The Peter Attia Drive - #175 - Matt Kaeberlein, Ph.D.: The biology of aging, rapamycin, and other interventions that target the aging process
Episode Date: September 13, 2021Matt Kaeberlein is globally recognized for his research on the biology of aging and is a previous guest on The Drive. In this episode, Matt defines aging, the relationship between aging, chronic infla...mmation, and the immune system, and talks extensively about the most exciting molecules for extending lifespan. He discusses the current state of the literature of testing rapamycin (and rapalogs) in animals and humans, including Matt’s Dog Aging Project, and provides insights into how we can improve future trials by conceptualizing risk, choosing better endpoints, and working with regulators to approve such trials. He also examines the connection between aging and periodontal disease, biomarkers of aging, and epigenetic clocks. Finally, they explore some of the biological pathways involved in aging, including mTOR and its complexes, sirtuins, NAD, and NAD precursors. We discuss: The various definitions of aging [3:25]; The relationship between disease and the biology of aging [16:15]; Potential for lifespan extension when targeting diseases compared to targeting biological aging [22:45]; Rapamycin as a longevity agent and the challenges of targeting the biology of aging with molecules [32:45]; Human studies using rapalogs for enhanced immune function [39:30]; The role of inflammation in functional declines and diseases of aging [50:45]; Study showing rapalogs may improve the immune response to a vaccine [56:15]; Roadblocks to studying gero-protective molecules in humans [1:01:30]; Potential benefits of rapamycin for age-related diseases—periodontal, reproductive function, and more [1:12:15]; Debating the ideal length and frequency of rapamycin treatment for various indications like inflammation and longevity [1:21:30]; Biomarkers of aging and epigenetic clocks [1:29:15]; Prospects of a test that could calculate biological age [1:37:45]; The Dog Aging Project testing rapamycin in pet dogs [1:42:30]; The role of the mTOR complexes [1:58:30]; mTor inhibitor called Torin2, mitochondrial disease and other potential pathways [2:09:45]; Catalytic inhibitors, sirtuins, and NAD [2:19:15]; NAD precursors: help or hype? [2:28:15]; and More. Learn more: https://peterattiamd.com/ Show notes page for this episode: https://peterattiamd.com/MattKaeberlein2 Subscribe to receive exclusive subscriber-only content: https://peterattiamd.com/subscribe/ Sign up to receive Peter's email newsletter: https://peterattiamd.com/newsletter/ Connect with Peter on Facebook | Twitter | Instagram. Â
Transcript
Discussion (0)
Hey everyone, welcome to the Drive Podcast. I'm your host, Peter Atia. This podcast, my
website, and my weekly newsletter, I'll focus on the goal of translating the science
of longevity into something accessible for everyone. Our goal is to provide the best
content and health and wellness. Full stop, and we've assembled a great team of analysts to make this happen.
If you enjoy this podcast, we've created a membership program that brings you far more
in-depth content if you want to take your knowledge of this space to the next level.
At the end of this episode, I'll explain what those benefits are, or if you want to learn
more now, head over to peteratia MD dot com forward slash subscribe.
Now without further delay, here's today's episode.
My returning guest this week is Matt Kiberland. Matt is recognized globally for his research
on the basic biology of aging, and Matt is clearly for me on the short list of people who I always reach out to when I have
questions about aging.
I consider him an amazing mentor and an amazing scientist.
Matt was a previous guest way back in episode number 10, circa mid 2018, when we dove deep
into his background and his interest in aging in the origins of the dog aging project,
which uses wrap up mice and to study companion
dogs. In this episode, we pick up that baton and go even deeper, but we also take an even
broader look at aging. Arguably the broadest look I've taken in any podcast because we
really start from the nine hallmarks of aging and talk about each of them. And then we tie
it into a framework for how do you think about aging?
Sort of a top-down view, a disease-centric view,
or a bottom-up view through the lens of biology.
And we talk about some of the pros and cons of each of these.
We then pivot our discussion to talking about rapamycin,
Matt, and I both speak very openly
about our personal use of rapappamysin and all
the trials both in animals and humans that have led us to our conclusions.
We then return to the Dog Aging project where Matt provides an update on some of the exciting
work that's being done there and some of the exciting work that's being done elsewhere
in his institution with respect to Rappamysin trials humans, and what they can be targeting in the short term
to better understand the impact of these drugs
and drugs like rapamycin, rapolog, so to speak,
how they can impact humans in a long-term way,
even though we can't really study humans in a long-term way.
We also discuss a relatively new molecule
that many of you may not have heard of called Torin II,
which is like
RAPA mice and another inhibitor of MTOR, but one that does so via a slightly
different mechanism. It looks quite promising and we discuss what that may or
may not imply. And we finish up our discussion with a another deep dive into
SirTuin's NAD, NR, and NMN, the precursors to NAD.
So I think if you have any interest in the topic of aging,
you're gonna find this episode probably riveting,
if not at least half as riveting,
as I did, which is still to say very riveting.
So without further delay, please enjoy my conversation
with Matt Cable. Hey Matt, awesome to have you back on the show.
It's been a while because you were one of the very, very initial and original guests on
this podcast.
So, been looking forward to having you back from almost the moment we finished our first
discussion.
Yeah, thanks Peter.
It's great to be back.
Unfortunately, we can't see each other in person, but this is the next best thing.
Yeah, yeah, I know. We certainly will and get together in person, but this is the next best thing. Yeah, I know.
We certainly will and get together in person soon, I'm sure.
There are so many things I wanted to talk about.
And I think the last time we spoke,
we focused mostly on our mutual favorite drug,
rapamycin and our favorite pathway, the mTOR pathway.
And certainly for those listening,
I don't want them to think we're not going to touch on that today.
We absolutely are.
But I want to start even broader than that because so often you and I are having these discussions about
all things that pertain to aging. And I find you to be one of the most thoughtful people across the topic. So I
sort of want to start with these broader questions about aging. A lot of people have different definitions of aging,
and truthfully, I'm not sure I even know how to define aging sometimes. It depends, I guess,
on the context in which I'm asked, right? I think if my five-year-old asks me about aging,
I would come up with one answer, and if I'm giving a keynote talk and somebody asks me about aging,
I'd have to give a different one. So, what are some of the ways that you really describe aging? That's a really good question. And I think I would probably say the same thing
that you just said, which is that I'm not sure I have a great answer, and it changes depending
on the situation. I think in the past, I often gravitated towards sort of a molecular definition
of aging. So what are the types of damage that occur during aging?
What are the consequences of that damage?
In some ways, a hallmarks of aging framework view, right?
Where we know that things like mitochondrial dysfunction, telomere shortening, cellular
senescence, things like that happen during aging at the cellular and molecular level, and contribute to many of the functional declines and diseases that go along with aging.
And so, given my training in biochemistry and molecular biology,
that sort of is the natural place where I go when I think about what is aging.
I would say over the last several years,
I have developed, I think, a greater appreciation
for a functional definition of aging.
And so, as I start to think more and more about translation of interventions that seem
to affect the biological aging process and laboratory animals outside of the laboratory
into the clinic, I spend a lot of time thinking about, well, what are some of the functional changes that go along with aging?
And we know that across every organ system in the body, we see functional declines that
go along with aging.
And so I've started to think more about things like frailty as an important component of
defining aging from a biological perspective when we're talking
about having an impact on health and longevity.
So I think it really depends on the context.
I guess the other thing I would say is both of those definitions and the way I almost exclusively
think about aging unless somebody sort of forces me out of my box is from a biological perspective.
There are other aspects of aging that intersect with biological aging, right?
That are probably as important as what I think of as fundamental biology of aging to quality
of life.
So social aspects of aging, for example, are extremely important in people, especially
right?
For quality of life, as you get older, I don't tend to gravitate towards that kind of a definition
of aging, but I do recognize that it's important. I think I'm naturally always thinking about
the biology of aging, and as I already said, I tend to focus on the molecular mechanisms that
drive the biological changes that go along with aging. So let's look at the sort of two extreme views
within the biologic and then try to figure out
where disease fits in.
So on the one hand, you referred to the hallmarks of aging
and there's a very famous set of papers
that have laid these out.
And if the day comes that I can actually recite all of them,
I'll be very impressed.
I can, I always have to look at that.
I think I can probably get seven.
Yeah, yeah.
We can do like, who can name the most hallmarks of aging?
Right, right.
So DNA damage is one of them.
Cellular senescence is another stem cell.
Fatigue is another protein misfolding is another
telomere shortening is another mitochondrial dysfunction.
Is it my favorite?
Yep.
Did I mention nutrient sensing issues?
Deregulated nutrient sensing out.
Deregulated nutrient sensing.
So that's my seven.
What am I forgetting?
There's an infrasalular communication.
Dingo.
And what are we missing here?
I'm going to be really ashamed
if it's something that I study.
Yeah.
So we got eight out of nine.
That's not bad.
So you've got those. And then at the other side, you talked about, okay, let's talk phenotype.
Let's talk about the outward expression within the organism and frailties, I think, of fantastic
example. Where does disease fit into this, right? Because with aging comes more cancer, with aging,
comes more cardiovascular disease, with aging, comes more dementia. And you could argue that those are functional.
Maybe less so cancer,
but certainly cardiovascular disease
and dementia are very functional forms of decline,
but of course, at their root,
they have a very cellular component
and a very strong set of cellular contributions.
Do you think of disease as basically the bridge
or the link between these fundamental
cellular declines and the ultimate phenotypic declines?
No, I would not say they're a bridge. So I think that I actually personally tend to think
almost the other way around where I think that the functional declines that often proceed
over disease or clinical diagnosis of disease are probably
as important or more important from a quality of life perspective as we get older.
And you know, this may simply reflect the fact that I'm getting older and you know,
I've noticed some of these functional declines in myself, right?
And I think these functional declines happen, as I already said, in every organ, every
tissue.
We don't always recognize them as such unless we sit down and think about and try to list
out all of the ways that we have changed as we get older.
And those often happen far before you get diagnosed with any age-related disease, right?
So I'm pretty fortunate.
I turn 50 in February.
I don't have any age-related disease, at least that I've been diagnosed with.
Yet I have a multitude of functional declines,
which fortunately don't impact me severely,
but that I recognize I'm not,
I'm functionally impaired to where I was 25 years ago.
I think we, you know, it's just a fact,
any 50-year-old is.
And so I kind of think of those as the first things
that you can observe that happened during aging often way before you
get an age-related disease.
The other thing I would say about disease, so there's actually two things, two points
I want to make.
One is, I think it depends on the disease, but we really don't have a good understanding
of when the pathology of the disease is no longer normative aging.
And what I mean by that is, you know, we've got some understanding of the molecular cellular
mechanisms that drive biological aging,
that contribute in some way to our risk of developing
Alzheimer's disease, cardiovascular disease,
all of the age-related diseases that are major causes
of death and disability.
But in most of those cases, there comes a point
where the pathology of the disease is not necessarily
at a molecular mechanistic level and extension of aging biology.
It becomes something different. And I think that's really important to recognize, because one of the implications of that is that
an intervention that affects biological aging, let's just say rapamycin, We can discuss whether rapamycin is really affects biological
aging in people. I think that's still a little bit of an open question. Let's just say for the sake of
argument that it does, it's not clear that that same intervention is going to be effective once a
pathology progresses to the point that it's not the same mechanism anymore. So I think that's a
really important point that sometimes gets lost in this discussion of aging in disease. So let's actually double-click on that using A disease. So pick one of the big three.
Athrouschlorosis cancer dementia. I think cancer is a really good example, right? So we know
MTOR, which, and I'll go back to Rapa Mison in part because, you know, it's, again,
something I think a lot about, but it actually, it's a really good, I think, example in this specific case,
because we know that MTOR,
which is the target of rapamycin, right,
the protein that rapamycin inhibits,
plays this fundamental role in regulating cell division
and cell cycle, right?
So if you inhibit in a non-cancerous cell,
if you inhibit MTOR enough, you will stop the cell cycle.
The cell will stop dividing, right?
But there are mutations that can happen that lead to cancer that cause the cell to no longer
pay attention to the MTOR break, right?
And so once that's happened, if that's the type of cancer you have that no longer responds
to MTOR inhibition, RAPA-MISON won't do anything to cell cycle in that case.
So that's a really, I think specific example
that you can point to, there are, you know,
sort of an infinite number of other examples
that we could use, but that's a really nice one
because there, rapamycin will be quite effective
at preventing cancer before that mutation happens.
But after that mutation happens
and the cell's not responding to rapamycin anymore
because it doesn't sense since the emtorbake, it's completely ineffective, right?
So that, I think, is a case where the mechanisms have changed,
the mechanisms that are important for preventing cancer before that mutation occurred
are different from the mechanisms that might deal with that cancer after that mutation has occurred.
Yeah, it's funny. This is a little off topic, but I've often contemplated this question
in the context of nutrition. Because in as much as there's an optimal nutrition to prevent
a condition, it might not be the same as the optimal nutritional strategy to treat the disease
once it's present. An example of that in an extreme sense might be a ketogenic diet.
I happen to believe a ketogenic diet is probably the best treatment for someone with type
two diabetes.
Because of course, type two diabetes by its very definition is a carbohydrate intolerance
disorder.
So once a person has it, you pull out the carbohydrates completely and you let them heal,
you basically let them recover and regain their ability.
But, you know, and again, we've seen that people
who have been on a ketogenic diet
for a long enough period of time
can resume some amount of carbohydrate consumption,
provided their other factors are changing,
such as exercise.
Does that mean one needs to be on a ketogenic diet
to prevent diabetes?
No, I don't think so.
So it's a little bit of
the same idea, though it's still something that's unclear. One thing I want to go back to on the
disease front is, and I believe it was Cynthia Kenyon who spoke about this once. I think I read it
in a paper. Something to the effect of using a diseased-based definition for aging is she didn't use
the word totology, but she effectively said it is a bit of a
totology because at what point is a disease, a disease?
It's only a disease when some people have it and some people don't.
If everybody has cancer by a certain age, then it's normative aging to your
point. It's no longer a disease. And then we get into, well, what does it
mean that something like why do you know, point zero, zero, zero, four percent of the population live to be a hundred, they've
managed to not succumb to a disease by the age of a hundred. And what does that tell us
about their normative aging versus everybody else? All of this is to say, I literally still
don't think I understand what age it is, which is unfortunate given my line of work.
We have to just accept that it's extremely complicated, right?
And so you're never, I shouldn't say never, I don't think I will ever understand aging
fully, and I don't think the field will, at least in any time frame that I can expect
to experience, right?
But I also believe that we don't have to understand it fully to be able to have an impact on the biology of aging through interventions.
And that's kind of where I'm at. I feel like I've got a conceptual flavor for what aging is.
And I have a some information about what the molecular mechanisms are.
And it's enough information that I can come up with rational approaches
to target those mechanisms with the prediction that those approaches should have an impact
on health and longevity as animals and people get older. And then we have to test those predictions.
That's kind of the way I think about it. I do want to come back to one point, though, which I
also think is often underappreciated in this relationship between disease
and the biology of aging.
Sometimes people get into this debate about whether or not the biology of aging causes diseases
of aging, right?
So does the biology of aging cause Alzheimer's disease, cancer, cardiovascular disease, right?
People get into debates about that. And I
personally think the data are pretty good that these, that what I think of is
the biology of age, and the molecular mechanisms, the hallmarks, whatever you want
to call them, contribute in a causal way to your risk of developing diseases.
But I also think it doesn't matter. And this I think is really important. From
the perspective of what is the best strategy to keep people healthy or longer, it just doesn't matter whether aging causes disease or it creates a permissive physiological state for disease. Biological age is the single greatest risk factor for every major cause of death and
disability in developed countries.
That is just a fact.
And whether or not biological age causes those diseases or creates a physiological state
that allows those diseases to manifest themselves doesn't matter from the perspective of what
is the most effective way to prevent those diseases. And I think that's where this debate is counterproductive, right?
Should we call aging a disease, does aging cause disease?
I think that those are not the right questions to be asking in my view.
Yeah, let me see if you would agree with my assessment.
I think you would, but I'll tell you how I think about this problem clinically.
So let's use atherosclerosis as an example.
And I want to highlight what you just said in case the person watching this or
listening to this missed it in any way, shape or form.
When I used to give talks, I'd always lead with a question like this, what is
the greatest risk factor for atherosclerosis?
Hand will go up.
Yes, smoking.
