Placeholder Image

Subtitles section Play video

  • Hi, this is Jim Wells again. I'm going to tell you about part two: from hit to pill.

  • Now, last time we left off, we had taken you up through the stages of these nine stages.

  • We'd taken you up from the target identification stage, to hit ID, and now we're at the hit to lead stage.

  • And the key thing about this stage

  • is that we want to identify compounds that don't just bind to the protein,

  • they actually work inside a cell. And they actually show selectivity in a cell.

  • And this is yet another elevated level

  • and gets us closer to what we want, which is a drug that's safe and efficacious in people.

  • The key aspect of hit to lead stage is an iterative process in which we

  • not only show that the compound works in a biochemical assay,

  • but we also demonstrate that it works effectively and selectively in a cell-based assay.

  • So, it can actually go through the cell membrane, reach the target inside the cell,

  • if it is an intracellular target,

  • and engage that protein in a cell-based assay.

  • So, in starting this process, the compounds start off with potencies

  • that are weaker than we would like.

  • As shown in this biochemical binding assay

  • what we're looking for is compounds that will make the medicinal chemistry

  • that will improve the potency of the hit compound at least a factor of ten,

  • ideally a factor of twenty, in the biochemical assay.

  • We'll also be looking for things that start off with, from the hit stage,

  • that have weak cellular potency, shown here,

  • but with medicinal chemistry that correlates with the biochemical potency above,

  • drives the cellular potency to be more potent in the cell.

  • And this is all toward, the goal here is to get potent compounds

  • that are cell active.

  • Now, we also look in this stage for several other important properties.

  • For instance, we don't want it to bind to other off-targets

  • that are related to it that may cause toxicity.

  • So, what we'd prefer is that the compounds have potency that are at least ten-fold weaker

  • to the closest related target.

  • We'll also be looking to see that there's chemical evidence that we can advance these compounds.

  • When we look at compound's structures,

  • you should see that there are compounds which both

  • affect the target and compounds which don't affect the target

  • that are fairly closely related

  • suggesting that they're binding to a single site.

  • Because one of the properties of a drug is that it bind to a single site

  • on the target protein.

  • We'd like to know the molecular target; Michelle mentioned

  • that you can start off these hit-finding expeditions

  • just looking for cellular activity.

  • In that case, we don't really know the target that's involved.

  • At this stage, we really would like to have a molecular understanding of what that

  • small molecule is engaging.

  • Lastly, and very importantly,

  • in this especially for formulations reasons,

  • we look to have compounds which have solubilities above a hundred micromolar.

  • And that's because we wish the compounds to be

  • soluble and dissolve well once they're administered to an animal.

  • Ok, having successfully passed this stage,

  • of a hit to lead stage,

  • we're now into this very important and pivotal stage,

  • called lead optimization.

  • This is where we're looking to see that compounds that we've created

  • can actually work in a whole animal, can reach the

  • protein target, through a cell, and through the circulatory system of the animal.

  • Ok, so I think at this stage it would be good to understand

  • what happens when you swallow a pill.

  • Here's our patient right here, going to take a pill,

  • there's the pills, they go down through the saliva

  • into the stomach, where they're

  • subjected to pH one, so they have to survive that.

  • They come out of the stomach and they go straight into the duodenum,

  • which is this arrow here, which you see goes straight into the intestines.

  • Now, the intestinal, the intestines have about

  • two hundred square meters of surface area to absorb.

  • That's about the size of a tennis court.

  • There's a lot of opportunity to absorb a drug,

  • but it has to have the right physical properties to get across that intestinal barrier

  • so it actually enters into the blood stream.

  • Once it does get across that, into the intestine,

  • and into the bloodstream, the first thing that is does is

  • it goes straight into the liver through the portal vein, here,

  • and then meets a series of very important obstacles.

  • They include a variety of enzymes that are in the liver that are meant to detoxify,

  • get rid of, these foreign small molecules.

  • So, they would be things like p450s, which oxidize the compound,

  • or hydrolases, protease, lipases, esterases,

  • that would be hydrolyzing compounds.

