Skip to Main Content

Identifying and Targeting DNA Repair Defects in Cancer

October 02, 2024

Yale Cancer Center Grand Rounds | October 1, 2024

Presented by: Dr. Shridar Ganesan

ID
12158

Transcript

  • 00:00A real pleasure to,
  • 00:02have our speaker, but be
  • 00:04we're here, and I will
  • 00:06introduce him in just a
  • 00:07second. I just wanna remind
  • 00:08everyone,
  • 00:10to fill out your
  • 00:12DEI climate survey.
  • 00:15They would very much appreciate
  • 00:17it as would I,
  • 00:19and so,
  • 00:20please do it.
  • 00:23So,
  • 00:24I stood up and I
  • 00:25said, you, I do without
  • 00:27notes because I've known Sridhar
  • 00:30Ganesan
  • 00:31for a long time.
  • 00:33And Sridhar has,
  • 00:36over the course of his
  • 00:37career, taken a trip up
  • 00:39and down the East Coast.
  • 00:41He He was an undergraduate
  • 00:42at Princeton, a Yale, MD
  • 00:44PhD student,
  • 00:46and we actually overlapped here
  • 00:48a little bit way back
  • 00:49in some time in the
  • 00:51past.
  • 00:52And then,
  • 00:54was a resident at the
  • 00:55Brigham and a fellow at
  • 00:57Dana Farber and stayed on
  • 00:58there
  • 00:59and worked for quite a
  • 01:01number of years in the
  • 01:02lab of the late David
  • 01:04Livingstone,
  • 01:05late and great David Livingstone.
  • 01:08And Sridhar is now professor
  • 01:10of medicine at Rutgers,
  • 01:13has assorted titles which are
  • 01:15up on the screen,
  • 01:17and has focused his career
  • 01:20on
  • 01:22DNA damage
  • 01:23repair
  • 01:25and related topics.
  • 01:28Back
  • 01:29now probably
  • 01:31close to twenty years ago
  • 01:33when we were both at
  • 01:35Dana Farber,
  • 01:36I pulled Schreuder out of
  • 01:38the lab.
  • 01:39He was totally focused in
  • 01:41the lab at the time,
  • 01:43and he and two other
  • 01:44people came to the clinic
  • 01:46half a day a week.
  • 01:48And I said, this is
  • 01:49the way I'm mixing up
  • 01:50the clinical people and the
  • 01:52lab people, and I think
  • 01:53it was a pretty successful
  • 01:54experiment. I said, I don't
  • 01:56care how many patients you
  • 01:57see. Just show up.
  • 02:00And to this day,
  • 02:02Shreedhar still sees patients with
  • 02:04breast cancer,
  • 02:05and is interested in both
  • 02:06breast cancer and
  • 02:08rare tumors as well,
  • 02:11which he may or may
  • 02:12not comment about. And,
  • 02:13today,
  • 02:14his topic is identifying and
  • 02:16targeting DNA repair mutations in
  • 02:18cancer.
  • 02:19Shreedhar, it is a pleasure
  • 02:21to have you here. We
  • 02:22actually have
  • 02:24a plaque for you,
  • 02:27which,
  • 02:28you can,
  • 02:30oh, and Roy's gonna take
  • 02:31a picture of of the
  • 02:32two of us with the
  • 02:33plaque.
  • 02:36Gotta have a butter here.
  • 02:40Expect to see it hanging
  • 02:41in your office next time
  • 02:42we visit. Awesome.
  • 02:44There you go. And I'll
  • 02:45keep this for you, Ricardo.
  • 02:47Thanks for being here. Alright.
  • 02:49Thanks.
  • 02:50That's a very kind introduction.
  • 02:51Thanks so much. It's such
  • 02:51a pleasure to be back
  • 02:52here at Yale. I did
  • 02:53spend eight years in lovely
  • 02:54New Haven, even though that
  • 02:55time I had long hair
  • 02:56in a ponytail.
  • 02:58Actually, that stayed actually doing
  • 02:59fellowship and when I first
  • 03:01met, Eric as well. And
  • 03:02I am incredibly indebted to,
  • 03:04both Eric and, Judy Garber
  • 03:06for really bringing me into
  • 03:07the clinic and, and and,
  • 03:12have started my career in
  • 03:13breast taking care of patients
  • 03:14with breast cancer, which has
  • 03:14been, you know, incredibly rewarding.
  • 03:17So today I'm gonna talk
  • 03:18to you it's a little
  • 03:19bit of history of kind
  • 03:20of DNA repair defects in
  • 03:21cancer, some stuff that I
  • 03:22did originally in in in
  • 03:23David's lab and some more
  • 03:25work right now that's we
  • 03:26hope is a little more,
  • 03:27clinically acable as well as
  • 03:29some of the underlying basic
  • 03:30science. And the idea is,
  • 03:31you know, I have to
  • 03:32tell this,
  • 03:34audience
  • 03:36alright. Ah, so these are
  • 03:37my disclosures. So I do
  • 03:39a lot of research. My
  • 03:39wife is an employee of
  • 03:41Merck.
  • 03:42And, you know, I just
  • 03:42wanna stop by. Yes. A
  • 03:43lot of my work is
  • 03:44done, with David Livingstone. This
  • 03:46is David Livingstone in his
  • 03:47natural habitat, which is surrounded
  • 03:49by his former trainees.
  • 03:50So there's,
  • 03:51you know,
  • 03:52Sharon Kanter who cloned,
  • 03:54BRIP one or Fang j,
  • 03:56Bing Shah who, cloned and
  • 03:58characterized PALB two in the
  • 03:59lab, and Lonnie Drapkin who,
  • 04:01you know,
  • 04:02contributed to identification of the
  • 04:04cell of origin of high
  • 04:05grade serous ovarian cancer,
  • 04:07in his lab. And, David
  • 04:09passed away unexpectedly a couple
  • 04:10years ago, and the Farber
  • 04:12is a,
  • 04:13much quieter and less interesting
  • 04:14place, I think, with his,
  • 04:17with his passing. But I'm
  • 04:18greatly, you know, a lot
  • 04:19of what I work was
  • 04:20really, he grabbed me when
  • 04:21I was a senior resident.
  • 04:23And,
  • 04:24I had no idea who
  • 04:25he was. And within five
  • 04:26minutes of meeting him, somehow
  • 04:27I'd agreed to work in
  • 04:28his lap.
  • 04:30Alright.
  • 04:31And so, you know, obviously,
  • 04:33you know, talk a little
  • 04:33bit of background of BRCA
  • 04:35and BRCA two. If you
  • 04:35kind of look at families
  • 04:37that have,
  • 04:38high I don't know if
  • 04:38there's a point here, who
  • 04:39have, high risk disease.
  • 04:41If you look at families
  • 04:42in which there's multiple,
  • 04:45members that have a breast
  • 04:46cancer, a small proportion of
  • 04:48them are due to Mendelian,
  • 04:49you know, high penetrance,
  • 04:51Mendelian genetics. So in those
  • 04:53families that have high penetrance,
  • 04:55about fifteen percent are associated
  • 04:56with germline mutations in BRCA
  • 04:58and BRCA two. There's a
  • 04:59small percentage of others.
  • 05:01PALB two is probably the
  • 05:02most predominant one. We're talking
  • 05:03about it's probably the closest
  • 05:04thing we have to BRCA
  • 05:05three in terms of being
  • 05:05a high penetrance,
  • 05:07gene. And then there are
  • 05:08SNPs and other protected SNPs
  • 05:10perhaps
  • 05:11contributing from many of them
  • 05:12and then unexplained or you
  • 05:14know, breast cancer is common,
  • 05:15so there can be clustering
  • 05:16by chance in a common
  • 05:17disease, that also can be,
  • 05:19seen in a bunch of
  • 05:20these. And for a for
  • 05:22a while, you know, the
  • 05:23I'm gonna talk a little
  • 05:24bit about the the the
  • 05:25BRCA one, cancer syndrome.
  • 05:28Okay. You know, so women
  • 05:30carrying a,
  • 05:32mutant allele BRCA one have
  • 05:33a greatly increased lifetime risk
  • 05:34of breast and ovarian cancer.
  • 05:36Actually, the relative risk is
  • 05:37much greater for what we
  • 05:38used to call ovarian cancer
  • 05:39than breast cancer. Alright?
  • 05:42Because breast cancer because ovarian
  • 05:43cancer or a fallopian tube
  • 05:44cancer is rare. The tumors
  • 05:46arises when we all have
  • 05:47undergone
  • 05:48lost heterozygosity. We've lost the
  • 05:50wild type,
  • 05:51allele, implying the BRCA one,
  • 05:52and we'll talk about this,
  • 05:53really functions as a classical
  • 05:54tumor suppressor. And men with
  • 05:56a interestingly, with the BRCA
  • 05:57one mutation have little or
  • 05:58no phenotype.
  • 06:00That's discernible.
  • 06:01Right?
  • 06:02And the cancers that arise,
  • 06:04you know,
  • 06:05tend to have early onset.
  • 06:06So this is kind of
  • 06:07the distribution of age of
  • 06:08onset. The bottom line here
  • 06:09is kind of the incidence
  • 06:10in the general population over
  • 06:12time of breast cancer. And
  • 06:13up here is BRCA one.
  • 06:14You see the instance starts
  • 06:15to rise in the thirties
  • 06:17and really peaks in the
  • 06:18premotor puzzle time. And then
  • 06:19after about age fifty, you
  • 06:21can see these these curves
  • 06:22are actually parallel. So then,
  • 06:23you know, the bulk of
  • 06:24the risk is really here.
  • 06:26Alright? This is BRCA two
  • 06:27and PALB two. So BRCA
  • 06:29one BRCA two and PALB
  • 06:30two have a slightly different
  • 06:31kind of age of onset.
  • 06:32It it and the risk,
  • 06:34occurs a little later and
  • 06:35continues to increase, and PALB
  • 06:37two is very similar to
  • 06:38BRCA two. Alright?
  • 06:40And these are high risk.
  • 06:41These are much less penetrant,
  • 06:42and we won't talk about
  • 06:43them all, not that much
  • 06:44today.
  • 06:45Alright? And so when BRCA
  • 06:47and BRCA two were first
  • 06:48cloned, everyone assumed
  • 06:50that because it was associated
  • 06:51with breast and ovarian cancer,
  • 06:52they would have some sort
  • 06:53of lineage specific role. That
  • 06:55it was somewhat to be
  • 06:56something with the biology of
  • 06:58the breast or the ovary.
  • 07:00And, when I was a
  • 07:01senior resident, we made the
  • 07:02first antibodies against BRCA one
  • 07:04that actually were worked and
  • 07:05were validated. And we found
  • 07:06to our surprise back then
  • 07:08in David's lab that BRCA
  • 07:10one instead was expressed was
  • 07:11not lineage restricted, but cell
  • 07:13cycle restricted. Any cell that
  • 07:15passed the g one transition
  • 07:17expressed BRCA one in these
  • 07:18beautiful little nuclear foci.
  • 07:20Alright? So it was cell
  • 07:22cycle restricted, not lineage restricted.
  • 07:24And for tumors to purposes,
  • 07:26it was kind of unusual
  • 07:27because you took out BRCA
  • 07:28one, the tumor didn't grow
  • 07:29bet cells didn't grow better.
  • 07:30They all crashed. And, partly
  • 07:33PFT dependent arrest within a
  • 07:34few cell cycles, we took
  • 07:35out BRCA
  • 07:36one. Alright? So it was
  • 07:38cell cycle dependent, and without
  • 07:40it, cells just crashed. Alright?
  • 07:43At that time, BRCA two
  • 07:44was also cloned and identified.
