Skip to Main Content

Therapeutic Targeting of Epigenetic Mechanisms in Cancer

January 29, 2024
  • 00:00Good morning everybody.
  • 00:01It's so nice to see everybody here.
  • 00:03So let's get started.
  • 00:04So this is a special occasion
  • 00:06and actually nobody better than
  • 00:08Doctor Armstrong to present the
  • 00:10Lecturer in honour of Lance Tallman.
  • 00:12So this series was established
  • 00:15in 2012 by Doctor Marvin Sears.
  • 00:17Dr. Sears was a long time chair
  • 00:19and founder of the Ophthalmology
  • 00:21and Visual Sciences at Yale,
  • 00:23and he established this lecture
  • 00:24series series in honour of his mother,
  • 00:27Lance Tallman,
  • 00:27who passed away from leukemia.
  • 00:29And it really was the first lecture series
  • 00:31dedicated solely to hematologic malignancies.
  • 00:33So hematologists are always
  • 00:35delighted and it's really intended
  • 00:37to bring to Yale pioneers,
  • 00:39you know,
  • 00:40who study malignant hematologic diseases
  • 00:43and then bring treatments to the patient.
  • 00:46There's actually nobody better than to give
  • 00:48today's lecture than Doctor Scott Armstrong.
  • 00:51Doctor Armstrong is the
  • 00:52President of Dana Farber,
  • 00:54Boston Children's Cancer and Blood
  • 00:56Disorder Center and the Chairman of
  • 00:59the Department of Pediatric Oncology
  • 01:00at Dana Farben Cancer Institute.
  • 01:02And since 2016,
  • 01:05he serves as the Associate Chief of
  • 01:07the Division of Hematology Oncology
  • 01:09at Boston Children's Hospital and
  • 01:11was previously the Director of the
  • 01:13Center for Abidinex Research at
  • 01:15Memorial Sloan Kettering Cancer
  • 01:17Center and Professor of Pediatrics
  • 01:19at Weill Cornell Medical College.
  • 01:21I'm not going to go back to
  • 01:23medical degrees and all this,
  • 01:24so good to have you here.
  • 01:25So Roger Armstrong really has you know
  • 01:28pioneered research in in epigenetics
  • 01:33and studying pediatric cancers,
  • 01:34right.
  • 01:35And we always learn that studying
  • 01:37cancer for example in Pediatrics can
  • 01:39then really enlightened mechanism
  • 01:41of disease also for adult patients.
  • 01:43And I think it's super,
  • 01:44super exciting to hear your talk
  • 01:46today really bringing basic mechanism
  • 01:48all the way from the lab to benefit
  • 01:50so many of our patients and we're
  • 01:52incredibly excited to have you here.
  • 02:01Thank you for the nice introduction
  • 02:02and and for the lectureship and
  • 02:04the plaque that's very nice.
  • 02:06And thank you for coming today.
  • 02:08It's not the most beautiful
  • 02:09day to be out walking around.
  • 02:10So I appreciate you you making it here.
  • 02:13And as Stephanie said,
  • 02:15I'm going to talk to you today
  • 02:17about work we've been doing over
  • 02:20the past couple of decades now
  • 02:23focused on originally a relatively
  • 02:26rare subset of leukemia and then
  • 02:28move to more common leukemias
  • 02:31and maybe even beyond leukemias.
  • 02:33And as many of you probably know
  • 02:35that the concept of targeting
  • 02:37chromatin or epigenetic based
  • 02:39mechanisms been around for quite
  • 02:41some time and there have been some
  • 02:43therapeutic advances in that regard.
  • 02:44But it's been it's stops and
  • 02:46starts I would say along the road
  • 02:48and hopefully I can convince you
  • 02:50that we're maybe finally starting
  • 02:52to make some significant go get
  • 02:54some significant insights there.
  • 02:56So these are my disclosures.
  • 02:57I do consult for a number of biotech
  • 02:59companies trying to convince them that
  • 03:01these mechanisms are relevant that's
  • 03:03and sometimes I'm able to do that.
  • 03:05And then this patent on amended inhibition,
  • 03:08NPM one AML.
  • 03:08I'm going to talk about NPM one AML.
  • 03:11And more important disclosure is these
  • 03:12are the people that do the work.
  • 03:14I don't do the work and I'm very lucky
  • 03:17to have tremendous fellows in the lab.
  • 03:20And actually all of these fellows other
  • 03:22than Emily who soon will leave the
  • 03:24lab to go start her own have started
  • 03:27their own independent lab based careers now.
  • 03:30So this is the outline of the
  • 03:31talk I'm going to introduce you.
  • 03:33Many of you probably know a lot of
  • 03:37this to the MLL or CAT or MLL or KMT
  • 03:41two ACI can't even remember what the
  • 03:44other name is for complex and MLL
  • 03:47rearranged leukemias and then move to
  • 03:50the therapeutic development of small
  • 03:52molecules that target those complexes.
  • 03:54Talk a little bit about the clinical
  • 03:56translation and resistance mechanisms
  • 03:57that we're starting to see to those
  • 04:00molecules and then talk about the
  • 04:02potential role in other cancers.
  • 04:03And as I already mentioned the,
  • 04:05the concept of the OR the
  • 04:08relevance of epigenetics.
  • 04:10And just for those of you that
  • 04:11are that are purists in the
  • 04:13epigenetic and chromatin space,
  • 04:14I will interchangeably use
  • 04:16epigenetic and chromatin biology.
  • 04:17Actually there there is a group of
  • 04:20people that think those two things are
  • 04:22not the same thing and the concept
  • 04:24that these mechanisms are relevant and
  • 04:26cancer has been around for quite some time.
  • 04:28This is not a new idea and epigenetics
  • 04:31really encompasses many different
  • 04:33types of modifications of chromatin,
  • 04:36DNA methylation,
  • 04:37histone modifications,
  • 04:38complexes have proteins that
  • 04:41read those histone modifications.
  • 04:43The nucleos,
  • 04:44there's nucleosome remodeling complexes
  • 04:45that you've probably heard about.
  • 04:47The bath complex also are
  • 04:49frequently mutated in cancer.
  • 04:50So we've known that this is relevant,
  • 04:52but what to do about it has been
  • 04:54a little bit harder to understand
  • 04:56and the the kind of simple
  • 04:58concept is, is these mechanisms
  • 05:00control developmental gene expression
  • 05:02and if we were smart enough we'd
  • 05:05figure out how to therapeutically
  • 05:06target them and reverse those cancer
  • 05:09causing gene expression mechanisms and
  • 05:11hopefully we're starting to get there.
  • 05:13There are some FDA approved drugs that
  • 05:15you probably know about H TAC inhibitors
  • 05:18and DNA methyl transferase inhibitors.
  • 05:19I would say whether or not those molecules
  • 05:22are working via epigenetic mechanisms,
  • 05:25still a little bit of a question,
  • 05:26but indeed those were the first ones
  • 05:29that could be working via these some of
  • 05:32these mechanisms that were FDA approved.
  • 05:34So this is the leukemia that I became
  • 05:37most interested in as a fellow
  • 05:39back in the late 1990s.
  • 05:40Now those are leukemias with
  • 05:43rearrangements of the KMT 2A or
  • 05:46or MLL gene and in Pediatrics as
  • 05:49mentioned I'm a pediatric oncologist.
  • 05:52This rearrangement when found in
  • 05:53infants with AOL predicts a very poor
  • 05:56prognosis and in pediatric leukemia
  • 05:58therapy we're actually not used to that.
  • 06:00We cure most of our patients with
  • 06:02AOL and we find a subset that has a
  • 06:05less than 40% long term survival.
  • 06:07We that's unusual and this subset
  • 06:09is that if an infant comes in that
  • 06:12has ALL and has a rearrangement of
  • 06:14this gene that it's probably in
  • 06:16the 4050% long term survival now.
  • 06:23And so back in the late 90s I joined
  • 06:26Stan course Meyer's lab to start to
  • 06:28learn about that and then obviously
  • 06:30ultimately to start my own lab.
  • 06:32So this is the the wild
  • 06:34type MLL or KMT 2A protein.
  • 06:37It's very large, it's in the nucleus,
  • 06:39it's about 500K Daltons,
  • 06:41makes it has made it difficult to study.
  • 06:44It has a number of different
  • 06:46domains and is bound to chromatin.
  • 06:48We've known that for over 20 years.
  • 06:50And when the translocation occurs that
  • 06:52in terminus of MLL is fused to the
  • 06:54C terminus of what's 100 different
  • 06:56fusion proteins also making a little
  • 06:59bit complicated to understand.
  • 07:00And this is just the history that
  • 07:02I won't go through in too much
  • 07:04detail other than to say that the
  • 07:06wild type MLL protein is was shown
  • 07:08in the mid 90s by Stan Course,
  • 07:10Mars Group and others to control
  • 07:13development of blood system of
  • 07:15hematopolysis through presumably
  • 07:16control of the homeotic or hox
  • 07:18genes that are important in many
  • 07:20types of development and but
  • 07:22in blood development as well.
  • 07:24And that was really actually
  • 07:26pointed to that concept by studies
  • 07:28done even before that in fly,
  • 07:30in fruit flies and Drosophila,
  • 07:32showing that the trithorax gene,
  • 07:33which is the Drosophila homologue is
  • 07:36important for development as well.
  • 07:38And then Mike Cleary and Terry,
  • 07:40rabbits showed in very nice mouse
  • 07:42studies in the late 90s that
  • 07:44the MLL fusion proteins indeed
  • 07:46do directly induce leukemia.