  • There would be glucaronidation enzymes and the like,

  • which tag them so that they can be rapidly excreted.

  • Of course, all of these things can reduce the potency and the availability of the drug

  • to have its effect in the peripheral tissues where it's probably got to act.

  • So, if really does, the drug has to get through this very important gauntlet here,

  • of the liver in order to go on from there, throughout the circulatory system

  • first into the right atrium, then into the lungs,

  • and then back through the lungs, into the left atrium,

  • and then through the arterial system all the way throughout the body.

  • So, understanding the pathway that a pill has to take

  • is actually very important in this drug discovery process

  • because even though a compound could be great in a biochemical assay or a cell based assay

  • if it doesn't work in this system over here

  • then it's not going to be a drug.

  • Ok, once, now understanding that

  • and having successfully passed the hit-to-lead stage we're now ready to do lead optimization.

  • And that again is an iterative process of chemistry and biology

  • paired together. But this time, at the animal level.

  • So, the very first thing that happens when you have compounds from the hit-to-lead stage

  • is that you'd want to determine how well do they survive in the body.

  • How long do they live in the body?

  • And to do this, you typically give either an injection of the compound

  • or feed an animal this compound

  • and then determine the PK of the compound

  • in other words, how well does that compound,

  • this is time along the x-axis here and amount of compound

  • that's in an animal along the y-axis

  • and as the compound is injected, you see that there's a rise

  • in compound levels as it reaches, as it goes into the circulatory system

  • and then it decays.

  • And this half-life and other parameters

  • are used to judge how well will that compound work in an animal

  • or how long it will last in an animal.

  • The next stage is called the pharmacodynamics part of this

  • and there we're actually looking to see how active is this compound

  • in an animal model.

  • This, I show here, is an example of a xenograph model for cancer.

  • This, for instance, is time along the x-axis and along the y-axis is tumor volume.

  • You can see if the animal is untreated with your drug,

  • your drug candidate, the tumor grows rapidly,

  • that's this blue line here,

  • and then, in the case of a compound that shows some efficacy,

  • if treated with the animal, treating the animal with that, you can see you can suppress the growth of the tumor.

  • That would be a good result.

  • Now, one would continue along this process

  • of lead optimization until one has gotten compounds

  • that show good PK and good animal efficacy

  • according to those guidelines that we discussed at the beginning of this lecture.

  • The notion of lead optimization is really one of

  • trying to push compounds from the vast amount, trying to identify compounds

  • from the vast chemical space that's out there.

  • We, through the hit-to-lead process, we've identified potent compounds,

  • both biochemically and in cell assay,

  • we use pharmacokinetics to identify those compounds that have both potency and good pharmacokinetics

  • so we take compounds within this area of the Venn

  • and then using our animal efficacy experiments, our PD or pharmacodynamic models,

  • we're really interested the compounds that are sitting right in this narrow area here.

  • So, it's this iterative process of testing these compounds

  • in pharmacokinetics, pharmacodynamics and cell and biochemical assays

  • that ultimately then identifies a compound that can be deemed a clinical candidate.

  • This would be a compound that would be ready for IND enabling studies

  • and hopefully on the pathway to being a drug.

  • So, the goals for an oral drug, then, after this process

  • again are that we're looking for compounds that can be dosed

  • once a day, at hopefully less than a hundred milligrams per day,

  • we want reasonable protein binding potency

  • and reasonable cell activity.

  • We want a decent half-life, so that it only has to be taken once a day

  • and we want good oral uptake

  • so that it can be taken orally.

  • Ok, all of this work I told you about,

  • this reminds me of this Greek myth of this character down here,

  • who you can see is exhausted

  • and that's how you feel once you get to the clinical candidacy stage.

  • You've just spent a lot of time, a lot of effort,

  • from target ID, hit ID, hit to lead and lead optimization,

  • you've just reached this plateau here and now you look up

  • and there's all of this stuff still to go

  • because that's the drug development stage that you need to get to to finally reach the nirvana.