  • 07:46And if you remember, BRCA
  • 07:47and BRCA two were identified
  • 07:48by classical genetics by Mary
  • 07:50Claire King. Alright? And, when
  • 07:53ultimately BRCA two is cloned,
  • 07:55I always say BRCA one
  • 07:56and BRCA two are, you
  • 07:57know, eighty percent identical by
  • 07:59name,
  • 08:00but zero percent identical by,
  • 08:02you know,
  • 08:03sequence or anything else. They're
  • 08:04completely different proteins.
  • 08:06Alright? And with little homology
  • 08:08to any other known proteins.
  • 08:09And it's another large gene.
  • 08:10And interestingly,
  • 08:12despite the fact that they're
  • 08:13completely dissimilar proteins,
  • 08:15some of the characters of
  • 08:16BRCA also, these are also
  • 08:17not lineage restricted,
  • 08:19but came up during s
  • 08:20phase in beautiful nuclear foci
  • 08:23that colocalized with BRCA one.
  • 08:25This is work by,
  • 08:28Daniel Silver and Junji Chen
  • 08:30in David Slab.
  • 08:32Alright? So, and people all
  • 08:34talk about Bracker, you know,
  • 08:35the BRCA1, but they're actually
  • 08:36very complete different genes. They
  • 08:37seem to they're just in
  • 08:38the same pathway. It was
  • 08:39not necessarily obvious that they
  • 08:40should they would.
  • 08:41Right?
  • 08:43And even though a lot
  • 08:43of work has gone into
  • 08:44what the fundamental problem function
  • 08:46in which BRCA one, works
  • 08:48in, so there's lots of
  • 08:49data on transcriptional regulation, genomic
  • 08:51regulation,
  • 08:52epigenetic regulation.
  • 08:54The fundamental thing is that
  • 08:55without BRCA one, if you
  • 08:57have a cell that goes
  • 08:58without BRCA one through a
  • 08:59couple of s phases, the
  • 09:00chromosomes fall apart.
  • 09:02Alright? So these are mouse
  • 09:03chromosomes in which we've taken
  • 09:04out BRCA one, and this
  • 09:06is after a few cell
  • 09:07cycles. The
  • 09:09normal chromosomes in mice should
  • 09:10look like these acrocentric. Alright?
  • 09:12But you take them out
  • 09:12also and the chromosomes fall
  • 09:14apart. You see quadradials,
  • 09:16chromatin deletions.
  • 09:17Alright?
  • 09:18So BRCA one is
  • 09:21incredibly important for normal s
  • 09:22phase,
  • 09:25maintenance. Alright?
  • 09:28Other thing is that, you
  • 09:29know, what we realized from
  • 09:30this is that BRCA one
  • 09:31is not as what I
  • 09:32consider an accidental tumor suppressor.
  • 09:35Alright? Its main function is
  • 09:36an s phase to keep
  • 09:37the chromosomes alive. A rare
  • 09:39consequence of its dysfunction
  • 09:41is cancer. If you look
  • 09:42at women,
  • 09:43with a BRCA one mutation,
  • 09:44they have about a fifty
  • 09:46to sixty percent lifetime risk.
  • 09:47So imagine that in their
  • 09:48in that,
  • 09:49a breast cancer. Imagine that
  • 09:51all trillion of her cells
  • 09:52are heterozygous.
  • 09:53There's only a fifty to
  • 09:54sixty percent chance that one
  • 09:55of those trillions of cells
  • 09:56will acquire the changes necessary
  • 09:58to get cancer. Right? So
  • 10:00this is not p fifty
  • 10:01three. You get multiple cancers
  • 10:02early in life. This is
  • 10:03not APC, where by the
  • 10:04time you're twenty or thirty,
  • 10:05your colon is filled with
  • 10:06premillion polyps. This is not
  • 10:08NF one.
  • 10:09Alright? This is a
  • 10:12per cell basis, very rare.
  • 10:13Obviously, with a trillion cells,
  • 10:14it's, it's inevitable.
  • 10:16So always consider BRCA one
  • 10:18a rare consequence of this
  • 10:19dysfunction is cancer. Right? I
  • 10:21tell my students actually, it's
  • 10:22like, you know, having a
  • 10:23shoelace. If you have your
  • 10:24shoelace is untied, the chance
  • 10:25of it catching in a
  • 10:26bicycle gear is greatly increased.
  • 10:29But the shoelace is not
  • 10:30a bicycle chain, you know,
  • 10:31catcher suppressor.
  • 10:33Alright? It is a rare
  • 10:34consequence of its dysfunction, you
  • 10:36know, unlike many other classical
  • 10:37tumors. That's the way I
  • 10:38think about it.
  • 10:41So, early on, some insight
  • 10:43to that role, of BRCA1
  • 10:46and DNA repair was actually
  • 10:47found in it was I
  • 10:48was lucky enough to work
  • 10:48with Ralph Scully early on,
  • 10:49again, as a resident and
  • 10:50fellow. We found that, hey.
  • 10:52One of the, that BRCA1
  • 10:53is really involved in esophage,
  • 10:56in part,
  • 10:57doing a DNA repair and
  • 10:58specific DNA double strand break
  • 10:59repair. We found that BRCA
  • 11:01mutant cells are very sensitive
  • 11:02to radiation therapy. And if
  • 11:04we look at the dynamics
  • 11:05of repair using old technique
  • 11:06called pulse field gel ultra
  • 11:09pulse field gel electrophoresis,
  • 11:11alright, in which we look
  • 11:12at the dynamics of how
  • 11:13DNA breaks are repaired. Normal
  • 11:14cells have a biphasic repair.
  • 11:16You get eighty percent of
  • 11:17the DNA breaks are resolved
  • 11:18in the first two to
  • 11:19three hours, and then the
  • 11:20rest of the of the
  • 11:21breaks are resolved in, the
  • 11:23next twenty four hours. It's
  • 11:24kind of biphasic annealing of
  • 11:26DNA double strand breaks. In
  • 11:28BRCA mutant cells, we noticed
  • 11:29early on that the original
  • 11:30phase of repair looked fine,
  • 11:32but it lagged
  • 11:34afterwards.
  • 11:34Alright? You had unresolved breaks.
  • 11:36And this gave us an
  • 11:37idea that this was involved
  • 11:38in fundamental aspect of DNA
  • 11:40doubles
  • 11:41that without BRCA one, you
  • 11:42get unrepaired DNA double strand
  • 11:44breaks during s phase.
  • 11:45Alright?
  • 11:47Work by a variety of
  • 11:48laboratories, especially Maria Jason, further
  • 11:50refined the role of BRCA
  • 11:51one to what we understand
  • 11:52today, our current story, which
  • 11:54is that BRCA one's involved
  • 11:55in homology mediated repair.
  • 11:57And to step back, imagine
  • 11:59that when you have a
  • 11:59DNA double strand break in
  • 12:01a cell, you have two
  • 12:02problems a cell has to
  • 12:03face. One is the DNA
  • 12:04strand is broken. You have
  • 12:06physical integrity of the of
  • 12:07the chromosome is now disrupted.
  • 12:09The second is informational loss.
  • 12:12Alright? So often DNA double
  • 12:14strand breaks,
  • 12:15or often single strand breaks.
  • 12:16When you have a single
  • 12:17strand break, you have little
  • 12:18resection at the end, and
  • 12:19you fill back in. DNA
  • 12:20double strand breaks are often
  • 12:21two single strand breaks that
  • 12:22are near each other. You
  • 12:23resect both areas and all
  • 12:24of a sudden, oops, you
  • 12:25have a DNA double strand
  • 12:26break. Alright?
  • 12:28And there, you've lost information.
  • 12:29The nucleotide sequence there is
  • 12:31gone.
  • 12:32Alright? And so there's an
  • 12:33informational challenge, which is to
  • 12:35restore that information that's lost,
  • 12:38at the edges of the
  • 12:39DNA double strand break.
  • 12:41And, so imagine the ways
  • 12:43to repair the DNA double
  • 12:44strand break, some which,
  • 12:45which dominate during s phase
  • 12:47oops. That was my pointer.
  • 12:50There we go. Some which
  • 12:51dominate during s phase. It's
  • 12:53basically just slam the ends
  • 12:54together. That's beautiful. It gets
  • 12:56rid of the structural problem
  • 12:57of the DNA double strand
  • 12:58break, but doesn't solve the
  • 13:00information problem.
  • 13:01Alright? Because in order to
  • 13:03restore the information problem, you
  • 13:04have to have a template
  • 13:06that has the missing information
  • 13:08as a template to restore
  • 13:09that information. The only time
  • 13:10you have that is an
  • 13:12s space with your sister
  • 13:13chromatid.
  • 13:14You also have your homologous
  • 13:16chromosome theoretically outside of s
  • 13:17space, but that'll that'll actually
  • 13:19not be truly error free.
  • 13:20You'll get loss of heterosic
  • 13:22acid, and that occurs occasionally.
  • 13:23Alright?
  • 13:24The other potential source of
  • 13:26information is RNA. And in
  • 13:28other organisms, there is RNA
  • 13:29templated repair, which also can
  • 13:31restore information.
  • 13:32The role of that in
  • 13:33mammals is still evolving and
  • 13:35whether that occurs or not,
  • 13:37is not clear. Alright. But
  • 13:39those so,
  • 13:41and so imagine that there
  • 13:43is, you know, all these
  • 13:44places, all these ways,
  • 13:46promote
  • 13:48resolution
  • 13:49of the structural problem. During
  • 13:50s phase, you can use
  • 13:51homology mediated recombination using the
  • 13:53sister chromatid to get error
  • 13:55free repair that also resolves
  • 13:56the informational challenge of a
  • 13:58DNA double strand break. Alright?
  • 13:59And what's interesting is is
  • 14:01that, you know, if you
  • 14:02impair
  • 14:04n h e j or
  • 14:05s single strand annealing, alright,
  • 14:07You actually don't have the
  • 14:08cells are okay. So you
  • 14:09you knock out fifty b
  • 14:10p one or ATM, it
  • 14:11caused a huge change in
  • 14:13the entrance. You get in
  • 14:14mice and humans, they're viable.
  • 14:16Okay? You can have humans
  • 14:17that live without ATM or
  • 14:18fifty b p one and
  • 14:19and mice that live without
  • 14:20them. They're small sensitive radiophone.
  • 14:22But if you mess up
  • 14:24the genes that are involved
  • 14:25in homogeneity of recombination, BRCM,
  • 14:27BRCA two, you get early
  • 14:29in mice,
  • 14:30and your chromosomes fall apart.
  • 14:32Alright? So that's really important
  • 14:34for normal man management of,
  • 14:36of s phase.
  • 14:39The interesting thing is, of
  • 14:40course, that,
  • 14:42somehow, cancers that lose BRCA
  • 14:44one in a way that
  • 14:45we still don't really understand
  • 14:47survive.
  • 14:48Or if you take any
  • 14:49normal cell, even most cancer
  • 14:50cells, and acutely get rid
  • 14:51of BRCA one, the cells
  • 14:52just fall apart. Apart. Alright?
  • 14:54But somehow, the BRCA one
  • 14:56and BRCA mutant cancer cells
  • 14:57survive and proliferate
  • 14:59in a in a reason
  • 15:00honestly, I don't think we
  • 15:01still really understand. But, interestingly,
  • 15:03they still they still carry
  • 15:05this profound defect in homology
  • 15:07mediated repair, And that makes
  • 15:08them highly sensitive to certain
  • 15:10forms of DNA damaging that
  • 15:11occurs during s phase.