  • 07:48And then David Alice's group showed
  • 07:50that in the wild type MLL is a histone
  • 07:53modifying enzyme modifies histone H3
  • 07:54on lysine 4 through this enzymatic
  • 07:56domain here at the C terminus.
  • 07:59So this was really the first well
  • 08:02characterized chromatin regulator
  • 08:03that is known to drive tumor genesis.
  • 08:07And so that was really why in the
  • 08:10early early 2000s a lot of labs
  • 08:12jumped on this to thinking maybe
  • 08:14this would give us some insight into
  • 08:16chromatin based mechanisms and cancer.
  • 08:19So to summarize a lot of work that
  • 08:21we did talk thinking about cells
  • 08:23of origin of of this type and
  • 08:25other types of leukemia.
  • 08:26Not going to get into that too
  • 08:27much today because I want to
  • 08:29get to the therapeutic part.
  • 08:30But we were able to show that the
  • 08:31MLO fusion when we put it into either
  • 08:33stem cells or progenitor cells,
  • 08:35either mouse or human,
  • 08:36that the MLO fusion can drive
  • 08:38the development of leukemia from
  • 08:40multiple different cell types
  • 08:41in hematopoietic development.
  • 08:43And these concepts have now been
  • 08:45shown in other types of tumors.
  • 08:46But at the time that was a
  • 08:48relatively new concept.
  • 08:49But what more importantly what it
  • 08:50let us do is really characterize
  • 08:52the gene expression program that's
  • 08:54driven by this MLL fusion protein
  • 08:57when we put it into in this case
  • 08:59a mouse progenitor cell.
  • 09:00And we could look very quickly to
  • 09:02see what types of gene expression and
  • 09:05chromatin based changes happened when
  • 09:07the MLL fusion binds to chromatin.
  • 09:09And our work and many,
  • 09:11many people's work across the
  • 09:13the world with this model
  • 09:14originally developed by Mike Cleary's
  • 09:16group have I would say that MLO
  • 09:19fusion driven leukemia now is about
  • 09:20as well characterized mechanistically
  • 09:22this as any type of leukemia,
  • 09:25probably any type of cancer
  • 09:27largely because of this model.
  • 09:28So we know where the fusion binds throughout
  • 09:31chromatin, which genes it controls.
  • 09:33We now have mechanisms of turning the fusion
  • 09:35off and we can see what genes get shut off.
  • 09:37We understand now quite a bit
  • 09:39about what the MLL fusion does.
  • 09:42Exactly how it does it is
  • 09:44still a little bit unclear,
  • 09:46but that is what we and others
  • 09:48are really working on now.
  • 09:50And of course that is what we
  • 09:51need to know and under in order
  • 09:53to develop hopefully therapeutics
  • 09:55that can target these mechanisms.
  • 09:57So this is a very simple actually
  • 10:00summary of how the MLL fusion works.
  • 10:03So here in Gray is the ML AF9 fusion,
  • 10:06the in terminus of MLL bound to
  • 10:08some of the proteins normally found
  • 10:10in the MLL complex Menon and Ledge
  • 10:13F here and those help localize
  • 10:15the fusion protein to chromatin.
  • 10:17And then the C terminal part of the
  • 10:19fusion brings in a number of complexes
  • 10:22is histone methyl transferase .1 L,
  • 10:23which is a histone H3 lysine
  • 10:2679 methyl transferase And this
  • 10:28so-called super elongation complex,
  • 10:30which is really a fundamental complex
  • 10:33for controlling transcription broadly,
  • 10:35not just in this setting.
  • 10:36Certainly the MLL fusion drags these
  • 10:40chromatin regulatory and transcriptional
  • 10:42control proteins and complexes to its
  • 10:45target genes to drive gene expression.
  • 10:48So with that level of understanding,
  • 10:51it became easier to go to pharma and
  • 10:53biotech and to get them interested
  • 10:55in developing small molecules that
  • 10:57might target these mechanisms.
  • 10:58Even though at the time no one knew
  • 11:01if these were mechanisms be relevant
  • 11:03beyond this relatively rare disease
  • 11:05which is probably A couple thousand
  • 11:07patients per year in the United States.
  • 11:09But I'll show you,
  • 11:10I think we think that indeed and
  • 11:12in fact we have now shown in in
  • 11:14patients that it it actually is.
  • 11:16So each, as I mentioned,
  • 11:17each of these labels in red is a small
  • 11:20molecule that's been developed to target
  • 11:22various components of this large complex.
  • 11:25And to summarize broadly on molecules
  • 11:28that target the complexes on the right,
  • 11:30the kind of general
  • 11:32transcriptional complexes,
  • 11:33the problem there has primarily been
  • 11:35toxicity that actually not too surprising
  • 11:38you turn off transcription broadly,
  • 11:40we can do that with some of
  • 11:41our chemotherapeutic drugs.
  • 11:42It's relatively toxic on the
  • 11:44left side and to date the problem
  • 11:47has actually been efficacy,
  • 11:49meaning we can do pre clinical studies
  • 11:51and I'll briefly load to this with
  • 11:53.1 actually get really impressive
  • 11:55changes in gene expression and such.
  • 11:57But then we go into patients with
  • 11:59the .1 inhibitor get a little bit
  • 12:02of clinical signal and but unable
  • 12:04to maintain that clinical response.
  • 12:07But but toxicity has not primarily
  • 12:09been a problem for the molecules
  • 12:11on the left side of this figures
  • 12:13and that's kind of where we've
  • 12:15focused. And I'm going to talk a
  • 12:17lot about this protein Menon here,
  • 12:19which is really a scaffolding protein
  • 12:22that's bound to the MLL fusion
  • 12:24and helps keep it on chromatin.
  • 12:25I'll show you more about that in a minute.
  • 12:28Men and as an important part of the
  • 12:30Amylo complex was first demonstrated
  • 12:32in 2004 by Michael Cleary's group.
  • 12:34And now there are many small molecules.
  • 12:36I'll talk about the one we've been
  • 12:38working on that disrupt this interaction.
  • 12:40I'll show you how in a minute.
  • 12:42And that really that concept,
  • 12:44the first chemical biology done around
  • 12:46this was done by Yolanda Grimbeck is due
  • 12:49in Michigan where they developed the
  • 12:51first small molecule to bind to minute.
  • 12:53So just quickly I'm going to
  • 12:55this is kind of jumping ahead,
  • 12:56but it's a concept that I think is
  • 12:59important in terms when we think
  • 13:01about targeting chromatin complexes
  • 13:02in leukemia or other diseases.
  • 13:04So if this is a even more simplified
  • 13:07view of the ML AF9 bound to chromatin
  • 13:10through its adapter proteins and
  • 13:11bring it in this case the .1 complex.
  • 13:14I told you that we worked actually
  • 13:16over a decade ago now with a
  • 13:19company called Epizyme to make small
  • 13:21molecule enzymatic inhibitors of .1.
  • 13:23We've shown with genetic studies
  • 13:25that that is an important component
  • 13:27of this complex and that histone
  • 13:29modification that it put places
  • 13:31is important for controlling MLO
  • 13:33fusion driven gene expression.
  • 13:35And that went into clinical trials and
  • 13:37we were able to a couple of patients
  • 13:39actually went into to remission
  • 13:41and many had some response but it
  • 13:43they were all pretty transient.
  • 13:44And I'm going to show you that
  • 13:47that's not the case with the min an
  • 13:49inhibitor and this summarizes why
  • 13:51we think that is that if you the .1
  • 13:54L inhibitor inhibits the enzymatic
  • 13:55activity of this protein,
  • 13:57but the whole complex remains stuck
  • 13:59on chromatin.
  • 14:00So it's actually doesn't take a
  • 14:02lot for the cell to figure out how
  • 14:04to deal with the fact that that
  • 14:06enzymatic activity is no longer there.
  • 14:08Whereas what I'm going to show
  • 14:09you with the MLL Menin inhibitor,
  • 14:10it actually disrupts the whole complex
  • 14:13and much of the complex lifts and
  • 14:15the fusion protein lifts off of
  • 14:17chromatin and we think that probably
  • 14:19is why the the Menin inhibitor
  • 14:22approach is more dramatic.
  • 14:24So in 2019 we published the small
  • 14:26molecule that we were working on which
  • 14:28was at the time called BTP 5O469.
  • 14:31The version of it that's in the clinic
  • 14:33is called Syndex 5613 or Revu Minib.
  • 14:35Get to that in a minute.
  • 14:37And this is a very potent small
  • 14:39molecule that binds to this pocket on
  • 14:41men and this is the men and protein.
  • 14:43This is the crystal structure of this
  • 14:45protein here in purple and blocks
  • 14:48the interaction between men and
  • 14:51and the MLL fusion and that leads
  • 14:53to as I mentioned the loss of this
  • 14:55complex on chromatin,
  • 14:56but interestingly enough only
  • 14:58does it at very selected sites.
  • 15:00This complex actually remains on on
  • 15:03chromatin and the wild type complex which
  • 15:06would also be potentially disrupted
  • 15:08by this molecule remains on chromatin
  • 15:11at many sites throughout the genome.
  • 15:13But there's a very selected group
  • 15:16of genes loci where it's lost.
  • 15:18The importance of that is,
  • 15:19is that we know genetic inactivation of MLL
  • 15:23broadly is toxic that to hematopoietic cells.
  • 15:26But there's some reason that this
  • 15:28mechanism seems to be only important
  • 15:31at certain developmental loci.
