Podchaser Logo
Home
Drag & Drop Genome Editing

Drag & Drop Genome Editing

Released Tuesday, 16th January 2024
Good episode? Give it some love!
Drag & Drop Genome Editing

Drag & Drop Genome Editing

Drag & Drop Genome Editing

Drag & Drop Genome Editing

Tuesday, 16th January 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

0:02

Hello and welcome to Raising Health, where

0:04

we explore the real challenges and enormous

0:06

opportunities facing entrepreneurs who are building the

0:08

future of health. I'm

0:15

Olivia. And I'm Chris. Today's

0:18

episode is with the co-founders of Tome

0:20

Biosciences, Rahul Kakkar, who serves as the

0:22

CEO of Tome, and John Fidd, the

0:24

CSO. They are joined

0:26

by Jorge Conde, General Partner of A16Z

0:28

Bio and Health. Rahul, John

0:31

and Jorge talk about the technology behind

0:33

Tome, known as PAST, a genome editing

0:35

technique. It's a combination of two

0:38

older technologies. What they

0:40

did is they used

0:42

a Cas9 nickase

0:44

with an RT enzyme that can

0:47

write small pieces. They

0:50

used that actually to programmably

0:53

write a landing site,

0:55

a beacon for

0:57

a highly specific

0:59

integrase protein. And

1:01

then they came in with the integrase protein

1:03

with a template DNA and then the integrase

1:05

does exactly what nature has evolved it to

1:07

do, which is to put that code

1:10

at that site. They also dive

1:12

deep into how this technology could be applied

1:14

to help patients, making cell therapy more accessible

1:16

for more people. Because

1:19

of the technical capabilities that John

1:21

just articulated, the ability to put

1:23

multiple pieces of code exactly where

1:25

you want them to go simultaneously,

1:27

we can bring design

1:30

make test cycle times that are

1:33

traditionally only thought of in the

1:36

biologic world to cell therapy. We've

1:38

never thought about being able to iterate cell

1:41

therapy design at speed and at scale, and

1:43

we can do that now. And so that

1:45

makes cell therapy look much less like a

1:47

niche product and much

1:49

more like a bonafide discovery platform. You're

1:52

listening to Raising Health from A16Z Bio and

1:55

Health. So

2:00

today I'm thrilled to be joined on

2:02

the A16Z Raising Health podcast by

2:04

Rahul Kakkar, who is the CEO

2:07

of Tone Biosciences and

2:09

John Finn, who is the Chief

2:11

Scientific Officer of Tone Biosciences. Tone

2:14

Biosciences is a portfolio company in the

2:16

A16Z Bio and Health portfolio. We're thrilled

2:18

to be a small part of the

2:21

tone story. I thought one place

2:23

where we could start is get

2:27

your views, your comments on where we

2:29

are on this journey of genomic

2:31

medicine. So where we stand today in

2:33

early 2024, we've just

2:35

come across a couple key milestones

2:37

in sort of the landscape of

2:39

genomic medicine. Maybe John, I'd

2:41

ask you to start first given that you've spent a lot

2:43

of time in this space. Just would

2:46

love to get your sort of take

2:48

on the state of the state. Yeah,

2:50

absolutely. So one of the

2:53

things just to actually let all

2:55

the listeners know upfront, one of

2:58

the things

3:01

about me that you'll be

3:03

able to tell is that I do have a

3:05

stutter. And I just want to let

3:07

everyone know what that means is that this

3:09

can take me a bit longer to say what I want

3:11

to say at times, but everything is fine. This is just

3:13

the way that I talk. Maybe one day

3:16

we'll be using CRISPR in the

3:18

brain for that. Not the first

3:20

indication, but anyway, everything is

3:22

fine. So when

3:24

I think about the state of the field

3:26

right now, it has come a

3:29

long way in a very short time. So

3:31

it was only about a decade ago that

3:33

the first publications came

3:36

out really on CRISPR

3:39

that really showed the ability

3:42

to site specifically target a

3:44

gene or a sequence

3:47

of code. That really

3:49

opened up the floodgates for everything

3:51

that we see now. And

3:54

I see that the CRISPR field has really

3:56

gone through a couple of different revolutions. The

4:00

first one was the ability to cut a

4:02

piece of DNA wherever you wanted.