  • 15:13And that includes this early
  • 15:14work from, Thomas Helen's group
  • 15:16and Alan Ashworth, which sound
  • 15:17that, you know, PARP inhibitors
  • 15:19were synthetically lethal to
  • 15:21BRCA1 and BRCA2 immune cell
  • 15:23lines. Important thing is this
  • 15:24was not even found in
  • 15:25cell lines. This is all
  • 15:25found in primary mouse both
  • 15:27both labs actually use primary
  • 15:28mouse ES cells to find
  • 15:30this. If they tried this
  • 15:30in cell lines, it wouldn't
  • 15:31have worked. We can talk
  • 15:32about why that is later.
  • 15:33Alright? They use, you know,
  • 15:34cancer cell lines. Alright. So
  • 15:36both these both these things
  • 15:37are found actually in primary
  • 15:38cells that would be our
  • 15:39same BRCA2 division.
  • 15:40Okay.
  • 15:42And the original reason rationale
  • 15:44for why this worked was
  • 15:46that, hey. You know, part
  • 15:47poly poly, ADP polymerase ribose
  • 15:49is this highly abundant enzyme,
  • 15:51and it turns on its
  • 15:53polymerase activity whenever it sees
  • 15:55single stranded DNA. So it's
  • 15:56active anytime in our genome,
  • 15:58single stranded DNA is generated.
  • 16:00And that is basically at
  • 16:02replication,
  • 16:03but also at
  • 16:06at transcription. Right? So PARP
  • 16:08is turned on every time
  • 16:09transcription fork is on because
  • 16:10you have single stranded DNA.
  • 16:11And you have also doing
  • 16:12DNA repair. So anytime you
  • 16:14have single stranded DNA, PARP
  • 16:15is turned on. Alright? It
  • 16:17causes this. And the original
  • 16:18theory for why BRCA and
  • 16:20BRCA may work is that,
  • 16:21you lose PARP. Now all
  • 16:22of a sudden you have
  • 16:23spontaneous single strand nicks with
  • 16:25PARP inhibitors. These are not
  • 16:26these are not repaired. And
  • 16:28so now, which is fine
  • 16:29because single strand DNA doesn't
  • 16:30cause anyone any harm. But
  • 16:32if these go into s
  • 16:33space, the single strand breaks
  • 16:34become DNA double strand break
  • 16:35equivalents, and these require BRCA
  • 16:37on two mediated pathway to
  • 16:38really restore the, the replication
  • 16:40for using homology immediate repair,
  • 16:42and you go on. But
  • 16:43now if you don't have
  • 16:44BRCA on BRCA two, now
  • 16:46this is now a catastrophe.
  • 16:48Alright? That was our original
  • 16:49story.
  • 16:50Alright? That it was really,
  • 16:51you know, unrepaired single strand
  • 16:53breaks that caused catastrophe.
  • 16:55Now we realize, actually,
  • 16:57it may be more complicated
  • 16:58than that. And what PARP
  • 16:59inhibitors really do is create
  • 17:01bulky adducts
  • 17:02by trapping PARP. And the
  • 17:04lesion that BRCA1 requires
  • 17:06is a,
  • 17:07is a a bulky adduct,
  • 17:09alright, that's caused by PARP
  • 17:11trapping by the inhibitor.
  • 17:12Alright?
  • 17:14That's our second story that
  • 17:15we had for why these
  • 17:16work. Now there's several more
  • 17:18stories that coming out which
  • 17:19we're going to that it
  • 17:20may be single strand gaps
  • 17:21or replication,
  • 17:24transcription interference that now coming
  • 17:25in to figure out how
  • 17:26PARP is. But it's showing
  • 17:27that, actually,
  • 17:28our story of why PARP
  • 17:29inhibitors work, alright, is still
  • 17:31in evolution
  • 17:35of,
  • 17:36in terms of how it
  • 17:37works. The other thing is
  • 17:38that it's clear that, at
  • 17:40least in our clinically active
  • 17:41drugs,
  • 17:42the activity of the drug
  • 17:43increases with the PARP trapping
  • 17:44ability. So talazaparib is and
  • 17:46and also the toxicity increases
  • 17:48with the PARP trapping ability.
  • 17:49So talazaparib is a much
  • 17:50better drug than voleparib in
  • 17:52terms of PARP trapping and
  • 17:53is actually much better single
  • 17:54agent in BRCA and BRCA
  • 17:55immune cells. This tells us
  • 17:57something very important in that,
  • 17:58actually,
  • 17:59other drugs
  • 18:01that cause bulky adducts are
  • 18:03also
  • 18:04very toxic to BRCA on
  • 18:06BRCA immune cells. So platinum,
  • 18:08topoisomerase
  • 18:09inhibitors, topo topotrappers,
  • 18:12alkylators, especially bifunctional alkylators are
  • 18:14also incredibly potent,
  • 18:17to BRC on BRCA2 mutant
  • 18:18cells.
  • 18:19Alright? So classical chemotherapy
  • 18:22is also in part
  • 18:24highly, you know,
  • 18:26we'll talk about this target
  • 18:27therapy for BRCA and BRCA
  • 18:28immune cells.
  • 18:30And so the cool thing
  • 18:31is this early,
  • 18:34identification of the sensitivity of
  • 18:35BRCA and BRCA immune cells
  • 18:37to,
  • 18:38to,
  • 18:39to PARP inhibitors translated from
  • 18:41a Nature paper in two
  • 18:42thousand and, I think, five
  • 18:44to a phase one study
  • 18:45that was published in New
  • 18:46York Medicine four years later
  • 18:47in two thousand and nine.
  • 18:49Alright? And we talked about
  • 18:50why PARP inhibitors are actually
  • 18:51already in development
  • 18:52as protectants for, ischemia because
  • 18:55they thought PARP was the
  • 18:56final pathway of death for,
  • 18:58perfusion injury in stroke and
  • 19:00heart disease. And so they
  • 19:01were already in development. But
  • 19:02they were repurposed to this,
  • 19:04and, they found that, hey.
  • 19:05This highly this is kinda
  • 19:06wild. This is a phase
  • 19:07one study in New England
  • 19:08Journal of Medicine in which
  • 19:09the entry criteria was a
  • 19:10genomic event,
  • 19:12alright, which is BRCA o
  • 19:13germline BRCA o one or
  • 19:14two mutation, alright, and showed
  • 19:16activity in a on a
  • 19:17variety of different things. And
  • 19:19this led
  • 19:20to
  • 19:21after a little bit, as
  • 19:22you guys some of you
  • 19:23might remember, after a little
  • 19:24bit of a misadventure with
  • 19:25inoperib,
  • 19:26which was ended up not
  • 19:27being a PARP inhibitor, alright,
  • 19:29and which, you know, kind
  • 19:30of sidelined the field for
  • 19:32a while. Once we recovered
  • 19:33from the inoperib
  • 19:34catastrophe,
  • 19:35finally, we got they got
  • 19:36back on track, and now
  • 19:37we have multiple approvals for
  • 19:39PARP inhibitors,
  • 19:40for BRCA and BRCA mutant
  • 19:41disease in a variety of
  • 19:43settings. Alright?
  • 19:45But
  • 19:46it's clear that none of
  • 19:48these in the setting, especially
  • 19:49advanced disease, are curative. And
  • 19:51there are multiple mechanisms acquired
  • 19:52resistant to PARP inhibitors, which
  • 19:53are very informative in us
  • 19:55both understanding the basic biology
  • 19:56of BRCA one, BRCA two,
  • 19:58and its role in tumorigenesis.
  • 20:00So if you take cells
  • 20:01that are BRCA two mutant
  • 20:03and bathe them in platinum,
  • 20:05ultimately
  • 20:06ultimately,
  • 20:07you, will,
  • 20:08get cells that are resistant.
  • 20:10Similarly, this is with Yas
  • 20:11Jankar. If you take mice
  • 20:13that are BRCA mutant and
  • 20:13treat them with olaparib, initially,
  • 20:15a beautiful responses, but then
  • 20:16you'll have outbreaks
  • 20:18of PARP resistant tumors that
  • 20:19pop up over time. Alright?
  • 20:22So
  • 20:23the mechanisms of acquired resistance
  • 20:24are actually quite informative
  • 20:26and frightening,
  • 20:27alright, and tell us a
  • 20:28lot about the biology of
  • 20:30of of BRCA1 as a
  • 20:31role in repair.
  • 20:33Alright? So the first thing
  • 20:34that was figured out was
  • 20:35by, both by Alan Ashworth
  • 20:36and by Toshi Taniguchi
  • 20:38was the concept of reversion
  • 20:39mutations. And I'll remind you
  • 20:40guys that, you know, most
  • 20:41of the BRCA1, this is
  • 20:42BRCA2 mutations, and BRCA mutations
  • 20:45are frame shift mutations
  • 20:46that introduce a premature stop
  • 20:47codon. Alright? So for example,
  • 20:49here's a classic one. There's
  • 20:50a frame shift here, which
  • 20:51theoretically will cause a truncated
  • 20:53protein by putting a premature
  • 20:54stop codon. Alright?
  • 20:55Both of these actually, most
  • 20:57of these mutations actually don't
  • 20:58create a truncated protein because
  • 21:00of nonsense media decay. Alright?
  • 21:02So you guys remember, if
  • 21:03there's a stop codon that's
  • 21:05near a splice acceptor site
  • 21:06in RNA, that RNA is
  • 21:07tagged as being bad,
  • 21:09and you get nonsense media
  • 21:10decay. So most of these
  • 21:11are functionally null.
  • 21:13Alright? Except unless you're in
  • 21:14the middle of a big
  • 21:15exon or the very terminal
  • 21:16exon where nonsense media decay
  • 21:18is you can escape from.
  • 21:19Alright? So these are all
  • 21:20functionally null. Alright? Most of
  • 21:22these.
  • 21:23Right? So then, what Alan
  • 21:25here Alan Ashworth did in
  • 21:26this paper was take cell
  • 21:28lines that had this truncating
  • 21:29mutation, bathe them in platinum,
  • 21:30got resistant cells, and then
  • 21:33took the resistant cells and
  • 21:34resequenced BRCA two. And what
  • 21:36he found was that in
  • 21:37the resistant cells, what happened
  • 21:38was the resistant cells had
  • 21:40acquired a deletion
  • 21:41that deleted the exon
  • 21:43that contained the frame shift.
  • 21:45And the edges of the
  • 21:46deletion were such that, actually,
  • 21:48the gene was put back
  • 21:49in frame.
  • 21:50Alright? And so we converted
  • 21:52a
  • 21:53functionally null frame shift into
  • 21:55an internal deletion
  • 21:57that created a, you know,
  • 21:59functional protein. So hypomorphic protein
  • 22:01that was functional in repair.
  • 22:02So it was a secondary
  • 22:03mutation
  • 22:04that actually fixed
  • 22:06the first mutation.
  • 22:07Alright? So reverse mutation. Frightening.
  • 22:09You see this in yeast,
  • 22:10but now we're seeing it
  • 22:11in humans.
  • 22:12Alright?
  • 22:13And really shows that, hey.
  • 22:14This is, the the,
  • 22:17so called reverse mutation that
  • 22:18really shows the mechanism required
  • 22:19resistance. So this is in
  • 22:20BRCA two in cell lines.
  • 22:22This is work from,
  • 22:23Toshi and,
  • 22:26Susan Swisher in human cells.
  • 22:27So this is a this
  • 22:28is in with BRCA one.
  • 22:29So this is initially a
  • 22:30patient with ovarian cancer that
  • 22:32had a truncating mutation, a
  • 22:33classic truncating mutation in BRCA
  • 22:35one two five nine four
  • 22:36del c. It was highly
  • 22:38sensitive to platinum initially. Upon
  • 22:39platinum resistance, they sequenced
  • 22:41the tumor, and this tumor
  • 22:42had, in addition to the
  • 22:44two five nine four del
  • 22:45c, had
  • 22:48an eleven nucleotide sorry.
  • 22:52No.
  • 22:53An eleven nucleotide deletion downstream.