  • 15:33And we think that may be why we're not
  • 15:35seeing tremendous toxicity that you
  • 15:37could imagine by lifting this chromatin
  • 15:39complexes off the genome broadly.
  • 15:41And this is just one example of
  • 15:43a preclinical PDX study that we
  • 15:46did with an MLL rearranged AML.
  • 15:48All of you probably know what
  • 15:49PDX studies are,
  • 15:50inject the human leukemia into
  • 15:52immunodeficient mice and treat the mice here
  • 15:55with two cycles if you will of of therapy.
  • 15:57And the beauty of this experiment is
  • 16:00from a technical standpoint is Syndax
  • 16:03actually supplies us with mouse chow
  • 16:06that's impregnated with the drugs.
  • 16:08So you don't actually have to dose the mice.
  • 16:11People in the lab love that fact.
  • 16:13So basically you inject the leukemia,
  • 16:15change the food and come back in
  • 16:16a month and see what happened.
  • 16:18And it's obviously not quite that simple,
  • 16:19but so we were able to do these experiments
  • 16:22with much more speed and much less pain
  • 16:25than they than they often can take.
  • 16:27And so you can see here that the the
  • 16:29mice that were treated with the the MIN,
  • 16:31an inhibitor essentially the AML
  • 16:33in this PDX model was eradicated,
  • 16:36which as most of you know who
  • 16:38do these types of experiments.
  • 16:40That's not the how these experiments
  • 16:42usually go.
  • 16:43You might see a prolongation of survival
  • 16:46but not this degree of response.
  • 16:49Same thing in a model that a lot
  • 16:51of people use,
  • 16:52this retroviral model of ML AF9
  • 16:54that we've used a lot as well,
  • 16:55which does lead to a very aggressive
  • 16:59mouse AML.
  • 17:00And you can see here,
  • 17:02we've treated mice that have gotten
  • 17:04these cells injected with the,
  • 17:06the min,
  • 17:06an inhibitor for this period of
  • 17:08time and in fact,
  • 17:08it eradicated the disease in this model.
  • 17:11So that is again very different from
  • 17:13what most of the graphs will look like
  • 17:16when you do an experiment like this.
  • 17:18So really indicating that
  • 17:20there's significant potential.
  • 17:22So I'm going to switch diseases or
  • 17:24switch subtypes of leukemia and tell
  • 17:26you a little bit about a different
  • 17:28subtype that one is interesting and
  • 17:31two actually kind of changed the
  • 17:33level of interest from biotech and
  • 17:35pharma because it's way more common
  • 17:38and that's NPM one mutant AML.
  • 17:40And I keep saying that because as
  • 17:43you can probably tell in paediatrics
  • 17:44and other rare cancers,
  • 17:46we're constantly and many of you
  • 17:48probably recognize this when we're
  • 17:50talking about drug discovery and development,
  • 17:52having to convince people to work
  • 17:54on the diseases we're interested in
  • 17:56because they're indeed quite rare.
  • 17:58When you go from 1000 to 2000 patients per
  • 18:00year in the United States to 15 into 17,000
  • 18:03patients per year in the United States,
  • 18:05you actually get a lot more interest.
  • 18:07And so that's one of the
  • 18:09reasons beyond just the the,
  • 18:11the mechanistic interest that
  • 18:12I think this is important.
  • 18:14So Michael Kuhn, when we were in New York,
  • 18:17a fellow at the time asked
  • 18:20the question of well, OK,
  • 18:21we know that the MLO rearranged leukemias.
  • 18:23This is a gene expression data for
  • 18:25the HOX genes and MIS ones from Tim
  • 18:28LAYS group in a bunch of AM LS,
  • 18:30and you probably know how to read these.
  • 18:31Each row here is AG in each
  • 18:33column with leukemia sample.
  • 18:34And here's the MLL rearranged group OX,
  • 18:37A cluster we know that's long known.
  • 18:39In fact, some of the microarray data
  • 18:41that I did as a fellow demonstrated
  • 18:43that the Hox A cluster is expressed in
  • 18:46MLL rearranged leukemias as is MIS one.
  • 18:48Those are two targets of the ML effusion
  • 18:51that have been worked on for many decades.
  • 18:54It turns out in the NPM one mutant
  • 18:56leukemia as you can see here on the right,
  • 18:58they have the Hox A cluster as
  • 19:01well and B cluster,
  • 19:02another Hox cluster and MIS one.
  • 19:04So this has also been known for some
  • 19:07time that this subset of AML and PM one
  • 19:09mutant also tends to express the Hox genes.
  • 19:11So Michael using CRISPR which in
  • 19:152016 was still relatively new did
  • 19:18a what's called a domain scan.
  • 19:19I won't get into the details of that,
  • 19:21but the point being that the inactivation
  • 19:24of MLL and ultimately I mean of men
  • 19:27and MLL led to differentiation and
  • 19:30ultimately death of NPM one mutant AML cells.
  • 19:33And the concept being at the time
  • 19:36that the disruption of MLL and
  • 19:38Menon in the NPM one state somehow
  • 19:41leads to a decrease in the Hox gene
  • 19:44expression and therefore ultimately
  • 19:46a enemies one expression and therefore
  • 19:49ultimately changes in gene expression.
  • 19:52Hannah Okleman,
  • 19:52who was a a fellow in the lab just
  • 19:54recently moved back to Germany
  • 19:56to start her own lab,
  • 19:57then demonstrated that similar to
  • 20:00the MLL fusions that NPM 1 mutations
  • 20:04in mouse models could transform
  • 20:06multiple hematopoietic cell types,
  • 20:09stem cells a little bit more significantly
  • 20:11more efficiently than progenitors,
  • 20:12but also could transform progenitors
  • 20:15as well and.
  • 20:17Needless to say,
  • 20:18there was still a question as to kind of
  • 20:21mechanistically how all this was working.
  • 20:23I'm going to summarize some chip seek
  • 20:26data here that really shows that at
  • 20:29least at this level of understanding,
  • 20:32it's quite similar to what we see
  • 20:34with the MLL rearranged leukemia.
  • 20:37So these are NPM one mutant cell
  • 20:39lines where we're doing chromatin
  • 20:41immunoprecipitation for men and for
  • 20:43example either in a control setting in
  • 20:47black PMSO treated cells or the min
  • 20:49inhibitor treated cells and you can see
  • 20:51that min and comes off of chromatin.
  • 20:53We know that when we treat
  • 20:54with the min inhibitor
  • 20:55min and lifts off of
  • 20:57chromatin MLL shown here.
  • 20:58Now this is wild type MLL because
  • 21:01it's a mutant in PM one setting
  • 21:03doesn't come off the Hawks loci.
  • 21:04We'd seen that in the MLL fusion setting
  • 21:07as well but does come off of MES 1.
  • 21:09So this is that concept that I show you
  • 21:12that certain loci respond differently to
  • 21:14the min and inhibitor and this is showing
  • 21:17the RNA SEC or the gene expression.
  • 21:20And you can see that when there's
  • 21:22a correlation between loss of MLL
  • 21:24occupancy and loss of gene expression
  • 21:26and this is that looking at that more
  • 21:28broadly by what's called a GSDA analysis.
  • 21:30So the point being that much like what we saw
  • 21:33in the MLL fusions in the NPM one setting,
  • 21:36you treat with the men inhibitor
  • 21:37men and comes off of chromatin.
  • 21:39The MLL protein itself comes off of
  • 21:41chromatin at about 100 to 150 loci
  • 21:43and those genes lose their expression
  • 21:46when the cells differentiate.
  • 21:48And then if we go into PDX studies to
  • 21:51NPM one mutant flip 3 ITD Co mutant,
  • 21:54these are relatively aggressive AM LS.
  • 21:57You can see again pretty impressive
  • 22:00response in the PDX setting.
  • 22:02And for the people who start do these
  • 22:04types of experiments in the audience,
  • 22:07we actually as you probably know the
  • 22:09standard way to do this is put in
  • 22:11leukemia into an immunodeficient mouse,
  • 22:13the first sign of any leukemia in
  • 22:14the peripheral blood of those mice,
  • 22:15you start treating them.
  • 22:17We actually waited in this experiment
  • 22:19on the right until the mice were ill
  • 22:21and started treating them and actually
  • 22:25we could recover essentially 4 out of
  • 22:27five of the mice and eradicate the disease.
  • 22:30So we stacked the deck against us and
  • 22:32we're still able to to make that or
  • 22:35extend the the survival of those mice.
  • 22:37So to summarize this at this point
  • 22:39we the MIN inhibitor and NPM one
  • 22:42mutant AML induces differentiation,
  • 22:44reverses leukemia,
  • 22:46genic leukemic gene expression,
  • 22:48certain genes like MIS one removes
  • 22:51MLL from those loci and we get
  • 22:54dramatic responses.
  • 22:54So that was exciting and that was
  • 22:57enough to get Janssen and Ichi
  • 22:59and the various other large
  • 23:01pharmaceutical companies interested in,
  • 23:03in small molecule development
  • 23:05of Menon inhibitors.
  • 23:06But it's still there was still
  • 23:08something here we don't quite or didn't
  • 23:12quite understand is why is it that
  • 23:14the NPM one mutant AML is depending
  • 23:17so much on the MLL Menon complex.
  • 23:20So Hannah dug into that as well.
  • 23:22You guys probably know this,
  • 23:23but this is just a little bit
  • 23:25about the mutant NPM one protein.
