Podchaser Logo
Home
Alex Telford - Unlocking Innovation in Pharma

Alex Telford - Unlocking Innovation in Pharma

Released Tuesday, 30th January 2024
Good episode? Give it some love!
Alex Telford - Unlocking Innovation in Pharma

Alex Telford - Unlocking Innovation in Pharma

Alex Telford - Unlocking Innovation in Pharma

Alex Telford - Unlocking Innovation in Pharma

Tuesday, 30th 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

0:00

This episode is brought to you by TIGUS,

0:02

the modern research platform for leading investors. Stretch

0:05

your research budget with flexible expert calls

0:07

you can trust. At a

0:09

fraction of the cost of traditional expert

0:11

networks, TIGUS customers pay only one of

0:13

the expert charges with zero markups and

0:15

no confusing call credits, netting an average

0:17

of 70% savings. Don't want to

0:20

conduct a full hour call? TIGUS offers the

0:22

ability to schedule 30-minute meetings, an offer

0:24

you won't find anywhere else. And

0:26

they don't stop there. With white

0:28

glove custom sourcing for every project

0:30

and robust compliance measures, including a

0:33

dedicated 50-plus person analyst team that

0:35

vets every call transcript, TIGUS ensures

0:37

your privacy and protection. As

0:39

the industry innovator for qualitative insights, TIGUS helps

0:41

you find the right experts you need at

0:43

a quality and speed that can't be matched.

0:46

For a limited time, as a listener, you

0:48

can trial TIGUS for free by visiting tigus.co.patrick.

0:52

I'm endlessly fascinated by people who are doing

0:54

their life's work and David Sendera gets about

0:56

as close as you can in his crafting

0:58

of the Founders Podcast. Each

1:00

week, David gives you ideas you can use

1:02

in your work revealed by the life stories

1:04

of history's greatest entrepreneurs. He connects

1:06

the legacies of the likes of Kobe Bryant to

1:09

Estee Lauder to Winston Churchill and the list goes

1:11

on. He recently put out

1:13

a new favorite episode on Anna Wintour. Founders

1:16

Podcast will undoubtedly light a spark of ambition

1:18

and aid all of us in our quest

1:20

towards our life's work. David

1:22

isn't stopping anytime soon. In fact, if you

1:24

ask him, he's just begotten. I'm

1:26

proud that Founders is part of the Colossus network and

1:28

you can find a link to this treasure trove in

1:30

the show notes. Hello

1:35

and welcome everyone. I'm Patrick O'Shaughnessy and this

1:37

is Invest Like the Best. This

1:40

show is an open-ended exploration of markets,

1:42

ideas, stories, and strategies that will help

1:44

you better invest both your time and

1:46

your money. Invest Like the

1:48

Best is part of the Colossus family

1:50

of podcasts and you can access all

1:53

our podcasts, including edited transcripts, show notes,

1:55

and other resources to keep learning at

1:57

joincolossus.com. Patrick

2:00

O'Shaughnessy is the CEO of Positive

2:02

Time. All opinions expressed by

2:04

Patrick and podcast guests are solely their

2:07

own opinions and do not receive the

2:09

opinion of Positive Time. This

2:11

podcast is for informational purposes only and

2:13

should not be relied upon as a

2:15

basis for investment decisions. Clients

2:17

of Positive Time may maintain positions

2:19

in the securities discussed in this podcast.

2:22

To learn more, visit

2:24

psuheap.com. My

2:29

guest today is Alex Stillford. Alex

2:31

is the founder of Convoke, a software

2:34

platform to help streamline drug development and

2:36

commercialization. He has also been writing

2:38

frequent blog posts on the biotech industry since 2019,

2:40

keeping a pulse

2:42

on the direction of innovation. He

2:44

joined me today to talk about the history

2:46

of the pharmaceutical industry and what's becoming possible

2:48

in medicine in the coming years. Alex

2:51

helped break down the complexities of investing

2:53

in new drug development, breakthroughs in gene

2:56

therapy on the horizon, and the dance

2:58

between timely progress and restrictive regulation. This

3:01

industry has a ton to unpack and Alex

3:03

thoughtfully lays out the landscape. Please

3:05

enjoy my conversation with Alex Stillford. Alex,

3:10

we're going to spend a ton of time

3:12

today to give out pharmaceuticals. Not just the

3:14

industry, some of the companies, but the process

3:16

by which the world has created incredible

3:19

technologies for our health and also the

3:21

impediments to creating more of them in

3:24

the future. Could you give

3:26

your version of just a

3:28

sweeping overview of the

3:30

process and evolution of how we find drugs?

3:33

Alex Stillford, MD Yeah. So, it

3:35

starts with academic research. This

3:37

is mostly funded by government research and

3:39

it occurs in universities. So, the

3:42

NIH is the biggest vendor of biomedical

3:44

research in the world and they support

3:47

scientists trying to understand the basic science

3:49

of how diseases work. What are

3:51

the mechanics of diabetes? What are the mechanics of

3:53

heart disease? What are the

3:55

different genes, proteins involved in those

3:58

pathological processes? to

4:00

some extent, how could we modulate

4:03

those processes to affect disease? But

4:05

that's only really a small part of the

4:07

story. So once you have an idea for

4:09

how you might intervene a disease, you then

4:11

have to do a lot of work to

4:14

convert it into a drug. So these initial

4:16

hypothesis is to use these target specific proteins,

4:18

RNA, whatever, so you can modulate to affect

4:20

the disease. I've then taken

4:22

up by mostly small profit

4:26

biotech companies who then do

4:28

all the hard work of turning this into

4:30

a drug, using a molecule that can modulate

4:32

these targets, either inhibit it or

4:34

increase the activity in a certain way.

4:37

And then running everything through

4:39

first these in vitro systems,

4:42

just basically cells in a Petri dish, then

4:44

up to animal models, so testing them in

4:46

mice, monkey models of disease

4:48

potentially. Once it's safe enough, then you

4:50

can go into human trials. You

4:53

have a steadily escalating sequence of trials

4:55

starting from phase one, which is just

4:57

basic safety trials in human volunteers and

4:59

dosing. Phase two, we try to

5:01

get a sense of initial efficacy of the drug. And

5:04

then phase three, which confirms the efficacy and

5:06

safety to the extent we can. And then

5:08

you have to go to the regulators, FDA,

5:11

EMMA, the MDA, and they'll

5:13

review the package of information and make a

5:15

determination for whether or not the drug can

5:17

be marketed for a certain specific use case,

5:19

which is called an indication. That

5:22

long chain produces the things we use. What

5:25

was so interesting to me reading all your

5:27

work was the rate of discovery and

5:29

the rate of development seems to have changed

5:31

a lot. You read about this guy, Paul

5:34

Jansen, maybe you can describe who he was,

5:36

but it seemed like in the early days

5:38

of this research, a single person or lab

5:40

could produce dozens and dozens of things that

5:43

get used for a long time. And now,

5:45

I think he said somewhere in there, like

5:47

the average researcher will not work on anything

5:49

that ever gets to production and use in

5:52

trials or practice. So give us a history

5:54

of how efficient we are at finding new

5:56

stuff and what the important timeline points are

5:58

in that development. So, if we

6:01

go back to, let's say, the end of

6:03

the 19th century, where you have

6:05

a really nascent pharmaceutical industry. So,

6:07

you have pharmaceutical companies that came

6:10

out of apocasaries, which were selling

6:12

extracts of plants like you might

6:14

still get today, naturalistic medicine, and

6:16

extracts of organs. So, there

6:18

was this idea that nature produces some bounty of

6:20

molecules that are somewhat useful to treat health. But

6:23

we don't really know what parts of

6:25

the natural extracts actually treat health. So,

6:27

you have some companies that are extracting

6:29

just those natural products and selling them

6:31

in extracts of cyberscranes, whatever. And then

6:33

you have another set of companies that

6:35

came out of the chemicals industry, so

6:37

the dye industry. And around when

6:39

our ability to understand chemistry

6:42

and use chemistry to develop our

6:44

own molecules was developing to

6:46

this back in the 1900s, some

6:48

of these dye manufacturers were figuring

6:50

out ways to just adapt dye

6:52

chemicals and use them for therapeutic purposes.