  • 22:55And so all of a
  • 22:56sudden eleven plus one is
  • 22:57twelve, twelve is divisible by
  • 22:58three, you're back in frame.
  • 23:00Alright? So it basically converts
  • 23:02a,
  • 23:07a frame shift mutation into,
  • 23:08again, a small internal deletion.
  • 23:11Alright? Putting the gene back
  • 23:12in frame
  • 23:14and having the cells,
  • 23:15you know, produce BRCA1 again.
  • 23:17Alright?
  • 23:18With a small internal deletion.
  • 23:20And if you look at
  • 23:21the edges
  • 23:23of the deletions,
  • 23:24they all have microhomology.
  • 23:26And so this is the
  • 23:27BRCA1 mutant cells inappropriately using
  • 23:29non homologous end joining to
  • 23:31slam together whenever they see
  • 23:32a break they just slam
  • 23:33together the ends. And so
  • 23:34this is in fact the
  • 23:35inappropriate repair mechanism that's generating
  • 23:37these reversion mutations.
  • 23:39Alright? So it's really wild.
  • 23:41Here you're getting putting back
  • 23:42in BRCA1 back in frame,
  • 23:45under selection.
  • 23:46Right?
  • 23:48The second mechanism of acquired
  • 23:49resistance, which was done by
  • 23:51our laboratory
  • 23:52together with,
  • 23:54Jass Jonker's laboratory as well
  • 23:55as found, by,
  • 23:58Andre Nusenzweig and Sam Bunting,
  • 24:01was compensating mutations. These were
  • 24:03acquired mutations in other parts
  • 24:05of the pathway that also
  • 24:06required resistance. This is kinda
  • 24:07wild. Let's just go do
  • 24:08a little bit of the
  • 24:08story,
  • 24:09which is what we did
  • 24:11with Yas
  • 24:13a while back,
  • 24:14was we said, you know,
  • 24:15what genetic events will allow
  • 24:17ES cells to tolerate BRCA
  • 24:19one loss? And what he
  • 24:20set up was an ESL
  • 24:21in which one allele of
  • 24:22BRCA1,
  • 24:25one allele of BRCA1,
  • 24:26you know, has a huge
  • 24:27deletion from exons three to
  • 24:28actually
  • 24:30thirteen here. And so it's
  • 24:31this giant deletion. And the
  • 24:33other allele has exons five
  • 24:34and six floxed.
  • 24:36Alright? And more importantly, not
  • 24:38only was it floxed,
  • 24:39but it was triggered in
  • 24:40such a way that there's
  • 24:41a pure micell resistant cassette
  • 24:42that was out of frame.
  • 24:43So when,
  • 24:45cui is expressed, it not
  • 24:46only excises exons five and
  • 24:48six, but puts the puro
  • 24:50cassette in frame. And so
  • 24:51the tumors the cells become
  • 24:52BRCO deficient and puro resistant.
  • 24:55And this is important because
  • 24:56what what the idea is
  • 24:57that you add on tamoxifen
  • 24:59and then add puramycin to
  • 25:00enforce that only the cells
  • 25:01have undergone recombination
  • 25:03are present. And when you
  • 25:04do that, you basically get
  • 25:05all the cells die. You
  • 25:06can recover this by putting
  • 25:08an exogenous BRCA one, showing
  • 25:09this is dependent on BRCA
  • 25:11one. So it's the thing
  • 25:11where we can get acute
  • 25:13deletion of BRCA
  • 25:14one in,
  • 25:15in this ESL.
  • 25:17And then the game was
  • 25:18then we use,
  • 25:20piggyback retrotransposons
  • 25:21system
  • 25:22to cause saturating mutagenesis
  • 25:25and say, let's turn on
  • 25:26piggyback retrotransposons.
  • 25:27So these are little retrotransposons
  • 25:29that can hop. They can
  • 25:30insert into an intron and
  • 25:31cause capture, or they can
  • 25:33insert in front of a
  • 25:33promoter and turn on a
  • 25:34gene. And the idea is
  • 25:36let's turn on a piggyback
  • 25:37retrotransposon,
  • 25:39add tamoxifen,
  • 25:40add puramycin,
  • 25:41and see if any cells
  • 25:42survive. If they do, what
  • 25:44happened?
  • 25:45Alright? What was the genetic
  • 25:46event there that caused allowed
  • 25:47these cells to survive?
  • 25:49And this was done initially,
  • 25:50Peter Baumann in in, a
  • 25:51lab that Amal Ali was
  • 25:53working on this. What we
  • 25:54found was that when Peter
  • 25:55Baumann looked at two hundred
  • 25:56and fourteen clones that came
  • 25:58out of this assay, Two
  • 25:59hundred of them had duplication
  • 26:00of the endogenous VRCA one
  • 26:02allele.
  • 26:03Alright? So they still had
  • 26:04VRCA one. They just had
  • 26:05one of the alleles had
  • 26:06the event. Alright?
  • 26:08And allowed pureomycin resistance.
  • 26:10The other
  • 26:11all had deletions and secondary
  • 26:14loss of of a gene
  • 26:15called fifty b p p
  • 26:15one, which is important in
  • 26:17non homologous
  • 26:18recombination.
  • 26:19And sorry. In, an in,
  • 26:21in end joining. Alright? So
  • 26:23this is kinda wild. Loss
  • 26:24of fifty b p one
  • 26:25allows cells to survive
  • 26:27BRCA one loss.
  • 26:29Alright?
  • 26:30And we thought, oh, maybe
  • 26:31this is just a checkpoint
  • 26:32effect. Alright? So just like
  • 26:34p fifty three, somehow p
  • 26:35fifty b p one is
  • 26:36involved in a checkpoint. And
  • 26:37so cells are tremendously bad,
  • 26:38but somehow they survive. But
  • 26:40to our surprise,
  • 26:42these double deficient cells actually
  • 26:44restored Raf fifty one foci
  • 26:45formation and restored homology dependent
  • 26:47repair. So homology dependent repair
  • 26:49was actually restored
  • 26:51in these cells. Alright? It
  • 26:53wasn't just some sort of
  • 26:54checkpoint loss. And so this
  • 26:55is highly, you know,
  • 26:57unexpected and said, you know,
  • 26:58how could two wrongs make
  • 26:59a right? Getting rid of
  • 27:00BRCA one, supposedly essential for
  • 27:02homologous recombination, heading getting rid
  • 27:03of a non homologous end
  • 27:04joining protein,
  • 27:06or something involved in that
  • 27:07pathway.
  • 27:08How did that restore?
  • 27:09Alright? Without going into the
  • 27:10nitty gritty details, it's worked
  • 27:12out both by,
  • 27:13Sam Bunting and Nusenzweig as
  • 27:14well as, our laboratories,
  • 27:16is this really tells a
  • 27:17lot about how BRCA one,
  • 27:20works in DNA repair. So
  • 27:22the story is really you
  • 27:23know, it's all depends on
  • 27:24how the ends are processed
  • 27:26after DNA break. So imagine
  • 27:27during,
  • 27:28fifty d p v one
  • 27:29is expressed during all phases
  • 27:31of cell cycle. So unlike
  • 27:32BRCA one, which is restricted
  • 27:33in s phase, fifty d
  • 27:34p one is present during
  • 27:35all parts of the cell
  • 27:36cycle.
  • 27:37When, you have a DNA
  • 27:38double ramp break, what fifty
  • 27:40b p one does is
  • 27:41basically it prevents end resection.
  • 27:43It prevents the ends from
  • 27:44being processed to chew back.
  • 27:46If you can't chew back
  • 27:47the ends, all you can
  • 27:49do is slam them together.
  • 27:50Alright? So,
  • 27:52it promotes anomalous end joining
  • 27:53during, outside of s phase.
  • 27:56During s phase, BRCA one
  • 27:58is
  • 27:59expressed. And in a mechanism
  • 28:01that we a lot of
  • 28:02play ups are still working
  • 28:03on, this basically suppresses this
  • 28:04function for the b p
  • 28:05p one and allows end
  • 28:06processing by the CTPI MRN
  • 28:08complex to chew back the
  • 28:09ends since we have a
  • 28:10free end that can now
  • 28:11be free for homology search.
  • 28:13Alright?
  • 28:14Now in cells that lack
  • 28:15BRCA one, what we think
  • 28:17is occurring is that fifty
  • 28:19fifty b p p one
  • 28:20is able to do an
  • 28:21s phase what it does
  • 28:22outside of s phase, which
  • 28:23is to block end processing.
  • 28:25And when you do that,
  • 28:26also now you no longer,
  • 28:28can do homology mediated repair
  • 28:30because you don't have any
  • 28:30ends to search for,
  • 28:32and genomic chaos ensues.
  • 28:34Right? If you lose fifty
  • 28:36b p one,
  • 28:37all of a sudden, actually,
  • 28:38CTFP m MRN complex, which
  • 28:40is turned on by,
  • 28:42cyclin dependent kinases,
  • 28:43can still do its job.
  • 28:44You actually get hyperresection, but
  • 28:45you allow HR to go
  • 28:47on. And so this shows
  • 28:48that BRCA one wasn't involved
  • 28:49in the nitty gritty of
  • 28:50DNA repair,
  • 28:52but was a manager that
  • 28:53was involved in repair choice.
  • 28:56Alright? And, the secondary mutation
  • 28:58in b r in fifty
  • 28:59b one
  • 29:01actually allowed compensation,
  • 29:03alright,
  • 29:04and allowed,
  • 29:05a homology media repair to
  • 29:07occur. Alright?
  • 29:09And in fact, so you
  • 29:10imagine, you know, with a
  • 29:11lot of work from multiple
  • 29:12laboratories, there are multiple mixes
  • 29:13required in the cyst depart
  • 29:14inhibitors.
  • 29:15Alright? And we kind of,
  • 29:16in this review, kind of
  • 29:17characterize this as,
  • 29:19several, as several independent potential
  • 29:22mechanisms. And understand the mechanism
  • 29:24resist is really important to
  • 29:25understand how to, you know,
  • 29:27kind of, deal with it.
  • 29:28So one is that there
  • 29:29are drug specific mechanisms that
  • 29:31I've seen on top. So,
  • 29:32for example, olaparib is a
  • 29:33beautiful PGP substrate. So upregulation
  • 29:35of drug pumps,
  • 29:36alright, is just like you
  • 29:37do for anthracyclines,
  • 29:39will actually is a well
  • 29:40recognized mechanism of acquired resistance.
  • 29:42If that's what's occurring, you
  • 29:43can just switch agents.
  • 29:45Alright? So these are drug
  • 29:46specific mechanisms. Also, some some
  • 29:48turning on a park, a
  • 29:49park can also do that.
  • 29:50And so if you switch
  • 29:51drugs,
  • 29:52you can actually still get
  • 29:54activity.
  • 29:55If you have reversion pathway
  • 29:57reversion, either by genetic reversion
  • 29:59or by,
  • 30:02compensating mutations,
  • 30:03now homology media repairs is
  • 30:06is restored,
  • 30:07and you have far fewer
  • 30:08options to be able to,
  • 30:10to handle that. Alright? Especially
  • 30:11with genetic reversion, now you've
  • 30:13lost the phenotypic difference between
  • 30:14the tumor cells and normal
  • 30:16cells in terms of repair,
  • 30:17and you don't have a
  • 30:18handle, to do it on.
  • 30:19So understanding the mechanism of
  • 30:21of of resistance is really
  • 30:22important to figure out what
  • 30:23we can do.