  • 23:27It's shown here.
  • 23:29Schematically,
  • 23:30it's found mostly in the nucleolus
  • 23:33in the wild type setting,
  • 23:35but when the mutation occurs,
  • 23:37it's a mutation in this nuclear
  • 23:39or localization signal that then
  • 23:41leads to a nuclear export signal.
  • 23:43So the mutant in PM one is largely
  • 23:45found in the cytoplasm and that
  • 23:47was recognized by the people who
  • 23:49initially described this mutation.
  • 23:51However,
  • 23:52there is some
  • 23:54that remains the mutant in PM one
  • 23:57in the nucleus and we've taken
  • 23:59advantage of a system that many of
  • 24:01you probably know about where you
  • 24:04can now by CRISPR mediated homologous
  • 24:06recombination actually tag if you will,
  • 24:10whatever gene or protein of interest with a
  • 24:13degradable version of FK PP12 shown here.
  • 24:16And actually this cell line was made
  • 24:18by Peggy Goodell's group in at Baylor
  • 24:21and you have a mutant NPM one protein
  • 24:23that has this degradable version
  • 24:25of F KBP 12 and you can treat them
  • 24:27with a small molecule Protac that
  • 24:29will degrade the whole thing.
  • 24:30So you can degrade the mutant oncoprotein
  • 24:33and look fairly rapidly after degradation
  • 24:35as to what's happening and here's
  • 24:37how rapidly you get degradation.
  • 24:39By 60 minutes you've got about half
  • 24:41of the mutant protein gone and by 120
  • 24:44minutes essentially all of us gone.
  • 24:45So it's relatively rapid and these
  • 24:47types of experiments are quite
  • 24:49illuminating because you really have
  • 24:51very tight control over over the system.
  • 24:56And what we see is that we get
  • 24:59differentiation when we degrade the
  • 25:01mutant onca protein and ultimately
  • 25:03apoptosis of the cells as well.
  • 25:05And this is just the Western blot looking.
  • 25:07We can actually separate the mutant
  • 25:09protein from the wild type because
  • 25:11it's tagged,
  • 25:11it's here and you can see that here.
  • 25:14This is the cytoplasmic prep,
  • 25:15the nuclear prep and the chromatin
  • 25:17prep and here's the mutant in PM one
  • 25:19and we can control that by degrading
  • 25:21it to show that that signal actually
  • 25:23is what we think it is.
  • 25:25And in fact there is a fair amount
  • 25:27of the mutant in PM one in the
  • 25:28nucleus and on chromatin.
  • 25:30Then if we do chip seek to say
  • 25:32where is it in the nucleus and
  • 25:34where are where is it on chromatin.
  • 25:36You can see here with with two different
  • 25:39antibodies in black that the NPM one mutant,
  • 25:42NPM one protein is bound to many
  • 25:44of the similar genes that we've
  • 25:46learned about with the MLL fusion,
  • 25:48the Hox cluster MIS one.
  • 25:50And when we degrade it that signal goes away.
  • 25:52And I keep saying that because
  • 25:54particularly with chip seek experiments,
  • 25:55the opportunity for background signal
  • 25:57is real and this is makes you feel
  • 26:00much better that the signal that
  • 26:02you're looking at is indeed the
  • 26:04signal that you are interested in.
  • 26:05And now you actually can go to primary
  • 26:07patient samples with those antibodies
  • 26:09and see the NPM one protein mutant
  • 26:11NPM one protein bound there as well.
  • 26:13And here's the list of the top 50
  • 26:15or so genes to which the mutant
  • 26:17NPM one protein is bound.
  • 26:18And you can see as some of these genes
  • 26:20that I've already talked about OX,
  • 26:22A&B cluster and a number of other genes
  • 26:24that we tend to pay attention to and
  • 26:26stare at when we're talking about MLL or
  • 26:29looking at MLL rearranged leukemias as well.
  • 26:33So,
  • 26:34but is it controlling gene expression.
  • 26:36So now we have mutant
  • 26:38NPM one protein bound to
  • 26:40interesting sites in on chromatin
  • 26:42and we can degrade it and show
  • 26:44that that signal is specific.
  • 26:46And now what happens to transcription?
  • 26:48So this was 24 hours later,
  • 26:50quite a bit later.
  • 26:51Most of those genes that I just showed
  • 26:53you where the NPM one protein is bound,
  • 26:55their expression is down and
  • 26:57this is an approach called pro
  • 26:59seek which I won't get into the
  • 27:00details as to how one does this.
  • 27:02So many of you,
  • 27:03some of you who work on transcription
  • 27:04probably know this technique.
  • 27:06But essentially it measures the
  • 27:08amount of bound RNA polymerase 2 out
  • 27:10throughout the length of the gene as a
  • 27:12surrogate for transcriptional activity.
  • 27:15And what we can see that as quickly
  • 27:17as 30 minutes after treatment of the
  • 27:19cells with the NPM one protein degrader,
  • 27:22you're already seeing a decrease in
  • 27:24transcription at those sites where
  • 27:26the NPM one was previously bound.
  • 27:28And and if you look at it across
  • 27:30the the the all express genes
  • 27:32you don't see those changes.
  • 27:33So in fact it's the NPM one protein's
  • 27:37bound there and somehow controlling
  • 27:40transcription of these genes.
  • 27:42And one of the ways it's doing
  • 27:44it is by keeping RNA Pol two,
  • 27:46CK nine that that super elongation
  • 27:48complex that I told you is critical for
  • 27:50transcription bound to those genes.
  • 27:52So when we treat with the degrader
  • 27:54NPM one comes off and then much
  • 27:56of the transcriptional apparatus
  • 27:58comes off of those genes as well.
  • 28:00So it's maintaining a state that allows
  • 28:03for those critical complexes including
  • 28:06pole two to to bind to those low side.
  • 28:09I'm going to go through the details of
  • 28:12this but just to kind of summarize it
  • 28:15that when we degrade mutant in PM one,
  • 28:17we lose RNA polymerase two occupancy
  • 28:19where the NPM one was previously
  • 28:21bound within an hour.
  • 28:23So off goes NPM one,
  • 28:24off comes pole two and a number
  • 28:26of other histone modifications
  • 28:28like H3K27 acceleration,
  • 28:29some of you know associated with various
  • 28:32types of gene expression decreases
  • 28:34and then the histone modification,
  • 28:36other histone modifications start
  • 28:38to decrease somewhat later.
  • 28:39For those of you interested in transcription,
  • 28:41we can talk more about this in detail later,
  • 28:43but it looks like it's like when
  • 28:45we degrade the mutant in PM one,
  • 28:47the decrease in gene expression
  • 28:48is actually biphasic.
  • 28:49There's so initially there's a decrease
  • 28:52of about 50% probably because pole 2
  • 28:55is not quite as there's not as much
  • 28:58pull two and other complex occupancy.
  • 29:00And then after about 3 days we
  • 29:02see a dramatic another dramatic
  • 29:04decrease in gene expression.
  • 29:06We think that's because now the
  • 29:09histone modifications are starting
  • 29:12to come in and and work together
  • 29:14with whatever the previous mechanism
  • 29:15was to fully shut off transcription.
  • 29:20So how does this connect to to Menon?
  • 29:26So
  • 29:31there we go. So we now if you
  • 29:33treat with the MIN inhibitor,
  • 29:36what happens to this chromatin bound in
  • 29:39mutant in PM one and I'll just quickly
  • 29:42summarize it by saying you can see here
  • 29:45here's the mutant in PM one we treat
  • 29:47with the MIN inhibitor doesn't come
  • 29:49off the hogs locus but it actually
  • 29:50does come off of the mis one locus
  • 29:52exactly where we're seeing MLL come off,
  • 29:54same thing down here and if you compare
  • 29:57that the gene expression again those are
  • 29:59the genes that are losing expression.
  • 30:02So to summarize what I'm saying here is
  • 30:04that when we degrade the mutant NPM one
  • 30:07protein with this degrader molecule,
  • 30:09we lose RNA pole two CDK 9 and
  • 30:12ultimately .1 at those loci.
  • 30:14When we treat with the min inhibitor we
  • 30:16do the same thing but at a subset of
  • 30:19the loci where the NPM one is bound.
  • 30:21So very similar to what's happening
  • 30:24with the MLL fusion.
  • 30:25However, we're now looking to see if
  • 30:28indeed the mechanisms are identical.
  • 30:30And it turns out that while some
  • 30:31of the complexes are overlapping,
  • 30:33these are the mechanisms are
  • 30:35not perfectly identical.
  • 30:37That is,
  • 30:37some complexes are important
  • 30:39in the MLO fusion,
  • 30:41not an NPM one and vice versa.
  • 30:42So we're trying to work through those
  • 30:45details because as you can imagine,
  • 30:47the next step that we want to do
  • 30:48is come in and target some of these
  • 30:50other complexes with small molecules.
  • 30:51So to summarize this part before,
  • 30:53now I move to the clinical translation.
  • 30:56There are a subset of leukemias that have
  • 30:59high level Hox gene expression MIS one,
  • 31:01another transcription factor
  • 31:02called PBX three.
  • 31:04This actually accounts for about
  • 31:0640% of human AML and it's ones
  • 31:10with these genetic abnormalities,
  • 31:13MLO rearrangement,
  • 31:14NPM 1 mutation and I'm actually more
  • 31:19relatively rare but more common than
  • 31:21in adults rearrangement called Newt 98
  • 31:24rearrangements in both pediatric and adult.
