Skip to Main Content
All Podcasts

Melanoma Awareness Month 2020

Transcript

  • 00:00 --> 00:03Support for Yale Cancer Answers comes from AstraZeneca,
  • 00:03 --> 00:14the Beyond Pink campaign aims to empower metastatic breast cancer patients and their loved ones to learn more about their diagnosis and make informed decisions.
  • 00:14 --> 00:17Learn more at lifebeyondpink.com.
  • 00:17 --> 00:20Welcome to Yale Cancer Answers with your host
  • 00:20 --> 00:21Doctor Anees Chagpar.
  • 00:21 --> 00:31Yale Cancer Answers features the latest information on cancer care by welcoming oncologists and specialists who are on the forefront of the battle to fight cancer. This week
  • 00:31 --> 00:36it's a conversation about Melanoma research with doctor Jeffrey Ishizuka.
  • 00:36 --> 00:41Doctor Ishizuka is an Assistant Professor of Medical Oncology at Yale School of Medicine,
  • 00:41 --> 00:45where Doctor Chagpar is a Professor of Surgical Oncology.
  • 00:45 --> 00:51Jeff, maybe we could start off by you telling us a little bit about yourself and what you do.
  • 00:51 --> 00:59I'm a physician-scientist and that means that I spend part of my time treating cancer patients and part of my time in the lab looking
  • 00:59 --> 01:02for new treatments for those patients.
  • 01:02 --> 01:09And so tell us a little bit more about the kinds of patients that you treat and the kinds of research that you do.
  • 01:09 --> 01:12I see Melanoma patients and I'm an immunologist
  • 01:12 --> 01:15by training. And that means I study ways to
  • 01:15 --> 01:19make the patient's immune system work better,
  • 01:19 --> 01:22to attack the cancer.
  • 01:22 --> 01:26Tell us more about that, we've talked on the show a little bit about immunotherapy and so on,
  • 01:26 --> 01:31but tell us a little bit more about the broad spectrum of immunotherapy.
  • 01:31 --> 01:35How exactly does it work and then the role that it plays in Melanoma.
  • 01:35 --> 01:38There are a couple of types of immunotherapy.
  • 01:38 --> 01:43And for a long time we knew that the immune system had the potential to control cancer,
  • 01:43 --> 01:46but I'd say the two big advances that are more recent are on the one hand,
  • 01:46 --> 01:54CAR T cells. And those are cells that are taken out of the patients body and reprogrammed to go back in and attack the tumor, and immune checkpoint
  • 01:54 --> 01:58blockade. And these are drugs that cut the brakes on the immune system.
  • 01:58 --> 02:02Those brakes stop the immune system from attacking the cancer.
  • 02:02 --> 02:03And when you get rid of them,
  • 02:03 --> 02:08the cancer is vulnerable to immune attack.
  • 02:08 --> 02:12Tell us about which of these you work on, and
  • 02:12 --> 02:15how exactly they work in Melanoma.
  • 02:15 --> 02:22In Melanoma, immune checkpoint blockade has really been one of the biggest developments
  • 02:22 --> 02:26really in the last, well maybe ever in the disease.
  • 02:26 --> 02:31And I think Melanoma was the first disease where
  • 02:31 --> 02:37these drugs were developed and remains one of the ones where they work the best.
  • 02:38 --> 02:41Can you talk about this immune checkpoint blockade.
  • 02:41 --> 02:45Is there more than one molecule that needs to be blocked?
  • 02:45 --> 02:49How does that work? Why does the immune system have brakes to begin with?
  • 02:49 --> 02:54These are great questions and they're really at the forefront of the field right now,
  • 02:54 --> 02:58so there are certainly at least a few molecules that are important.
  • 02:58 --> 03:02And all of the ones we learned about first are on the surface of T cells,
  • 03:02 --> 03:07which we know are one of the important cells for controlling cancer.
  • 03:07 --> 03:09There are a number of molecules that target
  • 03:09 --> 03:13PD-L1 and that's a major inhibitory pathway in the T cells,
  • 03:13 --> 03:15and also molecules that target CTL A4,
  • 03:15 --> 03:18which is another inhibitory pathway in T cells.
  • 03:18 --> 03:25And what we learned is when you block either one of these and sometimes if you block them both together it works even better.
  • 03:25 --> 03:28The T cells can get supercharged to attack the cancer.
  • 03:28 --> 03:30Why is it that
  • 03:30 --> 03:32they have breaks to begin with?
  • 03:32 --> 03:38The immune system is supposed to be able to identify foreign stuff in our bodies and get rid of it.
  • 03:38 --> 03:41So why doesn't that work in cancer?