  • 30:24So with this as a
  • 30:25background,
  • 30:26you know,
  • 30:27these
  • 30:28mechanisms of reversion
  • 30:29and resistance tell us a
  • 30:31lot about the basic biology
  • 30:32of BRCA one and BRCA
  • 30:33two. So what it tells
  • 30:34us that, hey. You know,
  • 30:35platinum,
  • 30:36you know, anthracyclines, alkylating agents,
  • 30:38all of those induce reversion
  • 30:40mutations in the clinic as
  • 30:41mexis required resistance.
  • 30:43That shows they're all on
  • 30:44target.
  • 30:45Alright? That
  • 30:47old fashioned alkylating agents are
  • 30:49like PARP inhibitors. It is
  • 30:51like topoisomerase inhibitors. They are
  • 30:53all acting on the BRCA1
  • 30:54dependent pathway because all of
  • 30:56them exert selection pressure for
  • 30:57reversion mutations. So they're all
  • 30:59on target. Right?
  • 31:01So,
  • 31:03if that's true, alright, and
  • 31:05you think about this, if
  • 31:06classical chemotherapy is in fact
  • 31:07targeted therapy for underlying defects
  • 31:08in the repair, our entire
  • 31:10dosing needs to be, you
  • 31:11know, maximum tolerated dose makes
  • 31:12no sense. It's a it's
  • 31:13a target agent when you
  • 31:14think about it as any
  • 31:15other target agent find the
  • 31:16extended exposure to the minimum
  • 31:18effective dose.
  • 31:19Second thing is you imagine
  • 31:20that BRCA one tumors can
  • 31:22tolerate reconstitution of BRCA one
  • 31:23in HR mediated repair. Alright?
  • 31:25So you can't cure a
  • 31:26BRCA one division tumor by
  • 31:28putting back in wild type
  • 31:29BRCA one. It's not like
  • 31:30p fifty three. If you
  • 31:31put back in BRCA1 into
  • 31:33a BRCA2 tumor, tumor is
  • 31:34happier.
  • 31:35Alright? This tells us strongly
  • 31:36that BRCA1 loss and the
  • 31:38repair defect is required for
  • 31:39initiation of tumorigenesis,
  • 31:41but not required for maintenance.
  • 31:43Alright? So,
  • 31:45we call this tumor suppressor
  • 31:46tolerance.
  • 31:47Alright? So this is not
  • 31:48p fifty three, but back
  • 31:49p fifty three, the cell
  • 31:49dies. This is,
  • 31:52you know, shows us really
  • 31:53quite. And also tells us
  • 31:54that second thing is, you
  • 31:56know, what is actually causing
  • 31:57the growth of b r
  • 31:58these BRCA and BRCA two
  • 31:59deficient tumors.
  • 32:00Alright.
  • 32:01It's not loss of BRCA.
  • 32:03Right? And we try why
  • 32:04this department plan is gonna
  • 32:06correlate.
  • 32:07With that background, what I'm
  • 32:08gonna switch next to is
  • 32:09that, you know, for not
  • 32:10right now, you know, next
  • 32:11generation sequencing of tumors has
  • 32:13now become commonplace for many
  • 32:14cancers. Alright? So now there's
  • 32:15a variety of hybrid capture
  • 32:16based methods in which now
  • 32:18where people are constantly,
  • 32:20doing tumor sequencing. In fact,
  • 32:21at Rutgers, we started doing
  • 32:22this since two thousand twelve
  • 32:24in which all patients, adult
  • 32:25pediatric, e malignancy,
  • 32:27were reviewed, and we have
  • 32:27a central molecular tumor board
  • 32:29that came up. And we
  • 32:29generate a lot of information
  • 32:31from this. And one of
  • 32:32the things I'm gonna talk
  • 32:33to you about is, you
  • 32:34know, how do we deal
  • 32:35with unanticipated,
  • 32:36unexpected
  • 32:37mutations in BRC on BRCA
  • 32:38two, other DNA repair tracks
  • 32:39that we find on tumor
  • 32:40sequencing? How do we figure
  • 32:42out whether they're real? You
  • 32:43know?
  • 32:44Are they actionable? How do
  • 32:45we figure this out? And
  • 32:46I'll start again with this
  • 32:47by giving you a case
  • 32:48because all our things start
  • 32:49with us puzzling this over
  • 32:50in our molecular report. How
  • 32:51do we deal with this?
  • 32:52And so our first case
  • 32:53is back in two thousand
  • 32:54and twelve when we first
  • 32:55started doing this. This is
  • 32:56when Foundation Medicine was first,
  • 32:58founded,
  • 32:59by friends of ours, and
  • 33:00so we collaborate with them.
  • 33:02It's a fifty nine year
  • 33:03old gentleman with a kidney
  • 33:05cancer. He presented in the
  • 33:07usual way, found to have
  • 33:07this large mass, underwent resection,
  • 33:09and unfortunately had rapid recurrence,
  • 33:11in both the nephrectomy med
  • 33:12and in the lung. He
  • 33:13was treated with what was
  • 33:14then the standard of care,
  • 33:16at that point and had
  • 33:17either, you know, rapid progression
  • 33:18or intolerance.
  • 33:19And so he was then
  • 33:20sent to our molecular tumor
  • 33:22sequencing protocol.
  • 33:23And in addition to the
  • 33:25alterations in NF two, CDK
  • 33:26and ARRAD one a, there
  • 33:28was a a classic BRCA
  • 33:29two truncating mutation. In fact,
  • 33:31this is a well known
  • 33:32founder mutation in BRCA two.
  • 33:34Alright? And so we sequenced
  • 33:36his germline and found that,
  • 33:37in fact, yeah, he's a
  • 33:38germline carrier of this BRCA
  • 33:40two mutation.
  • 33:41Right? So now the question
  • 33:43is, okay.
  • 33:45Here's a guy with a
  • 33:46germline BRCA two mutation. Just
  • 33:47because you're heterozygous or BRCA
  • 33:49two doesn't mean you're immune
  • 33:50to normal tumorigenesis.
  • 33:51Alright? So is this just
  • 33:52a sporadic renal cell cancer
  • 33:54occurring in a BRCA two
  • 33:55mutation carrier,
  • 33:57or is this a BRCA
  • 33:58two deficient cancer?
  • 34:00And with the underlying DNA
  • 34:02repair defect and the sensitivity
  • 34:04to agents we never treat
  • 34:05renal cell cancer with.
  • 34:07Alright? And can we figure
  • 34:08this out?
  • 34:10Right?
  • 34:12And so, interestingly,
  • 34:14you can.
  • 34:16So this is his,
  • 34:17the allele frequency of BRCA2
  • 34:19in his tumor specimen.
  • 34:21And then we have an
  • 34:22estimated tumor
  • 34:23of sixty two percent. And
  • 34:24we have an estimated tumor
  • 34:26purity in this specimen we
  • 34:26talked about how that's done
  • 34:28by genomics of about thirty
  • 34:30percent.
  • 34:31All right? And so can
  • 34:32we make a model in
  • 34:33which you figure out, you
  • 34:34know, if you imagine how
  • 34:35BRCA will lose a match,
  • 34:36can we figure out what
  • 34:36the expected allele frequency would
  • 34:37be in a mixed tumor
  • 34:39of wild type and normal
  • 34:40cells which have or have
  • 34:42not gone undergone or lost
  • 34:43the wild type allele. Alright?
  • 34:45And and so and the
  • 34:46cool thing is, you know,
  • 34:47with next, we have,
  • 34:49news this is done through
  • 34:50a hybrid capture based sequence.
  • 34:51And so imagine you make
  • 34:52a very simple model in
  • 34:54which
  • 34:54seventy percent of the DNA
  • 34:56that we sequence from this
  • 34:57tumor, it comes from stroma.
  • 34:59And like the rest of
  • 35:00his body, it will give
  • 35:01you one mutant allele of
  • 35:02BRCA two and one wild
  • 35:03type allele.
  • 35:04Thirty percent of the DNA
  • 35:06is coming from his tumor.
  • 35:07Alright? It'll certainly give you
  • 35:08one mutant allele.
  • 35:10And we know how BRCA
  • 35:11two, for the most part,
  • 35:12undergoes loss rise as a
  • 35:13gas. So you get large
  • 35:14deletions
  • 35:15around the,
  • 35:16around the locus. Interestingly, mechanism
  • 35:18unclear. Right? So if that
  • 35:20had occurred, you'd get no
  • 35:21immune no allele at all.
  • 35:23Alright?
  • 35:24And so then the estimated
  • 35:26tumor allele frequency in this
  • 35:27setting is the number of
  • 35:28mutant alleles over the total
  • 35:30alleles, so hundred over hundred
  • 35:31and seventy, or you'd expect
  • 35:32an allele frequency of fifty
  • 35:33eight percent.
  • 35:35Alright?
  • 35:36The null hypothesis here is
  • 35:37fifty percent. Right? That's, l
  • 35:39o that's just germline with
  • 35:40no l o h. So
  • 35:42can we possibly tell the
  • 35:43difference between fifty and fifty
  • 35:44eight percent?
  • 35:45Alright.
  • 35:46Cool thing is that with
  • 35:48hybrid capture based sequencing, we're
  • 35:50going to details, basically, because,
  • 35:52the depth there with hybrid
  • 35:53capture based sequencing, you randomly
  • 35:55fragment the genome so that
  • 35:57every region have a different
  • 35:58start site. Alright? If you
  • 35:59see a lot of reads
  • 36:00with the same start site,
  • 36:01it's actually a PCR artifact.
  • 36:02You can actually get numbers.
  • 36:04So when when they say
  • 36:04it's a depth of four
  • 36:05seventy one, that means four
  • 36:06seventy one different DNA molecules
  • 36:08from the samples are read.
  • 36:09With that, you can do
  • 36:10statistics,
  • 36:11okay, and actually put in
  • 36:13confidence intervals.
  • 36:14And to our surprise,
  • 36:16alright,
  • 36:16he had a allele frequency
  • 36:17sixty two percent. This with
  • 36:18the confidence intervals around it
  • 36:19for that depth. This rules
  • 36:21in LOH and rules out
  • 36:22lack of LOH.
  • 36:24Alright? And I didn't believe
  • 36:25this initially because I was
  • 36:26like, I expected this to
  • 36:27be carrier. And so because
  • 36:29we had no other we
  • 36:29had no other option
  • 36:36at that time, we treated
  • 36:36the patient with a platinum
  • 36:36based regimen, which we never
  • 36:36treat renal cell carcinoma with.
  • 36:37And the patient had a
  • 36:37dramatic response. Alright?
  • 36:38And so we said, okay.
  • 36:38Here's a way we can
  • 36:39start figuring this out on
  • 36:40expect you know, can we
  • 36:42figure out a little bit
  • 36:42of status? Well, this is
  • 36:43a one off. We literally
  • 36:44were doing this on the
  • 36:45back of a napkin to
  • 36:45figure out what this would
  • 36:46be like. And so we
  • 36:47say imagine that we should
  • 36:48be doing this for our
  • 36:48tumor sequencing. Imagine that in
  • 36:50a germline
  • 36:51setting, you can have, you
  • 36:52know, you if there's a
  • 36:53germline mutation, you can have,
  • 36:54you know, lOH by
  • 36:56gene deletion. You can have
  • 36:57LOH by gene conversion. This
  • 36:59is what happens with p
  • 36:59t three, or you can
  • 37:00have no LOH. And for
  • 37:02each of these, you can
  • 37:03write a relationship between the
  • 37:04mutant allele frequency and the
  • 37:05tumor purity.
  • 37:06Alright? So for example, here,
  • 37:08if there's no allergens in
  • 37:08the tumor, regardless of the
  • 37:09tumor purity, the mutant allele
  • 37:11frequency will be just fifty
  • 37:12five at least fifty percent.