  • 31:26So again accounting for
  • 31:28about 40% of patients.
  • 31:31So that all while all that was happening,
  • 31:35Syndax and another company called cure
  • 31:37oncology that many of you know about,
  • 31:40we're developing small molecules
  • 31:42here Syndax 5613 and here Cure's
  • 31:46what's now Zyftominib and these
  • 31:47I'll show you some of this,
  • 31:49the data from the Cindex trial
  • 31:52and some mechanism of resistance.
  • 31:54And as I mentioned essentially
  • 31:56when the NPM one story came out,
  • 31:59we were called by Janssen and actually
  • 32:01another couple of other pharmaceutical
  • 32:03companies and at least these three
  • 32:05now have MIN inhibitors that are
  • 32:06right on the tails if you will of
  • 32:08the cure of development and Cindex.
  • 32:11So this is so now we're into
  • 32:15patients with the Syndex 5613.
  • 32:17This is just date some of
  • 32:19the data from the phase one.
  • 32:21This is one of the first patients
  • 32:23that was treated at Dana Farber and
  • 32:26we were able to get the peripheral
  • 32:27blood and here are the blast.
  • 32:29And you can see this is with
  • 32:31Revumenib that day three,
  • 32:32not much has happened.
  • 32:33Day seven start to see a decrease
  • 32:35in peripheral blast,
  • 32:36day 14 further decrease and by day 30
  • 32:39at least the peripheral blood blasts
  • 32:41are in this case essentially gone.
  • 32:44Florian Perner is a postdoc
  • 32:45who was doing this.
  • 32:46He sorted these cells and looked
  • 32:48at gene expression and in fact the
  • 32:50gene expression changes that we see
  • 32:52here look very similar to what we
  • 32:53had seen in the preclinical studies
  • 32:56not being a clinical trialist.
  • 32:58I'm going to summarize the whole
  • 33:00phase one right here with a lot
  • 33:02of work from a lot of people and
  • 33:04this was published last year.
  • 33:07This is the Revue Minib Phase
  • 33:09one with Syndex 5613.
  • 33:10The other name for it,
  • 33:1268 patients with relapsed
  • 33:15refractory leukemia.
  • 33:16As you probably know,
  • 33:17many of these patients have had
  • 33:19tremendous numbers of cycles of
  • 33:21various types of therapies and the
  • 33:24CR rate depending on how you count
  • 33:26CRS and for those of you who do
  • 33:27clinical trials and A and all we can
  • 33:29talk about that is somewhere in the
  • 33:3040% range with an overall response
  • 33:32rate of about 50% and a median
  • 33:35duration of response about nine months.
  • 33:37So for in relapse refractory setting,
  • 33:39these are actually pretty impressive
  • 33:42numbers and the cure oncology
  • 33:44small molecule seems to be doing
  • 33:47something having similar activity.
  • 33:48And in fact some of that data from
  • 33:51Janssen was also just printed presented
  • 33:53at ASH and it looks like the activity
  • 33:55of that molecule is is similar.
  • 33:57So indeed there looks like
  • 33:59there's significant clinical
  • 34:01activity of this approach.
  • 34:02This lot slide just reminds me to
  • 34:05point out and and then now I'm talking
  • 34:08to people who are doing AML clinical
  • 34:10trials in the in the audience.
  • 34:12An interesting phenomenon that
  • 34:13you know better than I do,
  • 34:16but that is influencing how these
  • 34:17drugs are are able to be developed
  • 34:19and that is when you treat patients
  • 34:21with them in an inhibitor,
  • 34:23many of them will develop this syndrome
  • 34:25called differentiation syndrome.
  • 34:27But in this setting it looks a little
  • 34:30different clinically I'm told,
  • 34:31than the differentiation syndrome
  • 34:32that you usually see when you
  • 34:34treat patients with acute per
  • 34:36myelocytic leukemia with ATRA.
  • 34:37In fact patients have died from
  • 34:40this differentiation syndrome and
  • 34:42so that has prompted the FDA to
  • 34:44call this a dose limiting toxicity.
  • 34:46Think about what that means.
  • 34:47It means that your your your dose limiting
  • 34:51toxicity is actually occurring as a
  • 34:53result of efficacy of your molecules.
  • 34:56So we can talk about what So I think
  • 34:58the FDA is fighting against the the
  • 35:00the some things that they shouldn't be,
  • 35:02but that's a that's a whole other soapbox
  • 35:05that we can talk about if we want to.
  • 35:07That would be like for those of you
  • 35:08who treat patients with ALL saying you
  • 35:10start to see a little tumor lysis syndrome,
  • 35:11we'd better stop treating them
  • 35:13because that's bad. No, that's good.
  • 35:16So with all this in mind and the
  • 35:19clinical activity looking interesting,
  • 35:21we figured that it was much like
  • 35:23any single targeted agent,
  • 35:24there was likely to be some mechanism
  • 35:27of resistance to that targeted agent.
  • 35:29And right about the time we started
  • 35:31thinking about this the Broad Institute
  • 35:33developed this screening based on or
  • 35:36screening approach based on single
  • 35:38nucleotide base editing which in fact
  • 35:40what you can do is tile in this case
  • 35:44Menon the whole length of the gene
  • 35:47with guides that will mutate not
  • 35:49every nucleotide because of the way
  • 35:51that it's designed but where you can
  • 35:53mutate the majority of amino acids
  • 35:55across the length of that protein
  • 35:59to to basically do an in a
  • 36:02cellular mutagenesis screen
  • 36:03to see if you can phenotypes.
  • 36:06So what Florian decided to do is to
  • 36:08get that base editor library made
  • 36:10for the minute gene treat cells with
  • 36:13the min an inhibitor and see if there
  • 36:16were mutations that made the cells
  • 36:18resistant to the min an inhibitor.
  • 36:21And in fact there were there shown here
  • 36:23in two different MLL rearranged lines
  • 36:25and interestingly enough we we kind of
  • 36:29looking back this was probably silly.
  • 36:31We looked at this,
  • 36:32the new technique and there's a
  • 36:34little bit of noise and we didn't
  • 36:36know exactly what to make of
  • 36:38it looked kind of interesting.
  • 36:39Florian put it in the drawer and kind
  • 36:41of didn't do too much more with it
  • 36:43until we got a call from Ross Levine
  • 36:46and Etan Stein at Memorial Sloan Kettering.
  • 36:48Actually,
  • 36:49I got a like emergent text from Ross
  • 36:51which I thought something really bad
  • 36:52had happened and he said we have to talk now.
  • 36:55So I called him and he said we
  • 36:57found mutations in Menin in samples
  • 36:59from patients that have progressed
  • 37:01on the Menin inhibitor.
  • 37:03And great, you know,
  • 37:05what are they?
  • 37:06And in fact the first one was this
  • 37:08mutation 3 andine 349.
  • 37:10So we dug Florian's data out and
  • 37:13we're like holy cow,
  • 37:14the patients are getting the same
  • 37:16mutation that the base header screen
  • 37:18had suggested they might get even though
  • 37:21we weren't confident in our in our data
  • 37:23to go ahead and start studying that.
  • 37:25But Needless to say with that information,
  • 37:27we started studying this in quite a bit
  • 37:30of detail and we went to send X and
  • 37:32got samples from a number of patients.
  • 37:34And it looks like within two to
  • 37:37three months about 40% of the
  • 37:40patients had developed.
  • 37:41They weren't in fluorid
  • 37:42relapse or progression,
  • 37:43but had developed a clone with
  • 37:46this a min and mutation in them.
  • 37:49That's what this is showing here and
  • 37:50here is like and those mutations
  • 37:52were not present at screening.
  • 37:53So this is just a pie chart in
  • 37:55red here showing you the size of
  • 37:57the clone that has developed the
  • 37:58min and mutation.
  • 37:59So this is acquired selective
  • 38:02mutational resistance to the men,
  • 38:04an inhibitor which as most of
  • 38:06you probably know is considered
  • 38:08a validation of the therapeutic
  • 38:10targeting the kinase world when this
  • 38:12happens and essentially we think
  • 38:14it's saying the same thing here and
  • 38:16we found a both in patients with MLO
  • 38:18rearranged an NPM one mutant AML.
  • 38:20So this is the, if you will,
  • 38:22the gold standard for that validation
  • 38:24of a therapeutic target in patients
  • 38:26that that that you put so much pressure
  • 38:29on the target that the cancer mutates.
  • 38:31It's such that it's no longer effective.
  • 38:33I'll show you why in a minute.
  • 38:35And of course as most of you know,
  • 38:37we can have to have combinations
  • 38:39anyway and we were able to show that
  • 38:41we can do the same thing in PDX models.
  • 38:43So we take our MLL rearranged
  • 38:45or NPM one mutant models,
  • 38:46treat them with in an inhibitor and
  • 38:49in some cases but not all they will
  • 38:52develop the mutations that we see in
  • 38:55in the patients and mechanistically
  • 38:57we we know how this is working.
  • 38:59I won't go into all the details but
  • 39:01this is just one chip seek experiment.
  • 39:03In the wild type setting you can
  • 39:05see Menin comes off of chromatin,
  • 39:06this is chip seek increase in
  • 39:08concentrations of the Menin inhibitor.
  • 39:10But if you have mutated Menin in that
  • 39:12cell line, it no longer comes off.
  • 39:14And we know now biochemically it's
  • 39:17because the binding affinity of the
  • 39:19Menin inhibitor has been shifted
  • 39:21significantly as a result of those mutations.