6:55

Those companies eventually became more sophisticated

6:57

at the types of chemical

7:00

manipulations we could do, the type of extraction we could

7:02

do, and purification of these natural materials. And

7:04

that started to evolve into what

7:06

became the pharmaceutical industry around the

7:08

early 20th century. So,

7:11

then you had some

7:13

point, the golden age of antibiotics

7:16

between the two world wars became

7:18

the first great success story of the

7:21

pharmaceutical industry. So, moving away from products

7:23

like these natural extracts that didn't really

7:25

work in most cases towards antibiotics, which

7:27

had an obvious and noticeable effect on

7:30

the ability to cure these infectious diseases.

7:32

And a lot of these antibiotics were found

7:35

through just experiments. Go into the wild, find

7:37

soil samples, find drugs in these soil samples

7:39

from bacteria, and would purify them. And

7:43

that worked for a while, limited

7:45

to infectious diseases for the most part. And

7:47

then in the sort of 40s and 50s, you

7:50

start getting people like Jenssen

7:52

coming about. So, Jenssen

7:55

was a Belgian doctor. He trained in

7:57

medicine. And his dad was actually

7:59

one of these... importers of some

8:01

of these natural products, organ extracts. Janssen

8:04

recalled when he went to school that

8:06

in medical school, his contemporaries were making

8:08

fun of him or being part of

8:10

a family who sold these sort of

8:12

ineffective organ extracts. And

8:14

so Janssen had a bit of a chip on his shoulder, I think, from that. And

8:17

he wanted to figure out, adapt, how

8:19

can we actually improve on some

8:21

of these compounds that nature has given us? So he

8:23

went to the US for a while. He realized, I

8:25

think, when in 1948, there were some

8:28

labs that were springing up that were trying

8:30

to do more rational drug design, trade

8:32

their own new drugs rather than just take what existed

8:34

already in nature. And he brought that idea back to

8:36

Belgium and started his own lab in 1952 and started

8:38

trying to tinker

8:42

with natural compounds and use some of the

8:45

emerging tools of chemistry to just adapt some

8:47

of these existing compounds that had been discovered

8:49

already and find new uses for them. So

8:52

the 50s, there's a lot of opportunity in

8:54

the sense that you had these newly evolving

8:56

molecular tools, and you had a lot of

8:58

space to apply those tools to

9:00

discover new useful compounds. So you started with these

9:02

functional compounds that we found from nature. We had

9:05

a lot of starting points, morphine,

9:07

pettitine, and atropine, that could

9:09

be tinkered around with to find useful

9:11

things that are close to those natural

9:13

compounds, but not exactly the same. There's

9:15

some slightly different effects. And you had

9:17

an emerging suite of molecular tools. And

9:19

then you fast forward over time, the

9:21

kind of low-hanging fruit have, to some

9:24

extent, been exhausted with a lot of

9:26

those molecular tools were developed in that

9:28

period of time. So the pharmaceutical industry,

9:30

as it previously existed, has been declining

9:32

in its efficacy of finding new products

9:35

because you've exhausted the

9:37

opportunities to develop new drugs because we've

9:39

tinkered around a lot of what

9:42

is easy to find. And then you

9:44

have in the 80s, the biotech industry

9:46

arose. So just as you have this

9:48

declining pharmaceutical industry, which is small molecules,

9:51

medicinal chemistry, you have this rising

9:53

biotech industry that's built

9:56

on this idea that you can get bacteria to

9:58

produce any kind of product. of

10:00

arbitrary protein by inserting it into its DNA and

10:02

that's how you get a lot of those things like the

10:05

humanized insulin, the monofilamptibodies, there's all

10:07

these like biotech products that are now

10:09

somewhat still in the sentence. So

10:11

you have sort of two curves overlapping.

10:13

The fall off of the kind of

10:15

exhaustion of low-hanging fruits in traditional pharmaceuticals

10:18

and then the growth in what we

10:20

can do with biotechnology tools like recombinant

10:22

DNA and more recently CRISPR things like that. Yeah,

10:24

it'd be interesting to zoom all the way to

10:27

now and talk a little bit

10:29

about the best contenders for the

10:31

next explosion like what we saw in

10:34

small molecule pharma. What is the low-hanging

10:36

fruit of today? I love hearing you

10:38

talk about biologics or anything else that

10:40

you think about the major categories of

10:42

discovery or new enabling technologies that will

10:44

allow us to do more faster and

10:46

maybe have another explosion like what we

10:48

saw early in the industry's history. If

10:51

you look at the history of technologies in

10:54

biotech, it takes a long time to

10:57

commercialize something. Going back to the idea of people

10:59

discovering targets in academia, as soon as you have

11:01

an idea for a target, it takes something like

11:03

20 years for that to turn into a drug.

11:05

The best of time period 20 years plus

11:08

minus 10 is fairly consistent of how long

11:10

it takes to translate this sort of idea

11:12

of how you might treat a disease and

11:14

so much of what goes into commercializing and

11:16

developing a drug is turning it from this

11:18

idea into a product that is actually manufacturable,

11:20

scalable, doesn't trigger all these unwanted side effects

11:22

and many of these things you don't know

11:24

about until you've tried it out in humans.

11:26

Monoclonals now are the top

11:28

selling drug in the world, Keytruda,

11:31

previously with Humira. They're both monoclonal

11:33

antibodies and now the processes to

11:35

develop, produce, and sell those

11:37

monoclonal antibodies at scale are pretty established. It

11:39

took a long time to get there. I

11:41

think if you want to answer the question,

11:43

what is the next set of technologies that

11:45

are going to be really impactful in the

11:47

next 10, 20 years you need to look at

11:50

what is just nascent today

11:53

and getting approved. So things

11:55

like gene therapies, we have a few gene therapies

11:57

that have been approved. So-so, gentlemen, the treatment for

11:59

spiciness. It's just really devastating infant

12:01

neuromuscular disease. The patients who have that

12:03

disease would die before they are one

12:05

year old and the most serotype. But

12:08

now they can be seemingly almost cured

12:10

with these gene therapies and other modern

12:12

therapeutics. And that's one example of

12:14

an early success story, but we haven't had many

12:16

other examples of gene therapies being commercially successful. But

12:19

once you have one launch on success, you

12:21

can then iterate and refine the processes for

12:23

developing these drugs and you will have down

12:25

the lines future successes. So I think another

12:27

10 years, another 20 years, we'll see just

12:29

the process for developing manufacturing gene therapies.

12:32

All those things get worked out and

12:34

they'll become applied to many more conditions

12:36

at a larger scale. Also things like

12:38

cell therapies. So there's some

12:41

type of drug called CAR T-cell, which

12:43

is way more complicated than just a

12:45

typical pill. Immusals extract it from your

12:47

body. They're flown to a manufacturing site.

12:49

They're gene edited to have this sequence

12:52

protein inserted into their membranes that binds

12:54

a specific type of protein found on

12:56

cancer cells and it's reinfused into the

12:58

body and then it goes and eliminates

13:00

these pathogenic blood cancer cells. And

13:03

that's an extremely complicated manufacturing process.

13:06

You're taking these cells out as a human,

13:08

you're flying them to another country probably, you're

13:11

gene editing them and then you're flying them back and you have

13:13

to do that in a very short amount of time. One

13:16

because the viability of the product and

13:18

two because these patients have very severe types of

13:20

cancer and if you don't get it fast enough,

13:22

they're going to die. There's a lot of kinks

13:24

in those processes that need to be ironed out

13:27

before these technologies become scalable

13:29

beyond the niche use cases. One interesting

13:31

example is this company Bristol Myers Square,

13:33

they bought a company called CellGene who

13:35

are pioneers in this type of CAR

13:38

T-cell therapy. And they have

13:40

the same amount of employees working on delivering

13:42

these compounds as they have crazy patients. So

13:44

something like 4,000 each. The

13:46

high touch process expertise and scale you

13:49

need to deliver these super complex next

13:51

generation therapies like CAR T's, like gene

13:53

therapies that are somewhat personalized is just

13:56

way beyond traditional pharma where

13:58

you make a pill in a factory and then you put the the pill

14:00

on the shelf, you just have a pharmacist give it out.