4:04

This had a huge impact

4:06

both on life sciences, research,

4:08

understanding how life works, as

4:11

well as the first wave

4:13

of medicines. These

4:15

advances were all based on

4:18

the ability to site specifically break

4:20

DNA. Then came the

4:23

next wave of tools. You've got the

4:27

base editors, the prime editors. These

4:30

are fantastic tools that no longer break

4:32

DNA. If most of

4:34

the patients have the same mutation, this is

4:36

the right tool because if you make that

4:39

small change, if you fix that single mutation,

4:42

that will have a huge impact on that

4:44

entire population. What

4:47

we're doing here at home, I

4:50

see it as the last tool we've needed

4:52

in the editing toolbox. Now we can put

4:54

any sequence of code of any size in

4:56

any location. Yeah, and I

4:59

think when we take a look back, it

5:01

is remarkable that when we think about the

5:03

fact that this is a milestone, barely

5:06

a decade in the making given that the first CRISPR

5:08

papers came out just over a decade ago. Rahul,

5:11

first of all, you're a practicing physician. Second

5:14

of all, you're a repeat biotech

5:16

entrepreneur and executive. Of all

5:18

the things you could pick to work on, why

5:21

pick genomic medicine? Yeah, it's

5:23

a great question, Jorge. I did not

5:26

leave my prior company with the

5:28

intent to move into cell and

5:31

gene therapy or genomic medicines. My

5:34

clinical practice, as you mentioned, is

5:36

cardiology. My first company was a

5:38

cardiovascular-focused company. Second

5:40

company, Pandion, was autoimmune disease. I've

5:42

always focused on diseases

5:45

that have broad

5:47

societal burden and therefore where drug

5:50

development can have broad societal impact. And

5:53

to John's point, drug

5:56

development using either the first

5:58

generation or the first generation. of

6:00

CRISPR-Cas9 enzymes where we're breaking genes,

6:02

or the next generation, which is

6:04

really making small repairs, generally

6:07

lends itself to rare disease. And

6:10

it wasn't really a place, as much

6:12

as I don't want to take away from the gravity

6:14

of those diseases and the impact, particularly on children and

6:17

their families, just not where I've had my

6:19

focus. But becoming

6:22

familiar with the paste technology

6:24

as invented by our founders

6:27

struck me as something wholly different.

6:30

The augmentation of the CRISPR-Cas9 enzymes

6:32

with other enzymes so that it

6:34

can now not break genes or

6:37

make small repairs, but

6:40

wholesale reprogram DNA, allowing

6:42

us to really regard DNA

6:45

for what it is, which is software, and giving

6:47

us the tool to reprogram

6:50

a cell, which either

6:52

has a bug that nature unfortunately granted

6:54

it, or imparting new

6:56

therapeutic capabilities to a cell in

6:58

a dish struck me as

7:00

not just a step

7:02

forward, but

7:05

a quantum leap forward in creating a

7:07

whole new set of mature asset classes

7:09

of drug development. And

7:11

that potential societal

7:14

impact, which is broad by

7:16

definition, is really what attracted me to this technology

7:18

and to this company. So

7:20

I remember reading the

7:22

paste publication, and even as

7:24

a layperson thinking that

7:26

this seemed like a pretty

7:28

big step forward, like a pretty unique

7:31

innovation in and of itself. So

7:34

I guess the first thing that would be helpful is

7:36

if you could walk through what is

7:38

paste, what

7:40

was described in that publication. Now,

7:43

one of the remarkable

7:45

things about my story with Tome is that

7:48

I was not looking for a job. I

7:50

was very happy where I was. I was

7:52

working on next generation

7:54

delivery platforms. And so I took

7:56

the call with the founders, Omar

7:58

and Jonathan, as a favor. And

8:00

I thought they were doing something else

8:02

altogether. And when they said, no, actually,

8:05

Tom is doing programmable

8:07

genomic integration, I said, well, that's

8:10

been a holy grail of the

8:12

field since I started, and let

8:15

me see more. And so I read the

8:17

manuscript probably about two and a half years

8:19

ago now, honestly. And

8:21

as I read the manuscript, I had three

8:23

different emotions going on. So the

8:25

first one was I was super excited because

8:28

these guys solved the problem. And

8:30

the specific problem they solved was how

8:32

do you put a large piece of

8:35

DNA in a very specific location? My

8:37

second emotion was I was

8:39

actually jealous because it's such a good idea. And

8:41

next I'll actually talk about exactly what the

8:43

idea was. And then the third one,

8:46

I was getting sadder and sadder

8:48

because I love my old company, I love my

8:51

old team, but I knew if I

8:53

had the opportunity to be

8:56

a part of something like this, I

9:00

could not say no because this has been what I've

9:02

been trying to do

9:04

my whole career.