Nope. Hand will go up. High blood pressure.osis. Hand will go up. Yes, smoking. No, hand will go up. High blood pressure. No, hand will go up if it's an especially area diet audience, APOB or LDLC. No, and they'll just
keep rattling on inflammation. No, no, no, no, no, no. The number one thing is age. Hands down.
Number one thing is age, hands down. You take a 70 year old person who has everything perfect about them and you compare them to
a 20 year old train wreck who has not a single thing that is in their favor.
There is no comparison about their 10 year mortality prediction.
The 70 year old is in hands down a worse shape.
So you can't undo that.
Now, here's how I think about this problem clinically.
Anthroschlorosis is a great example
because it's the disease that we understand
the most of the big three, right?
Which is not to say we understand it completely,
but we have a far better understanding of what its drivers are
and how to prevent it than we do cancer
and Alzheimer's disease.
Lowering APOB, pharmacologically, nutritionally, etc. is arguably the most important
strategy you have to reduce it, along with probably improving metabolic health. So those two
things, right? So regulating glucose, insulin lowering APOB, all of these things can be done through
lifestyle, through drugs, etc. can dramatically reduce a person's risk of atherosclerosis. None of
those things are necessarily directly targeting the nine hallmarks of aging.
I think indirectly they certainly do, but when you give somebody a PCSK9 inhibitor,
which specifically targets a protein that allows the body to clear more APOB particles out
of circulation, it is by no means targeting one of those nine pillars,
but it's having a measurable impact
on reducing their risk of disease.
And in the end, that's the part that I think is hard
for some people to understand
within the aging community,
is that you can still target metrics of a disease
specifically without going after a hallmark.
I don't disagree with you that I think that is a concept that sometimes is underappreciated,
especially within the aging community.
I also think it makes sense, right?
So even if the hallmarks, we focus so much on the hallmarks because it's easy, right?
I think there are some reasons why the hallmarks are incomplete, but let's just keep using that term,
sort of as a surrogate for the molecular mechanisms
of aging, right?
If we accept that the hallmarks are at some level preceding the damage, whatever that
is that's causing the disease, it makes sense that you could intervene, sort of, if we
think about it in an upstream downstream perspective, right, the hallmarks being upstream,
the disease being ultimately downstream.
It makes sense that you could intervene at the level of the hallmarks being upstream, the disease being ultimately downstream. It makes sense that you could intervene at the level of the hallmarks.
It also makes sense that you could intervene
in whatever the bridge is that's connecting the hallmarks
to the disease, you wouldn't necessarily impact
the biology of aging at all.
And this gets back to what I was talking about before,
which in for many diseases of aging,
there comes a point where the pathology of the disease
is not normative aging anymore.
And so you can quite successfully treat or cure a disease of aging in an individual without
impacting the biology of aging.
In fact, I would argue that's almost exclusively what is done clinically, right, is to try
to either alleviate the symptoms of the disease or cure the disease, there's starting
to be more on the preventative side, but I still think that lags far behind, you know,
waiting until people are sick and then trying to do something about their disease.
But you don't have to impact the biology of aging to be successful at any of those things.
I would just argue that impacting the biology of aging is going to be a much more effective
and efficient approach from sort of the overall health perspective.
I think the other point that's obvious but is important to make, you can be quite effective
at treating an age-related disease without actually targeting the hallmarks of the biology
of aging.
But I think an important point to make is that if you don't actually confront the biology
of aging, that
you're really only impacting that one disease, say it's cancer, you can cure somebody's
cancer, you can take the tumor out, right, and they can go on and live a normal life.
But you haven't done anything to the biology of aging.
If you don't confront the biology of aging, right, you still have all of these other functional
declines and diseases of aging that are increasing
essentially exponentially as you're getting older.
And so the effect on health and longevity is quite small.
In fact, I think J.L. Shansky was the person who originally kind of did the map here,
right?
And it's really very striking.
So if you consider a typical 50-year-old woman in the United States and you say, what
would happen if we had a cure to all cancers, right?
Every type of cancer, we had a cure today and we implemented that.
Life expectancy for a typical 50-year-old woman in the United States would go up about three years.
That's it. So we've won the war on cancer, we get a plus three on life expectancy.
It's about the same for curing heart disease.
If you cure both of those diseases, it's roughly additive.
It's, I think, about seven years.
So the impact on life expectancy is actually, from curing disease is actually quite small.
If you compare that to the impact of targeting biological aging,
which, again, I think we have to be honest, right?
This is hypothetical in humans at this point.
So all we can really do is extrapolate
from what is done in laboratory animals,
but it's pretty routine now.
There are, I don't know, maybe a dozen,
15 different interventions that can increase lifespan,
slow aging in mice by between 15 and 30%.
If we just extrapolate that to the human condition, the impact on life expectancy is about 20 years
with the added value that because you've sort of slowed all of the functional declines of aging
simultaneously, those extra a couple of decades are spent in relatively good health, right? So I think it's just important to appreciate the potential difference
in health and life expectancy from targeting aging as opposed to what we do right now,
which is trying to cure individual diseases. And I've sort of adopted the term, you know,
I think of that as 20th century medicine, the disease first approach. I would even say 19th
century medicine. We've been doing this I would even say 19th century medicine.
We've been doing this for a long time.
And I contrast that to what I think of as 21st century medicine, which is approaching
health and longevity from the perspective of the biology of aging.
And we're not there yet, right?
But I do believe that this is happening.
There is a transition occurring, a slow appreciation among the broader clinical community of the
potential of this kind of approach.
And I do believe that we will get there in this century.
So it is my hope and expectation that 21st century medicine will really become targeting
the biology of aging to enhance health and longevity, hopefully by much greater amount
than we're currently able to do. health and longevity, hopefully by much, much greater amount than
than we're currently able to do.
Yeah, so I agree with a lot of that.
The one thing I would throw a little bit of a question at is,
and I'd love to have Jay on the podcast, because I don't know that I fully agree with his analysis.
I agree with the spirit of his analysis, so I completely agree that disease-based
whack-a-mole is not going to be nearly as effective as targeting the basic biology of aging.
So completely agree with your points. I don't know that I agree with the magnitude, right? The diminimus magnitude of it, because I really think that J's analysis is focused on an independent look at each disease, whereas in reality, if you eliminated cardiovascular disease,
by definition, you have reduced inflammation significantly,
you've reduced the burden of microvascular disease significantly,
those are going to play into other diseases.
So, you know, his analysis is very actuarial,
but it's not actually very biological.
That's my opinion.
Yeah.
I think that's fair.
I think you could make the case at least for cancer that it may be closer to just what
the straight math would suggest.
Yeah.
Now with all of that said, I couldn't agree more with the macro point here, which is delaying chronic disease
is probably not going to be as effective
as targeting something at the foundation.
Now that said, let's take a look at near data.
So when you look at near-barzalized work with centenarians,
the overwhelming statement here is they get chronic diseases
later, they don't live longer with them. filming statement here is they get chronic diseases later.
They don't live longer with them.
And I remember really being struck by that,
that would not have been my null hypothesis going
into a study of near work and Tom Pearls as well,
between near and Tom, you really have
the greatest assessment of the centenarians
and their siblings.
And you realize that they don't seem to have magical protection from a disease once
they get it.
When they get cancer, they're just as hosed as the rest of us.
When they get heart disease, they're just as bad off as the rest of us.
But they get a phase shift.
They get a 20 year, 20, sometimes 30 year phase shift in when they're going to get the disease.
So how does that factor into my thinking clinically?
My factoring to that clinically is the sooner you begin prevention,
the more you can mimic the centenary.
What we don't fully understand is molecularly, why is that the case?
Right? We've identified some of their genes.
We know that they're going to be more likely to have
APOE2 versus APOE3 or APOE4. They're going to be less likely to have APOC3, high regulation versus
low. I mean, there's, you have a lot of genes that produce phenotypes that are favorable,
but when you bring it back to our nine hallmarks, by the way, the ninth hallmark is epigenetic. Of course. Oh my god. It's the epigenetic modulation.
Now, the other point, I guess, that we're both well aware of, but maybe for the listener, is
how difficult it is to study aging in humans, targeting the basic biology, because you don't get to do what our clinical trials apparatus is set up
to do, which is pick a disease, a very clear endpoint and target it. If your endpoint is
nutrient sensing, that's a tough clinical trial.
Yeah, I agree. And I think we should definitely come back to that point because it's clearly a
major focus of the field right now is how do we start to test some of these interventions
that we know work in laboratory animals in the clinic? That's a challenge. I want to
come back to your comment about centenarians, though, because I think this, again, is a
conceptual area where there's a lot of, I think, misunderstanding both among people in the
field and also lay people who pay attention to the field.
So you're right, I think, that the bulk of the evidence supports the idea that centenarians do not live longer with multiple age-related diseases,
but they have genetic risk factors or genetic variants that put them at lower risk, at least for the
major killers.
So, there is a genetic component to being a centenary, and it's not huge, but it's significant,
probably somewhere I think around 25, 30%.
And they tend to not have the high risk genetic variants for Alzheimer's disease, certain
types of cancer, heart disease, things like that.
And then there's a luck or environment or something else we don't understand, right?
That comes into play.
But people often look at that observation and make the assumption that we don't see variance
in things like M-tore or Sir Tunes, not that there aren't variants that haven't been
talked about, but that have strong effects.
So I think the question becomes, why don't we see variants in cert 6, or M-tor,
or pick your favorite longevity gene, Foxo, right?
That caused people to live to be 180 years old,
because we can do that magnitude of lifespan extension
in a mouse or a sea elegant.
And so they go from that, so that's an observation.
We haven't found those variants yet.
Although Foxo is one of the genes,
not Foxo 3A.
No, it's not your right.
It's a very weak effect, but it's clearly a higher amount of data.
Yeah, as it's said, right?
And as it's emperor, there's evidence for all of those things.
You can find genetic evidence,
but the effects are relatively small.
But then the interpretation that people make
is the reason we don't see those variants
is because humans are fundamentally different from mice
or whatever your favorite laboratory organism is,
and none of those things will work the same way in people.
Now, I can't prove that they will work the same way yet
for aging in people,
but that's a, that's not a logical sort of interpretation, right?
I would argue the reason you don't see strong effect variants in MTOR, for example,
in people is because we know that strong effect variants in MTOR are incompatible with life
and development. Even in mice, right? You make an MTOR knockout mouse, it's dead.
I'd say, you know, so I think the reason why you don't see these strong effect variants in
people is because there is such a strong selective pressure. Either you don't see these strong-effect variants in people is because there
is such a strong selective pressure.
Either they don't make it through gestation, they don't make it through development, or
they're sterile.
All of those are incompatible with evolutionary success.
That's my hypothesis for why we don't see these very strong-effect genetic variants in people
that lead to 160, 170-yearespans. Again, it's a
hypothesis, right? It's hard to know what the explanation is, but it certainly
fits with what we see in laboratory animals. Any of these mutants in mice that
have 30, 40% lifespan extension, I mean, they are all significantly defective in from an evolutionary perspective, right?
They would not be selectively advantageous mutations.
And so I think that's at least a consistent explanation
for why we don't see these,
but we would think of as slower aging variants pop up
in people.
The other point that's important to make as well, though,
is that that doesn't mean that we can't intervene
in these pathways to have an impact on health and longevity.
And for me, this has been probably the most exciting aspect in development in the field
over the last probably 15 years.
You know, if you asked me 15 years ago, whether I thought that it would be easy to slow
aging in an old mouse, I would have said no. I would have said you probably have to start
at six months of age and treat them all the way through life
to get the benefits.
And that was honestly, largely based on caloric restriction,
which that seems to be the case for caloric restriction.
You know, what has emerged now is there are five or six
or seven or maybe even more different interventions
that can be initiated in middle age or even in late age in mice.
And you actually go, it's not only that you slow down
the declines, you actually reverse the declines, right?
And again, I come back to Rapa Mison
because in pretty much every tissue
where this has been looked at, you take an old mouse,
you give it Rapa Mison functionally,
it's younger in that tissue or that organ.
And lifespan is extended.
And by the way, to your point, Matt,
I was having dinner with a friend last night
who asked me, why don't we start giving rap amicin
to children?
And I said, look, I think rap amicin
is the most important, you know,
geroprotective agent out there today.
But you absolutely,
you wouldn't want to give it to a developing child, right?
So he said, if you had to guess
when would be the right age to start? I said, I have no honest clue, but it wouldn't be developing child, right? So he said, if you had to guess, when would be the right age to start?
I said, I have no honest clue,
but it wouldn't be before 25, right?
It just wouldn't be before about the age of 25,
which speaks to your point,
any genetic manipulation, or in this case,
naturally acquired genetic variation
that mimicked rapamycin would probably not be selective,
because you wouldn't be able to turn it on and off the way you would need to with the drug.
That's right.
Yeah, that's right.
I think that's a really interesting question about wrap a mison and other interventions.
As more and more interventions start to get to the same level of confidence that we have
about wrap a mison, when is the optimal treatment of treatment period, right, to get the biggest benefits.
And I think it's not something that often gets talked about in that conversation, but it's
really a balancing act, right?
Any intervention is going to have some risk associated with it, right?
The risk can be low, the risk can be high, but there's always some risk of side effects.
And so the question of optimal treatment is a balance between the beneficial
effects that you get and the risk of detrimental effects, side effects. And so I think we
still don't, even with rap and mison, we still don't have a great feel for what that risk
looks like, right? And that's in part because we haven't had long-term control, clinical trials,
and multiple doses, multiple strategies of intermittent versus continuous treatment, things like that.
So we just don't know, right? And it's interesting in the case of Rapa Mison, because my impression is
that, you know, there are a group of people, I think you and I both know some of them, we might even
be among them, who, you know, have self-experimented, right?
And my impression is that there has been a movement towards the idea that once a week
dosing with rapamycin is probably better than daily dosing with rapamycin for aging
effects and people and that you might want to do it for three months and then you stop
for six months and you do it again for three months.
I think that's where people are starting to coalesce around this idea.
There's not a lot of data to support that, right? It's a guess.
So it's just interesting to see how that is evolving in the absence of long-term, large-scale, controlled clinical trials.
And this is getting to the point that you raised earlier,
which is it's really hard and expensive
to do long-term, controlled clinical trials
for aging in people.
We really, I think still, as a field,
don't have a great strategy for how to address that challenge
of actually trying to answer some of these questions
in the way that at least traditionally, we would want to see them answered in a clinical setting.
I mean, I think there's an even bigger problem that which is, I don't think there's a regulatory
appetite to even do what would be necessary because we have the regulatory appetite to say,
you take an individual who is either at very high risk for a disease or already has said
disease and we will go ahead and accept the risk
of a clinical trial to assess it.
But by definition, if you truly want to understand
how geroprotective, metformin, or rapamycin,
or canagaflasin, or pick your favorite molecule is,
you really need to be testing it in people
before they have a disease.
So there's really two fundamental problems.
This is one.
We do not have a regulatory environment that accepts that risk.
So I would love to see the IRB submission that says, we're going to test RAPA MISIN in
healthy 50-year-olds whose 10-year risk of death is less than 1%.
And then secondly, because we can't logistically follow those people for the next 50 years,
which is what would be necessary to understand its true magnitude of zero protection,
we would need really good biomarkers of aging, of which I will pause it and you may disagree.
We have somewhere between zero and epsilon of those.
I mean, we don't have dittily squat in the way of meaningful biomarkers of
aging. By the way, I want to go back to your point on RAPA. So as you do know, and I'm very
happy to talk about, I mean, I've been taking RAPA Mice in for two and a half years now, and
I am one of those people who has coalesced around once weekly dosing. I have fiddled a little
bit with my dose. I've varied it from as little as five to as many as eight milligrams
once a week. I've also done the cycling on and the cycling off,
but I am completely flying blind.
Most of my dosing and frequency data comes from
Joan Mannack and Lloyd Clixteens 2014 paper
with Everolamus, which of course is not the same as
Rapamysin, but it's a pretty reasonable hand-drawn
facsimile of it.
And it was from that study that I concluded that 20 milligrams versus 5 milligrams versus
1 milligram, your sweet spot was at about 5 from a side effect standpoint and an efficacy
standpoint.
And the daily dose of 1 milligram didn't seem to produce as good an effect as the 5
milligrams once a week.
But how does that stand in comparison
to what we learned from all of the ITP studies where it's continuous administration of rapamycin,
and to your point, because we'll come back to these and talk about them, remarkable results
regardless of when it's initiated. I don't know what to make of that.