  • So, now having worked all, done all the work in the drug discovery part,

  • which is basically this area from target ID to, through lead optimization

  • to identify a clinical candidate,

  • that typically takes somewhere around three to four years to go through that process.

  • There's still a whole lot of work to go on.

  • From IND enabling all the way through registration and FDA approval.

  • This process is an even, much more expensive and longer process,

  • about four to eight years to get through that process.

  • And it's worth considering what are the,

  • what are some of the reasons that compounds, we should this as a funnel,

  • because there's a lot of attrition that goes on in this process.

  • In fact, when a compound enters the clinic,

  • here in phase one, only one in ten compounds will actually make it to being a pill.

  • What are some of the reasons that they don't make it all the way to being a pill?

  • About a third of the reasons are due to lack of efficacy.

  • So, this is, when a compound shown to be efficacious in animals

  • and all of the target ID experiments that were done before,

  • this really reflects the fact that we didn't have a validated target

  • to begin with. That there's more going on with the biology than we thought

  • and it's sort of back to the drawing board

  • about what is actually driving that.

  • Other reasons for compound failure

  • is about a third of them die because of toxicity in humans.

  • These are things that we couldn't really model in animals

  • and so they wind up being disqualified as, they may have failed in phase one.

  • Other reasons for failure are pharmacokinetics,

  • they just don't have the right clearance

  • and the right properties to be a once a day or twice a day medication.

  • And, so they can fail for those reasons.

  • There's another, and much less likely failure,

  • which is due to commercialization reasons,

  • and this may be because the company just feels that the market size is not what they thought,

  • the drug is really not going to be worth developing further.

  • And so they will stop that.

  • There are many other reasons that might happen,

  • but these represent the lion's share of the kind of failures that happen in the clinic.

  • So, we really need to get much better at doing this.

  • We know that healthcare costs are spiraling out of control

  • and this represents something like fifty to twenty percent of the GNP

  • and new drugs and new therapeutics can really

  • spare us a lot of money in our economy if we're able to find new compounds that can ameliorate a disease.

  • Because it's much better to treat it early than to have to deal with the symptoms

  • and the ramifications after.

  • This also highlights the fact that we need to get to

  • prevention and cure, versus crisis and symptom,

  • which is the process that we're in now.

  • It's much cheaper to prevent and cure

  • than it is to manage a crisis or just simply manage the symptoms of disease.

  • Now, how are we doing in this process?

  • Well, there's something like twenty thousand gene targets,

  • meaning twenty thousand genes that encode for proteins;

  • if each of those were associated with a disease,

  • there's got to be a one to one correlation

  • but you could imagine that we should be able to find a lot more drugs

  • because currently there's only two hundred drug targets that are addressed.

  • And that means we're a long way from really

  • saturating the genome, or the proteome, as it were, with potential compounds

  • that can affect disease.

  • The other thing is that these days

  • we're only adding, the FDA, while it approves something like

  • thirty to forty new drugs a year,

  • we're only adding three to four new targets addressed per year.

  • That's because many of the drugs that are approved these days

  • are what are called "me too" drugs

  • that are second generation drugs that may have better pharmacokinetics,

  • less toxicity, but they're really addressing a brand new mechanism.

  • We truly need to address new mechanisms if we're going to affect

  • our healthcare.

  • So, in the last little bit here,

  • I wanted to tell you why I think drug discovery is so exciting

  • from a scientific point of view.

  • And I'm going to do this, these next few slides

  • with the example of Gleevec.

  • Gleevec's a new drug to treat CML,

  • and other cancers.

  • And this was discovered from some very careful

  • molecular biology and oncology studies

  • and genome wide association studies, that showed that this target,

  • the ABL kinase, gets hyper activated when its fused in patients with

  • CML because of this fusion with this protein BCR.

  • So, BCR-ABL, when they're fused together

  • at the gene level and then encoding a protein,

  • leads to this hyperactive protein kinase

  • that phosphorylates its substrates much more active than it should

  • and that drives cellular proliferation in chronic myeloginous lukemias.