9:06

So what is it? Essentially,

9:09

it's a combination of two

9:11

older technologies. What they did

9:14

is they used

9:16

a Cas9 nickase with

9:18

an RT enzyme that can write small

9:21

pieces. They

9:24

used that actually to programmably

9:26

write a landing

9:29

site, a beacon for

9:32

a highly specific integrase

9:34

protein. And

9:37

then they came in with the integrase protein

9:39

with a template DNA and then the integrase

9:41

does exactly what nature has evolved it to

9:43

do, which is to put that code

9:45

at that site. And

9:47

so now for the first time,

9:49

the field could efficiently put large

9:51

pieces of code in very specific

9:54

locations without depending

9:56

on breaking DNA,

9:59

without depending on cells cycle. This works

10:01

in cycling cells, also works in non-dividing

10:03

cells, which is very important because most

10:05

of the cells in the body are

10:07

non-dividing cells. And so, when

10:09

I read this paper, it blew me

10:11

away. And when I had

10:13

the opportunity, I could not say no.

10:15

So Rahul, I want to hear your

10:17

sort of your story here because presumably

10:19

you came across the technology or the

10:21

founders maybe in that order and the

10:23

reverse order. And you know,

10:25

you had just come off of selling

10:27

your previous company. And so, there was

10:29

an alternative here which is to sit on the beach for a

10:32

little while and you didn't choose

10:34

that. So I just love to hear sort of

10:36

what got you interested and

10:38

engaged on this technology and

10:40

on these founders. Reading

10:43

the manuscript, I think I had

10:45

a somewhat

10:47

parallel epiphany, although for very different reasons

10:50

as John, which is here's

10:53

the chance to build a company

10:56

that can define an era in

10:58

medicine. And that kind of opportunity rarely

11:00

comes along. It comes along once

11:03

every few decades, if not once in a generation. And what

11:05

I mean by that is we can

11:07

think about the history of

11:09

our industry of

11:11

biopharma in certain eras.

11:13

The first era was really our ability

11:16

to create synthetic compounds. We

11:18

don't have to hunt and forage for

11:20

medicines anymore. And fast forward to the

11:23

rise of Genentech and

11:25

the cloning of insulin and

11:28

into the first fully human

11:30

antibody therapy which was Humira, which even

11:32

today is one of the largest commercial

11:35

successes in our industry. And

11:37

then there were three which is really

11:39

the rise of nucleotide therapeutics with companies

11:41

like Moderna and Alnylam. But

11:43

we have not seen the company

11:46

that will define genomic medicines

11:48

as of yet. For all

11:50

of the incredible scientific breakthroughs

11:52

that Sheppard-Pallier and Doudna, Fang

11:55

Zhang and David Lu had brought to the table, companies

11:58

including John alma mater, which

12:01

are developing very important drugs, the

12:03

ability to broadly mature an

12:06

entire technology

12:08

such that it impacts therapies

12:10

for a myriad of therapeutic areas. I

12:12

don't think we've seen the technology within

12:14

the genomic space that can do that.