14:03

So I think we're going to see a

14:05

lot more of these type of process-like products

14:07

that are way beyond just a standard pill

14:09

and it's much more

14:11

a whole complex, large,

14:14

the actual drug itself is a complex

14:16

product. There's also a whole process around

14:18

the drug of delivering it to patients in

14:20

a timely fashion, manufacturing,

14:22

which is very complicated. Another good

14:24

example is radiopharmaceuticals. So a lot

14:26

of companies have been investing in

14:28

this lately, buying up biotechs, developing

14:30

these drugs. And these are compounds

14:32

where you have a targeting element,

14:35

let's say cancer, and then you have a

14:37

radionucleotide, which emits radiation. You infuse these drugs,

14:39

it binds the cancer, it emits radiation, the

14:41

radiation kills the cancer cell in a targeted

14:44

way. And these are really complicated to deliver

14:47

because half-life assignment of these radioactive compounds is

14:49

something like seven days. You can manufacture and

14:51

deliver it to the patient within a few

14:53

days before they lose efficacy. So

14:55

these complex therapies are

14:57

one way of farmers trying

14:59

to build up moats to

15:02

distribution of products. Can you

15:04

say a few words? There's four categories there that

15:06

we'll focus on, three that are terms that I

15:08

think people have heard, but they may not know

15:10

exactly what it actually means. So

15:13

the first is monocle antibodies or

15:15

biologics. The second is gene

15:17

therapy. The third is cell therapy. Can

15:19

you just describe what those mean and

15:21

what they're doing as categories of drugs?

15:24

So I'll start with this antibody, right? So when

15:26

you get infected with some sort

15:28

of virus or bacteria, your

15:30

immune system will generate antibodies against

15:33

the invading threat. And these

15:36

antibodies are specific to foreign

15:38

proteins. Your body's always generating all the

15:40

time all these antibodies. And

15:43

when a virus infects you, then

15:45

if you have an existing antibody that

15:47

binds that virus, it'll multiply within the body, and

15:49

then it will bind to viral copies

15:52

and it will stop the infection by

15:54

just binding and neutralizing these viruses or

15:56

bacteria. And that was recognized

15:59

as Antibodies

16:01

can bind proteins on these

16:04

foreign microbes. It can

16:06

also probably be useful for binding

16:08

other molecules or other proteins

16:11

in the body that aren't necessarily foreign

16:13

but may impact disease in certain ways. So we can

16:15

use it to knock out proteins that we would want

16:17

to knock out to treat diseases. So

16:20

one example is

16:22

Humira, which is until recently the top selling

16:24

drug in the world, something like 20 billion in revenue.

16:26

It's an antibody that binds to a protein

16:29

called TNS alpha, which is

16:31

an inflammatory protein. People

16:33

who have arthritis in these inflammatory conditions

16:36

will have an excess of this protein

16:38

that just causes an immune response that

16:40

leads to inflammation and joint pain. If

16:43

you can put in this antibody that binds this

16:45

protein, takes it out, then you can treat some

16:48

of these diseases that are pathologically too many of

16:50

a certain protein. And if you want to treat cancer,

16:53

cancers have a different composition of proteins

16:55

on the surface than normal cells, you

16:58

can put in antibodies that

17:00

bind those cancer cells and kill them

17:02

specifically. So where the monoclonal comes in

17:04

is that a while back

17:06

there was some scientists figured out that

17:08

you could take these immune cells that

17:10

were producing antibodies and combine them

17:14

with a cancer cell essentially and

17:16

immortalize the cells that produces antibodies.

17:18

That's how they make them just

17:20

scalable, manufacturing process. You have these

17:24

cells that are artificially immortalized that produce the

17:26

antibody anyway, and they just produce tons and

17:28

tons of this antibody product that you can

17:31

just grow it up into a bioreactor vat

17:33

and then up to a certain level grow it and

17:35

then you skim off the antibodies. Gene therapy

17:38

is this idea that you can insert

17:40

genes into the body to compensate

17:42

for effective genes, which you have something

17:44

like this spinal muscular atrophy example where

17:47

patients who have that disease, they lack

17:49

a functional copy of a gene called

17:51

SMN1, which produces a

17:53

protein that your motor neurons need to

17:56

stay alive essentially. And if

17:58

you can package up that gene

18:00

and deliver it into a cell, the cells

18:02

that are missing this functional copy will then

18:04

produce the protein. You can restore that function.

18:07

That seems like quite a promising approach. But

18:09

then, of course, the difficulty with fixing these

18:11

mutations is actually getting the gene into

18:13

the cells. So a lot of the

18:15

innovation around gene therapy was figuring out how to deliver

18:18

genes to cells. And the approach that

18:20

we've landed on now in the industry, the

18:22

dominant approach is to use a virus,

18:25

essentially. So what Christmas is doing

18:27

is making specific let's

18:30

say cuts of the DNA to

18:32

either eliminate genes that we

18:34

want to eliminate or potentially down the

18:36

line to insert new functional copies of

18:39

genes. And then cell

18:41

therapy is this idea that you

18:43

can edit cells, immune cells in

18:45

the body, to program them in a way to

18:47

do something that you want them to do. They

18:49

might otherwise be inclined to do so. You

18:52

take immune cells that are very good at killing other cells, they're very

18:54

good at killing foreign cells and

18:56

microbes. But the body has

18:58

a lot of systems to prevent immune cells

19:00

from killing your own cells. So

19:02

what cell therapy is doing is taking

19:04

out some of these immune cells and

19:07

treating them so they'll bind and recognize

19:10

pathogenic cells like cancer cells that

19:12

may look to the unedited immune system

19:15

similar to a normal cell and then

19:17

redirecting the immune system towards removing

19:20

those pathogenic cells. You tell

19:22

me a little bit about the

19:24

potential speed of all

19:26

of this and the problems that slow

19:28

things down. I was really struck by

19:30

the AIDS example, your post, and

19:32

also just by this, I think it's Moore's Law backwards,

19:35

or E-R talk

19:40

about that episode with the AIDS epidemic and the frustration with those that

19:42

could have benefited from some drugs and just the slow

19:44

process of being able to get them and E-R and

19:47

the things that are slowing down the

19:49

iteration speed cycling of, can we make

19:51

a discovery? We've got patients in need.

19:53

How do we shorten the time between

19:55

a discovery and an implementation? The

19:58

big tension in how you

20:00

regulate drugs is how

20:02

do you balance the need to develop

20:04

drugs quickly while also keeping patients who

20:06

are participating in clinical trauma safe and

20:09

keeping people who take the drugs when it

20:11

comes to market safe. So it's really

20:13

difficult I think for regulators to find

20:15

an appropriate balance and it's almost an

20:17

impossible problem because if you come down

20:19

too hard on drugs and on

20:21

manufacturers and make them jump to too many hoops,

20:24

then it's just going to be uneconomical to develop

20:26

drugs and there's going to be a massive invisible

20:28

graveyard of people who could

20:30

have been saved had the drug come to

20:32

market faster but it was blocked by various

20:34

regulatory processes. But then if you're too lenient

20:36

on drug makers, some of them will take

20:39

advantage and will push drugs onto market that

20:41

shouldn't have been there. They may be acting

20:43

completely good intentions and they'll test according to

20:45

the process that regulatory agency demands and bring

20:47

the drug to market and it turns out

20:49

later that it causes some harm that we

20:51

could have foreseen if you had more rigorous

20:53

testing. So there's

20:56

no one size, that's all solution

20:58

to fix this tension. So

21:00

you have to always make some compromises

21:02

and coming back to the issue about

21:04

we're running out of low hanging fruit,

21:07

one of the biggest problems is that

21:09

we already have so many good drugs

21:11

for many conditions. So if

21:14

you have diabetes or something, then

21:16

we have insulin, we have many

21:18

varieties of insulin, we have the

21:21

GLP1 drugs now, we have a number

21:23

of other drugs that treat diabetes effectively

21:25

and they're very effective drugs. So it's

21:27

hard to make the case that you

21:29

need to bring another drug to

21:31

market with great haste to treat large

21:33

number of patients who are being well

21:35

treated by current drugs. So

21:38

under that situation, you

21:40

probably as a regulator are inclined to

21:42

put really high burdens on drug makers.

21:45

So you have to absolutely demonstrate that anything you bring to market

21:47

is really safe, you have to jump through all these tubes, you

21:49

can do mortality studies, all these really onerous

21:51

requirements. On the other hand, if

21:53

you have a disease where there's no effective standard

21:55

of care and patients are

21:58

dying or suffering greatly. from

22:00

their condition, then it makes

22:02

sense to potentially relax the regulatory burdens.