  • Now, turns out that there's a, this is a kinase that binds ATP

  • and using high through put screening, a group at Novartis was able to identify compounds

  • that would actually displace ATP from BCR-ABL

  • and thereby inactivate the kinase

  • so it that it would no longer drive CML.

  • And this was a spectacular discovery,

  • one of the first really highly-targeted kinase inhibitors

  • that showed dramatic effects on this really life-threatening disease.

  • Another aspect of drug discovery that's so fascinating

  • is the fact that it allows us to understand how small molecules bind to their proteins of interest.

  • These are, they use weak forces to carry out this binding

  • reaction and it's those weak forces that if we understood them better,

  • we could really make better drugs, discover drugs more rationally,

  • and more completely.

  • And so, by looking at the structures of compounds bound,

  • to here, we're showing Gleevec bound to BCR-ABL,

  • in the spheres here,

  • we can begin to understand the hydrogen bonding interactions

  • between the drug, the ion dipole interactions between the drug and the protein and

  • hydrophobic interactions between the drug and the protein

  • and we can make analogues, chemical analogues, of these drugs and see how that affects potency,

  • so we can really understand the molecular details of how these contacts drive potency

  • and that way we can then understand how we can build better ones

  • because by seeing the values of certain interactions,

  • we can encode those into the small molecule to drive potency.

  • Another aspect that I find particularly fascinating

  • is that proteins themselves are quite dynamic.

  • They're moving in solution, their side chains are rotating on the surface of them.

  • This is our friend BCR-ABL and when this protein

  • binds its substrate, it undergoes conformational changes

  • and when it binds the drug Gleevec, it involves other conformational changes.

  • So, maybe if we could show this movie here,

  • you can see here, as the protein binds the substrate,

  • there in this green region,

  • you can see a conformational change.

  • And then when it binds Gleevec, there's a much larger conformational change that ensues.

  • These conformational changes are very important

  • in understanding how proteins bind small molecules

  • and how they can be controlled

  • because they are moving, dynamic objects that can be trapped

  • in different conformations and drugs allow us

  • a way of studying these kind of phenomena.

  • Now, another thing that's very exciting about drug discovery

  • is it allows one to have, a chemical biologist or biologist, to have tools

  • that can allow you to effect a biology in a cell in a very rapid and dose dependent manner.

  • Small molecules will act, often, on the timescale of seconds

  • to minutes, and this is much faster than

  • such probative technologies like knockout mice,

  • which take the development of a whole organism,

  • or siRNA, which typically takes 72 hours before you've had a complete knockdown

  • of the protein of interest.

  • Another aspect of drug discovery that's very exciting,

  • and this is for the pharmacologists,

  • is that it allows them tools to understand, to relate the physiological

  • effect of inhibiting or activating a particular target,

  • on the animal itself.

  • And in how that relates to the disease.

  • And so, this really goes, provides, important tools for pharmacologists

  • and has for years, for understanding

  • how particular biologies are regulating the phenotypes that we see in animals.

  • And lastly, it provides tools for geneticists and clinicians to relate

  • pharmacogenomics and epidemiology to disease.

  • As I mentioned, the ultimate target validation is a drug itself,

  • an approved drug that is shown to be effective in a particular disease

  • indeed validates that target, that this is important.

  • And additionally, by looking at responders and non-responding patients

  • and the difference in their genomes, we can begin to understand what other factors modulate

  • the disease itself and what makes them sensitive or resistant to certain compounds.

  • And finally, one of the things that's so great about drug discovery and development

  • is that it provides the fundamental basis for a healthier society.

  • Drugs have made a huge impact on our

  • health and will continue to do so.

  • We have a lot more to do in this area, as I've shown you,

  • and this is something that's, I think, is particularly exciting and worthwhile doing.

  • So, I'd like to thank you for listening to this presentation and see you later!

Hi, this is Jim Wells again. I'm going to tell you about part two: from hit to pill.

Subtitles and vocabulary

Click the word to look it up Click the word to find further inforamtion about it