12:18

And reading the paper and

12:20

then interacting with Jonathan and Omar,

12:23

I realized that what we might be looking at is

12:25

the technology in the company that actually does that. And

12:28

I think the founders have been key to this, Jonathan

12:30

and Omar, not just

12:32

being brilliant, of course, but

12:34

they really are invested in the

12:36

company with their time, with their

12:38

energy. They're

12:41

really seeing this as their way of leaving a mark on

12:43

the world. I just want to

12:45

dig in on a point that both

12:47

of you made, which is this ability

12:49

to do programmable genomic integration is

12:52

a massive step forward, a quantum leap, I think your whole

12:54

is referred to it as. Why

12:57

is that such an important enablement? What

12:59

does that allow us to do that

13:01

other approaches to genomic medicine just haven't

13:03

been able to achieve? So

13:07

to me, when you

13:09

can move from small edits

13:11

to wholesale inserting DNA

13:13

without any size restriction and

13:15

certainly without creating double

13:18

strand breaks, in layman's

13:20

term, making mincemeat of the DNA. So

13:23

to insert very large pieces

13:25

of genomic code in a

13:27

relatively safe manner, it really

13:29

allows you to utilize all

13:31

the various elements that nature

13:33

has encoded into our DNA,

13:36

coding regions, regulatory regions. We

13:38

can fully choose what

13:41

gene goes where, which

13:43

therefore dictates how much of the

13:45

genetic product is made, under

13:48

what regulation, in what cell, at

13:50

what time. It gives

13:52

us complete flexibility. For

13:55

lack of a better word, it allows us to hack the genome.

13:57

Yeah, if I could just add, I mean, one of

14:00

One of the applications

14:02

that I'm most

14:05

excited about is if a patient

14:08

has a broken gene, now we can put

14:11

a functional copy of the gene

14:13

in the right location. What's

14:15

funny is that when I told my wife that she's like,

14:17

well, what have you been doing for the past 20 years?

14:23

That's what everyone thought gene therapy was.

14:25

But we had to use Epizomes. We

14:27

had to use random integration, etc. But

14:29

now with this ability, not only can

14:31

we put the gene in the right

14:33

location so that it stays under its own

14:36

endogenous expression. Not too much, not too

14:39

little, not the wrong cell type,

14:41

etc. But we can

14:43

now have one product

14:45

for all the patients because we don't

14:47

have to go in and fix each

14:49

individual mutation. And for

14:52

most of the applications

14:54

that we're going after, there are

14:56

tens, hundreds, if not thousands of

14:58

separate mutations. And so with this

15:00

approach, we can cover most,

15:02

if not all, of the patients with a single

15:05

drug. That to me is

15:08

a total game changer. I wanted to highlight

15:10

something John just said, which is because of

15:12

the technical capabilities that John just articulated, the

15:15

ability to put multiple pieces of code

15:17

exactly where you want them to go

15:20

simultaneously, we can bring

15:23

design, make, test cycle times that

15:25

are traditionally only thought of in

15:27

the biologic world to cell

15:30

therapy. We've never thought about being

15:32

able to iterate cell therapy

15:34

design at speed and at scale. And we

15:36

can do that now. And so that makes

15:38

cell therapy look much less like a niche

15:41

product and much more like

15:43

a bona fide discovery platform. And

15:45

so to me, one of the

15:47

things that that tone would leave behind is

15:49

a legacy other than finally bringing gene

15:52

therapy into its mature form

15:55

is bringing cell therapy into its

15:57

to fulfill cell therapies potential as

15:59

a therapeutic. Four minutes? All right? I

16:01

think you just described our in terms

16:03

of what you can do for genetic

16:05

medicine in in the body is extraordinarily

16:07

profound. Because I just heard you say

16:09

there's. There's. Two fundamental things

16:12

that this technology enables that

16:14

become incredibly important: The. First

16:16

one. Is to be able to fix. A

16:19

broken gene regardless of what mutation broke

16:21

that team. So now we traditionally have

16:23

thought of a lot of these genetic

16:26

medicines as you need it, you know

16:28

and added to fix a specific mutation.

16:31

And. Hear what you're saying is you can

16:33

insert the corrected copy so you go

16:35

from one medicine permutation. To. One

16:37

medicine per jean. And I get

16:39

to very, very profound shift. But. The

16:42

second one I just heard you say,

16:44

which I think a lot of people

16:46

probably under appreciate. Is. The fact

16:48

that. Every gene in

16:50

our body is very carefully regulated

16:53

by the cells that they inhabit.

16:56

And. Soaps Different cells turn on

16:58

and off or turn up and

17:00

down different sets of jeans. And.

17:03

So if you can programatic, we insert

17:05

the corrected gene at exactly the right

17:08

spot. That. Means not only have you

17:10

corrected the problem. But. You've

17:12

also ensured that it is used

17:14

correctly within that cell type because

17:16

it's been regulated by that cell

17:18

type as nature intended. We.