I kind of agree with everything you said about why, you know,
if you're gonna pick one regimen for Appa Mison,
that makes sense.
And like you said, it's really based on two clinical trials.
I mean, really.
And so it's a limited data set,
but at least for immune function in people,
it seemed to work.
We could spend hours talking about this,
because that's a whole nother interesting
and informative
sort of experience was the failure of the phase three clinical trial at RestorBio.
And what happened there? And I don't think we actually, it's at least not in the,
it's not published yet. Well, let's let's talk about this because it is really interesting. So
let's back up for a moment and give people the context. So when Joan was at Novartis and both
Joan and Lloyd were at Novartis in 2014,
the Evalymas study was done. Do you want to just tell people really briefly what that study
looked at? So Evalymas is a derivative of rapamycin and biochemically, it has exactly the same
mechanism. So it's, at least I conceptually think about them as essentially the same molecule.
So anything that we see with Evalymas, we would expect to see with rapamycin.
And so, that was a phase 2 clinical trial in healthy older adults to look at whether or
not a six-week treatment with ever-alimus could boost vaccine response.
So, the ability of older adults to respond to an influenza vaccine.
And that actually was based on one of my favorite papers from Penn-Jeng's lab at the University
of Michigan who showed that in mice, I think it was four weeks of rapamice.
It was either four weeks or six weeks of rapamice and an old mice boosted the ability of a flu
vaccine to protect those mice against a lethal dose of influenza.
If anybody hasn't looked at that paper, I would just encourage you to go check it out.
I think it was a 2009 science signaling paper from Penn Jings Lab.
The tile has something about hematopoietic stem cells in rapamycin.
We'll link to it in the show notes for sure.
Okay, yeah, it's an amazing study, but my favorite experiment there was this experiment
where they took young and old mice and they gave them
influenza vaccine and then they waited two weeks and then they gave them a leaf, what would be a
lethal dose of influenza if they hadn't got the vaccine. And in the old group half the mice either
got rapamysin or they got a control. So the first thing that struck me about that experiment was if
you look at just the difference between young and old mice who got the vaccine, all of the young mice were protected. 100% vaccine response. I think it was about
65%. So two thirds of the old mice did not have a sufficient response to the vaccine that they
were protected against influenza. I don't think the numbers are exactly the same in people, but there is definitely that same trend where older adults tend to not respond to vaccines as robustly as younger adults.
Unless those old mice got Rapa Myson, if they got six weeks of Rapa Myson, they were 100% effective at responding to the vaccine. So at least for that measure of immune function,
RAPA mice and fully restored the immune system back to that of a youthful immune system. So that was the mouse data that sort of served as support for testing this in humans, in this clinical
trial that we're about to get to. And by the way, Matt, do you recall if the immune response
in question, i.e., the immune response that was better in the RAPA group versus
the non-RAPA group of older mice. Was this a B cell response, a memory B cell response,
or was it a T cell response?
I don't recall the data. I know they have a lot of data in there looking at the antibody
titers and different immune cell types and how they responded. As I said, the title of
that paper and the model that they proposed was that rapamysel
was acting at the level of hematopoietic stem cells to rejuvenate hematopoietic stem cell
function in some way.
I don't recall the details of the immunology.
I've tried multiple times throughout my career to become fluent in immunologist speak and
have failed miserably every time.
So that's a weakness of mine that will likely never be addressed adequately.
So I don't know the answer.
It's just got such amazing implications for what we're seeing with COVID vaccination,
because we are still in such a nascent stage of truly understanding.
I mean, we're seeing people who naturally acquired COVID who within six months have no more circulating
IgG.
And we have no clue if they still have immune function.
For example, do they still have memory B cells in their bone marrow that when challenged
will make antibodies?
Do they still have memory T cells that will show up and immediately respond?
And so we're actually involved in a study that's looking at that.
But my intuition is you can probably still have an immune response,
or an immune response that's ready to go absent circulating IGG.
It'd be interesting to see where a rapper plays.
Yeah, the one thing I will say about rapamysen and, and ever-alimus is that it,
it, it seems like it's not one simple
mechanism at play.
So Joan, for example, has published data that one of the effects of Evalymas and another
drug called RTB 101, which is also an emitor inhibitor, is to boost antiviral gene expression.
And we know also that rapamycin, at least in mice, and almost certainly in people, tamps
down on the sort of chronic sterile inflammation that goes along with aging through mechanisms
that still are being worked out.
So I think it's probably multiple things that are going on that can impact immune function
in different ways when you treat with rapamycin, especially in the context of an old animal or an old person.
So it's probably not going to be one mechanism is my guess.
So based on this study, so Joan and colleagues take a group of,
I believe, 65-year-olds, I think they're, you know,
320 of them, so divided into four groups of 80.
You've got a placebo group, one milligram a week group,
20 milligrams once a week
and five milligrams once a week, yeah?
That's right, yeah.
And so they did a six,
I think it was a six week treatment period.
And then they also waited, I think, two weeks.
And then they gave them a flu vaccine.
And what they showed,
I mean, obviously it's people,
so you can't do the mouse experiment
and then give those people a lethal dose of influenza.
So they were sort of stuck with looking at things
like antibody titers and to try to get an assessment
of did the people who got ever-alimus respond better
to the vaccine than the people
who got the vehicle control?
And it looked like they did.
And it was, you know, you can argue about the strength
of the data.
I think it was pretty clear that they had
at least at the five-meg once a week,
and I think the one-meg daily,
I can't remember, maybe it was the 20-meg once a week,
but the five-meg once a week look pretty convincing
that they had a better response to the vaccine.
I think the other thing that's really important
about that paper is that the incidence of side effects
was really
hardly different at all between the different Evalymas groups and the placebo, and particularly the 5-meg once a week, there really was no significant increase in adverse events in that group,
which gets back to how we kind of started on this tangent, which is that that's part of the reason
why I think people are moving towards the idea that you can still get efficacy from
once weekly dosing and that the side effects are reduced at once weekly dosing to essentially zero or close to zero.
That was the first phase to trial.
And before we leave this trial, Matt, I think it is important for someone listening to understand why people like you
and I and Dave Sabatini and all the people who live in EmTorland, I'm not putting myself
in the same categories as you guys, but you know what I mean, I'm an EmTor fanboy.
Why was that such an interesting and landmark paper?
Well I think it's because up until that point, the only human application for rapamycin was
immune suppression. Rapamycin is a drug I have known about forever
because as a surgical resident I was giving rapamycin to kidney transplant patients, heart transplant
patients, and liver transplant patients, along with a cocktail of cyclosporin, prednisone,
MMF, all of these other really nasty drugs. And it was one part of that in their ability to suppress
the immune system.
So, I think prior to that paper, you have the ITP, the first ITP that came out in 2009,
suggesting this is a remarkable tool for longevity, at least in the ITP mice.
But then on the other hand, you're saying, well, but it can't really work in humans,
because it's going to be really horrible
to the immune system.
So you had this sort of dialectical dilemma,
which all of a sudden it became,
well, maybe that's not the case.
So how do you reconcile the data that we saw
in Jones paper with the fact that
Rapa Mison is at least in theory
an immune suppressor as it pertains to organ transplantation? with the fact that rapamycin is at least in theory
an immune suppressor as it pertains to organ transplantation.
Yeah, and I think, you know, you're absolutely right.
That was one important aspect of that paper.
And I unfortunately think that the old view,
which in my room is the wrong view that rapamycin
is an immunosuppressant still is prevalent
in the clinical community.
I think most clinicians still think of rapamysin
and rapologs as a immune suppressant
because that's often how they're used clinically
to prevent organ transplant rejection.
So I think the simple answer is it's all about dose dummy.
I mean, we know every drug has a dose response, right?
And that you can get different effects,
different outcomes, different side effects
depending on the dose. And so, you know, different side effects depending on the dose.
And so, you know, when you back off on the dose of rapamycin or everalymus, in the context
of an aged physiology, so there's, you know, there's multiple things going on here in
that study.
They used lower doses, they tested once weekly dosing, and these are old people who show a functional
decline.
You're not going to see a functional improvement in vaccine response if you did this experiment
in young people, right?
You're only going to see it in the context of older people where they already have a functional
deficit.
And that's, again, it's an important conceptual point that I think often gets lost in these
discussions.
The other thing to appreciate about how rapacin has been used traditionally and how it was first approved
is these are people who had an organ transplant
taking high doses of the drug.
And also, I think always, I'm not an organ transplant position,
but my impression is these people are always taking
other immune suppressants in combination
with emtorenhibitors.
So in that context, yes, it does seem to be the case
that MTOR inhibitors can help reduce the chance
of organ rejection in transplant patients.
I don't, I could be wrong.
I think there's preclinical data.
I don't know of any strong clinical data
that by itself, rapamycin, even at higher doses, has substantial immune
suppressing effects.
It wouldn't shock me if it does at high doses, but I don't know that that's really ever
been shown clearly in healthy people taking higher doses of the drug.
Yeah, that's interesting.
I don't think I've ever gone back and looked at the FDA approval process in 99 because
obviously it was approved for that use,
and I think that's the only use that it's been approved for.
So I wonder what trials led to that approval,
whatever trials would have taken place in the mid 90s.
Yeah, another point that I think is worth making is,
we use, and I'm as guilty of this as anybody else,
we use sort of broad terms when we're talking
about the immune system suppression, you know, lower function.
We say the immune system doesn't function as well
and older people is compared to younger people,
which is true, but I think when we say doesn't function
as well, we tend to think of as it's functioning
at a lower level, right?
There's just less activation,
which actually isn't necessarily the case. In old mice, in old people, there's just less activation, which actually isn't necessarily the case.
In old mice, in old people, there's a lot of immune activation that shouldn't be happening,
right?
There's a lot of sterile inflammation that's occurring, so it's not necessarily that the
immune system is functioning less, it's functioning inappropriately.
And I think there's a ton of evidence that many of the benefits that we see in mice
from rapamycin occur because it tamps down on that sort of sterile inflammation that goes along
with aging, right? The inappropriate activation of the immune system. So in that sense, you could
think of it as an immune suppressant, but through mechanisms that I don't understand at all,
and I don't think anybody really understands, it seems to be more effective at targeting the bad part of the aberrant immune
response in an aged physiology, which might be the reason why you're able to then have more of
the good part. And here I'm, you know, clearly showing my ignorance of immunology because I'm
referring to it as bad and good, right?
But I think in some ways,
it's useful to kind of think about it at that level
because it's easier to appreciate that it's not,
you know, not immune function isn't intrinsically good or bad,
right? It is.
And there are different types of immune responses.
And if you get the wrong type of the immune response
at the wrong time, that's bad. If you get the wrong type of the immune response at the wrong time, that's
bad.
If you get the right type of immune response when you need it, that's good, right?
And I think in the context of aging, we just see a lot more of the bad and probably a decline
in function of the good.
And I don't know if rapper Mison is affecting both of those, but I think it's definitely
affecting the bad and bringing it back down, which might just be enough to allow the good to come back up where it's supposed to.
Yeah, which, you know, look, I think that's a great point to make, and it's very similar
with reactive oxygen species, right?
I mean, you have to have them.
They are vital signaling molecules, and yet if they run a muck, they cause damage.
And so, yeah, a lot of biology is in the Goldilocks framework of not too much, not too little.
Can I just throw one more thing out there as well,
which is, and this is, again, I'll admit,
if you'd asked me 15 years ago,
like how important did I think that, you know,
chronic inflammation and immune function would be an aging?
I probably would have said, you know, I don't know,
but in my guess is it's not gonna be the major player, right?
Because, you know, my background came from working in yeast
and worms, which, you know, worms sort of have an immune system, but it's not
really, right? I would have pointed to things like translation and autophagy and mitochondria,
all of which are clearly important for affecting inflammation and immune function, but I wasn't
a bit, I wasn't really bullish on inflammation at that point, and I have become very bullish
on inflammation as, you know, critically important for many of the functional
declines and diseases of aging that we see in people. The point that I want to
make here is that maybe I shouldn't say every. All of the interventions that I
know about that I'm enthusiastic about translationally seem to hit
inflammation in mice. And they seem to tamp down on the chronic sterile
inflammatory signaling that we see go along with aging,
which makes sense.
It's encouraging that they all seem to have this shared mechanism.
But the flip side of that is people talk a lot about,
they're people are very excited now about, you know,
senilelletics or reprogramming now
is the new senilelletics, right?
It's the thing everybody's excited about.
I'm not sure that those are fundamentally different from
wrap a mison in terms of the way that they're working.
And, and I think we, obviously, we need to find out because, you know,
it would be nice to know whether combinations of these interventions are going
to do better than one alone.
But, but to me, that the underlying theme that seems to be similar about all of these things that work in
mice is if you look in tissues of aged mice at inflammatory cytokines, P16, P21, markers of
senescence, they seem to be tamped down by all of these interventions, which might explain the
functional improvements that we see from using
these interventions in age mice.
I think that's actually a really interesting point.
I would kind of say I'm in the same boat.
I have become probably more convinced at the importance of inflammation, certainly in Alzheimer's disease and certainly in atherosclerosis, because
I've seen enough people who either develop these conditions without otherwise clear reasons
for it and vice versa, right? People who do have other risk factors and don't go on to
develop, but have at least have a demonstrally low measured amount of inflammation.
Again, the question becomes, is there a direct target of inflammation? Or do you simply reduce inflammation by targeting these other mechanisms,
which we'll get to?
Let's go back to the everolimous progression.
You were just about to talk about RTB 101.
So what is that?
And how is that the next stage
in the evolution of this rapologue?
Right.
So there was the first phase two trial was done.
They hit their endpoint.
It looked like it worked.
And they didn't see any, I mean, this was a phase two trial.
So the important thing was they didn't really
see any substantial adverse effects as well, right?
So then for the next trial, they added in another drug
called RTV 101, which is one of these
drugs that hits multiple kinases in the cell.
So MTOR is a kinase, which means that the biochemical activity is to put phosphate groups on
other proteins.
But you can think of it as a signaling sort of activity.
MTOR senses the environment and regulates the output based on this signaling
function of being a kinase. This drug, RTV 101, inhibits MTOR, the catalytic activity of MTOR,
it also inhibits other kinases. So it's talked about as sort of a dual kinase inhibitor, but in reality,
it hits multiple kinases. So it's a dirtier drug. That's maybe the way to the point to make. It's a
dirtier drug than rapamycin, which has a very specific biochemical effect on M-tork.
And M-tork-1 specifically, which we haven't dove into the M-tork-1 versus M-tork-2 situation yet.
And was the rationale for using a dirtier drug simply to create a new drug, to have a molecule
that is novel and therefore protectable by IP, or was it designed
to be dirtier without thinking of it as being dirtier, but thinking of it as being sort
of pluripotent across several kinases?
I obviously wasn't involved in designing this trial, so I can't answer that question as
to what exactly their thought process was.
I think it is true that RTB 101 had and has a longer existing
patent life than ever alignment.
So there is that component, certainly, to moving a drug through the approval process.
And if you talk to Joan, she will tell you that they had unpublished data that RTB 101
at the doses that they were using in this trial appears to be specific for mTOR complex one like
RAPA MISON. So I think their thought process, I mean in reality it was
probably twofold, right? The IP life was greater and there's a biological
rationale for testing RTB 101 either alone or in combination with ever
alimus. And so that was really the design of the phase two trial. It was
structurally very similar to the first Phase II trial, right?
So you have older adults, same sort of age range.
I think they had to be 65.
They couldn't have a pre-existing age-related condition,
significant age-related condition.
They had a control, placebo arm, ever-alimus alone,
RTB-101 alone, and both.
And I'll be honest with you,
I don't remember the dosing on the RTV 101.
And so they looked at vaccine response.
And then they also looked at, in this study,
upper respiratory tract infections over the next season.
To look beyond just, you know,
if there's an impact of the M-Tore inhibitor
on immune function, is it specific to vaccines
or does it look like it's broadly boosting
in immune function? You know, you can look at the data and not be completely convinced,
but certainly for the Evalymas plus RTB 101 and the RTB 101 alone group, I think the
Evalymas alone group in this case didn't reach statistical significance, but for the combination
and the RTB-101 alone group,
they saw improved vaccine response, and I think what was really striking was a lower risk of upper
respiratory tract infection in the people who'd gotten the M-Torinhibitor over the next season.