  • 39:23And in fact,
  • 39:24we know this now at the crystal
  • 39:26structure level and we know
  • 39:28exactly why that's the case.
  • 39:29So here's revuminib bound to Menin.
  • 39:32You can see over here on the right,
  • 39:35these amino acids
  • 39:39M327T349-G331, all these are mutations
  • 39:40that have been found in patients.
  • 39:44Interestingly enough,
  • 39:45the wild type MLL protein does not
  • 39:49use those amino acids to anchor
  • 39:51and that's actually an important
  • 39:53concept because if you develop
  • 39:55the mutation that where men and
  • 39:58the MLL can no longer interact,
  • 40:00those cells won't survive that.
  • 40:02So that that that's not an
  • 40:03option for the cells.
  • 40:04They have to mutate something that
  • 40:06doesn't affect MLL but does affect
  • 40:08the the binding of the inhibitor.
  • 40:10And in fact that's exactly what they've done.
  • 40:12They've mutated this region of men and
  • 40:15that has plays no role in MLL binding.
  • 40:18And the way that that happens is
  • 40:20essentially right here the yellow
  • 40:22is the the min inhibitor bound to
  • 40:24wild type min and the purple is the
  • 40:27min inhibitor bound to mutant min.
  • 40:28And and you can see essentially what's
  • 40:31called a steric clash which so the the,
  • 40:33the min inhibitor is pushed out a
  • 40:35little bit here because of these
  • 40:37changes in the amino acid and that
  • 40:40leads to a 10 to 100 fold decrease
  • 40:42in affinity of this molecule.
  • 40:44So this is I find this amazing
  • 40:47because it's rare that you get to see
  • 40:50molecularly the difference between
  • 40:51response and resistance which is really
  • 40:54essentially a few angstroms here of
  • 40:57this Menon inhibitor binding to to Menon.
  • 41:01So this we're we're continuing to work
  • 41:04on mechanisms of resistance in the past
  • 41:06five or last five or 10 minutes here.
  • 41:08I'll tell you about some others,
  • 41:11but just getting back to a little
  • 41:13bit to that comment I made about
  • 41:15dose escalation and stopping your
  • 41:18dose escalation before you perhaps
  • 41:20get to full potential efficacy.
  • 41:22And now we see that we're developing or
  • 41:25patients are developing mutations that
  • 41:27all they do is shift the curve a little bit.
  • 41:31It does make you wonder if you'd had a
  • 41:33higher dose and a higher concentration
  • 41:35earlier on if you might have prevented
  • 41:38the cells from developing those mutations.
  • 41:40So we with send X luckily providing
  • 41:43us food now with varying amounts
  • 41:45of the min an inhibitor.
  • 41:47We were able to do a dose response
  • 41:49experiment in a PDX model with increasing
  • 41:52concentrations of the min an inhibitor.
  • 41:54And you can see here that at
  • 41:56the lowest concentration point,
  • 41:58O3 3%, you see no response.
  • 42:01At the minimal the medium concentration
  • 42:03here you do see a response and the
  • 42:06leukemias progress and the vast majority
  • 42:08of them will have developed the mutation.
  • 42:10If you then go threefold
  • 42:12more of them in an inhibitor,
  • 42:14you get a much longer response.
  • 42:16In fact,
  • 42:16maybe some of them here are cured of the
  • 42:19disease and when the resistance occurs,
  • 42:21it occurs without the min and mutations.
  • 42:23So in fact,
  • 42:24a higher concentration does at
  • 42:26least in this model prevent the
  • 42:28development of those mutations.
  • 42:30So you can see if you stop your dose
  • 42:32escalation right here because you're
  • 42:34getting differentiation syndrome
  • 42:35and somebody tells you you have to,
  • 42:38you're actually setting up a
  • 42:39situation where you're going to
  • 42:41get acquired resistance mutations.
  • 42:44Having said all that,
  • 42:45we're still getting resistance way out
  • 42:47here with the single agent at higher doses.
  • 42:49So what's that all about and
  • 42:52I'll quickly summarize this.
  • 42:54Essentially what we're seeing here
  • 42:55is that the leukemia cells and
  • 42:57this is another phenomenon that's
  • 42:59known in other settings now are
  • 43:01not mutating the men and they're
  • 43:03actually changing their state
  • 43:04significantly to lead to a state that
  • 43:07we don't understand completely yet,
  • 43:09but where they're now no longer
  • 43:11dependent on that Hawks niece program.
  • 43:14And and in fact interestingly they
  • 43:16look much more differentiated.
  • 43:17The leukemias themselves almost
  • 43:20look like monocytes in terms
  • 43:22of their their flow cytometry,
  • 43:24but definitely will transplant the
  • 43:26disease from 1 bow to the next.
  • 43:28So they're not monocytes,
  • 43:30but so we're trying to understand
  • 43:32this mechanism mode of adaptive
  • 43:33resistance a little bit better.
  • 43:35Now have developed a model,
  • 43:37a cell line model for it and have
  • 43:40developed some PDX models as well.
  • 43:41But the cell line model actually
  • 43:44lets us move to what everyone
  • 43:47likes to do now which is a genome
  • 43:49wide CRISPR screen to say OK,
  • 43:51how did the dependence do the
  • 43:53dependencies change when you
  • 43:54go from one state to the next?
  • 43:56And to summarize,
  • 43:57a lot of analysis in fact they do
  • 44:00appear to and this is on a subtle
  • 44:03transferase CAT6A or MAZ which
  • 44:05also rearranged rarely in some
  • 44:07leukemias now becomes seems to become
  • 44:11relevant in this in this setting.
  • 44:13So here is just an experiment
  • 44:15showing you that.
  • 44:17So we developed resistant leukemia cells
  • 44:20that don't have the MIN in mutation.
  • 44:21They're resents to the MIN inhibitor.
  • 44:23Here it is in blue.
  • 44:24But if you treat with the cat
  • 44:266A in this case guide,
  • 44:28it re sensitizes the the cell
  • 44:31line to the MIN inhibitor.
  • 44:34But interestingly and interestingly enough,
  • 44:36the CAT 6A by itself in the
  • 44:38absence of the MIN inhibitor has
  • 44:40a little bit of an effect.
  • 44:41It's really something about the relationship
  • 44:45between CAT6A and Menon that is important.
  • 44:48So what's CAT6A?
  • 44:49It's a histone has still transferase
  • 44:51as well modifies histone H3
  • 44:53on various lysine shown here.
  • 44:55And if you do now chip seek in either O
  • 44:58sensitive leukemia cell lines or resistant,
  • 45:01the MLL Menon and CAT6A chip seek
  • 45:06data looks very similar and in so here
  • 45:08both in the sensitive or the resistant
  • 45:10and this is just showing that more
  • 45:12broadly so men and Catsix is there,
  • 45:14it's on the scene and it becomes seems
  • 45:17to become much more important when the
  • 45:20cells adapt to the men and inhibitor.
  • 45:23Needless to say,
  • 45:24we're now doing the experiments to
  • 45:26see if this combination in mice will
  • 45:29reverse the resistance in patient samples.
  • 45:33It won't reverse the resistance
  • 45:35to the mutations,
  • 45:36but it might reverse the
  • 45:38resistance to the adapted form.
  • 45:40But the men inhibitor works well enough,
  • 45:42it's hard to generate that adaptive form,
  • 45:44so it's taking us a little while.
  • 45:47I'm going to skip this just for time's sake,
  • 45:50but to and just to summarize saying that
  • 45:54these complexes here, the .1 complex,
  • 45:57the Super elongation complex,
  • 45:58I've been talking about MLL and CAT6A.
  • 46:02If you look broadly,
  • 46:04some of you probably know that Broad
  • 46:06Institute's been doing broad CRISPR
  • 46:08screens on five 600 cancer cell lines,
  • 46:10and they make all this data publicly
  • 46:12available in many different ways.
  • 46:14You can search that data.
  • 46:15If you ask what genes have a
  • 46:18similar dependency to Menin
  • 46:20throughout all of cancer space,
  • 46:23.1 is the next thing on the list.
  • 46:26And then and then these other
  • 46:28proteins here E&L which is part of
  • 46:30the Super elongation complex CAT6.
  • 46:33A point being these,
  • 46:35it's clear that these complexes
  • 46:37are working together somehow
  • 46:39throughout many cancer cell lines,
  • 46:41Leukemia for sure,
  • 46:43but also probably others as well.
  • 46:46And in fact that led us to this
  • 46:50publication for actually for
  • 46:51about a year and a half ago now.
  • 46:54Matt Hemming,
  • 46:56a paediatric or medical oncology
  • 46:58fellow was interested in
  • 47:00gastrointestinal stromal tumors
  • 47:01and just did a genome wide CRISPR
  • 47:04screen because that's how you start
  • 47:06every project these days it seems.
  • 47:08And in fact cat 6A was one of
  • 47:10the top hits in that screen.
  • 47:12And then we looked a little bit more in more
  • 47:16detail .1 and Menin were in there as well.
  • 47:18And the bottom line is,
  • 47:19is it appears that this CAT6,
  • 47:23A .1 Menin complex cooperation is
  • 47:28important in this type of cancer as well.
  • 47:32And we don't understand,
  • 47:33you might ask why should ask why?
  • 47:36We don't completely understand
  • 47:37why it's the case.