17:20

Can harness any promoter in in

17:22

the body and so now I

17:24

shall we concert sort actually to

17:26

to have logic gates and gates

17:28

if this gets express and this

17:30

us to and that you know

17:32

that really might open up a

17:34

whole nother Rains have some suture

17:36

applications and so we're not limited

17:38

to wear discounts. What is kind

17:40

of remarkable was at at the

17:42

at a very fundamental level this

17:44

technology. It's a program and technology

17:46

is a programming language effectively as

17:48

what you are you working. On So.

17:51

In the case of a genetic disease, You.

17:54

Can essentially. Provide

17:56

patch software to replace a broken gene

17:58

with the corrected version. It

18:00

would really neat as you describe and it in the

18:03

case of cell therapy. What? You have

18:05

is the ability to fundamentally reprogram

18:07

cells. And reprogram sell to

18:09

behave in ways that perhaps weren't previously

18:11

possible. And. Can do things

18:13

that it with sophistication that weren't

18:15

previously achievable. In. The in the course

18:17

of cell therapy. And. So as you're describing

18:20

it, we get to a point where we have

18:22

a discovery platform of get to the you know.

18:24

To. The design build test time scale so

18:26

we can in a rapidly advanced technology.

18:29

A. Presumably that brings down cost

18:32

increases Access. That's obvious

18:34

he got to be seen, but that's that's a

18:36

potential promise. So. You can do

18:38

all of this both on. The.

18:40

Genetic Disease Front. In

18:43

terms of replacing broken jeans. And

18:45

on the cell therapy from in terms

18:48

of reprogramming cells to do new things.

18:51

You. Can do anything. But

18:53

you can't do everything. What? His

18:55

tone gonna do. So. When.

18:58

When. John and I first met you

19:00

before we decide to join the company.

19:03

Even. From are very first cup of

19:05

coffee together it was very clear that

19:08

were certain philosophical alignments. That. By

19:10

the end of that conversation my camera how

19:12

long it lasted. Maybe an hour, maybe two

19:14

hours. We both realized we had to work

19:17

together because your point where he this technology.

19:20

Can. Be the basis will be the

19:22

basis of a company that will last

19:24

decades and have a very large pipeline

19:26

in the future. But one of the

19:29

arts of Biotech start up is. Where

19:31

do you start when you're cost of capital

19:34

is very very high. And you

19:36

need to prove your technologies quickly as possible.

19:38

The As: every minute is hyper. And.

19:41

So remote concepts. Together we we actually

19:43

created a set of what what I

19:45

call first principles on how we're going

19:48

to select where we go. First.

19:51

Let's. Not take any more technical risk than

19:53

we have to write the tech. T.

19:56

G I and specifically the

19:58

instance of integrate. mediated PGI

20:00

or IPGI is complicated enough. It's one

20:03

of the most complicated drug products our

20:05

industry has ever envisioned. So let's make

20:07

sure we're not also taking for instance

20:10

delivery risk. Let's make sure

20:12

we're not taking inordinate clinical development risk

20:14

or regulatory risk. The

20:16

other place and this is again from my

20:19

experience at my prior to the basis, let's

20:21

make sure we go into clinical indications to

20:23

the point I just made where

20:25

the clinical risk is minimized. And

20:28

that means that there's some degree of target

20:30

validation. In the gene therapy world,

20:33

that's relatively straightforward. All you're doing is choosing

20:35

for diseases where there's a good genotype phenotype

20:37

correlation. But in the cell therapy world, that's

20:40

much harder. When I spoke with

20:42

the founders, when I you know read

20:44

the manuscript, I was

20:47

already working on new delivery systems. You

20:49

know how to get into new muscle,

20:51

CNS, etc. What I realized

20:54

that we already

20:56

have clinically validated delivery

20:59

systems available today that we can

21:01

use with this technology. And so

21:03

that's where we want to focus.

21:05

So the initial focus is going

21:07

to be on genetic diseases

21:10

of the liver? Yes. And

21:13

on autoimmune diseases? Yeah,

21:15

that's exactly right. So as we

21:18

think about not taking excessive technical

21:20

risk on the integrative gene therapy

21:22

side, it's really talking about focusing

21:24

on areas where delivery

21:26

of our components, which are

21:28

both lipinataparticle and viral vectors,

21:31

are already de-risked, which is the

21:33

hepatocyte specifically. Because even within the

21:35

liver, obviously, there's numerous cell types.