So that suggested that not only is it boosting vaccine response, but it's also broadly
conferring protection against a variety of immune
challenges in this older population. And again, very little in the way of adverse events, which
which gets back to the point, you know, you hear people talk about how mTOR inhibitors, you know,
you could never use them for aging in people because the side effects are so bad. And you know,
it just gets frustrating over and over and over again to say, go read the
data. Go look at the data, right? That's just not true at the doses that have been tested
so far. That's just blatantly false, right? There's no evidence actually for significant
side effects from RAPA Mison Monotherapy, EverLimus Monotherapy, or RTB 101, at the doses
that people are talking about using
in this context.
Yeah, I mean, statins have far, far greater side effects
than rapamycin.
I mean, and it's not even close.
And I think statins are a very important type of drug,
but to think how ubiquitous they are,
and that we accept, hey, 10% of people,
I mean, I think the literature says 5%,
but really clinically, 10% of people have I mean, I think the literature says 5%, but really clinically, 10% of people
have debilitating muscle aches from statins
that render it impossible to take them.
Some people have elevated liver function tests
that are otherwise unexplained.
I mean, it is really frustrating for me to hear this,
especially when people talk about,
well, we could never really study rabomisin
because it's just too unsafe.
It's like, I'm not sure what date or you're pointing to, but when you have it, let me know.
Yeah, I agree.
Anyways, so that was the second phase 2 trial, which was successful.
Then they went on to a phase 3 trial to hopefully get FDA approval for improving vaccine response
and immune function in elderly people.
In that phase three trial,
I'm trying to remember what they called that trial.
I don't remember the trial,
but now what was Rad001?
What was that agent?
That's ever-alimus, that's the same drug.
That's what they were calling ever-alimus, okay.
Yes, yeah.
And they had a name for the all clinical trials have names,
so they had a name, I don't remember what it was.
But in that phase three clinical trial,
they only tested RTB 101.
So they took out the RAPILOG ever-alignness and only tested RTB 101.
So there was no Rad001 plus RTB 101?
That's right. No combination, no individual RAPILOG. That's right.
And that clinical trial failed to hit the endpoint point. And it was terminated halfway through.
So this is my understanding.
They were going to do one group in the fall season and one group in the spring season or
something like that.
And they got halfway through.
They weren't hitting their end points.
So they terminated the trial early.
Restor Bio was the company that was doing this clinical trial to try to move RTP 101 through to approval,
the board basically merged them with a Carti cell therapy cancer company and they gave
up on RTP 101 because of the failed clinical trial.
So why that clinical trial failed, I think still the data has not seen the light of day
yet.
I don't think it's come out yet.
So we don't know for sure what happened.
I think we can observe that the rapologue wasn't in there anymore, right?
That is one difference between the two successful trials and the one that failed.
And I believe Joan has talked about this publicly that they also, in conversations with the FDA,
were required to change the endpoint
from laboratory confirmed infections.
That was the endpoint, one of the endpoints in the phase two trial
to something else, which involved patient reported symptoms
as the endpoint, and they didn't hit it.
And so I think that, I think my understanding
from talking to Joan is that data will come out at some point
and, you know, we'll be able to really take a look and see what the drug was doing and what it wasn't doing
in that third clinical trial.
My intuition is that it probably worked and they probably got screwed by being forced
to change the endpoint of that clinical trial.
If that's the case, then I think we really do need to have a conversation around
the way that FDA, I mean, we need to have this conversation anyway, but the way that FDA is
approaching clinical trials in the aging space, what needs to be shown for an appropriate endpoint, and what is the acceptable level of risk when the goal of the
trial is to test whether or not an intervention is affecting a functional decline that goes
along with aging or aging itself, right?
And that's a bigger question.
I certainly have plenty of thoughts around that.
I know there are lots of people in the field who are thinking about this and working on
it and some have talked to FDA. It's a real challenge. I don't want to blame
anything on FDA, right? I think that they are the people at FDA want to do their jobs
to the best of their ability. I think there are constraints around the way that FDA is
required to work and the culture that does not, in my view, appropriately evaluate risk reward.
I think that there is a culture of the risk has to be extremely low in people who are of normal health status for their age.
I actually am trying very hard. I still slip.
But I'm trying very hard not to use the word healthy when we talk about
aging or older people. I'm relatively healthy for my age. I would say I'm probably in the top
10% health wise for my age group. I am not as healthy as I was 20 years ago. We already talked about
this, right? So I'm of, you know, normal to upper health status for my age, but I would not call a 65-year-old,
a typical 65 or 70-year-old healthy.
They're not.
They're functionally impaired.
And we really need, we being, you know, the regulators, society, policymakers, really need,
I think, to start taking a realistic look at what normal aging is.
It is a progressive chronic decline in function
that at some point will lead to overt disease
and with 100% probability will lead to death, right?
And so if we can intervene in that process
to slow it down or reverse it,
there should be some level of risk that is
acceptable for that potential outcome.
And I think that because we tend to think of 65, 70-year-olds as healthy as opposed to
functionally impaired, it makes it really hard to have a rational discussion on what the
appropriate level of risk is.
Matt, I think that is so astonishingly well said.
I've had so many discussions with near and to a lesser extent Steve Ostead about the
choice of metformin over rapamycin in tame.
It always comes back to this point, which is the regulators will absolutely positively
not consider rapamycin, which of course comes back to this question, which is,
what are we claiming to be studying and in whom?
The other thing is, the studies are being designed around disease, right?
Progression of disease or outcomes of disease.
So this really comes back to a broader theme, which is, where are we on the spectrum of understanding aging in the way
that you're defining it and getting it further away from the discrete definition that involves
a disease.
Because until we really get people to center around that, your very eloquent explanation
of there being no such thing as a healthy 70 year old
until we realize that at the regulatory level, right, at the policy level, even at the scientific
level, it's going to be very difficult to study the things that will have an opportunity
to give step function changes in longevity.
Let me start by pushing back on on what you said about
tame and metformin versus rapamycin because I think that's a myth that regulators would not
allow you to do a clinical trial with rapamycin, right? We just talked about three clinical trials
that were done with amtor inhibitors, two of which were with ever-alimus, which is essentially
rapamycin. So I think that is talked about as to why
Metformum was chosen for tame.
I don't think that is true,
and I don't actually think that is the reason at all.
I've pushed, by the way.
I've pushed.
I needle near constantly about it.
I mean, honestly, I think there are good reasons
why Metformum makes sense to test in that context.
And there's data in people that support that.
So I'm not trying to say they should have used Rapa Mison.
I think that people with expertise in the field
can come to differing opinion as to what the best
shot on goal is, I guess.
You're just saying, let's just say we pick Metformin
because of these reasons, but not because we thought
that Rapa wouldn't be safe enough,
or the regulators themselves would decline it.
Yeah, I think they'd let you do the trial.
Obviously, they would pay attention to adverse events,
and there would be concerns around adverse events,
but they would certainly let you do the trial,
and that's been proven, right?
I mean, Restor Bio did the trial.
And it wasn't because of adverse events
that it got shut down.
I think that's the other important point to make.
I think the regulators at FDA are doing the best job
that they can within the constraints
of how they are required to regulate drugs.
I have a real problem with the way we regulate drugs in this country, but I don't necessarily
layman on the people at FDA.
And I do think if you came to them with a clinical trial where you had an endpoint that was quantitative
and functional and related to quality of life in people, they would let you do that clinical trial with Rapa Mison.
I'm 100% certain of that.
The challenge is, I think the reason why this hasn't happened,
one, Rapa Mison's off patent, nobody's gonna make money off of it.
And two, there is a misplaced concern about side effects,
which we've already talked about.
It's just not reality that the risk is
significant at doses of rap and my son that would be tested. So I think the real challenge,
though, is identifying the right end points for a clinical trial in aging. You're not going to do
lifespan in people. We can do it in dogs. You're not going to do it in people. I think we just have
to accept that. So what is the, I don't even want to say the right approach, because I don't think there's one.
But what are some approaches that one might consider
if your goal is to move FDA towards approving a drug
in people of normal health status
to prevent age-related functional declines in disease,
to target aging.
That's what I mean when I say prevent
age-related functional declines in disease, to target aging. So if your goal is to get there, so what is that clinical
trial look like? So the tame trial is a specific example of one strategy, which is to take
people who already have one age-related disease and ask whether your intervention met
form and in this case can delay the onset of the second age-related disease. It's a comorbidity trial.
That makes a lot of sense because we know with pretty good precision how long that timeframe
is on average, from the first age-related disease to the second age-related disease.
And so you can quantitatively assess, does your intervention increase that length of time?
That's the rationale behind tame. In my personal view, the limitation of
that approach is it's not a true healthy aging study. It's not taking people who are of normal
health status for their age and asking whether the intervention can improve or extend the healthy
period of life. It's a different design, But I totally get why tame was designed that way.
And I like the design.
I think there's a reasonable chance it'll work.
But it's different from the way I would think about
a clinical trial in this space.
What I would do is I would try to identify
the best single endpoint or set of endpoints
that correspond to a significant functional deficit that impacts
quality of life in older people and assess whether or not my intervention improves that.
And optimally, that endpoint would have quantitative markers that you could measure to show that
you've improved it. So, RestorB bio went with the immune function and they went with vaccine
response initially and then ultimately I think in the phase three trial,
they were also looking at respiratory tract infections right over the next
season. That was their endpoint. It's quantitative. The problem is it's really
hard, right? It's a really noisy endpoint and I'm not at all criticizing them.
In fact, I think as you know, Joan Manack is one of my favorite people. She's a good friend. I have an amazing
amount of respect for her. I think they went for it. And for reasons that were
probably beyond their control, you know, that clinical trial failed. But I have so
much respect for what they tried to do. I'm just saying it's a tough clinical
trial. It's a tough endpoint. It's noisy. I think there are other endpoints that
you could consider that might not be other end points that you could consider
that might not be as noisy, that you could consider doing a clinical trial for. So my, one of my favorites at this, at this moment, is
periodontal disease. And that's because, you know, my lab has published that in, that aged mice get periodontal disease, that eight weeks of treatment with
rapamysin reverses the clinically defining features of periodontal disease and mice. We know that something like two thirds of older adults have periodontal disease
or we'll get it. And those who have periodontal disease are at higher risk for dementia, cardiovascular
disease, diabetes. So it's connected in some way to other age-related diseases.
And by the way, in a recent podcast of mine explored this topic and it may in fact be causally
related through that inflammatory access, right?
I mean, I think that that's probably the strongest line of evidence connecting oral disease
with systemic disease.
It's through this immune inflammatory pathway.
So I think that's actually an elegant approach.
So the reason why I like parodonal disease, right, is the endpoints are extremely quantitative,
right? So what we looked at in the mice are gingival inflammation, bone around the teeth,
which can be measured, you know, crudely by pocket depth, more quantitatively by x-ray,
and microbiome. That's really the, at least in my understanding. I'm not a dentist,
but I've learned a lot about oral health over the last few years. That's really the at least in my understanding. I'm not a dentist, but I've learned a lot about
oral health over the last few years. That's my understanding of, you know, if you have
gingable inflammation, you've lost enough bone around the teeth, you don't even have to have
the dysregulation of the oral microbiome, but it always goes along with it. You've got
periodontal disease, right? So there are nice quantitative endpoints that can be looked at
in people,
and it's extremely non-invasive.
The way that you do this clinical trial is you have people come in for a dental exam
before and after treatment.
So you've got a shot at seeing changes in quantitative endpoints
that we can not just delay but actually reverse the declines and mice in people in a reasonable
time frame. So you could easily imagine something similar to the structure of the Restor bio trial with
Rapa Mison or Everolimus or RTB101 or pick your favorite intervention where you treat people for
eight weeks, three months, they have a dental exam before, a dental exam after, a dental exam, six months later. And you just look and see what was the impact
of the intervention. So it's a pretty straightforward clinical trial. And I hope we will get this
off the ground. We're actually trying to get some funding to do a clinical trial now.
Inhumans are indoors. In people. Yeah. In people. And this is, this is really, the person
who deserves all the credit for this
crystallizing in my mind is a gentleman named Jonathan On who was a DDS PhD student. So he had already
got his dental degree. He did his PhD with me. And he, you know, before he came to my lab, he came to
me one day. I vividly remember this conversation because I had never thought about oral health. And he goes, you know, people get periodontal disease
when they get older in much the same way
that people get Alzheimer's disease or heart disease.
If you look at the risk profile as a function of age,
it looks strikingly similar to these other age-related diseases.
You know, what do you think maybe the biology of aging
is contributing?
And, you know, in hindsight, it's like, oh yeah, that makes a lot of sense. But I had never thought about that before. And so he
came to my lab and showed that we could do this in mice. We could, we could see age-related
parodontal disease. We could see bone loss around the teeth. We could see inflammation of
the gums. And that we could reverse that with rap and mice. And so he's actually now a
faculty member at the University of Washington. And he's really the one who's trying to push
this clinical trial forward. So, so I'm sort of peripherally involved, but John is really the guy in this space.
He's going to be a rock star.
And this would be a phase two.
Yes, right.
If it gets funded, I mean, we're just submitting the grant now.
So, I don't know, you know, it's very early, right?
What would be the budget for this study?
So it's going to be a three-year grant.
You know, I don't know.
I think it's RO1 size, a couple hundred thousand a year, three years. But I want to go back to something. If money were no object, what secondary endpoints
would you add to that study? In other words, if you could power this study to hit that as your
primary, but also go after multiple secondaries, let's just throw in immune function. There'd be no
reason not to repeat what was done with
Rad001. What else would you add to that? Right. So if you look at the mouse data, I mean, I think the mouse data is it's a reasonable place to start again Obviously mice are people. It's a reasonable place to start. So where does rapamysin
Reverse functional declines associated with aging hearing right? Yes. Wasn't there that study that just came out,
yeah, it just came out two weeks ago.
It reversed age-related hearing loss,
which that got me very excited.
Yeah, I agree.
So that, and that's again,
in very easy and quantitative endpoint, right?
It wouldn't be amazing if you could improve age-related
hearing loss with rapid mice.
And so that's a no-brainer.
I mean, function, I mean, I agree.
If money was no option, sure.
But even in the context of, let's say, the periodontal disease clinical trial that I talked about,
if you could also measure hearing in that clinical trial, right, it's a big, big bang for the
buck, right?
It costs you almost nothing, and you get potentially another endpoint that you could
hit on.
Where else?
So muscle function, there's pretty good evidence that at least rapid miceen can prevent sarcopenia. nothing and you get potentially another endpoint that you could hit on. Where else?
So muscle function, there's pretty good evidence that at least RAPA-MISON can prevent
sarcopenia.
I don't think there's a lot of data yet on improvements in muscle function, but you
could do things like, you know, grip strength, walk speed, things like that.
Cognitive function, that's hard.
Again, I put that in the sort of the same category as a immune function as an endpoint.
It's a tough endpoint, but it would certainly be interesting to look at cognitive function
in this elderly population of normal health status.
So some of these people are going to be on the road to dementia.
They won't have dementia yet because that's an entrance criteria to get into the trial.
Heart function is another place where we see reversal of age-related declines.
This would be hard.
This is a different clinical trial, but also potentially cool.
Reproductive function.
You wouldn't do that in that same patient population, probably.
But in mice, there's pretty good evidence that you can reverse, at least in females, reproductive
declines that go along with aging or at least delay them.
Let's just pause on that for a second Matt. That is staggering when you consider where we are today from a standpoint of reproductive
medicine.
Women are having children later and later and I can't tell you the number of just patients
in my practice, either male or female, for whom this isn't a top of mind priority.
It's simply unbelievable.
And to think that there would be a, you know, I never really had thought of this honestly,
because I don't think I paid attention to that subset of the rapa, mice, and literature.
If you were to, and again, this is guessing, but how much of an impact you think you could
have.
So if you had a woman who was 40 years old,
she's obviously still pre-menopausal, but her AMH is low, and or it's reasonable,
but she's got significant aneuploidy for the listener, meaning whenever her eggs are produced,
they don't evenly divide into the right number of chromosomes. So they don't show up with one of each chromosome.
And that's an enormous cause of infertility as a woman ages is this aneuploidy.
So they either omit a chromosome or include two, and those almost universally lead to an
early miscarriage.
Some of them like trisomy, 13 or 21 will make it.
Do you have a sense of what type of magnitude of an effect this could have?
I think the honest answer is no.
So I think there's a couple things to say on this.