22:04

Compared to the AIDS issue, when you

22:07

had the AIDS epidemic, there

22:09

were a number of drugs that seemed to

22:11

be promising. The pharmaceutical

22:13

companies were developing at

22:15

speed, like they did with COVID, right? A

22:17

lot of companies were developing all their anti-COVID

22:19

drugs and vaccines. And the AIDS patients were

22:22

quite rightly saying that we're going to die

22:25

anyway. So we have a death sentence. You

22:27

should let us try these drugs

22:29

and see if they're effective because

22:32

the alternative is death. And

22:34

the FDA, until that time, didn't

22:37

really have a process to deal

22:39

with these differences in need between

22:41

conditions. So they would treat

22:43

diabetes similar to how they would treat

22:45

AIDS. So after a lot

22:47

of protesting by the AIDS community and

22:49

a lot of patient advocacy, the FDA

22:51

ended up speeding a number of these

22:54

compounds through development, so AZT and

22:56

DDI to the drugs. These turned out to

22:58

not be particularly effective drugs,

23:00

but you could argue that potentially by

23:03

starting off that process of defining a pathway

23:05

for how you treat AIDS, getting some drugs

23:07

on the market, getting some initial revenue, you'd

23:09

then spurred the process to iterate and invent

23:11

drugs that are now very effective. So now

23:13

there are very effective treatments for AIDS. This

23:16

initial speeding of the regulatory process during

23:18

the AIDS epidemic turned into

23:20

what became the accelerated approval pathway.

23:23

And that is for drugs that have

23:25

seen promising in disease a very high

23:27

unmet need like AIDS was, like

23:29

a lot of cancers are. Drugs can get

23:32

through regulators with much

23:34

less evidence than they can in other conditions

23:36

like diabetes where the standards might be higher.

23:38

So now what you've seen in

23:40

the industry is regulatory arbitrage, where a lot

23:43

of companies are investing in things that are

23:45

easier to get past the regulators. You don't

23:47

need to do all these really huge phase

23:49

three trials because there isn't a good standard

23:51

care, where there's not so many patients. So

23:54

you see a lot of this investment in

23:56

cancers and greater genetic diseases.

23:58

The requirement for evidence is... lower and

24:00

the need is greater. One of the things that

24:02

you write a lot about is just

24:04

the discovery process itself. And there's a

24:06

very clever video game analogy to Super

24:08

Mario and what we can learn about

24:10

speed running video games and relating that

24:12

to the process of discovering things that

24:14

might be useful in the pharma world

24:16

and the biotech world. Maybe you

24:18

flush that idea out of just your interest in

24:21

the process of discovery itself and what we can

24:23

learn from other domains like games. Yeah,

24:25

so I think if you look at

24:27

how things get discovered in the pharma

24:29

industry, it's often very

24:31

serendipitous. So you'll have

24:34

these stories of people tinkering away on

24:36

some idea they think is promising for

24:38

years and years and then eventually

24:40

that becomes a drug. So like the GLP ones

24:42

is a good example where you

24:45

had decades of the scientists at

24:47

Novo Nordisk pushing for this idea,

24:49

tinkering around the boundaries of GLPs

24:52

and trying to figure out how to

24:54

actually take this idea from just

24:57

a concept to an actual drug. So

25:00

I think one way to help

25:02

make the industry more effective is to

25:05

try and find ways of

25:08

promoting that artful tinkering

25:10

because we just haven't proven to be very

25:12

good at developing drugs

25:15

to a pre-described positioning if you

25:17

like. The really important and powerful drugs

25:19

get discovered from this bottom-up process where

25:21

you have scientists who are really passionate

25:23

about an idea, they push through their

25:25

management saying you should stop this, it's

25:28

clearly not working and they try to

25:30

find ways to work on this without

25:32

management finding out. They try to

25:34

get resources wherever they can and champion

25:37

despair, loss of opposition and then eventually

25:39

it turns into this drug that becomes

25:41

a mega blockbuster and helps a huge

25:43

amount of patients and then the management

25:45

thinks they'll say that we do it

25:47

all along or something in retrospect. But

25:49

you can't prospectively figure out what drugs

25:51

are going to be really impactful a

25:53

lot of the time. So

25:55

I think it's quite dangerous in an industry

25:57

like pharma which is so innovation. driven

26:00

and science driven to impose these

26:02

top-down, pre-described notions

26:05

of what people should invest in, you're very

26:07

likely to be wrong. Until a few years

26:09

ago, I think Novo Nordisk was regarded as

26:11

a pretty boring European

26:13

pharma company, not very innovative,

26:16

just doing insulin. But now

26:18

they're like the 16th

26:20

biggest company in the world or something and

26:22

everyone's excited about the GLPs. You would have

26:24

necessarily predicted that would come out of Novo

26:27

Nordisk, but Novo Nordisk was able to produce

26:29

GLPs because they had this really long-standing interest

26:31

in a specific area, edible diseases, they support

26:33

the scientists there and they've just been tinkering

26:36

in this area for a long time. So

26:39

to come back to the food moray

26:41

idea, that post is about this

26:43

idea that one of the

26:45

ways you might productively use

26:47

technologies like artificial intelligence and

26:49

simulation tooling is to have

26:52

an intuition about what areas of

26:55

science might be promising and then

26:57

use automation to do scalable

27:00

tinkering around that nucleus of

27:02

an idea. So the example

27:04

in Super Mario was that there is this idea

27:07

that you could do a specific type of

27:09

jump to get through a level in Super

27:11

Mario much faster than people have been able

27:13

to do so far, but no one knew

27:15

it was possible. So until a software developer

27:17

developed this tool called Scattershot, which simulates millions

27:20

and millions of Marios jumping up this level in

27:22

every possible configuration of jumps and starting positions, he

27:24

managed to figure out there actually was a way

27:26

to get up to the top of the castle.

27:28

They were trying to jump up and finish a

27:30

level much faster than previously possible. It's changed this

27:33

whole way that speedrunning is approached for that game.

27:35

Speedrunning itself is probably out of interest to that

27:37

many people, but the general principle I think is

27:39

interesting that you have this intuition

27:41

that something is possible. For Jansen, when he

27:44

was developing his drugs, he had an intuition

27:46

that you could improve this compound

27:49

pethidine to make

27:51

a better drug, but he didn't exactly know

27:53

how to modulate it to make it the

27:55

best drug it could be. He was just

27:57

tinkering around. So if you could use technologies

28:00

to automated scientists to feel the

28:02

process of tinkering, trying out solutions,

28:04

testing them, then you can tighten

28:07

the feedback loop around the

28:09

industry and maybe develop drugs a lot faster

28:11

than you might otherwise be. I think

28:13

one of the big problems with pharmaceuticals and industry is

28:16

that the iteration speed is super low

28:18

compared to something like tech. Like in

28:20

tech, if you have

28:22

an idea for a product, you can

28:25

spin up a demo in

28:27

a few days and you

28:29

can show it to some customers, you

28:31

can get some feedback on that, you

28:33

can then iterate it, layer instantly in

28:35

code effectively, and then show the next demo to

28:37

customers, get more feedback, you can do these A, B

28:39

tests at massive scale. You can move super fast and

28:41

learn very quickly. But pharma is

28:44

a very far domain to learn quickly

28:46

because everything takes so long. Clinical

28:48

trials will take around 10 years to get

28:51

everything through clinical trials. Even

28:53

do animal experiments, it takes weeks

28:55

or months to do animal experiments in certain ways.