  • 47:39But it does appear that when we
  • 47:42inhibit CAT6A and men and in cell
  • 47:44lines or in mice or in PDX or
  • 47:48xenograft models that a program
  • 47:50that Matt had described earlier
  • 47:52driven by transcription factors
  • 47:54like one called hand one,
  • 47:56which is known to be important
  • 47:58for controlling lineage associated
  • 48:00gene expression in this cancer,
  • 48:03certain goes down pretty rapidly.
  • 48:05So there's something about the
  • 48:07developmental program and this
  • 48:08cancer as well that seems to be
  • 48:11dependent on these complexes,
  • 48:12but you don't really see it as
  • 48:15dramatically as in leukemia until you
  • 48:17start to combine the the small molecules.
  • 48:19Many will do a little bit of it,
  • 48:20Mos cats, XA will do a little bit,
  • 48:22but when you combine them,
  • 48:23you really get a a dramatic response.
  • 48:25So the point being that we're
  • 48:27looking at this in leukemia,
  • 48:28but we're starting to move into
  • 48:30some other cancers as well to see if
  • 48:34indeed these developmental regulators,
  • 48:36if you will, might be relevant there.
  • 48:39I just summarized this data and
  • 48:42I'll end with this.
  • 48:44Pfizer just published a paper
  • 48:46about six months ago.
  • 48:48They've now developed a clinical
  • 48:50grade CAT6A inhibitor and in fact
  • 48:52this small molecule is in phase
  • 48:54one clinical trials in estrogen
  • 48:57receptor positive breast cancer.
  • 48:58And it looks like from this paper
  • 49:01and we've now done a number of
  • 49:03experiments as well that somehow
  • 49:05Menon is supporting the ER driven
  • 49:08gene expression program.
  • 49:09And Needless to say, I mean sorry Cat 6A.
  • 49:13Needless to say, we're now looking
  • 49:14at this Cat 6A men in combination.
  • 49:16And again as much as like that we saw
  • 49:18in GIST, it looks like this the ER
  • 49:21driven program is highly dependent on
  • 49:23those two complexes and we're trying
  • 49:25to work through that now as well.
  • 49:28So to summarize what I've told you,
  • 49:31the MEN in MLO complex is a relevant
  • 49:34therapeutic target and snippet subset of
  • 49:37AM LS and that acquired somatic mutations
  • 49:40in men and are a mechanism of resistance,
  • 49:43not the only mechanism of resistance
  • 49:45but that clearly validates men and has
  • 49:48a therapeutic target in this disease.
  • 49:50We're continuing to work on the various
  • 49:52types of resistance and we do think
  • 49:54that rational combinations like men
  • 49:56and CAT6A or men and other things with
  • 49:58a lot going on in terms of trying to
  • 50:01understand which combinations may
  • 50:02prevent development of resistance.
  • 50:03And perhaps the most exciting in the
  • 50:05longer term is if we can understand
  • 50:08where these mechanisms might be
  • 50:09important even beyond leukemia.
  • 50:11And I think they're going to
  • 50:12be opportunities,
  • 50:13but we certainly still have work to do to,
  • 50:15to prove that.
  • 50:16So I've talked much about the people
  • 50:18in in the lab that have done the work.
  • 50:21These are our collaborators at Dana
  • 50:23Farber actually Nathaniel Gray now
  • 50:25at Stanford and Ross I mentioned and
  • 50:28Chang and Richard who work with Ross
  • 50:31at MSK and some of our collaborators
  • 50:34throughout HMS community.
  • 50:35So thank you,
  • 50:37happy to take any questions and
  • 50:39thanks for thanks for staying.
  • 50:41All
  • 50:49right, absolutely spectacular grand
  • 50:51rounds really going from basic science to
  • 50:54the patient and back and forth and it's
  • 50:56absolutely spectacular. Thank you. Yeah
  • 50:59and and great talk. So I think
  • 51:01the main issue and you know as a
  • 51:04clinical investigator in my mind
  • 51:05with all epigenetic therapies is,
  • 51:07is the therapeutic window as you were
  • 51:09saying like how do you actually disrupt
  • 51:11translational or transcriptional
  • 51:13programs that are relevant to the
  • 51:15leukemia but not to the normal tissue.
  • 51:18So for example with this index
  • 51:20particular drug was this as a result
  • 51:23of screening of thousands of molecules
  • 51:25and because as you mentioned it,
  • 51:27it seems like to disrupt only where
  • 51:29it's relevant to the leukemia,
  • 51:30but it's not disrupting the MLL
  • 51:32interactions that are important for
  • 51:34normal hematopoiesis and other functions.
  • 51:36So that how did this transition happen?
  • 51:38It's just a matter of luck or is
  • 51:40it tons of screening of other yeah
  • 51:42you know the old saying
  • 51:43better lucky than good.
  • 51:44I I think that that's what we found here,
  • 51:48meaning that for some reason and
  • 51:50we're looking into this men and is
  • 51:53only critical for MLL wild type.
  • 51:55Obviously this mechanism probably wasn't
  • 51:58developed during evolution for MLL
  • 52:00fusions to localize to certain loci.
  • 52:03So MLL as I mentioned is a monstrous protein.
  • 52:06It has many domains that combine chromatin.
  • 52:09So it's very likely and there's some
  • 52:11data to support this that different
  • 52:13domains or different binding partners
  • 52:16determine localization to different
  • 52:18places throughout chromatin.
  • 52:19And it just so happens in this it
  • 52:21kind of was predicted by some of the
  • 52:23early Cleary work that Menon was a
  • 52:25unique dependency in these leukemias.
  • 52:27Well that's because it's really
  • 52:30intersecting exactly with the
  • 52:34important MLL fusion driven targets.
  • 52:37We don't know the molecular
  • 52:39mechanism for that yet,
  • 52:40but basically it's a long way of
  • 52:42saying we think there's a multi
  • 52:43valent interaction between MLL and
  • 52:45chromatin and Menon is only important
  • 52:47for a subset of that interaction.
  • 52:50Scott, fantastic talk.
  • 52:51And the the question about
  • 52:53the solar cancer part,
  • 52:54so you alluded to that you are it's
  • 52:56great to see that you and other
  • 52:58companies are looking into this aspect.
  • 53:00So, so we know that oxygens are
  • 53:02often deregulated in solar cancer
  • 53:04as well in addition to leukemia.
  • 53:06So what happens to men inhibitor,
  • 53:09the men and MLL inhibitor monotherapies,
  • 53:12do they have any efficacy in
  • 53:14solar cancer or you have to
  • 53:15really using combinations before
  • 53:16you can see something happening?
  • 53:18Yeah, it's good question.
  • 53:20So actually it's a good point.
  • 53:21There are a number of say subtypes of
  • 53:24lung cancer that express HOX genes.
  • 53:26We actually haven't looked
  • 53:27at that probably should,
  • 53:29but in the both in the gastrointestinal
  • 53:32stromal tumors and the ER
  • 53:34positive breast cancer cell lines,
  • 53:36the men inhibitor will slow their growth.
  • 53:40So they have some effect and that
  • 53:42is it looks like through somehow
  • 53:44modulating the ER driven program,
  • 53:46but it's much more dramatic both
  • 53:48the gene expression changes and
  • 53:50the inhibition of proliferation if
  • 53:52you combine the minute inhibitor
  • 53:54and and the cat 6A inhibitor.
  • 53:56So how that works, we don't,
  • 53:58we don't completely understand yet.
  • 54:01It's a way of saying and and predicting
  • 54:04and hopefully get the word out
  • 54:06before all the trials get shut down,
  • 54:08that the single agents might
  • 54:10have some activity,
  • 54:11but I suspect they won't be home
  • 54:13runs and the companies have to
  • 54:15have the wherewithal to actually
  • 54:17move forward to the combinations.
  • 54:19And those of you who've done this
  • 54:22before know that can be difficult.
  • 54:23So we're going to try to get the
  • 54:26word out that you should move
  • 54:28the combinations quickly before
  • 54:30people lose interest.
  • 54:32There's a lot of psychology and
  • 54:34sociology that goes into keeping
  • 54:36the drug companies interested,
  • 54:38so that's a little bit of a soapbox to
  • 54:40say the single agents do something.
  • 54:42The combination definitely looks better.
  • 54:46Manoj, a great talk.
  • 54:48My question is about the specificity
  • 54:50of both the MLL fusion proteins and
  • 54:52the NPM 1C that you alluded to.
  • 54:53And Amar was also asking.
  • 54:56So I think you probably worked on this
  • 54:58on the cancer discovery latest paper,
  • 55:00but most of them also seem to be
  • 55:02overlapping with like say PRC 2
  • 55:04targets or you know are there other
  • 55:06mechanisms you think are relevant to
  • 55:08why these are so tightly overlapping
  • 55:10the fusion proteins and the
  • 55:12yeah, so, so it's a good point.
  • 55:14So they do overlap significantly with
  • 55:16PRC 2 targets and you know as you may
  • 55:20remember the the this has been predicted
  • 55:23for 3 decades from the Drosophila work.
  • 55:25The initial Drosophila work show
  • 55:28Polycom and Trithorax actually are
  • 55:30known to be genetically genetic
  • 55:32antagonists of one another and the the
  • 55:35trithorax slash MLL complex controls
  • 55:38developmental genes that the PRC two
  • 55:40or Polycom complex wants to shut off.
  • 55:43So in absolutely this is A these
  • 55:46proteins complexes MLL probably
  • 55:49Catsix A are inventing.
  • 55:50We've shown this in some other settings
  • 55:53are preventing the Polycom complexes from
  • 55:55coming in and repressing gene expression.