21:38

And going after diseases where

21:41

they're driven by genetic

21:44

mutations in or mutations

21:46

in genes that are expressed by the

21:48

hepatocyte, those diseases are

21:51

driven primarily by those mutations in the

21:53

hepatocyte itself, on the cell therapy

21:55

side, I Think

21:57

we take a step back and say, where does cell therapy.

22:00

The in the pantheon of the various

22:02

modalities that we have as we've talked

22:04

about already at again taking the lessons

22:06

of oncology are really the definitive area

22:08

where. Cell. And cell

22:10

therapies. Are proving themselves to be

22:12

superior to any other modalities. They can

22:14

really wipe out a cell with high

22:16

specificity and hi efficacy. On and

22:19

so that's really where we're focusing cell therapies and

22:21

and a blade of approach were there really isn't

22:23

good other option and we're starting with auto immune

22:25

disease. To the point to me before my first

22:27

industry job with Astra Zeneca. I'm had

22:30

acquired Human genome Sciences than list of Than and

22:32

A from a map. Great. The

22:34

state of the art was. Disease

22:36

Control and ten percent more patience

22:38

than Placebo. And that was. right?

22:40

That's like billion dollar plus revenue in the

22:43

autumn you space and fast for ten years

22:45

were talking about securing these diseases. This is

22:47

the kind of power that cell therapy can

22:49

bring, the tables, curing diseases that we thought

22:51

were barely controllable. past. As.

22:53

You all know better than I do.

22:55

It takes a long time to go

22:58

from paper the patience to two questions

23:00

for you. Number one. You recently announced

23:02

that you have acquired a company a

23:04

company called Replace Therapeutic. So.

23:06

At a good be helpful to understand

23:09

what replace brings to the table is

23:11

complimentary with the paste approach. And.

23:14

Number Two, I think it be helpful

23:16

for our listeners to understand where you

23:18

were. Tome is on the journey from

23:21

from paper to patients and one of

23:23

the saying things I think that was.

23:27

Surprising to me as I when I

23:29

first. Started. Started

23:32

at home movies from

23:34

from the. Manuscript.

23:37

Into medicine was a lot harder than I

23:39

had for spot. We

23:42

were able to replicate the founders

23:44

work no problem with a get

23:46

good efficiencies in cancer cells with

23:48

it gets seventy percent efficiency integration.

23:50

Fantastic! When we started moving into

23:52

primary human cells, non dividing human

23:55

cells, the actual cells we need

23:57

to actually it's actually worked with.

24:00

With. Reagents that look like medicine

24:02

or assistance used to have

24:04

to below one presets in

24:06

A took us a long

24:08

time to sue understand what

24:10

knobs we had to turn

24:12

to actually sissies, the clinically

24:14

relevant efficiency sees actually that

24:16

we have now and that

24:19

that was a lot of

24:21

different things on the guide

24:23

our in A the format,

24:25

the architecture, the chemical modifications,

24:27

the sequences that were targeting

24:29

with the enzymes. The architecture the

24:31

it's the the actual versions of

24:33

the enzymes excess are. So we

24:35

had to do a lot of

24:38

innovation there and along the way

24:40

we we've actually undercover some interest

24:42

in biology their think other people.

24:45

In a are no publishing on

24:47

but one of the reasons why

24:50

we're very excited about of. About

24:53

the acquisition of of for

24:55

places that brings on an

24:57

entirely new message of making

25:00

small others including like see

25:02

I'm. Reading. Beacon

25:04

were no longer news in and

25:06

sense to write and sequence were

25:09

actually providing the physical sequence we

25:11

want written and now we use

25:13

ally against actually to like a

25:15

that into the right place and

25:17

so if it's a completely different

25:19

mechanism to add to a John

25:21

said. I think about.

25:24

Crisper, Cast nine like a microprocessor.

25:27

It is a core piece

25:29

of technology that enables various

25:32

different products. right?

25:34

And if. The an

25:36

initial original embodiment of cutting

25:38

genes is akin to a

25:40

desktop computer. I. See

25:43

the. Modification.