28:58

Making all these chemicals takes a long time. You're

29:00

testing even in the very earliest stages. So

29:03

you learn at a very slow rate about

29:05

what works. And if you could

29:07

just find ways to speed that up across

29:09

the whole process, then you'll do a

29:12

lot to, I think, address some of these

29:14

problems with the pharma industry like drug

29:16

prices, just a huge healthcare

29:18

spend, things like that. I was

29:20

AI-figured to all this, obviously alpha-holed within

29:22

the mainstream consciousness. I think Jensen Wong,

29:24

the CEO of N videos, recently talking

29:27

about how much explosion of

29:29

activity there is using AI in the

29:31

world of medicine and biology. So talk

29:34

about the role that this new set

29:36

of tools might play in both discovery

29:39

and speed and tinkering

29:41

and all these different ideas. I

29:43

think I'm short-term, pessimistic, long-term optimistic

29:46

when it comes to AI for

29:48

drug discovery. And I'm much

29:50

more optimistic in the short-term about using AI

29:52

to automate some of the processes along with

29:54

drug development, not necessarily the designing of drugs

29:57

itself, but all the steps to bring a

29:59

drug to mind. market. So if

30:01

we start with drug discovery, like finding a drug, I

30:04

think it's just very difficult to see

30:06

how AI is anything more than just

30:08

another tool in the expanding toolbox that

30:10

drug developers use to produce and

30:13

test molecules. AlphaFold's an

30:15

example where now you can predict the

30:17

protein structure from a protein sequence, which

30:19

is a problem that we didn't know

30:21

how to do to a high degree

30:23

of accuracy until before AlphaFold. And

30:26

a lot of people are saying, oh, AlphaFold is

30:28

going to revolutionize drug discovery. But

30:30

in reality, it's just one little piece

30:32

of the hugely complex puzzle where sometimes

30:34

you'll have situations where you don't know

30:36

the structure of a protein you're trying

30:38

to make a drug against. And

30:40

AlphaFold can be a helpful starting point for that

30:42

specific circumstance. But

30:45

actually, AlphaFold has problems

30:47

with the very granular

30:49

predictions of the configuration

30:51

of the sites where drugs might bind. So

30:53

it's not completely accurate. So we need more

30:55

accurate models for it to be really useful

30:57

for drug discovery. And

30:59

to improve AlphaFold, we need more

31:01

data. It's just taken a long time to collect

31:03

all this data. So AlphaFold has trained on

31:06

decades of protein structures that have

31:08

been collected painstakingly by researchers growing

31:10

crystals in dark rooms. It's

31:12

definitely speeded up parts of

31:15

that process, but a very small part.

31:17

And I think the way AI is going to impact

31:19

the pharma industry looks similar to that. A lot of

31:21

little tools that are going to be very

31:24

useful and accurate, but they're going to take a long time

31:26

to deploy and figure out how best to use them and

31:28

work them into the process. One

31:30

thing I should also mention is that there's this

31:32

idea that drug discovery is rate limited

31:34

by our ability to design

31:36

molecules and design new drugs. And

31:39

that's not really true. Our ability

31:41

is much more rate limited by the downstream

31:44

clinical development and

31:46

testing these drugs and gathering information. We

31:49

need to actually justify approving these drugs

31:51

for sale and working them. While

31:53

I'm optimistic about over the

31:56

long term using things like AI to

31:58

predict what drugs might be

32:01

effective to do drug discovery and

32:03

drug candidate selection. I'm

32:05

much more optimistic about using AI in the

32:07

short term to help drugs to market faster

32:09

than they otherwise might have been. So things

32:12

like can we use AI

32:14

to help prepare

32:16

regulatory documents quicker? Can

32:18

we use AI to do things like

32:21

help companies figure out what

32:23

the markets for their drugs are and prioritize what

32:25

opportunities they should invest in? How can we use

32:27

AI to help identify patients who might be a

32:30

good fit to enroll in a certain trial? So

32:32

things like trial enrollment just takes away

32:35

too much time. So just

32:37

better tooling to identify patients who might

32:39

be a good fit for a trial

32:41

and enrolling them seems pretty impactful. If

32:44

you were just a czar of this

32:46

entire universe and godlike powers, what two

32:49

or three changes would you

32:51

make regulatory, technology-wise, industry structure?

32:53

Anything that you could change.

32:56

What would you change that you think would

32:58

most benefit patient outcomes or the overall

33:00

system? I would make regulatory

33:03

changes. I think I would want to

33:05

formalize the idea that different diseases require

33:07

different standards of evidence and then pre-publishing

33:09

the degree of evidence that we require

33:12

for each different disease. So something like

33:14

diabetes should have quite a high standard

33:16

of evidence. So you should have to

33:19

enroll very large numbers of patients and

33:21

test mortality because we have

33:23

extremely good drugs. And in other cases,

33:25

we should have a lot of diseases where our standard of

33:28

care is not as effective. We should

33:30

formalize this idea of smaller trials, maybe approved drugs

33:33

with a weaker standard of

33:36

evidence. And we are seeing some moves

33:38

towards that with rare diseases. So

33:40

you have a lot of these conditions that

33:42

are too small to really be able

33:44

to tell. They maybe have

33:46

too few patients, but they may have too few patients plus the disease

33:49

evolves over too long of a time for you to reasonably

33:52

be able to figure out the standard

33:54

drugs are really working to any high

33:56

level of confidence. So I think in those conditions,

33:58

you'll have to be able to say... Look,

34:00

we're just not going to be able to determine

34:02

this in a trial. We have to, based on

34:04

some principle of the reasons, evidence we've

34:07

collected about how the drug works in animals

34:09

and maybe some initial human testing and

34:11

some preliminary biomarker evidence, we can then

34:13

approve this drug and roll it out

34:15

and then collect information more rigorously once

34:18

on the market. Because you're just never

34:20

going to get drugs for certain diseases,

34:22

just uneconomical to achieve this vision of

34:24

hyper-personalized medicine where you're

34:26

developing a drug for everyone's individual

34:28

condition and biology without some ability

34:30

to make inferences about what is

34:32

likely to work based on early

34:34

evidence and genetics and reasoning

34:36

about the mechanism of action. So

34:39

I think the second thing I would do

34:41

is just be much more rigorous about data

34:43

collection post-marketing. So you have a lot of

34:45

drugs that go on to the market and

34:47

then they're approved and then we're not really

34:49

following up sufficiently to be able to

34:51

tell us they really work. So there's been some instances

34:53

of drugs that have got this extolibrated approval on

34:56

some fairly flimsy evidence but they seem promising.

34:58

But then the company is meant to do

35:00

a confirmatory trial to determine, okay, actually, we

35:02

have a good reason to believe this drug

35:05

is going to work but we

35:07

don't actually know that until we've collected this evidence

35:09

over five, ten years or whatever. So

35:12

companies drag their heels on doing the confirmatory

35:14

study. I think the regulators should be much

35:16

more stringent in forcing companies to collect that

35:18

data and report it and take it out

35:21

of the market. That is a combination of

35:23

being more open to approving

35:25

drugs based on a lower standard evidence when

35:27

there's no good treatments and when it's just

35:29

unfeasible to do with standard clinical trial coupled

35:31

with more rigorous data collection and then taking

35:34

things off the market when they're not working.

35:36

I think that will help increase the sort of range of learning

35:38

in the regulatory process. Surrogates as

35:40

well, I think, are really important. So

35:42

surrogates are measures like biomarkers that you

35:45

can use as a proxy to tell whether a

35:48

disease is being treated effectively or not. And so

35:50

for cancer, what you really care about when someone

35:52

has cancer is stopping them from dying but you

35:54

can get a proxy for that by looking at

35:56

the size of the tumor and it's not always

35:59

a perfect correlation. you shrink the tumor.

36:01

Most of the time, that's a good

36:03

thing. Sometimes you shrink the tumor, but it

36:05

doesn't actually help the patient survive longer in the

36:07

end. So you're exposing them to toxicity and

36:09

you're not helping them survive. So it's actually you

36:11

doing that harm with that drug. The idea

36:13

of a surrogate is that you have a

36:16

measure like tumor shrinkage that

36:19

predicts benefit down the line and that

36:21

helps you develop a drug much faster. A lot

36:23

of drugs have been, not to market, much faster

36:25

in cancer than they otherwise would have

36:27

been many very effective drugs because they've

36:29

used these surrogate markers of efficacy like

36:31

tumor shrinkage. But it's harder to

36:33

apply this principle in other conditions like

36:36

more complex chronic conditions where you don't

36:38

have an objective measure that's as simple

36:40

as does a tumor get smaller or

36:42

not. If you're treating a disease that's

36:45

eventually fatal, if you see that this

36:47

biological response happens early on, then that's

36:49

very highly predictive of improved survival down

36:51

the line. And just finding more of

36:53

these surrogates is a really, I think,

36:55

valuable activity that something like the NIH

36:58

and government agencies should invest a lot

37:00

more money in developing because as soon

37:02

as you have an established surrogate that

37:04

the regulators could use, you really speed

37:06

up how quickly you can iterate on

37:09

developing drugs. You can take it to the clinic, you can

37:11

see if it works in the surrogate, and then you can decide whether

37:13

or not to continue or discontinue based on that. You don't have to

37:15

wait five, 10 years to just

37:17

survive or not. So something like aging, we're

37:20

never going to get drugs to prolong

37:22

people's lives until we figure

37:25

out a surrogate for aging. Can

37:27

you talk about RCTs, randomized controlled

37:29

trials, and something like vitamin D?