  • 37:13Alright? And the same thing,
  • 37:15you do this for, you
  • 37:16know,
  • 37:17somatic mutations too. Somatic mutations
  • 37:19can either be, you know,
  • 37:22heterozygous in in the in
  • 37:23the patient or can or
  • 37:25for tumor suppressor can convert
  • 37:27to, homozygosity
  • 37:28either by gene conversion or
  • 37:30by gene deletion. So for
  • 37:31example, p fifty three often
  • 37:33does this. Right? You it
  • 37:34it undergoes LOH by gene
  • 37:36conversion.
  • 37:37Alright?
  • 37:38And, again, you can write
  • 37:39the equations. And for our
  • 37:41for our clinicians, we develop
  • 37:43nomograms to help them understand
  • 37:45this and use this. Alright?
  • 37:46So imagine here's, the correlation
  • 37:48between true purity and the
  • 37:49specimen and the mutant allele
  • 37:50frequency.
  • 37:52Alright? And, in in blue
  • 37:54are all the lines that
  • 37:54look at somatic alterations. In
  • 37:56red are all the different
  • 37:57models, you know, no LOH,
  • 37:59LOH by,
  • 38:01gene deletion, LOH by gene
  • 38:02conversion
  • 38:03for,
  • 38:04a germline mutation.
  • 38:06And if we just use
  • 38:07this, we just plot where
  • 38:08our patient was, thirty percent,
  • 38:09tumor purity, mutant low frequency
  • 38:11of sixty one percent, and
  • 38:12put in the confidence intervals.
  • 38:13You can see, ah, it
  • 38:14already matches one of these.
  • 38:16And if you don't understand
  • 38:16what the tumor purity is,
  • 38:17you can put in you
  • 38:18can slide the tumor purity
  • 38:19around. You can say, hey.
  • 38:20What models does it correlate?
  • 38:21And if we knew nothing
  • 38:22else, we could tell already
  • 38:24that this is gonna be
  • 38:25a germline alteration
  • 38:26with likely l o h,
  • 38:27gene deletion.
  • 38:29Alright?
  • 38:30The other mutations that are
  • 38:31seen here, like ARD one
  • 38:32eight, should also fit in
  • 38:33that true impurity in one
  • 38:34of these models,
  • 38:35And they do. It's actually
  • 38:36a way to figure,
  • 38:38this out. Alright? And in
  • 38:40fact, we started doing this
  • 38:41for all our tumor sequences
  • 38:42to understand what the allelic
  • 38:44status is. Alright? This is
  • 38:45why p p three always
  • 38:45has a higher allele frequency
  • 38:46because it has undergone LOH
  • 38:48by gene conversion. Alright? Because
  • 38:49there's two copies in every
  • 38:50cell. Not more cells have
  • 38:51it. Alright?
  • 38:54And so, you know, with
  • 38:55this, with work from Nahed
  • 38:57Jalul and Hussain Chibani, who's
  • 38:59now at Genron,
  • 39:02We actually made, you know,
  • 39:03models, calculators online. We call
  • 39:04logic for LOH and germline
  • 39:06inference calculator, alright, in which
  • 39:08you can take all the
  • 39:09the allele frequencies and turn
  • 39:11for a given term purity
  • 39:12will, a, help you identify
  • 39:14the term purity and give
  • 39:15you an estimate of what
  • 39:16the model is for each
  • 39:17alteration, you know, whether it's
  • 39:19somatic, germline,
  • 39:20undergone LOH or not. Alright?
  • 39:23And also depending on the
  • 39:24local ploidy d two, which
  • 39:25we won't, go on to.
  • 39:26And, we validated this so
  • 39:28that we'd this is with
  • 39:29Judy Garber, actually, from the
  • 39:31Brigham. We had a large
  • 39:32dataset in which they had
  • 39:33done, somatic tumor sequencing and
  • 39:34germline sequencing and showed that
  • 39:35we can, in fact, highly
  • 39:37predict germline sequence germline status.
  • 39:39But just by looking at
  • 39:40tumor sequencing,
  • 39:43for every tumor except for
  • 39:44p for every gene except
  • 39:45for p t three. And,
  • 39:47I would like math versus
  • 39:48I will I will leave
  • 39:49that as an exercise so
  • 39:50that we know why it
  • 39:51fails for p fifty three.
  • 39:52Alright. I can talk about
  • 39:53it later. Alright.
  • 39:56Alright.
  • 39:58But then we said, okay.
  • 39:59We have these kind of
  • 40:00tools.
  • 40:01Can we now sit there
  • 40:02and go to a large
  • 40:03dataset and sit there and
  • 40:04go, how often are there
  • 40:05pathogenic BRCA and BRCA two
  • 40:06mutations seen in tumor sequencing
  • 40:08across
  • 40:11tumors.
  • 40:12And so we collaborated with,
  • 40:14Foundation Medicine, and this is
  • 40:16work for Eden Sokol and
  • 40:17Hossein Kivani in here. And
  • 40:18we said, okay. If you
  • 40:19look at their initial dataset
  • 40:20of two hundred thousand cancers,
  • 40:22how many times are there
  • 40:23true truncating mutations in BRCA
  • 40:24or BRCA two in different
  • 40:25cancer types? And you can
  • 40:27see that, hey. You know,
  • 40:28there's a reasonable frequency in
  • 40:29ovarian cancer, interestingly unexpected at
  • 40:32that time in prostate cancer,
  • 40:35squins skin squamous cells, which
  • 40:36we'll talk about, breast and
  • 40:38these others. Alright? And you
  • 40:39can see how in addition
  • 40:40to what we expected,
  • 40:42alright,
  • 40:43there's also things like, squamous
  • 40:44cell skin cancer and some
  • 40:46of these others. And, can
  • 40:47you guys guess why squamous
  • 40:48cell skin cancer has a
  • 40:49high allele high percentage of
  • 40:51bad BRC on two mutations.
  • 40:54It's the highest mutation burden
  • 40:55cancer we have, so every
  • 40:56gene is mutated,
  • 40:59in there. Alright?
  • 41:02But now if something goes
  • 41:03instead of this, let's now
  • 41:05use our approach and say
  • 41:06which ones can we call
  • 41:07this biallelic.
  • 41:09Alright. And what's interesting is
  • 41:10is that all of a
  • 41:12sudden,
  • 41:13ovary,
  • 41:14breast,
  • 41:15pancreas stays well.
  • 41:17Okay. Everything else drops to
  • 41:19one or two percent.
  • 41:20Alright. So most of the
  • 41:21alterations we see in other
  • 41:22tumors are somatic and heterozygous,
  • 41:24often the setting of high
  • 41:25mutation burden. So for example,
  • 41:26mismatch repair, endometrial cancer. You'll
  • 41:28see lots of truncating mutations,
  • 41:29BRCA one or PALB two,
  • 41:31other things, and they're bystander.
  • 41:32If you look at them,
  • 41:33they're somatic and heterozygous.
  • 41:35Alright? And if you look
  • 41:35at where the mutation is,
  • 41:36they're in a polynucleotide
  • 41:38repeat in the middle of
  • 41:38BRCA one because that's where
  • 41:39mismatch repair causes mutations.
  • 41:41Alright?
  • 41:42So those are bystanders they
  • 41:43should be activated on. Right?
  • 41:46And,
  • 41:47but interestingly, there's still a
  • 41:48low level of cancers that
  • 41:50really have bialylic
  • 41:51LH, like one or two
  • 41:52percent. But, you know, one
  • 41:53or two percent of lung
  • 41:53cancer is a lot of
  • 41:54cancer.
  • 41:55You know, it's almost like
  • 41:56more than fifteen percent of
  • 41:57ovarian cancer. Alright?
  • 42:00That's there. Alright. And so
  • 42:02so if you look at
  • 42:03kind of, you know, when
  • 42:04you see germline or somatic
  • 42:05mutations, how often do we
  • 42:06see LOH or BRCA one
  • 42:07or BRCA two? So interestingly,
  • 42:09in high grade serous ovarian
  • 42:10cancer,
  • 42:11if you see a predicted
  • 42:13germline or predicted somatic alteration,
  • 42:14either BRCA or BRCA two,
  • 42:16over ninety five percent of
  • 42:17tumors have clear genomic evidence
  • 42:19of loss fedoras egosy, loss
  • 42:20of the wild type
  • 42:23allele. Alright? In breast, it's
  • 42:25about eighty five percent, alright,
  • 42:27have undergone LOH. And we
  • 42:28can talk about why that
  • 42:29is, you know, because, obviously,
  • 42:31as you get older, there
  • 42:32are in fact,
  • 42:34you know, wild type tumors
  • 42:35that that progress in through.
  • 42:36You know? You know, not
  • 42:38every tumor that are a
  • 42:39woman who has a germline
  • 42:40BRCA one, two mutation is
  • 42:41not immune to normal tumorigenesis.
  • 42:43It's an additive effect. Right?
  • 42:46Prostate is very interesting. If
  • 42:47you have a germline BRCA
  • 42:49two mutation, the cancer that
  • 42:50arise almost always have undergone
  • 42:51LOH.
  • 42:52BRCA one, not so much.
  • 42:54Even if your germline BRCA
  • 42:56one, most of those are
  • 42:56not under LOH. So those
  • 42:58are so BRCA two is
  • 42:59a prostate cancer gene. BRCA
  • 43:01one, not so much.
  • 43:04Alright.
  • 43:05From this analysis. And you
  • 43:06can see pancreas cancer, you
  • 43:07also see, again, BRCA germline
  • 43:09there, somatic alterations are harder
  • 43:11to pick out. And if
  • 43:11you're outside of those four
  • 43:12genes, even if you're germline,
  • 43:14most have not undergone l
  • 43:15o h. Alright? So they're
  • 43:17bystanders. There's a way to
  • 43:18pick up, hey. Is these
  • 43:19bystander mutations? There's really are
  • 43:20there are are there really,
  • 43:22alterations going on? Next question
  • 43:24you can ask is, okay.
  • 43:25We we show this, you
  • 43:26know, biolytic alterations in a
  • 43:28small subset of cancers outside
  • 43:29of the ones we expect.
  • 43:31Alright?
  • 43:32Do they,
  • 43:33really have an underlying DNA
  • 43:34repair defect, and will it
  • 43:35be sensitive to these agents?
  • 43:37Is it just some sort
  • 43:37of statistical anomaly? Is there
  • 43:39a way we can find
  • 43:39out? Right? So one way
  • 43:41you can look at that
  • 43:41is actually look at you
  • 43:42know, there are these markers
  • 43:43which aren't great, but okay
  • 43:45in the setting of BRCA
  • 43:45and BRCA two, which says,
  • 43:46hey. You know, when you
  • 43:47have BRCA and BRCA loss,
  • 43:48the whole genome falls apart,
  • 43:49and then you have clonal
  • 43:50selection. And so you can
  • 43:51see the pattern of genomic
  • 43:52changes, gross chromosomal alterations
  • 43:55that you can you can
  • 43:56impute by,
  • 43:58by targeted sequencing. And so
  • 43:59there are these features like
  • 44:00c m ary choice. We
  • 44:01have, you know, global LOH,
  • 44:04telomerelic
  • 44:05allelic imbalance, large scale transitions,
  • 44:07which are all different ways
  • 44:08that these are chromosomal abnormalities,
  • 44:09kinda like that original karyotype
  • 44:11I showed you of cells
  • 44:11falling apart, without BRCA one,
  • 44:13BRCA two. We can actually
  • 44:15measure global LOH,
  • 44:17using
  • 44:18high,
  • 44:19quantitative that, especially in the
  • 44:21high-depth sequencing that's done for
  • 44:22foundation medicine, but also in
  • 44:23certain other high depth
  • 44:27hybrid capture based sequencing approaches.
  • 44:29And so the question we
  • 44:30ask is, okay, What is
  • 44:31the level of global LOH
  • 44:33in the tumors that we
  • 44:33say are monoelelic
  • 44:35versus biallelic for BRCA one?