One is there's not a lot.
There have been I think two or three studies in mice looking at this, right?
And so there's not a lot of data on magnitude of effect, even in mice.
So my guess is that in people,
so I don't really want to comment on magnitude of effect,
because I really don't know.
I think the way that you would design a clinical trial, though,
would be very similar to what we've talked about
with rap and mice and already,
with the immune function or parodonal disease.
You would take a woman premenopausal,
let's say early 40s,
a group of women and do eight weeks of treatment
and then a wash out, and then you could look at your endpoints
or if they are going through IVF, for example,
look and see whether or not you get an improvement
in outcome using that functional measure.
Now why the eight weeks?
Do we really believe that that would be a sufficient amount
of time? I mean, are we just coming up with eight weeks because that's where we saw the vaccine response
in the Evalymus trial?
Like, do we think we, you know, if you were shooting for the moon and cost for not an issue,
what would be the downside of longer treatment?
A couple of things.
So, one is increased risk of side effects, right?
I mean, I think that's the longer you go with any treatment, the more risk there is for
an adverse event, even though I think the risks are pretty low,
here's a really interesting piece of data that we don't have with the immune function studies,
and in mice or people. In all of those studies, the treatment was stopped, and then there was a
two-week or so period before the vaccine was given. So what was the rationale for that wash out?
I think it's because people think and thought
about rapamycin as an immune suppressant. So and this is where I was going with that. What we
don't know is if they were still taking the drug, would you have gotten the same effects for
immune function? I don't know. So it's an important question that I think we just don't know the answer
to, but the other risk is that at least for some of your functional measures, if it requires,
let's say, hyper activation of MTOR to get the response, you might impair that by doing
continued treatment with RAPA MISON.
And at least all of the data that I've seen, limited in people, extensive in mice, eight
weeks is enough to give you essentially that full benefit for whatever the functional output
is that you're looking at.
Maybe not for lifespan, right?
Maybe multiple eight week transient interventions would be better than one.
Only thing that I know of for data there is our study where we did three months of rapid
mice and treatment, between 20 and 23 months of age, and then let them go to the end of
life.
The magnitude of effect was pretty similar to what the ITP saw.
So it was reasonably close to continuing treatment.
But we don't know.
But eight weeks in mice is, you know,
what would that be?
There's a thing, yeah, I know where you're going with that,
right?
If you were to linearly extrapolate that,
that would be a few years of writing people.
And I agree with that.
I think that the immune trials that we've talked about
suggest that that might be long enough. But I agree. We don't know, we don't know, is the answer.
I'll just say, so I haven't talked about, you know, my experience with rap and my son,
and I really don't talk about this publicly, but I'll do it here.
So I've tried eight to ten week courses of rap and my son a couple of times.
The most recent time, it was because this was probably spring of 2019.
So I'm pretty active. I play softball in the spring, and I noticed this was probably spring of 2019.
So I'm pretty active.
I play softball in the spring.
And I noticed I had a lot of shoulder pain.
And by the end of the season, it was to the point where I couldn't throw a football.
Like I couldn't go play catch with my son.
Actually, that's one of the hardest personal sort of aging experiences I had was when we went
across the street to the park.
And I was going to play catch with my son.
And I couldn't do it because my shoulder hurt so much as my right shoulder.
And I thought I must have a rotator cuff tear. I went in to see the specialist. Finally got
diagnosed with frozen shoulder, which is inflammation of the shoulder capsule, which happens to people.
Some people as they get older, extremely painful, completely limited my range of motion.
And the doctor was like, well, I could give you a shot of cortisol, but I don't really recommend it. people as they get older, extremely painful, completely limited my range of motion.
The doctor was like, well, I could give you a shot of cortisol, but I don't really recommend
it.
That can, that can degrade the cartilage.
Really, there's not a lot you can do.
Some people, it goes away after a year, some people just, you just learn to live with
that.
I was pretty depressed by that diagnosis, right?
I was like, at least if it was a rotator cuff, I could get surgery, get it fixed.
So I go home and I'm sitting there and I'm thinking, and so I'm looking on the internet and I see, okay,
it's an inflammation of the shoulder caps,
so I'm thinking to myself, what do I know
that has anti-inflammatory effects,
in the context of aging, rapamycin.
So I got some rapamycin and I did eight weeks.
And I mean, again, placebo effect is real,
but this was so painful.
I don't believe it was placebo effect.
Within two weeks, I had probably half my range of motion back
within eight weeks.
I was back 195%.
Were you just dosing once a week?
Once a week, eight migs once a week.
And I want to be careful, because I want to say,
I'm not encouraging other people to go do this,
but I am a true believer after that experience, right?
I don't think it's placebo effect.
I don't see how it could be with how painful it was,
how real the limitations were on range of motion.
So that goes to this eight week question.
At least for that indication,
which I believe this was a real effect,
eight weeks was plenty, and it hasn't come back, right?
Which actually kind of makes sense with frozen shoulder,
I think when people recover from it,
it doesn't always come back.
So I think that again, a lot of these age-related conditions that are inflammatory driven, you can kind of reset that with an eight-week treatment and rapamysin.
I suspect senolytics, if we had good senolytics, would do pretty much the same thing.
Do you think that rapamycin is itself a
centelitic?
I don't think it's a centelitic in the sense that at least the classical way people
have thought about centeletics were kills the senescent cells.
I absolutely think it turns down the chronic inflammatory signaling that is driven by P-16,
P-21, NF-capably.
Yes, we see that in mice, multiple tissues, no question about it.
I still think the senescence field is a little bit messy
in the terminology.
I think a lot of what people call senescence
isn't truly being derived from senescent cells,
the way that we think about them.
It's P16, P21 mediated inflammatory cytokines, right?
Doesn't necessarily have to come from senescent cells. No question. Rappamysin shuts that off and it shuts it off within eight
weeks, at least in mice, in a lot of tissues. And do you think it's doing that independent
of what it's doing at the mTOR or do you think that it's doing that through mTOR?
I would be shocked if it's not doing that through mTOR. I don't know of any good evidence
that Rappamysin has off-target effects through M-Tor.
Any activity, yeah.
Okay.
Yeah.
So really it's these sasps getting whacked that is how it would act through via the Sinesan pathway
as opposed to targeting a Sinesan cell directly.
That's by the way, that's been my reading of the literature.
Would you agree with that?
I think that's right.
I think in some way it shuts off that those chronic inflammatory
signals. The secretories, yeah. Yeah. And maybe other stuff, right? I mean, we always look at the
SAF because that's what we know. I mean, I think, you know, this is natural in science. We look
under the lamp post. We measure what we know to measure, right? It would not shock me at all if
there are other things that go along with the canonical sasp, that
senolytics or rapamycin or chaloric restriction.
I think that's a big part of fasting.
Fasting does the same thing, right?
Tamps down on that chronic inflammatory signaling, maybe through mTOR, maybe through other
mechanisms.
So I think that what we know about is part of it.
It wouldn't shock me if there were things we don't know about yet that also are contributing.
Yeah, but but getting back to this this eight week, right? That's how we got started
So I tend to think based on my personal experience and the little bit of data from these two clinical trials that that's probably long enough for at least
Some endpoints that are driven primarily by immune dysregulation and chronic
inflammation. I think two and a half years ago when I started,
I was very strict about eight on six off, eight on six off,
eight on six off.
And I don't know, a little while ago,
I just sort of said, yeah, I'm just not coming off.
And I wish I had a biomarker to point to,
I wish I had some way of measuring whether this is the right thing to do or if you know eight four eight four eight four.
There's a symmetry to eight five because you'll get through exactly four cycles a year. Maybe I do eight five eight five eight five eight five.
But it really frustrates me that we don't have a biomarker for this.
Yeah, or or aging in general.
For aging general. Or aging in general.
So let's talk a little bit about that.
So what does that look like?
I mean, when I had Eileen White on the podcast,
gosh, it's been, whoo, maybe a year and a half now,
we had a really interesting discussion
about why we don't even have biomarkers for autophagy.
I mean, something that is so important,
and we can't measure it.
And this was important in the context of people who choose to
chlorically restrict or fast is fasting for a day long enough to
generate a meaningful amount of autophagy in a human, in a
mouse it clearly is, but in a human is it?
No idea.
It's two days, three days, seven days.
Seven days is almost assuredly enough.
It's a big difference between fasting for a day and
fasting for seven days.
Why don't we have biomarkers for that?
Why don't we have a biomarker that can assess
nutrient sensing better?
Why don't we have a, you know, I mean,
you could argue we have some biomarkers.
We can measure telomere length,
but you know, my feelings on this, Matt,
I'm in the camp that thinks measuring telomere length
is not helpful at all for aging.
And I think there's plenty of data to suggest
that while telomereK is a very important marker
of cellular division, it really speaks very little
about the organism's state of aging.
Despite the popularity of that biomarker,
even the epigenetic clocks, I don't find to be helpful.
I find them to be far too, and I'd like you to push back
on this if you feel as much,
but I've seen how
easily they can be manipulated by short-term interventions that don't seem biologically relevant.
I'll start with the epigenetic clock because everybody, that's a big area of interest in the field.
Let's explain to people what that is. Let's let's let's start from the beginning.
Assume people don't know what an epigenetic clock is.
The epigenetic clock refers to typically chemical marks on DNA that regulate gene expression, whether or not,
you know, the gene that is located at specific points in your genome gets turned on or off.
And what has been observed is that those marks change with age in pretty much every organism
where it's been studied, and that you can identify patterns of change at specific locations in the genome.
So specific changes in these chemical marks with age that correlate very strongly with chronological
age.
And so that has led to the idea that you can create clocks that look at specific changes
in chemical marks in the DNA, the genome, that are telling you something about how long
that organism has been alive.
And then what sort of has emerged from that,
there are two things that have emerged from that.
One is you may be able to use that chronological
aging clock to find individuals whose marks
don't fall on the line that you would expect it to fall on
based on their chronological
age. In other words, they have marks that make them look older or younger than their chronological
age says that they are. And so you would hypothesize that those individuals biologically, if those
marks are really reflecting biological age, might be aging more slowly or more quickly. And
what's been shown is that indeed, those individuals who tend
to be off the line, depending on whether they seem to be aging more slowly or more quickly,
are at lower or higher risk for specific diseases. So that adds some level of confidence that
this epigenetic clock, chronological epigenetic aging clock, is actually reflecting biological
age. And so the idea is maybe we can use that information to develop epigenetic clocks that will, in a
predictive way, tell you how old you are biologically.
So you can get tests now.
There are plenty of companies now that are selling these things where you can go buy your
epigenetic blood test.
Mostly this has been done in blood cells.
That's one limitation to think about is almost all of the literature in humans
is developed on epigenetic clocks from blood and it's and it's still I think a little bit of a question
even if this is reflecting biological age, it's the biological age of your blood
which may or may not reflect the biological age of your entire body. But you can buy tests now that based on your you give them some of your blood
they will tell you your
epigenetic
biological age or some number.
They're looking at PBMC, I assume.
I don't know, honestly.
I'm not, I'm not involved in any of this stuff.
So I don't, I don't know exactly what, yes, most of the studies that have been published
are PBMCs.
I don't, they may even have saliva tests now.
I don't know, honestly, how these commercial companies are doing it.
I mean, some of the clocks I've seen
where I've just immediately discounted them is
when some of their inputs are things like glucose level,
vitamin D level, which are things that vary so much
from day to day.
And by the way, are so easy to manipulate.
Like you can take a vitamin D supplement
or not take a vitamin D supplement.
You can have a high cortisol spike one morning
and your glucose is 110 versus have a good sleep the one morning and your glucose is 110 versus have a good sleep
the night before and your glucose is 95.
So something that's that malleable,
I just don't think makes sense
as an iron clad marker of true biological age.
Yeah, let me take a step back.
So the epigenic clocks are probably the one
that people talk about the most
and have gotten the most traction in the field.
And I guess I'm a little bit of a skeptic, but I believe these clocks, I believe the one that people talk about the most and have gotten the most traction in the field. And I guess I'm a little bit of a skeptic,
but I mean, I believe these clocks, I believe the data
and I believe that the correlations are extremely strong.
I'm a little bit worried still that there are
so many data points in the epigenome
that you can find a pattern that will fit anything
you go looking for.
So I'm a little bit worried about the dimensionality of the data
and whether or not it's pattern matching in some cases,
rather than it's really truly going to be a robust predictor
of biological age.
That probably reflects what is admittedly my limited understanding
of the mathematics behind a lot of the epigenetic clocks
that have been built.
So I don't view that as a strong criticism. It's just a personal sort of concern that I have.
But I think these clocks are telling us something.
So you're basically saying without doing the complex mathematics to correct
for so many, the multiple looks that you can take at the data, you could be tricked.
And I have not spent enough time looking at that either. I would like to have Steve Horvath on the podcast
at some point, because I think Steve could speak to that
probably better than anyone else.
And for sure.
But the other point I wanted to make is what you alluded to
is now what people are doing is going beyond
the epigenetic clocks to try to look at every possible thing
you could measure, sometimes combining that with the
epigenetic clock to build these super clocks
or multi-clox or multi or multi-omic clocks, right?
And I think there's huge power in that, but it also increases that dimensionality problem
that I just mentioned, because you know, all of the sudden now you've got, if you're doing
omics stuff, you've got tens of thousands of additional data points that you can measure
and you can fit a pattern where a lot of this, and I think even Steve and
other people who are in the epigenetic clock field would agree with this, where a lot of this has
yet to really mature is in getting us to biological explanations for what the patterns are telling us,
right? What genes are they that these marks are located at and are those in any way causal for biological
aging?
I think if you get to the point where you can understand mechanism, it's going to be much
more powerful.
I also think, though, this gets to the fundamental challenge with biomarkers.
I think this is where you're dissatisfied.
We have a lot of biomarkers of aging.
We just don't have any validated biomarkers of aging, right? And this has been a problem,
you know, since I was a graduate student, everybody's wanted biomarkers of aging. The NIA
had a huge program where they did all this funding to identify biomarkers of aging. I think
it was back in the 80s before my time, 90s maybe. You can identify all sorts of things that correlate with age.
How do you get to the point of convincing yourself first and other people second that these
things are actually telling you something about biological aging that can then be used
to understand whether an intervention is working, first of all, at the population level,
but ultimately where we want to get to
is at the individual level.
So what we all want is a test that you can take,
and you can fast,
you can do your fasting regimen,
you can do your rapamysin,
you can take metform and whatever,
and you can come back and find out,
is it working from this set of biomarkers?
And that's where we want to get to.
And we're not there yet.
I think everyone would agree.
I'm not sure when we're going to get there.
So who's the natural owner of getting there?
Because I had this discussion with Steve Austin recently
and he made the same point you did, which is,
look, the NIA tried to do this a long time ago
and tried, you know, validly, right?
They put a lot of money into it.
You could make the case that the technology simply
wasn't mature enough to do this.
30 years later, we have a lot more tools at our disposal.
You've got the entire world of omics
at your disposable, plus you've got machine learning,
plus, plus, plus, is there any reason
this couldn't be done today?
And if so, this strikes me as a project that's almost too big for academics,
because it's too disjointed.
But at the same time, it's not a particularly interesting commercial problem to solve,
because it's far too big an investment before you could get to why you would care about it.
A commercial problem is, give me a drug.
But I'm arguing, you can't develop a drug really well
without this. So who's, like there's a bit of a cart and a horse thing, which is someone's got
a pony up a lot of money to develop the foundation of a pyramid that will ultimately become a great
tool for drug discovery and a much more streamlined manner in which we could do
clinical trials around this. Yeah, I think the answer to your question is it depends on whether
you're talking about doing this preclinically or clinically, right? I actually think this is a
problem that is- To think it has to be done both? Well, eventually yes. It's a problem that can be
solved today preclinically. Like like there is no real barrier to doing what
you just said.
Multiomic analysis of aging in mice with interventions, applying machine learning to identify patterns
that predict the effect of interventions and individual outcomes for longevity.
You obviously have to think a little bit about, you know, what can you measure?
If you want to do this longitudinally, you can't kill the animals, right? So you could,
you're sort of restricted to blood. But so there are some practical aspects, but there's
no, there's no technical barrier to doing that now. Who should do it? Who might be doing it?