37:31

I love the idea that this

37:33

tool, RCTs, is an incredibly important

37:35

process that's helped us learn a

37:37

lot. What, if any, limitations are

37:39

there in your mind to RCTs

37:41

and learning? So the big

37:44

limitation to RCTs is they

37:46

take time, they're complex to run, and then people

37:48

don't want to be in the control group, essentially.

37:50

So you have to expose patients to something that

37:52

they don't want to take really because you're entering

37:55

an RCT for a drug, you want the active

37:57

drug. But maybe take a step back. So the

37:59

idea behind... an RCT is that

38:02

you have patients

38:04

or people who are going to take

38:06

some intervention and you want to figure

38:08

out does this intervention really work

38:11

or not. And you have to find some

38:13

way of assigning the intervention randomly because if

38:15

you rely on anything other than randomness, there's

38:17

going to be some bias that creeps into

38:19

the study. Let's say you have this

38:21

new drug and you take it to doctors and you say,

38:23

I want to test this new drug, then the doctors

38:27

may be more inclined to give

38:29

the drug to patients who are sicker, who

38:31

really need it. And they're less inclined to give

38:33

the experimental drug to patients who seem like they're

38:35

going to recover anyway. So if you just do

38:38

that non-random allocation, you're going to see that

38:40

people who take the drug die at a much higher

38:42

rate or have a much worse illness than people who

38:44

don't take the drug because you have

38:46

this selection bias of doctors giving the drug to

38:48

people who are less likely to have a good

38:51

outcome. You need to find a

38:53

way to eliminate those biases and you can do that

38:55

with just total randomization. And so coming back to the

38:57

vitamin D example, one thing in

39:00

data and retrospective data that

39:02

looks at vitamin D levels

39:04

and outcomes is that

39:06

you see people with low vitamin D levels

39:08

have generally worse health outcomes.

39:10

So they have high rates of mortality,

39:13

they have cardiovascular disease. I think it's

39:15

probably been correlated with every bad thing

39:17

you can probably get. And there's a

39:20

natural inference there that we should just

39:22

supplement vitamin D to the people

39:24

who are low levels and then they'll have less

39:26

rates of all these bad diseases. So then people

39:28

do the trials and they do

39:30

a randomized control trial when they assign

39:33

people randomly to groups and they

39:35

give supplementation of vitamin D and then they

39:37

measure outcomes in all these things like mortality

39:39

and health. And then what they find is that

39:42

actually vitamin D has very

39:44

minimal or no effect on both of these

39:46

outcomes, even though some of the

39:49

data from retrospective analysis looks pretty strong. So

39:51

how did that happen? What

39:53

you find out is something like vitamin D

39:55

is actually a marker for poor health in

39:57

general. So vitamin D is produced by your

39:59

boss. body in response to sunlight.

40:02

And if you are ill

40:04

or poor health generally, you're

40:06

less likely to go outside or you

40:09

may be less mobile in general, so you may

40:11

get less exposure to sun.

40:13

And then you'll see the correlation between just

40:15

general poor health and vitamin D. It's like

40:17

with pushups. So they say, if you can

40:19

do less than 10 pushups, then you're much

40:21

more likely to die. Not literally the ability to

40:23

do pushups determines how likely you are to die

40:25

is just being able to do pushups

40:28

is a decent proxy marker for general health.

40:30

So RCTs are just the most

40:33

effective way we know of eliminating all

40:35

these biases that you don't know exist

40:38

from the start. Because otherwise, it's just everything

40:40

you try gets really confounded and

40:42

you don't even know how it's confounded. Can

40:45

you imagine an alternative or do you

40:47

think this will be and needs to

40:49

be the method by which we learn

40:51

truth and efficacy across medical interventions? So

40:54

I think RCTs are always going

40:56

to be the gold standard. And

40:59

I think when we can do RCTs

41:01

recently, we should do them because

41:03

they just eliminate so many of these

41:05

problems that you're just unaware of. That's the biggest

41:07

issue if you control for the sources of bias

41:09

that you think are likely to exist. There'll be

41:11

hidden sources of bias that you just can't control

41:13

for. So I think we want

41:15

to keep RCTs as a control inventory method.

41:17

But there are other tools in

41:19

these instances where RCTs aren't feasible that have a lot

41:21

of promise. So things like if

41:23

you do a single arm study, which is

41:26

just one treatment group, and you compare the

41:28

progression of a disease with a

41:30

matched digital twin of these patients, you

41:32

can train with information from natural history

41:35

data. We can make a good assumption

41:37

about the rate at which these patients would

41:39

have progressed had they not had

41:41

the drug. So there's been a few approvals

41:43

that have used some of these matched natural

41:46

history data as an alternative cohort. And those

41:48

have been in cases where there's been too

41:50

few patients to really do largely

41:52

randomized trials. And then you have

41:54

these instances of super rare diseases where you

41:57

may have just a handful of patients

41:59

in the US, for instance, is the

42:02

same possible thrown RCT and have any

42:04

reasonable statistical outcome. You need to use

42:06

things like digital twins or

42:08

surrogates, single arm trials, you

42:10

can't use RCTs. There's a lot of innovation

42:13

in this digital twin idea and natural history

42:15

cohorts. Obviously, a big driver

42:17

of all this is profits. We

42:19

talked about how people don't like pharma,

42:22

in part because they charge so much

42:24

for really valuable drugs and they make

42:26

big profits. And that bothers people as

42:28

it relates to their health. But profits

42:30

and revenue drive. The motive

42:32

for discovery and the US has always been a

42:34

leader here. Can you talk about

42:36

what you've learned about blockbuster drugs specifically? You

42:39

can define what you mean by a blockbuster

42:41

drug, but it seems like the biggest drugs

42:43

that represent, there's sort of a mini power

42:45

law here that the top handful

42:47

of drugs represent a huge percent of the

42:49

entire industry's revenue. So talk about the business

42:51

side here, the distribution of revenue, the role

42:54

of blockbuster drugs, the good, the bad, the

42:56

ugly. Love to hear what you've learned there.

42:58

Yeah. So a blockbuster drug is a drug

43:00

that makes a billion dollars in annual revenue.

43:03

So blockbusters are really have an

43:05

outside influence in the pharmaceutical industry

43:07

because they account for a

43:10

huge proportion of the overall revenue

43:12

of the industry. Well, the way

43:14

the economics operate is pretty analogous

43:16

to venture capital and these long

43:19

tail models where you have many

43:21

losers who don't make much money or don't recoup the

43:23

money that has invested in them and

43:25

a few really huge winners that will go

43:28

off and generate billions of billions

43:30

and billions of dollars and pay for all

43:32

the failures many times over. So the industry

43:34

is in many ways worth

43:36

investing in for investors and

43:38

for companies because there's always this potential

43:41

of hitting it really big and having a mega

43:43

blockbuster like Humira or Kritrudar, the COVID vaccines that

43:46

generate tens of billions of dollars in revenue a

43:48

year. Because if you look at all the drugs

43:50

that are getting launched, it's sad that you have

43:52

all this work that goes into developing a drug

43:54

and it takes me 10 years to

43:56

get it to market or 12 years to get it to market. And

43:59

it... launches and then it stops. 55%

44:03

of drugs that launch, that get to the left process,

44:05

they don't even recoup enough money to

44:07

pay the average development cost of

44:09

a drug. And then you

44:11

have a very small number of drugs, blockbusters, that

44:14

30% to 40% of revenue of the

44:16

whole industry is made by these blockbuster drugs, which

44:19

are quite a small fraction of all

44:21

drugs. But when I look at the numbers, something

44:23

like 170 blockbuster

44:25

drugs that were actively

44:28

generating revenue, and then the

44:30

whole long tail of other drugs is

44:32

making very small sums relative to blockbusters.