  • 44:37Okay. And what we can
  • 44:39see is that in multiple
  • 44:40tumor types,
  • 44:41when there is biallelic
  • 44:43status, the global LOH is
  • 44:45elevated.
  • 44:46Alright? But it's either monolelelic
  • 44:48or wild type or low,
  • 44:50suggesting that in these rare
  • 44:51cancers
  • 44:52that when we impute biolelic
  • 44:53loss, there's in fact, gene
  • 44:56evidence of a global global,
  • 44:58you know, genomic instability pattern
  • 45:00underlying this, suggesting that these
  • 45:02are really, you know,
  • 45:04affecting the genomic instability of
  • 45:05the term and maybe there.
  • 45:06So imagine that, you know,
  • 45:08biolig loss is associated with
  • 45:10evidence of an HRD signature
  • 45:12in multiple cancer types, and
  • 45:13maybe a better biomarker
  • 45:15or even chemotherapy sensitivity,
  • 45:18than just the presence of
  • 45:19the mutation alone. Alright. You
  • 45:20have to see what's the
  • 45:21allelic status, and is it
  • 45:22really is it really an
  • 45:23early clone of it?
  • 45:25Alright? And it's important because,
  • 45:26you know, is we actually,
  • 45:28Ethan published this with, others,
  • 45:30that are here that, you
  • 45:30know, for example, we see
  • 45:32this in a classic things.
  • 45:33We see lots of monolithic
  • 45:34somatic mutations, BRCA and BRCA
  • 45:36two, microsatellite unstable tumors. And
  • 45:38these are a side effect
  • 45:40of the of the underlying
  • 45:41mismatch repair and not a
  • 45:42driver.
  • 45:43Alright? And so, we won't
  • 45:44go to this data, but
  • 45:45that's just, you know, it's
  • 45:46important because you wanna be
  • 45:47distracted by BRC and BRC
  • 45:48mutations if they're not biallelic.
  • 45:51Alright? And in fact, in
  • 45:53many high mutation burden cancers,
  • 45:55you will see point mutation
  • 45:56burden cancers, you will see
  • 45:57this. Now the question is,
  • 45:58hey. You know, can PARP
  • 45:59PARP inhibitors work
  • 46:01for mutations in other DNA
  • 46:02repair genes? Now we developed
  • 46:03a method to look at
  • 46:04allelic loss and a a
  • 46:06way to look at,
  • 46:09a global Oh, at least
  • 46:11in this kind of data.
  • 46:12Imagine that there are a
  • 46:13lot of genes which have
  • 46:13been imputed to be associated
  • 46:15with sensitivity to,
  • 46:17pop inhibitors. They're core HR
  • 46:19genes, which clearly are probably
  • 46:20in the process. BRCA one
  • 46:22is partner BARD one, BRCA
  • 46:23two is partner PALB two,
  • 46:25and the RAFF two on
  • 46:26paralogs. These are core. Lose
  • 46:28any of these genes as
  • 46:29lethal. It's s these are
  • 46:30all s phase dependent. These
  • 46:31are all the same biology.
  • 46:32There are others which are
  • 46:33kinda HR ish,
  • 46:35I say. You know, ATR
  • 46:37kind of is Birch and
  • 46:38HR and CHK one, the
  • 46:40zemia genes, which really involve
  • 46:41more in interest rate and
  • 46:42cross link repair than really,
  • 46:44homology media repair. BRIP one,
  • 46:46which is kind of funny
  • 46:46in of itself, CDK twelve,
  • 46:48possibly,
  • 46:49in here. Then there are
  • 46:50others which really have no
  • 46:51no role in HR, but
  • 46:52are, you know, DNA repair
  • 46:53genes itself like ATM, CHEC
  • 46:55two,
  • 46:56MBN, and others which have
  • 46:58been imputed but not clear
  • 46:59that they're in the in
  • 47:00this pathway.
  • 47:01And so what we could
  • 47:02do is we actually looked
  • 47:04at,
  • 47:05together with, with colleagues of
  • 47:07Foundation Medicine and a bunch
  • 47:08of colleagues in in Europe.
  • 47:10We did a pan cancer
  • 47:11analysis using the tools that
  • 47:13we developed,
  • 47:14and asked, you know,
  • 47:16when
  • 47:17when
  • 47:18is by biolig loss of
  • 47:20which genes is associated with
  • 47:21evidence of genomic instability?
  • 47:23Alright? And we found that
  • 47:25there was incredibly strong associates
  • 47:26in the biolytic status and
  • 47:28evidence of global o h,
  • 47:29interestingly,
  • 47:30in BRCA one, BRCA two,
  • 47:32RAF fifty one, PAL b
  • 47:33two, and RAF fifty one
  • 47:34c, the core HR genes.
  • 47:36Alright?
  • 47:37There was weak associations from
  • 47:39other genes, and some of
  • 47:40the weak associations like in
  • 47:41RAF fifty one b,
  • 47:43and, HR is that the
  • 47:44numbers were low, so the
  • 47:45error bars are high.
  • 47:47Alright?
  • 47:48And there was no association
  • 47:50with things like ATM,
  • 47:52BAP1, check two. And here
  • 47:53we have lots of good
  • 47:53numbers.
  • 47:54Alright? So even in bialylic
  • 47:56ATM loss does not cause
  • 47:57the kind of genomic stability
  • 47:59pattern that's associated with BRCA
  • 48:00on BRCA two. And so
  • 48:01we predict that ATM, as
  • 48:02we well know, is not
  • 48:03an HRG. ATM mutants shouldn't
  • 48:05be sensitive to the department
  • 48:06editors. Alright? I'll I'll understand.
  • 48:10And in fact, if you
  • 48:11look across
  • 48:12the,
  • 48:13the genome, you know, kinda
  • 48:14look at, you know, where
  • 48:15are the biologics, what kind
  • 48:16of cancers have them. In
  • 48:17addition to ovary, prostate, breast,
  • 48:19and pancreas, where, again, prostate
  • 48:20is driven mostly by BRCA
  • 48:22two, alright, Not BRCA
  • 48:25one.
  • 48:26There is a low level
  • 48:27of bioelectrical alteration of variety
  • 48:29of the core HR genes
  • 48:30in other cancers.
  • 48:31Small percentages, but these may
  • 48:33be appropriate to think about
  • 48:35those are the ones that
  • 48:35need to target with, the
  • 48:37HR pathway.
  • 48:38Alright?
  • 48:40And then I will
  • 48:41end with you know? So
  • 48:42imagine that, you know, HR
  • 48:44is present with all disease
  • 48:45exam, more common BRCA one
  • 48:46syndromes, but they are present
  • 48:47at low level and other
  • 48:48other types. So imagine that
  • 48:49we really understand the allelic
  • 48:51status of BRCA1 and BRCA2,
  • 48:53all these other core HR
  • 48:54repair genes to really understand
  • 48:55when they're potentially actual,
  • 48:57alright, and to understand the
  • 48:58biology.
  • 48:59Alright?
  • 49:01Last thing I'll do is
  • 49:02this really important because, you
  • 49:03know, recently, there was a
  • 49:05yeah. Not not so recently
  • 49:06anymore, but, you know, there
  • 49:07was this, you know, large
  • 49:09approval for PARP inhibitors for
  • 49:11a set of fifteen
  • 49:12genes involved in DNA repair
  • 49:14based on, the study. And,
  • 49:16yeah, I'll just take you
  • 49:16through it just to understand.
  • 49:17It's kinda wild that this
  • 49:18was done. Alright?
  • 49:20So this is a labra
  • 49:21for metastatic castration matrix americans,
  • 49:23and they analyzed two cohorts.
  • 49:24Cohort one was BRCA one
  • 49:26sorry. It's just BRCA two
  • 49:27and then ATM. These are
  • 49:28grouped into was one cohort.
  • 49:30And you can see that
  • 49:31those three of the labra
  • 49:32did better than standard care.
  • 49:33Care. Then there was a
  • 49:34combined cohort a and b,
  • 49:35which is BRCA one, BRCA
  • 49:37two ATM, and then twelve
  • 49:38other genes. And this is
  • 49:39the,
  • 49:40the result of this combined
  • 49:41cohort. And based on this,
  • 49:42FDA approved elaborate for castaways
  • 49:44and prostate cancer with mutation
  • 49:46any one of fifteen, quote,
  • 49:47unquote, HR genes.
  • 49:50Alright? Alright. This is the
  • 49:51current FDA approval. If you
  • 49:53look at the data, alright,
  • 49:54this
  • 49:55cohort a and cohort b,
  • 49:57there's no effect in cohort
  • 49:58b. Just twelve other genes.
  • 50:00Alright? And this is this
  • 50:01is just pulled from the
  • 50:02supplemental data. Alright? That's there.
  • 50:04This is cohort a, BRCA
  • 50:06one two plus ATM.
  • 50:07Let's look at that. This
  • 50:08is BRCA one two versus
  • 50:10ATM.
  • 50:12Alright. No effect in ATM.
  • 50:14It's a BRCA and BRCA
  • 50:15two. And in fact, if
  • 50:16you look what's here, it's
  • 50:17all BRCA two.
  • 50:19Alright.
  • 50:21There's very few BRCA one
  • 50:23in there. Alright. So the
  • 50:24entire cohort was driven by
  • 50:25the BRCA two mutant population
  • 50:27causing the effect. Alright?
  • 50:29And so it's really important
  • 50:30to understand this. So if
  • 50:31you actually this is the
  • 50:32relative increase in survival. You
  • 50:34can see the only signals
  • 50:35seen in BRCA two, RAFT,
  • 50:36EVO one b, and fifty
  • 50:37four d. It might be
  • 50:37a little thing for CDK
  • 50:38twelve, but the errors are
  • 50:40error bars are overlapped. And
  • 50:41BRCA one, of course, there's
  • 50:43no effect.
  • 50:44And you guys now know
  • 50:45why because even when there's
  • 50:46a BRCA one mutation in
  • 50:47prostate cancer, most of the
  • 50:48time, there's no LOH.
  • 50:50So it's a bystander.
  • 50:51Alright?
  • 50:52There's a subset that may
  • 50:53respond, but not all. Just
  • 50:54better.
  • 50:55Alright? And so with that,
  • 50:57you imagine that sequencing data
  • 50:58is a lot more information
  • 50:58than just a list of
  • 50:59mutations. You have to really
  • 51:00make a model to figure
  • 51:01out what's going on.
  • 51:02Alright?
  • 51:03And that they're,
  • 51:05understanding true impurity and the
  • 51:06variant allele frequency. You can
  • 51:07build models that sit and
  • 51:08go, what's going on in
  • 51:09the tumor? What's going on
  • 51:10in the stroma? What's going
  • 51:11on, in the allelic state?
  • 51:13So especially for tumor suppressors.
  • 51:14Alright?
  • 51:15Oncogenes is reasonable. There it's
  • 51:17there. But tumor suppressor, many
  • 51:19of them, you know, the
  • 51:20most don't have as a
  • 51:21clear haplo infusion, phenotype, which
  • 51:23we would talk about. You
  • 51:24need to understand the locus
  • 51:25test. And not all DNA
  • 51:26repair genes are the same.
  • 51:27You can't lump an ATM
  • 51:28with BRCA one, BRCA two.
  • 51:30Alright?
  • 51:32ATM mutants will respond to,
  • 51:34like, for example, ATR inhibitors.
  • 51:35They're synthetic lethal with a
  • 51:36variety of things, but different
  • 51:37than what the BRCA and
  • 51:38BRCA two mutants will will
  • 51:39respond to. Right?