I mean, I think this would fall maybe in the realm of what Calico could do. They've got
the resources, they've got the expertise. Is there any evidence that Calico is interested in
this type of a problem? I think so. I don't know. I don't honestly don't know anything about the
inner workings of Calico these days. I think conceptually they are interested in multi-omics
signatures of different aging processes. I don't know if they've done this particular experiment. They certainly have the resources and expertise to do it.
They're not the only ones, but they're the first ones who come to mind and they
sort of fit this space between true academia and industry, right, where they're
kind of this interesting beast in the middle. So I think it could be done
preclinically and you could actually then once you let's just say you have this
test, right? You get to the end of day, you say, okay, these are the
most predictive, I don't know, whatever, 24 things that give you, you know, 95% confidence on
remaining life, whatever your endpoint is. Then you get that test and then you show whether
it works or not in an independent study. And if it does, I'd be pretty convinced, right?
If you can show me that you create this test and then you go do a separate experiment and
you can predict when the mice are six months old, how long they're going to live at an individual
level, I'm impressed.
And if you can show that this intervention, when you treat them, makes the signature go
in the way that you think it should go and you can predict they're going to live 30%
longer, I'm even more impressed.
I'll believe it at that point.
That's not easy, but I think it's doable.
I think we know enough now, and we've got enough things
that we could measure that you could certainly build the test,
and then whether it would work in the validation step or not,
I don't know, but I think you could probably get it to work.
You can't take exactly that same approach to people.
This gets back to that, you know, the same issue that we talked about as clinical trials,
right?
Takes a long time to do the validation step and know that you have actually changed
somebody's biological state so that as they get older, they are at lower risk for disease
and are likely to live some x percent longer.
So you're almost obligated to have some level, you have to have some level of faith in
the test at that point, right?
And I don't know, it's going to be different for everybody.
And I honestly don't know what the regulatory step has to be before you could convince regulators
that you can actually go out and tell the general public
that this test works.
Although I will say there are already people doing that and the regulators aren't doing
anything about it as far as I can tell.
Let's come back to your dogs.
We spoke about them at length three years ago when we sat down to talk about rapamycin.
But again, let's assume a clean slate and folks might not be familiar
with some of your work specifically around dogs and how working with companion dogs offers
many advantages over working in mice, beginning with some of the obvious, like they're far
more genetically similar to us, they live in our environment. And they also seem to die
of things that more closely replicate how we die.
They die of heart failure.
They die of cancer, but not the same type of cancer
that a mouse gets that's almost genetically predetermined.
Just to start from ground zero, we at the University
of Washington and Texas A&M and several other institutions
have a large project called the Dog Aging Project.
The goal of the Dog Aging Project is really to understand
the biology of aging in companion dogs or pet dogs.
Some people, when I say companion,
some people think I mean, like seeing eye dogs.
Seeing eye dogs, yeah, yeah.
Pet dogs.
And there are really two aspects of this project.
One is a large scale longitudinal study of aging, completely observational. The goal there is really just to understand what are
the most important genetic and environmental factors that influence healthy
aging in dogs. And part of that, this is getting back to the conversation we just
had, is to measure as much as we can about those dogs every year as they go through their lives in order to be able to
identify patterns that are associated with health outcomes during aging, lifespan, disease incidents.
So in some respects, it's a similar approach as to what you would do if you wanted to create
biomarkers of aging, right? So that's the longitudinal study of aging. The second goal, so one way to
think about that is that's to try to understand aging in dogs. The second goal, so one way to think about that is that's to try to understand aging
in dogs.
The second goal, and this is really where I'm, you know, focused largely, is to do something
about it.
So can we slow or reverse biological aging in pet dogs to increase healthy lifespan?
So that ultimately, I hope, will be a series of veterinary clinical trials to test interventions
to figure out can we slow aging, reduce disease, and increase lifespan in pet dogs.
The first clinical trial is with rapamycin.
And so that's our first shot on goal, but I hope it won't be the only one.
And so you talked about so why dogs, why pet dogs in particular.
And I think you hit on, I think most of them,
the most important reasons, right?
So they've got this really interesting and powerful genetic architecture.
We have a couple hundred purebred breeds of dogs,
which you can almost think of as inbred strains, right?
And then on top of that, we have this mixed breed population.
And that's coupled with phenotypic diversity.
So for almost any trait that you think about, dogs are more diverse than people are even.
Body sizes are really easy one.
Everybody can just think about the difference between a great day and a chihuahua.
So that combination of unique genetic architecture with phenotypic diversity is really powerful
for mapping genotype onto specific
traits.
And lifespan is another case where you have this strong diversity.
A great dain will grow old and die maybe in 8 to 10 years, whereas a chihuahua often will
live to be 16, 17 years old.
So we're talking 100% difference in lifespan.
So that's really powerful for mapping genotype onto lifespan.
Dogs share the human environment is another big one, right?
And that's, for me, one of the most important, because we cannot model that in the laboratory.
In fact, we do exactly the opposite.
We really try to limit variation in environment to extreme measures.
Dogs share our environment
with the exception of diet, share almost all aspects of the human environment. And so that's
a bridge in some ways between laboratory studies and human studies. And as I've already alluded
to, they age more rapidly than we do. Their lifespan is substantially shorter. We all are familiar
with the idea of one human
year is about seven dog years, right? That's just another way of saying dogs age about seven
times faster than people do. And we can talk about that. It's interesting. If you actually look
at the epigenetic clock, it's not a linear seven-time rate, but they, but I think it's close enough,
right? It's close enough. And as you suggested, they age very similarly to the way that people do.
They get essentially all of the same age-related diseases and they're all age-related.
And they show the same functional declines that people do.
So dogs get arthritis as they get older, right?
You know, an arthritis is, I guess it's a disease, but it also starts as a functional decline, right?
Anybody who's ever had an old dog, you will notice that your old dog doesn't move around as much, doesn't walk
as fast, so they are going through the same changes with aging at the functional level
that we are. Again, it just happens seven to ten times more quickly. So they're a very
powerful animal in which to understand aging and test interventions for that reason. And
you can do it in a time frame that's feasible.
I think we've talked about the challenges with doing clinical trial for lifespan in people.
Even if we believe Metformin is going to extend lifespan in people,
you're not going to do a clinical trial to prove that.
You can do that clinical trial in dogs.
And so we've designed the RAPA MISON, the test of R test of rapamycin in aging dogs or triad.
That's what we call our clinical trial.
We've designed triad so that we'll be powered,
statistically powered, to detect a 15% change in lifespan
within a three year window.
So we can do a three year clinical trial,
reasonable cohort sizes to see an effect on lifespan.
That's comparable to what rapamycin does in mice. That's a pretty high bar, Matt. Are you at all
worried you've underpowered that given that you only have, and they're being
administered the drug for the entire three years? Yes, I'm worried that the study
is underpowered. I will say from my experience now we've done two safety
clinical trials, and this is our big clinical trial.
So this is my third veterinary clinical trial. I've learned that there are many reasons to be
concerned when you do a clinical trial. Clinical trials are a lot of guesswork. You have to take an
educated guess about a lot of different things. You can't test every dose, you can't test every duration,
you know, you can't test an infinite number of study subjects.
So, I'm worried about this clinical trial for many reasons.
I think we've done the best that we can,
given what we know, and given the constraints that we have to work under
to give ourselves a reasonable shot.
15% might be a high bar,
but it's consistent with the lower end of what people see
in mice.
So the first study at the low dose of rapamysin in mice from the ITP had a 14% effect, I
think, in females and a 9% effect in males.
Subsequent studies at higher doses had larger effects.
So it's reasonable.
It's a reasonable place to start.
Do you expect to see a sex difference in dogs?
I don't know. I don't know.
I don't know.
So, people still don't completely understand why female mice seems to respond to a given
dose of rapamycin better than male mice.
It's correlated with blood levels, so I think the simple idea is that male mice either take
up rapamycin less effectively or break it down more quickly.
There's no evidence for that in dogs.
We will be measuring rapamycin levels in the dogs,
so we will see if there is a difference in blood concentration
in females versus males.
From the limited data that we've got so far,
we don't have any evidence that that's the case in dogs.
And I don't know of any evidence in people
that that's the case either.
So that might be a mouse specific thing.
I think there's this misperception that rapamysin works better in female mice than in male
mice.
That's not true.
At a given dose, especially the lower doses, we don't know if it's true, right?
Given dose in the food at lower doses, females show a bigger lifespan extension.
When you go to higher doses, the males catch up.
If you push it too far, you can actually find a dose where female mice, the lifespan
extension starts to go back the other direction and male mice actually get a bigger lifespan
extension.
I think it's more about effective concentration than it is of a male versus female.
Truly sex-specific response.
In the dogs, you give it daily in their food?
No, no, so it's once a week.
So this is now, you know, guesswork, right?
What's the best way to do it?
No, we're back into the alchemy.
So we've tested three times a week and once a week
and we decided to go with once a week for triad
and you know, it's a guess.
And how many makes per gig are they getting?
Is it one dose?
So it's 0.15 mix per gig once a week.
And that was based on our observations
from 0.05 mix per gig three times a week.
That's how we get, you know, 0.05 times three.
So lifespan is our primary endpoint,
which is important because I think if,
I think this is the first clinical trial that has lifespan in a healthy or normal health status population
as the endpoint, I will say I also it's funny because I get pushed back from clinical
people.
You can't call this a clinical trial.
It's a, it's a, it's a dog.
It's not in people.
And I just simply respond that a veterinary clinic is a clinic.
It's a veterinary clinical is a clinic. It's a
veterinary clinical trial, but it is a clinical trial. And I think this is the first clinical
trial with lifespan as the endpoint. I don't know if you can hear my dog. You want me to
stop. I can. No, no, it's funny. It's totally appropriate that as we're talking about dogs,
we can hear the environment. So the point I want to make though is that lifespan is our primary
endpoint, but we are tracking multiple secondary endpoints to give us a picture of is rapamycin broadly impacting the aging
process. So we're looking every six months, the dogs get echocardiograms to look at heart function.
The dogs will be fitted with activity monitors periodically to look at spontaneous activity.
Every six months, the dogs will get cognitive assessments to look at spontaneous activity. Every six months, the dogs will get cognitive assessments
to look at cognitive function.
We're getting blood chemistry, serum, metabolism,
fecal microbiome, and of course, we'll be tracking
disease incidents as these dogs get older.
So, you know, over this three-year window,
I hope that even if we don't see that magnitude
of lifespan extension and we don't reach statistical significance,
if there is a broad effect on multiple age-related outcomes, that we will be able to detect changes in
other secondary endpoints.
What's the sample size?
You're going to have two groups, placebo versus dose.
Right, half placebo, half treatment.
The intention is to randomize 350 dogs equally split between the two groups, so 175 of each.
And do you know roughly if you had gone with a 10% effect size, how many that would have
required, how many more?
I don't recall up top my head.
I think probably around 500.
In your previous work, you had already got a sense of what was happening in animals with
heart failure.
Rying people a little bit about that if they don't remember the first episode.
Sure.
So, there's really good data.
At least three, maybe four now studies from different labs in mice showing that if you
look at age-related declines in heart function, particularly left ventricular function.
So, the several measures of left ventricular function decline with age,
that 8 to 10 weeks of rapid miceen is enough to reverse those changes
and make the young heart, by echocardiography, look functionally like a,
make the old heart look like a young heart.
There's also data in mouse models of a few different types of heart failures.
So, dilated cardiomyopathy is work that we've done
in particular where you can reverse dilated cardiomyopathy
in mice with rapamycin treatment.
So I think there's really strong evidence in mice.
In our first clinical trial,
which was designed to really only be a safety trial,
there were 24 dogs, 16 got rapamysin,
eight got the placebo,
but we had the dogs also get echocardiograms
before and after the treatment period.
And in that study,
there were statistically significant improvements
in two measures, two of the three measures
of left ventricular function by echocardiography
in the dogs that got rapamysin compared to the placebo.
What was, I think, to me most interesting in that data was that the improvements that we saw
were exclusively found in the dogs that came in with lower function.
Now, one thing to note is none of these dogs had function solo that it would be clinically diagnosed as heart failure. So it was normal age-related declines in function.
And it was exclusively the dogs with the lowest function
that showed the improvements from rap and mison.
That is an interesting observation, which my gut feeling
is real, again, a small cohort.
So I don't want to make it out to be stronger than it is.
But it also is intuitive, right?
We know that individuals have individual trajectories of aging and develop a unique spectrum of
functional declines.
And it makes sense that some of these interventions that are restoring function would primarily
be effective in people who've lost function.
That's just intuitive. So I think that's probably what we'll see in our long-term study
in Triad where we follow the dogs for three years,
is that there's going to be variation at baseline,
and it might be the case that those individuals
that have the lowest function at baseline
are the ones that are going to see the biggest benefit
if there is a benefit at all.
Now in Triad, will you be trying to create
somewhat of a homogeneous sample in size?
By that, I mean the size of the dogs,
or are you going to be completely heterogeneous
with respect to the size of the animal?
And also, what is the age of the animal?
What are the exclusion criteria
around the age on either end, lower high? Right, so age, they have to be at least seven years old to come into the animal. What are the exclusion criteria around the age on either end, lower high? Right. So, age, they have to be at least seven years old to come into the study. So, that'd be
middle-aged. And we do have a size range. So, the dogs have to be between 40 and 100 pounds
to be randomized into the study. So, it's not for little dogs. That's right. And the reason for
that is not because we have any reason to think rapamycin will work differently
or better or worse in little dogs,
it's because big dogs age faster than small dogs.
Let me come back to that in a minute
because that might not be,
that might be an oversimplification.
But they certainly live shorter than small dogs
and develop many age-related diseases and functional
declines at an accelerated rate compared to small dogs.
So we need a population that's already middle-aged and that will be aging rapidly to be able
to see potential benefits from rapamycin in the time frame of this study.
And that was all factored into the power calculations, the demography of dogs in that
age and weight range.
I said that it may not be completely accurate to say that big dogs age more rapidly than small
dogs because there's a growing body of evidence, which we have some preliminary data in support of
as well, that for brain ageing and cognitive function, that might not be the case, that the rate
of cognitive decline in big dogs looks from a chronological sense very, very similar to small dogs.
Even though the big dogs are, you know, dying at an earlier age,
they don't seem to show accelerated cognitive decline,
which is interesting, and I think there's a little bit of data in people,
although obviously people don't show the same diversity in body size that dogs do,
so that's harder to see.
It doesn't really seem to be the case that body size accelerated aging due to increased
body size is reflected in brain aging.
And the mechanisms there might turn out to be really interesting.
So just an observation that I've noticed recently and I think might be important.
You're using once a week dosing, explain to people, because we just mentioned it briefly
and then said we'd come
back to it now.
We're going to sort of bifurcate MTOR into MTOR complex, one MTOR complex, to give people
a sense of how those function and why it, on the one hand, is leading you to do what you're
doing, me to do what I'm doing, and why in some ways it makes it a little bit interesting why the
ITP found what it did with daily dosing.
Let's tie all of that together, but first with an explanation of how MTOR works.
Sure.
So MTOR, of course, is a protein.
It's a kinase.
We already talked about that.
But it acts in a complex with other partner proteins.
And so the MTOR protein acts in at least two, I think there's only two that we know of,
two different complexes called MTOR complex one or MTORC one and MTOR complex two or MTORC two.
And the difference between those those complexes is that they have different partner proteins for
MTOR. So there are a couple of things that are the same across both complexes, and then there are a set of
partner proteins that are unique.
And the two complexes do functionally different things
in the cell.
So MTOR complex one is largely thought of as the MTOR
complex that is most responsive to nutrient levels.
So when nutrient signals are low, that leads to lower mTOR complex one activity,
and mTOR complex one downstream is known
to regulate things like autophagy, mRNA translation,
effects on metabolism, and mTOR complex two
does different things.
And I think we know a lot about what mTOR complex one does.
We know much less about what MTOR complex two does,
although people are studying that and learning more and more
about what MTOR complex two does.
From the perspective of aging biology,
people have focused almost exclusively on MTOR complex one.
There's a little bit of data and CLEGans on MTOR complex two
affecting lifespan, but outside of CLEGans,
almost all of this, the data for RAPAMISans on MTOR complex to affecting lifespan. But outside of C elegans, almost all of this,
the data for rapamycin or MTOR as a regulator of aging,
is thought to be mediated by inhibition or reduced signaling
through MTOR complex one.
So that's what people almost always think about
when they think about effects of MTOR on aging.