44:35

So because the industry operates like

44:37

a lottery type model where you just want

44:39

to really hit a big, you have a

44:41

venture capital, it distorts all the incentives. So

44:44

you see a similar thing in

44:46

biotech investments that you do with VCs

44:48

where the farmer is only really interested

44:50

in developing a drug if it can

44:52

potentially become one of these blockbusters. Because

44:54

anything less than that is just unlikely

44:56

to recoup the investment. So you get

44:58

a lot of these small markets that

45:00

patients really would like to have drugs

45:02

for. They could be really beneficial and we

45:04

know that we could potentially develop a drug against this

45:06

disease. We have a good understanding of

45:09

the mechanism, like many small genetic diseases, but it's

45:11

just not worth big farmer's time or even biotech's

45:13

time to invest in developing these drugs because the

45:15

economics don't work out. Even if

45:18

you get to the whole process of developing this drug and

45:20

it works and you get to clinical trials, you

45:23

maybe make a few tens of

45:25

millions a year and that's just absolutely not worth it because it

45:27

will cost you 100, 200, 300

45:29

million just to get the drug through the whole development process.

45:32

So there's a lot of, I

45:34

think, alpha that could be unlocked

45:36

in making the process much cheaper

45:39

in how drugs are tested and validated because

45:41

it would mean that it's actually

45:43

worth developing all these drugs that are not

45:45

worth developing. What do you

45:48

think about drug pricing as a key variable

45:50

in all of this big equation that blockbuster

45:52

drugs are driven by a price times an

45:54

amount? What drug pricing sometimes comes up

45:56

all the time as predatory

45:59

or strange or... or the US system

46:01

subsidizing a lot of the rest of

46:03

the world. We make an outsize percent

46:05

of the discoveries and you want to be

46:07

compensated for that. It seems like a very complicated equation.

46:10

So what, if anything, have you learned

46:12

about drug pricing that you feel is different than

46:14

the norm or how would you change things? So

46:17

I think drug pricing is truly difficult. One thing I

46:19

guess to say about the US is the US has

46:21

by far the highest prices in the world. I think

46:23

people recognize that. It's about twice as high as Europe

46:26

on a net price basis. And the

46:28

US market for drugs is something like

46:30

40% of the global market and it's something like 60%

46:33

by revenue of newer drugs. So drugs

46:35

launched in the past 10 years or so. So

46:37

the US, it's kind of where outside share of

46:40

the revenue that drug companies make. And part of

46:42

that is because the US system is, for the

46:44

most part, some of this is changing, but it

46:46

is mostly a free pricing system where you can charge

46:48

whatever you want. So when you're pricing

46:50

a drug for the US market, you'll often, as

46:52

a company, just try to charge whatever you think

46:54

you could get away with, what if you think the market

46:56

will bear. And in a reasonable world where

46:58

all the incentives are aligned, what the

47:00

market will bear is close to the actual

47:03

value of that drug. In healthcare,

47:05

you have all these strange incentives in markets that don't

47:07

function properly. But in the rest of the world, you

47:09

have systems where you

47:12

have governments doing the

47:14

negotiation on behalf of the population.

47:16

So they'll make assessments of the value

47:18

of a drug based on metrics like how

47:20

many quality adjusted life years does this drug

47:22

give us, what's the benefit of this drug

47:24

over the existing standard of care, and how

47:26

much better is it, and how much for

47:28

price premium can we give it on the

47:30

existing drugs. So both systems have flaws. I

47:32

don't think it's possible to find a perfect

47:34

system to satisfy everyone. If you look

47:37

at the profit margins of drug makers, they make

47:39

something like 10% to 20% profit

47:41

after you take out the cost. And the growth margins are something like

47:43

80%. Really,

47:46

that's a distorted picture of the

47:48

actual profitability of

47:51

drug companies because the distortions between

47:53

when R&D expenses are paid and when the drug revenue comes

47:56

in. So it's not necessarily a good picture of how much

47:58

of a profit you take out of the drug. drug

48:00

companies getting for their investment. And if you look at

48:02

how much drug companies are getting for their investment, actually,

48:05

most of them are pretty near zero or very

48:08

low return on invested capital. The

48:10

industry as a whole is not that great of a business. It's

48:12

very much like a lottery model that

48:14

is just type of lottery that's

48:16

attractive to people with biomedical PhDs,

48:18

where most people are not

48:21

making that much money, but

48:23

a small number of drugs and companies are

48:25

making supernormal profits. And you can

48:27

point to the examples of people

48:29

who have very high prices and making supernormal

48:31

profits like Humira, 20 billion seems

48:34

like an incredible amount of money for a drug

48:36

to make. And I think if you

48:38

look at that isolated example, you can say,

48:40

just seems unreasonable that anyone is making 20

48:42

billion off this drug per year. It's way

48:45

more than what someone should reasonably

48:47

get for producing this drug. But you

48:49

have to think more about the system

48:51

of incentives. You want to be worth

48:53

investing in developing drugs. And so the

48:55

incentive needs to be very strong. And

48:57

because such a small number of drugs

48:59

produce so much of the revenue, like

49:02

a real Pareto distribution in revenue,

49:04

you need these lottery winners

49:07

to make it worthwhile investing in the

49:09

system, which is an

49:11

unfortunate reality that because drug development

49:13

is so inefficient, you need high

49:15

prices. And you need these

49:17

superblockbusters to make the economics work out.

49:19

When you're doing R&D, you're paying money

49:22

that you have now for revenues in

49:24

10 years. So from a temporarily discounted

49:26

point of view, you need to think

49:28

you're going to get a really huge

49:30

amount of money in the future

49:32

to be worthwhile you paying hundreds of millions of dollars

49:34

in the next few years. I think

49:37

it's a really difficult problem. If you put

49:39

pressure too much on drug prices, you can

49:41

very easily remove the incentives people to invest

49:43

in the industry. Quite a fragile ecosystem that's

49:45

built up once you start cutting down the

49:47

tall poppies, you actually

49:50

are removing a lot of

49:52

the incentives to even develop any

49:54

drugs. And there's just a few things that

49:56

you can do outside of damage by cutting

49:58

down even just a small

50:00

number of these huge lottery winners,

50:02

if you like. You have to accept a bit of

50:05

a trade-off where you are trading off

50:07

innovation for how much you're spending on

50:09

drugs. What about prevention?

50:12

We've talked entirely about

50:14

interventions where something bad happens

50:16

in the body and we've

50:18

developed ways of treating that thing or making

50:20

it better. What about similar

50:23

research that could go into, whether

50:25

it's lifestyle or other things that we

50:27

do before these bad things happen to

50:30

us that would prevent them from happening

50:32

in the first place? Any thoughts

50:34

on that side of the ledger? Yeah,

50:36

I think prevention is difficult. People

50:38

in the US often say that the

50:40

reason why the US doesn't invest in

50:42

prevention is because a lot of people

50:44

are on these employee insurance plans and

50:47

there's a lot of insurance plan turnover.