  • 51:41And reversion events, I didn't
  • 51:42go into subclonal events, even
  • 51:44clonal hematopoiesis can be picked
  • 51:45up by this kind of
  • 51:46analysis. So we can pick
  • 51:46up what genes but things
  • 51:48that fall off the model,
  • 51:48we can pick up clonal
  • 51:49hematopoiesis in solid tumors and
  • 51:51other things which we published.
  • 51:53And other things to leave
  • 51:55you about, which I wanna
  • 51:55is classical chemotherapy really may
  • 51:57act as targeted therapy aimed
  • 51:58at underlying defects and repair.
  • 52:00And so we really need
  • 52:00to think about how we
  • 52:01approach chemotherapy.
  • 52:02Alright? We could I think
  • 52:04PARP inhibitors is nothing but
  • 52:05chemotherapy, but given as an
  • 52:06oral agent,
  • 52:08side effects of PARP inhibitors
  • 52:10are nausea, vomiting, hair loss,
  • 52:11and neutropenia,
  • 52:12you know, just like etoposide.
  • 52:14Alright?
  • 52:16And I think, you know,
  • 52:17we need but we need
  • 52:17to rethink how we, how
  • 52:19we dose that if we
  • 52:19really understand the DNA repair
  • 52:21effects. Alright? With that, I
  • 52:23will, stop. I'm happy to
  • 52:24take questions.
  • 52:28Are these some funding sources?
  • 52:30While people ask questions, I
  • 52:32will
  • 52:33formulate questions. I'll ask you
  • 52:34two quick somewhat related questions.
  • 52:37Okay.
  • 52:37First,
  • 52:39are we sure that mono
  • 52:41monorhemic
  • 52:42loss does not increase risk
  • 52:44at all? And
  • 52:46I'll give you the second
  • 52:47one too, which is when
  • 52:48we pick up somatic
  • 52:51BRCA one and two mutations,
  • 52:52are they always biolabel?
  • 52:55Yeah. So, so when we
  • 52:57I think the germline you
  • 52:58know, there's a lot of
  • 52:58thing. Hey. This, you know,
  • 53:01epigenetic silencing of BRCA one,
  • 53:03etcetera. That may play a
  • 53:04role in a very small
  • 53:04subset,
  • 53:05of cancer. So, you know,
  • 53:06obviously, the LH events will
  • 53:07pick up if there's an
  • 53:08inversion or there's epigenetic silencing,
  • 53:09it won't pick that up.
  • 53:10It look like there's no
  • 53:11LH. But interestingly, in ovarian
  • 53:12cancer,
  • 53:13alright,
  • 53:14we don't need to that.
  • 53:15That explains ninety five percent
  • 53:16of the events we can
  • 53:17see by genomic. We don't
  • 53:18have to pick another explanation
  • 53:19for that. That makes me
  • 53:20think that is the main
  • 53:21mechanism of LH. Interestingly, the
  • 53:23few that have undergone like,
  • 53:25in BRCA1 carriers,
  • 53:27there's never epigenetic silencing of
  • 53:28the other allele as a
  • 53:29mechanism
  • 53:30of of LOH.
  • 53:31There's always deletion, which is
  • 53:33interesting. There are patients who
  • 53:34have upfront dilute epigenetic silencing
  • 53:36in which they undergo LOH
  • 53:37by deletion of the of
  • 53:38the other allele, alright, which
  • 53:39I haven't talked about. So
  • 53:40if you have I think
  • 53:41you either have germline mutation,
  • 53:43somatic mutation, or epigenetic silencing
  • 53:45first event. The second event
  • 53:46is always deletion, which is
  • 53:47intriguing.
  • 53:48That suggests there's some sort
  • 53:49of unstable area in that
  • 53:50locus,
  • 53:51that's there. Somatic alterations. I
  • 53:54think most somatic alterations, VRCA
  • 53:55or VRCA two, outside of
  • 53:58ovarian high grade serous ovarian
  • 53:59cancer are mostly somatic
  • 54:01are are are mostly I'm
  • 54:02sorry. Are mostly,
  • 54:03a monolipid.
  • 54:05And yet in breast cancer,
  • 54:07they respond those patients on
  • 54:08metastatic breast gets respond to
  • 54:10part of the No. No.
  • 54:11So so in sorry. Breast
  • 54:12and ovary Right. And okay.
  • 54:14Breast and ovary. That's right.
  • 54:15Ovary is hundred percent. And
  • 54:16breast two, you can saw
  • 54:17we saw that even when
  • 54:18you have somatic alteration. So
  • 54:19breast, ovary, prostate for BRCA
  • 54:21two,
  • 54:22pancreas,
  • 54:23the there is some sort
  • 54:24of selection for, bilevel. So
  • 54:26we can see bilevel alterations
  • 54:27absolutely in the somatic setting.
  • 54:29That's there.
  • 54:30Questions?
  • 54:31So, for guacamutin
  • 54:34patients that are treated with
  • 54:35platinum invasive therapy, often do
  • 54:38you see,
  • 54:39reversing event? Oh, that's the
  • 54:41most common mechanism to resist
  • 54:42this. It's what it's when
  • 54:43we look for them. Often,
  • 54:44we don't look.
  • 54:45Alright?
  • 54:47Alright. When you look, it's
  • 54:48about fifty percent of the
  • 54:49time.
  • 54:50Alright? And the other ones
  • 54:51we miss is probably because
  • 54:52we're missing new versions or
  • 54:53other mechanisms.
  • 54:56Alright? And, I'll say I'll
  • 54:58I'll say it somewhere. I
  • 54:59don't believe in. I think
  • 55:00there's BRCA one and BRCA
  • 55:01two. They're very different genes
  • 55:02with very different underlying things.
  • 55:04We shouldn't my thing is
  • 55:05we shouldn't con we shouldn't
  • 55:06conflate them together. They just
  • 55:07happen to be interesting in
  • 55:08the same way.
  • 55:11You know?
  • 55:13That's
  • 55:14my personal lives.
  • 55:21Do you have
  • 55:22a preference of sequencing then?
  • 55:25What? In? When you have
  • 55:26a broken mutation, how do
  • 55:27you sequence them with what
  • 55:28the perception part inhibitors in
  • 55:30other eight? Yeah. I think,
  • 55:31you know, it's hard. Right
  • 55:32now, our standard are NCCN
  • 55:35guidelines.
  • 55:37Well, we treat them the
  • 55:38same as anyone else with
  • 55:40the addition of, of olaparib
  • 55:41in the in the in
  • 55:43the early stage set. Right?
  • 55:45In the advanced setting, we
  • 55:46have no ideas. Alright?
  • 55:48Because they respond well to
  • 55:49a variety of agents,
  • 55:51because I think they're all
  • 55:52hitting the same possible, pathway.
  • 55:54I think the PARP inhibitors,
  • 55:55especially,
  • 55:56are are well tolerated. It's
  • 55:58and some women are are
  • 56:00well tolerated in oral dosing.
  • 56:02Some PI patients who prefer
  • 56:04platinum to
  • 56:05to all you know, couldn't
  • 56:06tolerate all elaborate. You know?
  • 56:08And I give them either
  • 56:09oral or liposide to load
  • 56:10this platinum and respond.
  • 56:11You know? But I think
  • 56:12it's very complicated, and I
  • 56:14think in some patients, there's
  • 56:15audio version even by the
  • 56:16time you see them, you
  • 56:17know, depending on when
  • 56:20I
  • 56:21see it. And I think
  • 56:22in some ways, there are
  • 56:24many cancers in which I
  • 56:25think a repair effect
  • 56:26occurred early in the form
  • 56:27of tumorigenesis
  • 56:28and then reverts
  • 56:30by the time you get
  • 56:30a well established cancer. BRCA
  • 56:32was an unusual setting where
  • 56:33we we don't see we
  • 56:34see that history occurring in
  • 56:35front of us through selection.
  • 56:37You know?
  • 56:38But it's there. But I
  • 56:39think, you know, we have
  • 56:40to, another thing we don't
  • 56:41understand is what the what's
  • 56:42the commutation profile? What are
  • 56:43the rearrangements that are present
  • 56:45that are driving the biology?
  • 56:47You know? And that's something
  • 56:48we know nothing about.
  • 56:53Yeah. I mean, it's just.
  • 56:54I just have a question
  • 56:56following up on what you
  • 56:57just said,
  • 56:58which I noticed you had
  • 56:59a lot of I mean,
  • 57:01really, they're great.
  • 57:02You
  • 57:03know, you're talking about mechanisms
  • 57:05where they have a select
  • 57:07it has a selective impact
  • 57:08on this. It has a,
  • 57:11an effect on the pharmacogenic,
  • 57:14process of the cell that
  • 57:15you the cell
  • 57:16because
  • 57:18But another thing that these
  • 57:20DNA repair mutation do is
  • 57:21just talk mutations. Right? So
  • 57:23this is a completely separate
  • 57:25kind of phenomenon that you
  • 57:26haven't mentioned it. So I'm
  • 57:27just wondering what context you
  • 57:29might put for separating out
  • 57:30the new cache out of
  • 57:31sex. So for instance, early
  • 57:33on in the generation of
  • 57:34cancer, maybe they're just playing
  • 57:35a role in creating a
  • 57:36suite of mutations that then
  • 57:38can be able to care.
  • 57:39Absolutely. I think I think
  • 57:40it's clear that BRCA1 loss
  • 57:42is required for the initiating
  • 57:44event of tumorigenesis, which is
  • 57:45probably the two populations.
  • 57:47And the big question to
  • 57:48new was I call it
  • 57:49the missing drivers. What the
  • 57:51hell is causing the growth
  • 57:52for
  • 57:52a BRCA1 or BRCA2 mutations?
  • 57:55Okay?
  • 57:56You know, what is actually
  • 57:57causing,
  • 57:59growth? Because you sequence them,
  • 58:00you're showing sequence, and you
  • 58:00get fifty three and up
  • 58:01in house, and then, quote,
  • 58:02unquote, copy to go all
  • 58:03places. You know? Ten percent
  • 58:05of the genome. Whatever gene
  • 58:06whatever gene you find will
  • 58:07be, quote, unquote, amplified in
  • 58:08ten percent of high rates
  • 58:09of.
  • 58:10Alright?
  • 58:13And so, sure, there's lots
  • 58:14of genome gaps. But underlying
  • 58:15them, there's probably real drivers.
  • 58:17And we actually have a
  • 58:17large effort trying to find
  • 58:19this, and what we see
  • 58:20is there's not lots of
  • 58:21infraim regions. There's this entire
  • 58:22story we did with about
  • 58:24out of frame reagents,
  • 58:25the tiresome kinases
  • 58:27that are present that are
  • 58:28actually drivers.
  • 58:29Alright?
  • 58:30And I think that's what's
  • 58:31gonna happen. It's gonna be
  • 58:32a classical drivers,
  • 58:33but altered in ways that
  • 58:34we don't that we don't
  • 58:35currently understand this DNA driving
  • 58:37multiples,
  • 58:39by the mechanism mutagenesis.
  • 58:41Alright? Because the mutagen mechanism
  • 58:43in BRCM and BRCM mutant
  • 58:44cancers is not.
  • 58:46It's genomic rearrangements and deletions.
  • 58:49And we're able to prepare
  • 58:50to actually interrogate that without
  • 58:52currency.
  • 58:55Right? I think hidden there
  • 58:56is the bias. This is
  • 58:57my hope. Alright? And I
  • 58:59think that these will be,
  • 59:00like, non small cell of
  • 59:01cancer. Like, two percent of
  • 59:02this, four percent of that,
  • 59:03five percent of this, it
  • 59:04gets you caused mutagenesis
  • 59:06and then selection,
  • 59:08okay, or some driver events
  • 59:10that are if we believe
  • 59:11one.
  • 59:16Thanks for a great talk.
  • 59:18Thanks for being here. I
  • 59:19believe.