And rapamycin as a drug biochemically is a specific inhibitor of m-tore
complex 1. So the way rapamycin works is it actually, it's a small molecule that binds to
another protein called FKBP12 or FPR1 in yeast. And once rapamycin binds to FKBP12, that complex
of rapamycin with FKBP12 goes to MTOR complex1, and you
could think of it as sort of just messing it up, breaks it apart.
So inhibits MTOR complex1 when rapamycin is bind to FKBP12.
So biochemically, rapamycin is an extremely clean drug, and that as far as I know, and
I haven't really seen any good data otherwise, there's no direct inhibitory
effect of rapid miceen on anything other than mTOR complex 1.
What people have observed is that chronic long-term inhibition of mTOR complex 1 can have
feedback effects on mTOR complex 2.
And it's kind of confusing because there's actually some data, both directions, that
chronic inhibition of mTOR complex 1 can lead to activation of mTOR complex 2 in some
context. Mostly, I think the data supports the idea that chronic inhibition of mTOR complex
1 with rapamycin can lead to inhibition of mTOR complex 2 in the long term. And that's
definitely true in mice at higher doses of rapamysin. At lower doses of
rapamysin, it's not completely clear to me how much effect on MTOR complex 2 there is. So,
so the reason why that's interesting from the perspective of aging is I just said that almost all
of the data for lifespan at least is that it's inhibition of MTOR complex 1 that leads to lifespan
extension. Work from David Sabatini and Dudley Lamming
when he was in David's lab,
led to the development of a model
which I think is still the sort of preferred model.
I will say upfront, I think it's at least partly wrong,
but it's the preferred model
which is that the side effects associated
with rapamycin, particularly the metabolic side effects
associated with rapamycin, particularly the metabolic side effects associated
with rapamycin are due to this chronic effect
of inhibiting mTOR complex 2.
So people will talk about rapamycin
as if it induces something like diabetes,
a pseudo-diabetes.
And this is true in mice, there's evidence for it
in humans as well, that chronic long-term treatment
with rapamycin leads to glucose intolerance.
So if you give a mouse who's been on rapamycin for, you know, a year, a glucose tolerance
test, they will not clear that glucose as rapidly as a mouse that never saw rapamycin.
And the model is that that's due to the chronic effects of rapamycin on MTOR complex
2.
Most of the evidence in support of that model comes from
genetic experiments with MTOR complex two deficient mice. So I have yet to see a really clean experiment
showing that that's what accounts for the rap and mice in effects on glucose tolerance. I think
it's a reasonable model. In other words, that evidence comes from creating genetic mice where you
manipulate MTOR quantum MTOR 2 rather than experiments where you give the mice rapamycin.
You know, I didn't ask Rich Miller this question because I'm pretty sure the answer is they didn't do it,
but in all of the ITPs where the animals are getting rapamycin every single day, did they see impaired glucose tolerance?
Despite longer life.
Yeah, my recollection is they did not look, but there have been other studies.
I think mostly in the C57, black 6J mouse strain,
so that's a different strain than the ITP.
We don't need to get into it,
but it's a different genetic background.
But in the C57 background,
you do see the same changes in glucose tolerance test
at the, I think at the 42 part per million
for sure, rapamycin dose, so the higher ITP dose in older mice. So I think it's a real
effect. I think you see that effect with rapamycin. Like I said, there's evidence
in organ transplant patients for impaired glucose homeostasis as well. So I
think it's a real effect. There's a couple of things that I, and I actually, my
intuition is it probably is due to M-Torque 2. I don't have any reason to doubt
that. I just don't think it's been shown cleanly
that that's the mechanism.
Where I differ a little bit, certainly from Dudley,
I'm not sure what David's view on this is right now.
Where I differ from Dudley's interpretation
is that I am less convinced that these effects
that we see for glucose homeostasis are bad.
I think there's at least as much likelihood that what this really reflects is an underlying
change in metabolism that might actually account for part of the beneficial effects of
rapamycin, where they shift away from primarily relying on glucose as the preferred carbon
source and switch over to fat metabolism and maybe even ketogenesis
to some extent. In that context, so it's a different physiological metabolic state. In that context,
when you challenge them with a non-physiological amount of glucose, they don't respond the same way.
So I don't know that it's actually a defect in glucose homeostasis. I think it may reflect the test that's done, and in some ways it's an artifact of that
test that you get a different response, which is in the context of diabetes would be interpreted
as a bad response, it might just reflect a different underlying physiological state.
I haven't seen anybody really try to address that.
And what makes me believe that might be the case
is we and others have seen that rapamycin treatment
has pretty profound effects on fat mobilization,
fat metabolism, adipogenesis,
and at higher doses, ketogenesis.
So it would not surprise me at all
if that metabolic adaptation is accounts for some of the
beneficial effects of rapamycin and is also leading to this apparent, apparent response
to a glucose tolerance test.
Yeah, it's interesting.
When I've spoken with one other physician who uses rapamycin, although he uses it very
liberally in many of his patients, I do not.
I think his patients are coming in much older
and much more metabolically deranged.
And the results that he's seeing in an uncontrolled manner,
meaning you simply have no idea
what the performance effect of rapamycin is.
So when you give it to somebody who's expecting to get better,
they may go and change many other behaviors.
But he's shared with me some of his data,
and it's quite profound, right?
So, triglycerides falling from unhealthy levels
of 200 milligrams per desoleter to 70 milligrams per desoleter,
and actually they're seeing the opposite,
they're seeing improved glucose homeostasis.
Again, these are people starting
who are pre-diabetic and in some cases diabetic.
And I think your point's a fair one about oral glucose tolerance tests are very unnatural.
And you can see a physiologic insulin resistance when you do them on people who are either calorie
restricted or carbohydrate restricted because that initial form of muscle insulin resistance
is actually a protective element there. So all of that said, you've decided to go with once a week,
rather than daily,
despite all of the other animal models
that have shown great success with daily dosing.
Yeah, so that a hedge?
I don't know if I would define it as a hedge.
I mean, I think that,
so the rationale there is both based on the human data,
which again is limited, right?
But we've talked about the daily versus weekly dosing for immune function.
And again, we've also talked about why I think that readouts of immune function
are probably telling us about the underlying inflammatory state,
which I think is a big part of what RAPA-MICEN is doing.
So that is suggestive that weekly dosing is at least as good as daily dosing. And it's also suggestive that weekly dosing has fewer side effects. And because this is a challenge with human
clinical trials, as we've already talked about, it's also something you have to be absolutely
aware of when you're talking about doing a clinical trial in companion dogs.
These are people's pets.
There is an extremely, rightly so, an extremely low tolerance for significant side effects
when we're talking about people's pets.
I love my dog and I would be devastated if I hurt anybody else's dog in this clinical
trial.
You want to do everything you can to reduce the
likelihood of side effects. And there's reason to believe that once weekly dosing is likely to have
fewer side effects. And it's also the pragmatic aspect of we're asking owners to give this medication
to their dog in the context of a clinical trial. We expect that there will be better compliance and fewer mistakes with once a week
dosing versus daily dosing or three times a week.
So, let's pivot a bit from RAPA to Torrent 2.
You recently, you and I have shared a couple of emails on this topic.
Tell people a little bit about what that is and why you're excited.
So, Torrent 2 is a different version of an M-Tor inhibitor.
So, we just talked about M-Tor complex I and M-Tor complex II
and how Rapa Mison, at least biochemically,
is a specific inhibitor of M-Tor complex I.
Torin II is what's called a catalytic inhibitor
or an ATP competitive inhibitor of M-Tor,
which, at least in theory,
versus what we call allosteric.
Allosteric inhibitor, which is R in theory versus what we call allosteric.
Allosteric inhibitor, which is rapamysin, meaning it interacts not through the catalytic
site, yes.
At least in theory, torrent 2 and torrent 1 and other catalytic inhibitors will equally
inhibit both M-tore complex 1 and M-tore complex 2.
And for reasons that I still am not sure about, as far as I know, nobody has tested
Torin II, Torin I, other catalytic inhibitors
for effects on aging in mice.
Why has this not, but this, yeah, this seems like something
that needs to be submitted to ITP tomorrow, right?
For next, or at least for the next cycle.
Yeah, I don't know.
I don't know why it hasn't been done, to be honest with you.
And maybe it has, and I just haven't seen the data,
that certainly is possible.
But I don't think, I don't know of any data,
not just for lifespan, but for, you know,
other functional measures of aging.
I don't know of any data.
I can tell you why I think there's additional reason
to test it beyond, you know, sort of the rationale
that seems obvious.
But I don't, I don't know that I would say
I'm excited about Torrentu.
It seems like an obvious gap in knowledge and would be actually a nice way to test the question we
were talking about previously, if Torque 1 versus Torque 2, good bad, all of that stuff.
It seems like an important set of experiments to do, and as far as I know, nobody's done it.
So part of the reason, additional reason why I think it's an interesting
set of experiments to do is you know in addition to studying aging my lab also works on mitochondrial
dysfunction and mitochondrial disease and we have worked for many years in a mouse model of a
childhood mitochondrial disease called leesandrum. So this mouse is defective in in complex one of the
electron transport chain of the mitochondria,
and it is very short lived.
It lives about 55, 60 days,
and it develops many of the same molecular phenotypes
and neurological phenotypes as kids who have leysendrum,
this childhood mitochondrial disease.
And these children typically live how long?
It's variable onset, but it's anywhere from
infants to, you know, eight, nine years old. It typically, kids with leesundrum don't make it to
be teenagers. It's a horrible, horrible disorder. So we found out many years ago that rapamycin could
roughly double or triple the survival of these mice and basically prevent the neurodegeneration
and brain lesions that are thought to limit
lifespan in both the mice and the kids with leesendrum.
Presumably, Matt, that's because when you knock out complex one, you're destroying oxidative
phosphorylation and presumably the neurons are going to be the most sensitive to that.
Yeah, it's an interesting question.
We don't really know.
I mean, I'll tell you the sort of current thinking in the field.
So first thing is to recognize is this particular component of complex one
is an accessory to stabilizing factor.
So the mice are not 100% deficient in complex one.
They have a low level of complex one.
I think you'd probably be dead if you didn't have any complex one.
And the same thing's true in the patients. So it's a deficiency in complex one. I think you'd probably be dead if you didn't have any complex one. And the same thing is true in the patients. So it's a deficiency in complex one. And you're right.
So one idea, there are actually several things reasons why being deficient in complex one
could be a problem. One could be, you just can't generate enough ATP and neurons or a subset of
neurons are particularly sensitive, right? Another could be that you're generating high levels of reactive oxygen species. We know subsets of neurons are especially sensitive to that.
And you could come up, there's also an inflammatory component to this disease where we see a lot of
neuroinflammation in the brain regions where delusions occur. And we don't quite understand what's
causing that inflammation. So there are multiple ways that this could be causing, you know,
the symptoms of the disease.
An interesting body of work from the MCMuthus lab and Isha Jain,
who was in his lab and now has her own lab,
shows that hypoxia can also rescue these mice.
And hypoxia actually rescues these mice to a greater extent
than rapamycin.
So that at least is consistent with an oxidative stress model, right?
So one idea would be that when you're deficient in complex one, those neurons are not using as much
oxygen because they've switched over to non-oxidative glycolytic metabolism and fermentation. So you have
higher oxygen levels in the brain. That leads to higher oxidative damage. When you reduce oxygen
through hypoxia, you've suppressed that.
I don't have any reason to doubt that model.
By the way, have you ever tried the experiment where you give them excessive amounts of lactate,
which neurons like lactate, and if you gave them lactate in theory, you would also come
up with a way to bypass the ETC.
I mean, it would be an interesting control or way to test that hypothesis.
We've never tried that. So what I can tell you is they have higher levels of lactate in their blood.
That's common in mitochondrial disease because they switch over to fermentation.
But that doesn't mean that your experiment wouldn't work. It's just an observation.
And RAPA mice and suppresses that high level of lactate, interestingly, as well as the accumulation
of all the glycolytic intermediates upstream of lactate. interestingly, as well as the accumulation of all the glycolinetic
intermediate upstream of lactate. So anyways, oxidative stress is, there's at least some reason to
believe that oxidative stress is important. Another phenotype that these mice get is a dramatic loss
of fat. So they are extremely lean and rapamycin suppresses that. So we think that there is some,
we know there's something going on with fat mobilization,
adipogenesis, when we treat with rap and mice, and that spares those mice from presumably using
up all their fat, presumably they're trying to burn the fat in some way, and that's why they become
so lean. So that's another observation. So how do we get to M-Torke 2 and Torren 2? So we did a
phosphoprodiumic study with Judith Villan, who's at the University of Washington
in genome sciences, to try to just take a sort of a global picture and ask, what is the
effect of rapamycin on the proteome and the phosphoprodiome in the knockout mice compared
to wild type and then with rapamycin treatment?
There was a lot of interesting stuff in there.
This work is published.
It was published in Nature, Metabolism,
I don't know, several months ago.
So people are interested.
They can look at that paper.
There's a lot of interesting stuff
in that proteomic data set.
But one of the striking things that we saw
was that MTORC 2 components were,
at the protein level, decreased,
which actually fits with the idea that
high-dose rapamycin chronically leads to inhibition of MTORC2. And associated
that with that was an inhibition of protein kinase C, which is another kinase
that's regulated by MTOR complex 2. So that led us to hypothesize, and protein
kinase C is known to regulate some aspects of inflammation. So that led us to hypothesize that maybe some of the effects of rapamycin, at least in
this mouse model, are through protein kinase C and MTORC 2, as opposed to, you
know, what we had assumed was all MTORC 1 related effects of rapamycin. So we
tested a few drugs that are known inhibitors of protein kinase C, and they rescued part of the lifespan
of the mitochondrial disease mice. So they weren't as good as rapamycin, but they did give significant
increases in survival and delayed some of the neurological symptoms of the mice experience.
So that is consistent with the idea that this inhibition of MTORque 2, inhibition of protein kinase C, is part of the
effect of rapamycin.
So now we've gone back and we've tested torrent 2 in this mitochondrial disease model,
seems to work, you know, as well as rapamycin, which is interesting.
No negative side effects that we can see from inhibiting torque 2 in that context.
And then we're also working in another mouse model of a severe metabolic disease.
And I think I'm not going to talk about it because this is a collaboration and it's not
published yet.
And so I don't want to give away what the initial observation was really the observation
of our collaborators.
But let me just say in another childhood metabolic disease, which is on the surface completely unrelated to complex
one of the mitochondria, we see dramatic rescue from rapamycin and torrentu. So this makes me
wonder, first of all, I thought for a long time that mitochondrial disease might be a good
model for normal aging in some respects.
We know that, I mean, we talked about mitochondrial dysfunction as one of the hallmarks of aging.
So severe mitochondrial dysfunction, I would not argue, is accelerated aging.
But I think interventions that are effective in a model of severe mitochondrial disease
might also be effective in the context of normative aging.
Everything we've seen so far is consistent with that.
And it makes me wonder if torrentu or other catalytic inhibitors of m-tore
might be as effective as rapamycin in the context of aging,
and maybe even more effective in the context of some age-related indication.
So to me, it just seems like a gaping hole in the literature
that really needs to be explored and will learn
from doing that sort of exploration.
The other point that's interesting to make is RTB 101, the drug we talked about previously
in the Restor Bio Phase III clinical trial, is a catalytic inhibitor of M-TOR as well as
other kinases.
So it would fall at least biochemically into the torrent 2 class as opposed to the
rapamycin class. How is torrent 2 discovered or synthesized? Was it synthesized directly to be a
catalytic inhibitor? Was it discovered and modified? I, you know, David Sabatini would be the
person to ask. I should know the answer to that. My recollect, so rapamycin is interesting,
right? It was bound on raponoeve made by bacteria.
I think torrentu came out of a more traditional sort of chemical screening as a mTOR inhibitor
and then was modified to be a more potent catalytic mTOR inhibitor.
But I honestly don't know that literature or recall it.
And does it already have an IND?
Is there already a molecule that's in a pipeline, then an FDA pipeline? That's a good question. I have looked a little bit and I don't know if anybody who's developing
Torin II or Torin I, which is another catalytic inhibitor for FDA approval. And I've
wondered about this, and honestly, I just haven't had the bandwidth to really dig into it.
Is it because people tried and there were side effects? I don't know. But I don't know of anybody who's actively developing
the Torrens, at least, for FDA approval.
RTB 101, again, we talked about that's being developed
or has attempted to be developed for FDA approval.