50:50

So any insurance company that invests in

50:52

prevention for its members is unlikely to

50:54

reap the benefits of those

50:56

investments when they switch to another employer and

50:58

get a different plan. But the

51:01

problem is that if you look at other countries

51:03

that do have these nationalized healthcare systems like the

51:05

NHS, other European countries, they actually don't

51:07

invest in that much in prevention either

51:09

even though they should in theory because it's a

51:12

whole nationalized health system. They should be

51:14

wanting to invest in those things. So is the

51:16

problem really that there's a lack of supply of

51:19

preventative treatments or is it really there's a lack

51:21

of demand for preventative treatments? And

51:24

I feel like the issue is probably more

51:26

on the demand side where people unfortunately don't

51:29

have a strong demand for preventative

51:31

medicine and they don't take up

51:33

preventative medicine in many cases when

51:35

it's offered to them. So it's

51:37

the whole thing with the argument

51:39

about othempic and why don't

51:41

people just practice diet and exercise. That's

51:44

a more sustainable way of dealing with obesity. The

51:46

reality is that people just don't do that. So

51:48

they would rather just take othempic when

51:50

they are obese and have that treat

51:52

their obesity. So I think this whole

51:55

consumer attitude shift that needs to happen

51:57

before preventative medicine ever becomes really established

51:59

needs to be demanded. So you think

52:01

about like the healthcare system, who is really the

52:03

end consumer of pharmaceuticals and healthcare system? I think

52:05

you can make the argument that it's actually healthy

52:08

people who are enrolling in the plans and are

52:10

paying the majority of the money that goes into

52:12

the system. And they want that

52:14

system to represent value for money for when they

52:16

do get sick. So you have to have

52:19

the people who are healthy have

52:22

a greater demand for products of

52:24

prevention that then incentivize companies

52:26

to invest in them. And that's a really important

52:28

thing to do is to provide the services. I'd love

52:30

you to talk about two extremes. The

52:32

things that have you in this entire

52:34

world, the most excited for the future,

52:36

and the things that have you the

52:38

most worried. I think what I'm

52:40

most excited about is not a specific technology,

52:43

but more this Cambrian explosion

52:46

if you like those modalities

52:48

and biotech. So

52:50

for a while, I think

52:52

back maybe 10, 20 years ago,

52:54

there were that many different types of different treatment

52:57

classes. And since the rise

52:59

of biotech, you have all these different ways

53:01

of treating disease that are becoming established and

53:04

have a lot of potential. So you

53:06

have CRISPR, RNA interference, these CAR T

53:08

cells we talked about, base editing, different

53:10

variations of CRISPR. Our ability to control

53:13

our biology is getting much

53:15

better than it was even just a

53:18

few decades ago. So

53:20

this increase in our ability to control

53:22

our own biology and intervene in biological

53:24

systems and very precisely

53:26

use these molecular scaffolds to push

53:28

a system into a desirable state

53:31

is really interesting. And just

53:33

this flywheel of improving on the

53:35

modalities that we do have. So

53:38

you look at something like CRISPR, we just got our

53:40

first CRISPR-approved therapy in the UK and the US. But

53:43

CRISPR is already in some ways becoming a bit of an

53:45

outdated technology. You have a lot of investment in what's next.

53:47

So instead of making cuts, you're going to have these

53:50

base editors that are changing specific

53:52

letters in the DNA sequence. And

53:55

that's a more effective way of precisely treating

53:57

certain genetic diseases than making cuts at CRISPR.

54:00

cuts. And then maybe after these base editors you

54:02

have prime editing, it's even more versatile in the

54:04

type of edits it can make. And you have

54:06

different types of editing. So just a lot of improvements

54:09

in tooling for how we intervene in

54:11

biological systems to treat diseases and

54:14

seemingly like an increase in the rate

54:16

at which we're developing these tools and

54:18

applying them to the clinic to solve

54:20

biological problems and to treat diseases. Like

54:23

general explosion in technology, our ability to

54:25

control systems is really interesting. I'm interested

54:27

in just general new business models of

54:30

the industry. So one

54:32

trend you've seen over the past, I

54:35

think it started probably in the 2000s,

54:37

is this greater externalization research.

54:40

So pharma companies, big pharma companies used to do

54:42

a lot of research in-house. They've

54:44

mostly externalized that to smaller biotechs

54:47

that are venture capital funded. So

54:49

the big pharma companies are really

54:51

just like commercialization machines all the time. And

54:53

they do maintain some research labs, but the

54:55

purpose of the research labs is in many

54:57

cases just to validate external opportunities and test

54:59

them in-house and have the expertise to actually

55:01

meaningfully evaluate these external opportunities they're going to

55:04

bring in. Maybe you'll see greater and greater

55:06

externalization of more and more functions. Now

55:08

we have innovation R&D externalized to

55:10

little biotechs. We have running

55:13

clinical trials being externalized to these

55:15

clinical research organizations that

55:17

specialize in that. You can argue whether that's been

55:19

a good thing or not, but it's just a

55:21

hard externalization trend and you have more and more

55:23

just pieces being externalized. Every commercial analysis

55:25

will be externalized in the future as

55:28

well. So you may end up with

55:30

pharma companies just being very specific IP

55:32

holding companies, plus just

55:34

managing the finances of actually selling and distributing

55:36

these drugs. Every little piece of

55:38

the ecosystem is focused on some specific component

55:40

of the process and maybe that

55:42

will help to be more efficient. So I'm interested in

55:45

that. I think you can argue that it

55:47

may not be helpful. Externalization hasn't been completely helpful.

55:49

There's been some trade-offs there. But there's

55:51

a lot of redundancy in the industry. And if

55:54

you're in a biotech who's getting ready to

55:56

launch, people having to build up redundant capacity

55:58

to run and execute trials in an efficient

56:00

way. And then what I'm worried about is

56:03

there's a genuine worry that it's

56:05

not going to be worthwhile

56:07

for a lot of companies to continue

56:10

to invest in developing new drugs. So

56:12

I'm worried that if we don't get

56:14

the balance, rewards innovation and incentives to

56:17

innovation, we'll end up with a system

56:19

where companies will decide, okay, I'm not

56:21

actually incentivized to really invest in doing

56:24

meaningful fundamental research and spending 30 years

56:27

tinkering on some opportunity that may

56:29

eventually bear fruit. I'm actually just

56:31

going to market the drugs I already have

56:33

that are old. I'm going to invest in

56:35

drugs that are very hard to copy. So

56:38

even though once they do go generic, no one's going

56:40

to be able to copy me. So things like these

56:42

radio, radio fair treats, talk about Cartes, they

56:45

have a whole process of sojourn with them, so they're

56:47

quite hard to copy. And then you're just going to

56:49

sit on existing treatments and try to milk

56:51

them as much as possible and use

56:53

tricks and techniques to extend the patent life

56:56

and make it hard as a copy and

56:58

try and extract as much value for as long as possible. And

57:00

maybe not invest in this fundamental

57:03

research that really drives meaningful products

57:05

and has this element of unpredictability to it.

57:08

One thing I think is quite dangerous in

57:10

the pharma industry is just this over-reliance on

57:12

an accounting type mindset. Rather than a recognition

57:15

that there's a science-driven industry, there's

57:17

an innovation-driven industry, the commercial

57:20

parts of big pharma should be in

57:22

service to the R&D portion of the

57:24

organization or the ecosystem. And

57:27

you should really be hiring good

57:29

people with good intuitions about what's worth

57:31

developing and letting those scientists tinker

57:33

for as long as they need to on some

57:36

of these really challenging ideas at the forefront of

57:38

what is possible and give them enough time, 10 or 20

57:40

years, for these ideas to mature into

57:43

actual products and then you can commercialize them.

57:45

And if the returns innovation are down too

57:47

much and maybe you get too many fully

57:49

managerial positions who are focused on the accounting

57:51

aspect of it, there's one of milk-existing products

57:53

and copy the big ones that we don't

57:55

have an existing market, then you're not going

57:57

to get a kind of meaningful process like...

58:00

like Keytruda, these immuno-oncology drugs, the GLP-1

58:02

agonists that we're seeing now. So

58:04

yeah, that's one thing I worry about. So just a

58:06

totally fascinating overview of one of the most

58:08

interesting parts of the world and of the

58:10

business world, of just the technology and innovation

58:13

world. I've so appreciated everything you've written and

58:15

sharing so much here with us today. In

58:18

these interviews, I always ask the same traditional closing question.

58:20

What is the kindest thing that anyone's ever done for

58:22

you? I think this has a

58:24

pretty obvious answer in my case. Many

58:26

people have done many kind things for me, right?

58:28

I think I just have to go with probably

58:30

what is the most common answer, which is my

58:33

parents have always been an unending well of support.

58:35

And they bailed me out of many low points

58:37

in my life. So I'm very thankful for that.

58:39

And now while other people have done me many

58:41

kindnesses, nothing really compares to what

58:43

my parents have done. Alex, thank you

58:45

so much for your time. Thanks. If

58:49

you enjoyed this episode, check out

58:51

joincolossus.com. There you'll find every episode

58:53

of this podcast complete with transcripts, show notes,

58:56

and resources to keep learning. You can also

58:58

sign up for our newsletter, Colossus Weekly, where

59:00

we condense episodes to the big ideas, quotations,

59:02

and more, as well as share the best

59:04

content we find on the internet every week.

59:28

Thank you.

Unlock more with Podchaser Pro

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