  • 55:57So the the way we think that this
  • 55:59is working is during hematopoietic
  • 56:00development as you go from stem cells to
  • 56:03progenitors to fully developed myeloid cells,
  • 56:06the Polycom complex at least for a
  • 56:08subset of developmental loci are
  • 56:10shutting those programs off and the
  • 56:12MLL fusion won't let them do that.
  • 56:14So they're antagonizing and then
  • 56:17Newt 98 fusions and probably NPM
  • 56:19one are antagonizing PRC 2 section.
  • 56:22Yeah,
  • 56:23awesome. I'm going to bring
  • 56:24it over to you in a second.
  • 56:26We have an online question which I
  • 56:27think you probably partially answered
  • 56:29and that is what is the mechanism of
  • 56:31gene specific targeting of MLL EF9
  • 56:33and similarly what you think is the
  • 56:35underlying mechanism for the gene target
  • 56:37specificity of men and inhibitors.
  • 56:39So for the online person, yeah.
  • 56:40So it's a good, it's a very good question.
  • 56:43We do it. I don't the bottom line is,
  • 56:45is we don't know the answer
  • 56:46to the second part of that.
  • 56:47I mean that's the that's at the moment.
  • 56:50Probably the most critical question
  • 56:52is why is it that Menon's only
  • 56:54important for localization of the
  • 56:56MLL compacts to certain loci.
  • 56:58So Needless to say,
  • 56:59we're looking at various aspects
  • 57:00of those loci to try to understand
  • 57:03what that's what that's all about.
  • 57:04MLL targeting to chromatin broadly has many,
  • 57:07probably has many mechanisms,
  • 57:09some of its direct there's a domain
  • 57:11on MLL that binds to what's called
  • 57:13a CPG island which is upstream of
  • 57:16many transcriptional start sites.
  • 57:17Menon plays a role.
  • 57:19There are other accessory proteins
  • 57:20that play roles.
  • 57:21So I think the cell has just given
  • 57:23itself many options to figure out
  • 57:25where to put MLL and and each of
  • 57:28those mechanisms slightly different.
  • 57:31We have a trainee question.
  • 57:34So my name is trainee,
  • 57:37I just wanted to follow my name used
  • 57:38to be that at some point you graduated.
  • 57:40I just want to follow up on your
  • 57:42comment about combination therapies.
  • 57:44So I specifically wanted to ask is there
  • 57:46any thought that men and inhibition
  • 57:47could convert these resistant like
  • 57:49subtypes especially in the pediatric
  • 57:50setting to a chemosensitive form if
  • 57:52there's thought of combining with
  • 57:54chemo to then re sensitize them and
  • 57:56potentially cure those patients. Yeah,
  • 57:57it's it's a good short answer is
  • 58:00we don't know the answer to that.
  • 58:02I I think there's a lot to be learned
  • 58:05and the beauty of having now multiple
  • 58:08small molecule selective small molecules,
  • 58:11we can do those types of experiments,
  • 58:14but the short answer is we
  • 58:16haven't haven't gotten there yet.
  • 58:20Yeah, great talk.
  • 58:21Have you seen any phenotypic
  • 58:23differences in the fusion
  • 58:24partners with MML or MLL?
  • 58:26You mentioned there's you know,
  • 58:27100 different ones.
  • 58:27Do they all have the same kind of,
  • 58:29you know, break points?
  • 58:30Does it change expression?
  • 58:31Do you see any variability
  • 58:32in the kind of those fusion
  • 58:33partners? Yeah, it's a good question
  • 58:36that's been asked for many decades
  • 58:38and not been answered very well,
  • 58:41at least in patient samples because
  • 58:42it's hard to get enough patient
  • 58:44samples of these subtypes to to ever
  • 58:46really do that experiment. Well,
  • 58:49at least for the fusion,
  • 58:51the different fusion AM LS we've
  • 58:53assessed and that have been
  • 58:54assessed in patients, it doesn't.
  • 58:56It's not clear that the fusion partner
  • 58:58is determining men and responsiveness.
  • 59:02Is the fusion partner influencing
  • 59:04the phenotype of the leukemia?
  • 59:06I think that question still still
  • 59:10open and there's aren't enough good
  • 59:12models to really answer that question.
  • 59:18If you delete the at least for AF9,
  • 59:21if you delete the C turn much of the AF9,
  • 59:23it will no longer be transforming.
  • 59:25So that fusion partner is important
  • 59:29and and in in the AF9 setting,
  • 59:31we think it's important because
  • 59:32that's the anchor to drop to pull
  • 59:34all those other complexes in.
  • 59:35But it's that's a nice simple answer.
  • 59:37It's not that simple because
  • 59:39some of the fusion proteins
  • 59:40don't bind to those complexes.
  • 59:42So what they're doing is, is less clear,
  • 59:47beautiful talk. Thank you.
  • 59:48Obviously there are lots of
  • 59:50other chromatin complexes,
  • 59:52switch, sniff, polychrome, etcetera.
  • 59:54Any any thoughts on those?
  • 59:56Are you looking at any exploring
  • 59:58any of those other chromatin?
  • 01:00:02Yeah, accessibility.
  • 01:00:04Epigenetic complexes?
  • 01:00:05Sure. So we have over time looked at
  • 01:00:11the Polycom complex mostly in leukemia
  • 01:00:15and it for whatever reason and this is
  • 01:00:18going to be right contradictory to how
  • 01:00:19I answered one of my previous questions.
  • 01:00:21The Polycom complex does seem
  • 01:00:24to be important in the continued
  • 01:00:27proliferation of many types of leukemia.
  • 01:00:30How that's working and why hard to know,
  • 01:00:34but the small molecule PRC 2
  • 01:00:37inhibitors or or EZH 2 inhibitors
  • 01:00:40don't have tremendous activity.
  • 01:00:42So whether or not it's the enzymatic
  • 01:00:44activity versus some other part of the
  • 01:00:47complex at least in leukemia anyway,
  • 01:00:49I don't know.
  • 01:00:50And I think that probably it brings
  • 01:00:53up a good point that and we've
  • 01:00:56done this in the past as well,
  • 01:00:57but we have to be careful about
  • 01:00:59the thought process that enzymatic
  • 01:01:02inhibition of a protein in one of
  • 01:01:04these complexes is the same thing as
  • 01:01:06complete interactivation of the protein.
  • 01:01:08It's not and we now seen that
  • 01:01:09many different times.
  • 01:01:10So the enzymatic part of the
  • 01:01:13proteins is important,
  • 01:01:14but there's probably a structural
  • 01:01:16component to this that when you
  • 01:01:18take the protein completely out and
  • 01:01:19this is the same way for EZH 2 the
  • 01:01:22the changes are much more dramatic.
  • 01:01:23So it gets to the question as to what
  • 01:01:25his some modifications are doing and
  • 01:01:27that gets to even more deep and complicated.
  • 01:01:30But so long answer to yes,
  • 01:01:32we've looked at the complexes,
  • 01:01:33we haven't looked much at the
  • 01:01:36chromatin remodeling complexes.
  • 01:01:37Segal Kadosh who some of you may know is
  • 01:01:40at Dana Farber and we just let her do that.
  • 01:01:42She they're doing a lot in that regard.
  • 01:01:46I'm sure they're playing a role here.
  • 01:01:48What but how and what we don't know.
  • 01:01:51We have one last question and after this
  • 01:01:54is actually a session for the trainees,
  • 01:01:56very private was Doctor Armstrong.
  • 01:01:59Yeah, one last question.
  • 01:02:00This is actually related
  • 01:02:01to the fusion partners,
  • 01:02:03but as you know we often get,
  • 01:02:05you know when we do the genetech
  • 01:02:07sequencing we get MLL deletions,
  • 01:02:08MLL mutations,
  • 01:02:09sometimes Trisom 11 or you know PDD.
  • 01:02:14And is your sense that beyond
  • 01:02:16the fusion MLL Fusion's those
  • 01:02:19alterations also have susceptibility
  • 01:02:21to an inhibition or yeah,
  • 01:02:23as you probably know since you've been
  • 01:02:25important in running some of the trials,
  • 01:02:27the MLLPTD subtype of AML for
  • 01:02:31some of the trials is included.
  • 01:02:33There's been an assumption to some extent
  • 01:02:36that they should be responsive the PD.
  • 01:02:38So we have generated some MLL.
  • 01:02:39So MLLPTDS are actually a partial
  • 01:02:41tandem duplication of a part of the
  • 01:02:44MLL protein and that subset of AML is
  • 01:02:46incredibly difficult to treat and it's
  • 01:02:48also found in MD's those mutations,
  • 01:02:52but that almost tells you immediately
  • 01:02:54it's also found in MDSMLL.
  • 01:02:56Rearrangements are not found in MDS that
  • 01:02:58they're probably something different
  • 01:02:59going on in the minute inhibitor
  • 01:03:01doesn't have at least in our PDX
  • 01:03:03models the same type of activity in
  • 01:03:04those PDX as it does MLL rearranged.
  • 01:03:07So I think it gets back to this issue
  • 01:03:10that we've been discussing that in
  • 01:03:12that setting either because of that
  • 01:03:15duplication or otherwise the that MLL
  • 01:03:18Oncoprotein sticks on chromatin through
  • 01:03:20a different mechanism than Menon.
  • 01:03:22So we're got a project looking
  • 01:03:25at exactly that,
  • 01:03:26trying to understand what that is.
  • 01:03:28We haven't looked at the deletions.
  • 01:03:30I wouldn't predict they would be.