There are other dual kinase inhibitors
that are used clinically that hit M-TOR
in addition to other, like a PI-3 kinase,
a different class of kinase.
So there are molecules that have similar types of activity,
but I don't know of anybody who's really trying hard
to develop the more specific M-torch catalytic inhibitors.
And again, I'm not sure why.
Let's change gears for a second and talk about NAD,
NR, NMN, all these things.
I've had David Sinclair on the podcast a couple of times. He's very
eloquently explained what Sir Tuenzaar, how they work, why they require NAD. So for folks
who want to get up to speed on that, you can do so in great depth. You want to give the
30-second answer as to why Sir Tuenza matter and why they need NAD?
Sure, but I would start by saying that I'm not sure that sertoins are the only or most important reason why
NAD is important.
Sertoins are a class of NAD dependent,
mostly deacetylases, they also can do some other activities.
But basically one way to think about it is that
sertoins take a settle groups off of other proteins.
That's their activity and that requires NAD. And it
actually consumes NAD. So NAD is a cofactor for many metabolic reactions where it gets converted
between NAD, which is the oxidized form of the coenzyme and NADH, which is the reduced form.
Many, many different metabolic reactions use NAD in that way.
Including the electron transport chain. Including the electron transport chain,
anglicolysis and fermentation.
Yes.
Certunns are fundamentally different
in that they use up NAD, right?
And so NAD is required for their activity.
NADH, the reduced form of NAD,
is actually an inhibitor of Certunns.
So the NAD to NADH ratio can be used as a proxy
of likely
Sertuan activity.
So Sertuan's are important in the aging field.
You know, this really goes back to my work as a graduate student in yeast when I was a grad student with Lennie Grantee.
When we first showed that you could overexpress the yeast Sertuan, which is called Sertuan, that's where Sertuan comes from.
So the yeast protein is called Sertuan. If you overexpress Sir 2,
you increase lifespan in yeast. And since then, other people have shown that
that activation or overexpression of Sir 2 and's and worms or flies or mice can have
interesting effects on aging. You and David must have overlapped, right? And the you guys
must have both been in mind, at the same time. Even my overlapped in the money's lab. Yeah, so he was
a postdoc when I was a grad student.
And I got to give David, I mean,
David and I have had our scientific disagreements
over the years, but I got to give David a ton of credit.
As a postdoc, he mentored me in important ways.
And I think actually guided me to the project,
looking at Sir 2, which is what I just talked about
when we over-expressed Sir 2.
So David was a very important early influence on my scientific career. And the Grantee Lab at that 2, which is what I just talked about when we overexpressed Sir 2. So David was a very important early influence on my scientific career.
And the guarantee lab at that time was full of really smart.
I'm sure it still full of really smart people, but it was just a really great environment.
It was a powerhouse.
Yeah, with lots of really fantastic scientists.
So that was the first, and it was really Lenny's lab that established certuans as important
in aging in multiple model systems.
So, in mice, David might disagree with this a little bit, but I think if you're being
honest, right, the evidence that certuans are potent regulators of lifespan in mice is
mixed.
It's not strong.
There are a couple of studies out of probably, you know, a dozen that have been published,
and there's probably two dozen that are unpublished where people saw no effects on lifespan
from manipulator or activating sertuins.
There are a couple of studies that show, in one case, a brain-specific activation of one
of the sertuins called sert1 could slightly extend lifespan, and another that over-expression
of a different sertuin, sert-6 could slightly extend lifespan, I think,
only in males.
But nowhere near the reproducibility or magnitude of a fact
of other things, including rapid miceen.
So the data on CER2 ends is broad, but the absolute effects
on lifespan, at least, are in my personal view,
unconvincing.
Like, it hasn't been broadly replicated, like but my son has, and they're not big.
What we do see with sertuins is abundant evidence
that metabolic markers of health can be improved
by activating sertuins, and in a few other disease-specific
models, pretty good evidence, heart disease,
in particular, some evidence for cognitive function as well,
improvements in age-related outcomes.
So there's a lot of smoke there, but I think there's a lot of confusion in the field about the
relative strength of data for different interventions. And at least in my view,
there's really no comparison between the effects you get from inhibiting amtore and the effects,
at least so far, that people have reported from activating sirtuans. Amtore is head and shoulders
above sirtuans when it comes to magnitude of effect. I think it would be impossible to that people have reported from activating sertuins. It's like, M-tore is head and shoulders above sertuins
when it comes to magnitude of effect.
I think it would be impossible to dispute that.
I don't think there could be any dispute of that.
What's interesting is if I were to tally up the number
of questions I get per month about NR and NMN versus
rapologues, the ratio is the exact opposite to the magnitude.
So if the effect size of rapamycin and the importance of MTOR is 10x that of SirTuin's,
it's flipped in the number of questions I get about it and just the pop culture awareness of that.
So let's put the marketing of that aside
and talk about the chemistry of it for a moment.
Right, so Sertunns are NAD dependent enzymes, right?
So they need NAD to do their action.
And we've already talked about,
in general, the model is that turning up Sertunns
is a good thing, right?
That you're gonna get,
if you're gonna get benefits in the context of aging,
that's gonna happen from activating sertuins.
Again, that's probably a pretty massive oversimplification
because there are seven sertuins, right?
And they do different things and different things
and different tissues,
but that's kinda where the field has gotten stuck.
The idea is that activating sertuins is good.
And so if you accept that,
and an NAD is an activator of sertuins,
then more NAD is good.
And there's good evidence that NAD homeostasis becomes impaired with aging.
That the ratio of NAD to NAD to NADH, the oxidized to reduced form of NAD, in many tissues,
at least, shifts towards more NADH unless NAD, right?
And so that with age, and so the prediction would be
that you would have declining seroton activity
due to that metabolic shift.
And I will also note,
because I think this is important,
that in mitochondrial disease,
you see the same shift,
it's just much more pronounced.
You see a shift towards NADH and less NAD.
And that's exactly what you see when mitochondria are less functional because the cells will switch over to glycolysis and fermentation, right?
Firmmentation to lactate.
And the whole reason why we ferment to lactate is to restore NAD levels.
Firmmentation to lactate takes NADH and turns it back into NAD.
So this probably reflects an underlying metabolic defect,
which could be mitochondrial in origin
that leads to this shift towards the reduced form
of NAD with age.
So those two observations, less NAD, bad,
sirtoins good, the prediction is that if we could boost NAD,
that would be good, because that would then restore
sirto certain activity and
have effects on aging.
And so this led to the development and popularization of these molecules called NAD precursors or
NAD boosters, the two of which get talked about the most are nicotinamid riboside NR.
That was kind of the first to gain popularity.
And then nicotinamid mononucleotide NMN. Both of those are precursors of NAD
that within cells can be converted into NAD. And so, you know, there's a large body of literature
in a variety of model organisms showing that treatment with NR or NMN sometimes leads to benefits that are associated with healthy aging and in one study,
lifespan extension and mice.
I say sometimes because there's also a large body of literature that doesn't reproduce those
results.
Some of it published, a lot of it unpublished.
Including the ITP.
Including the ITP, that's right.
Which is, I think, has to be considered the gold
standard for at least my state. I think that's true, although, you know, as we talked about
before, it's a different genetic background than C57 Black6. And I would argue it's a much
better genetic background. Well, I think you can make that argument and there are good reasons
to believe that argument. Nonetheless, I think it's important to note that that could be why it worked in one context and didn't work in another context.
And we've struggled with this as well in my lab.
So, you know, it's been reported in this mitochondrial disease mouse by a collaborator of ours who I trust their data, right?
That NMN could increase lifespan in that mouse model.
We've tried multiple times with both NR and NMN in that mouse model and been unable to get these effects.
So I think these drugs are tricky from a biological efficacy perspective to there's something we don't understand about delivery or uptake.
Do you think the temperature is stable?
Probably, well, I mean, I don't know. I'm sure there are people who know the answer to that.
I've read conflicting things, right?
I've read that, and I need to go back to the sources on it, but I've read that at least
through the lens in which they're provided as supplements, the likelihood that by the time
that thing arrives at your door, it still has the biological activity that it would have
had in a refrigerated manner is low.
Now, I don't know that the companies that sell them recommend
refrigerating them, but they might not recommend refrigerating them
because then it would imply that they're being shipped
in a refrigerated manner, which sort of nullifies the whole
benefit. But I don't know if you've looked at any of that.
We haven't. So definitely in our mouse studies,
we are careful to keep the food in the refrigerator until
we put it in the mouse cage. I suspect that there certainly might be some truth to the idea that the biological activity
goes off over time at room temperature.
But I think that reflects a bigger problem with the NAD precursor field, which is that there
is a lot of controversy even among the two camps.
So I mean, it's sort of funny, right?
Because there's an NR camp of researchers who really think NR is the tool that we should use.
And then there's an NMN camp.
And they both say that the other camp
that their molecule doesn't work
because it's not biologically available
or all sorts of reasons, right?
So there's a lot of lack of clarity
around biological availability and efficacy
with these molecules in the preclinical literature, right?
Where, you know, in theory,
people should be able to exactly reproduce the way
that other people do the work and get the same results, right?
There are lots of reasons why scientific results
don't get replicated.
It's my impression that in the NAD precursor field,
that's a bigger problem than in some other areas.
And I don't know the reasons for that,
but all I can say is we've experienced that in my lab as well.
So I've tried to stay on the fence here because I think there's a ton of smoke, right?
There's a ton of smoke with certunes.
There's a ton of smoke with NAD precursors that they can if you do the experiment the right
way, have positive effects that look a lot like what we would expect for something that's
impacting the aging process.
And as I said, I mean, it's funny because people
who are in the field, I think sometimes think of me
as this anti-sertuin guy, which is absolutely not the truth.
I'm the guy, I'm the guy who first showed
that you could overexpress a Sertuin
in an increased lifespan.
If anybody's gonna be pro-Sertuin, it's me.
I think the problem is that I've seen a lot of data
that people have struggled to reproduce.
And I just honestly don't know how to interpret that.
Whereas with Rapa Mison, it works for everybody.
It's robust, and everybody gets the same result over and over and over again.
So I'm less enthusiastic, I would say, about Sir Tuen's and NAD precursors as opposed
to some other interventions in the field.
But I think there's a lot of data that suggests that these molecules and
that Sertunans are important for aging. I think what we haven't done yet is figured
out how to tweak the system in exactly the right way to get robust and reproducible experimental
results that I personally would feel comfortable moving forward with clinically.
And it might be that the answer is you have to hit this system with two prongs.
You have to provide more of a precursor and you have to activate the Sertuan in the way
that Resveratrol attempted to do but wasn't doing.
So maybe the answer is it's both.
Yeah, right.
Sure, that's a possibility.
Yeah.
The other thing that I find weird about the NAD precursor literature and the limited work that's
been done clinically is often, I mean, in principle, it should be trivial to determine whether
or not you have boosted NAD levels, right?
If you treat somebody with an NAD precursor, we know how to measure NAD.
It's not hard.
We know what the biomarker is, at least that far in this case.
And oftentimes, that's not done. So if you're treating somebody with NR or NMN
and you're not increasing NAD levels in the blood
or in your target tissue,
that should tell you something important, I would think.
Well, and the other question is,
is increasing it in the blood sufficient?
That's a different question,
but it is an important question.
I agree, that's why I said target tissue.
And we can't biopsy certain tissues,
but we can biopsy some tissues, yeah.
It's super messy.
Look, I get asked about it at least once a week
by patients, and I usually point them
to something I've written on the subject matter.
But in the end, I say, look, I will say,
I think it's very safe.
I really don't see a downside other than to your pocketbook
of taking NR or NMN.
So they check the first box of any intervention,
which is the downside sufficiently low.
And I think the answer is yes.
I'm just having a hard time seeing upside.
Yeah, and I'll tell you honestly,
I would love to test NR or NMN or both in dogs,
for exactly that reason, right?
Because there is essentially no risk,
and we could actually find out, does it work? Three years
from now, five years from now, I could tell you, do NAD precursors slow aging at least in pet dogs?
And I think if I saw NR slow aging increase lifespan and or improve multiple functional measures
of aging in dogs, I'd be much more bullish on taking NR myself. It wouldn't prove it's going to work
in people, but it gets you a part way there and a pretty big part of the way there.
And I would put that on the short list of things I would like to test.
The fact is nobody's going to do the definitive clinical trial in people,
because they don't have to, because they can sell that stuff to people now, right?
They aren't required to do the clinical trial to show that it works.
Matt, you probably don't remember this, but there was one night, oh, four, four, five
years ago.
Are you making comments about my cognitive decline with aging?
No.
I just don't think you'd remember some random night of us having dinner in New York, but
it was about four years ago.
Yes.
Okay, wow.
We do have the cognitive function.
So the three of us were having dinner at my favorite not my favorite
but a decent Persian place on the Upper East Side and I don't know if I said so at the time
But I might have told you after it was after that dinner that I decided I got to do a podcast
Because the three of us had such an enjoyable. She's at Einstein, isn't she?
She was.
Yeah, she's actually moved to Columbia.
So now she's running a reproductive aging center
at Columbia now.
Apropos with our discussion today.
But we just had such an amazing discussion, which really
means me asking the two of you guys non-stop questions.
And I remember thinking after that, God damn,
why didn't I record this dinner?
Why didn't I have my phone sitting on the table
to record it?
And it was literally that moment that made me realize,
like this happens the same time.
Every time I go out with Sabatini,
I have the same feeling.
Every time I go out with someone,
so I have the same feeling.
I gotta just do this podcast thing.
So for anybody listening to this,
who is appreciative of the podcast,
they owe you personally a great
dead of gratitude for.
And you shin, I gotta say, I'm sure it was you shin who made most of the really insightful
comments at that dinner.
I think I think I think I think it was both of you guys, but it was, it is always such an
awesome time to be able to sit down with you.
I think as the listener appreciates here, the breadth of topics within the space of longevity
that you can cover is broad.
So you're one of the few people who can go very wide
and very deep.
And I think today's discussion demonstrated that.
So thank you very much, Matt.
Really, as always, enjoy this discussion.
Sure, yeah, anytime.
I enjoy the discussion as much as you do, I think.
So it's been a lot of fun.
All right, and best of luck with the triad study.
I know a lot of people are probably more interested in that
than any of the human stuff because the dog owners,
I know care far more about what rapamycin can do for their dogs
than they care about what it can do for them.
So, hopefully we'll have those results
by the next time we speak.
Great, thanks Peter.
Thank you for listening to this week's episode of The Drive.
If you're interested in diving deeper into any topics we discuss,
we've created a membership program that allows us to bring you
more in-depth exclusive content without relying on paid ads.
It's our goal to ensure members get back much more than the price
of the subscription.
To that end, membership benefits include a bunch of things.
One, totally kick-ass comprehensive podcast show notes, the subscription. To that end, membership benefits include a bunch of things. 1.
Totally kick ass comprehensive podcast show notes that detail every topic paper person thing
we discuss on each episode.
The word on the street is, nobody's show notes rival these.
Monthly AMA episodes are asking me anything episodes, hearing these episodes completely.
Access to our private podcast feed that allows you to hear everything without having to listen to spills like this.
The Qualies, which are a super short podcast that we release every Tuesday through Friday,
highlighting the best questions, topics, and tactics discussed on previous episodes of the drive.
This is a great way to catch up on previous episodes without having to go back and necessarily listen to everyone. Steep discounts on products that I believe in, but for which I'm not getting paid to
endorse.
And a whole bunch of other benefits that we continue to trickle in as time goes on.
If you want to learn more and access these member-only benefits, you can head over to peteratiamd.com
forward slash subscribe.
You can find me on Twitter, Instagram, and Facebook, all with the ID, Peter Atia MD.
You can also leave us a review on Apple Podcasts or whatever podcast player you listen on.
This podcast is for general informational purposes only.
It does not constitute the practice of medicine, nursing, or other professional healthcare services,
including the giving of medical advice.
No doctor-patient relationship is formed.
The use of this information and the materials linked to this podcast is at the user's own risk.
The content on this podcast is not intended to be a substitute for
professional medical advice, diagnosis, or treatment. Users should not disregard or
delay in obtaining medical advice from any medical condition they have and they
should seek the assistance of their healthcare professionals
for any such conditions.
Finally, I take conflicts of interest very seriously.
For all of my disclosures and the companies I invest in
or advise, please visit peteratia-md.com
forward slash about where I keep an up-to-date
and active list of such companies. you