25:46

Or. The augmentation. Of

25:48

Crisper cast nine with or without

25:50

reverse transcriptase his for be surprising

25:52

being a laptop. What?

25:56

Pz. I is still uses a microprocessor,

25:59

but it's been. so augmented and

26:01

enhanced with other enzymes, that

26:03

it's now an iPhone. And

26:05

it fundamentally enables a whole

26:07

new product class, asset class,

26:09

therapeutic class in this case,

26:13

and a whole new way of approaching

26:15

disease, a whole new application. To

26:18

me, replace then is the Apple Watch. It

26:22

has standalone functionality, but it

26:24

is also complementary to what we're already doing.

26:27

And so at Tome, we really think about

26:29

innovation in two different ways. There's internal

26:31

innovation and there's external innovation. We're

26:33

not going to spend, again, your

26:36

dollars, Jorge, and those of

26:38

you out there in the country, inventing things

26:41

that are being well-invented in the academia

26:44

or in delivery companies. That's

26:46

where we partner or acquire, depending

26:48

on the nuances of the relationship.

26:51

But there are areas where we are

26:53

coming across technical challenges that

26:55

really nobody else is working on. Integraces

26:58

is a good example. There are very

27:00

few places in this world, academia or

27:03

otherwise, that are really focusing on integraces.

27:05

And the amount of progress we've made

27:07

on increasing both the accuracy and

27:10

the efficiency of integraces, not

27:12

only have we created IP and

27:15

some of those powerful integraces in the world, I would

27:17

actually argue that we are probably the

27:20

most advanced integrase scientific group in

27:22

the world right now. Same

27:25

thing for some of the delivery technologies. We're not going to go

27:27

invent new lipids. But where there are viral

27:29

vectors that allow us to put 30,000 base

27:32

pairs into a cell, a problem that nobody else really

27:35

needs to even think about, that's where

27:37

we put our innovation dollars. So

27:40

our innovation is really the combination of partnering

27:42

and acquiring, but also inventing

27:45

internally where we

27:48

need to unlock certain technical barriers to

27:50

exploit the full potential of PGI. So

27:53

John, when I met you, you had

27:56

this glorious beard. Today

27:59

I see you. You have this

28:01

glorious beard. But. We've run into each

28:03

other a couple times and you had no beard. Do.

28:06

You have an exploration for that. Yeah,

28:09

thanks. So. It's

28:11

funny now and a pass pass a

28:13

it. Plaza you know I had

28:15

a seasonal baird and people can tell

28:17

with season a was based on my

28:19

face was ah hair and then at

28:21

home early days of tom someone someone

28:24

actually made a bet and said if

28:26

we head to certain milestones more you

28:28

do. I said I will save my

28:30

beard if we had this milestone and

28:32

in our i don't set easy milestones

28:34

because I like my beard and my

28:36

wife likes my beard and my kids

28:38

hate it when I sit. And

28:41

I'm actually happy to say over the

28:43

course of tom. Over the past

28:45

two is years I've I've

28:47

now save my beard three

28:49

times. That. I did propose a leadership

28:51

team at one point that when we cure our first

28:54

child using Pg either we all shave our heads, but

28:56

it didn't go over very well. That's

28:58

I would gladly do that. So

29:02

we all aspire to have a clean

29:04

shaven Johnson for all of the authors

29:06

his sister to the thank you both

29:08

for for being here today. Thank

29:19

you! Been listening to Raising It. Says

29:21

it introduced. I mean preset. Years. In and

29:24

Me Olivia Lab with the help of the

29:26

bio and health team and a sub Kinsey.

29:28

The show is edited by Still Heads as

29:30

if you want to suggest topics are future

29:32

shows you can reach us at Raising Health

29:34

at a sixteen the.com Finally please rate and

29:36

subscribe to our some. The

29:40

content here is for informational purposes only

29:42

should not be taken as legal, business,

29:44

tax or and doesn't advice or be

29:46

used to evaluate any investment Our security

29:48

and is not directed at any investors

29:50

are potential investors and any and sixteen

29:52

season. Please note that a sixteen the

29:54

and it's affiliates naming him investments in

29:56

the company's discussed in the status. for

29:59

more details including a link to our

30:01

investments, please see a16z.com slash distributors.

Unlock more with Podchaser Pro

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