

46:31

can also subscribe to our Never

46:33

Spammy , always insightful monthly

46:35

newsletter at bioprocessonlinecom

46:38

. Backslash B-O-B . If

46:40

you have feedback or topic and guest suggestions

46:43

, hit me up on LinkedIn and let's chat and

46:45

, as always , thanks for listening .

Unlock more with Podchaser Pro

  • Audience Insights
  • Contact Information
  • Demographics
  • Charts
  • Sponsor History
  • and More!
Pro Features