  • 03:41 --> 03:45Why is it that we need to take off these brakes?
  • 03:45 --> 03:47Why do they have breaks to begin with?
  • 03:47 --> 03:51This is the foundational question of immunology,
  • 03:51 --> 03:54the distinction between self and nonself.
  • 03:54 --> 03:59All cells need to be able to get rid of foreign things just as you said,
  • 03:59 --> 04:00but at the same time
  • 04:00 --> 04:09they need to have mechanisms to avoid attacking the normal cells in the body that are healthy and so
  • 04:09 --> 04:15why is it that the immune system thinks that these cancer cells are normal?
  • 04:15 --> 04:26I think it's really because cancer cells arise from normal cells as normal cells become dysregulated as they acquire genetic errors called mutations.
  • 04:26 --> 04:29And eventually you develop cancer.
  • 04:29 --> 04:33And because the cancer cell arises from a backdrop of normal cells,
  • 04:33 --> 04:40doing normal cell things, the immune system has to find specific signals of damage or mutations that
  • 04:40 --> 04:43look abnormal in order to recognize cancer.
  • 04:43 --> 04:46So you're telling me that normally it won't do that?
  • 04:48 --> 04:54Some tumors are recognized by the immune system and the immune system can actually get rid of them,
  • 04:54 --> 05:02and other aren't. And really what we're trying to understand and at the heart of the field is how can we take tumors that are not well recognized by
  • 05:02 --> 05:07the immune system and turn them into tumors that the immune system can see and destroy?
  • 05:07 --> 05:10And so it seems to me that if you take that problem just at its face,
  • 05:10 --> 05:13there are two ways of doing that.
  • 05:13 --> 05:18One is to make the tumor look more abnormal so that the immune system
  • 05:18 --> 05:27realizes, I need to attack it and get rid of it without actually revving up the immune system or getting rid of the
  • 05:27 --> 05:36brakes and the other is to supercharge the immune system as you put it to make it more sensitive to recognizing what might be abnormal.
  • 05:36 --> 05:41Yeah, that's right, and I think people are working at both sides of that problem.
  • 05:41 --> 05:49We and others in the lab, are thinking of strategies both to make tumors put out signs for the immune system, saying,
  • 05:49 --> 05:55come get me and also looking for new ways to charge the immune system to be more aggressive against cancer.
  • 05:55 --> 05:57Tell us more about the first,
  • 05:57 --> 06:04because I think that we've heard a little bit about checkpoint inhibitors,
  • 06:04 --> 06:11but we really haven't heard a lot about the work that's going on to have tumor cells put out those signs that
  • 06:11 --> 06:22say, come get me. And it seems to me that might be a way to allow the immune system without getting supercharged to eat up or
  • 06:22 --> 06:24get rid of these cancer cells,
  • 06:24 --> 06:26because one of the problems,
  • 06:26 --> 06:35as you point out, of having a supercharged immune system is that it can then attack its own cells.
  • 06:35 --> 06:37Yeah, that's a great point and
  • 06:37 --> 06:42we've been thinking, and others as well,
  • 06:42 --> 06:49that it comes down to tricking the tumor cell into making inflammatory signals,
  • 06:49 --> 06:56tricking it into making kind of an antiviral response that recruits anti tumor immune cells into the micro environment,
  • 06:56 --> 06:57and I think
  • 06:57 --> 07:06you can go about that by infecting the tumor with a virus or making it think it's infected with a virus or triggering certain danger signals in the micro environment
  • 07:06 --> 07:10directly around the tumor.
  • 07:10 --> 07:15Tell us more about that work, is that actually something that's being done?
  • 07:15 --> 07:19Is it in clinical practice?
  • 07:19 --> 07:20How do we do that?
  • 07:20 --> 07:29There are a number of clinical trials now using stimulators of viral pathways that look like DNA or RNA,
  • 07:29 --> 07:36things that viruses make and that ourselves have dedicated sensors in order to detect,
  • 07:36 --> 07:42and I think none of them has proven to be the Magic bullet for cancer yet.
  • 07:42 --> 07:46But there are still some technical hurdles to workout.
  • 07:46 --> 07:49And I think we're getting there though.
  • 07:49 --> 07:56Why has that not proven to be as successful as supercharging the immune system?
  • 07:56 --> 08:03I think one of the challenges is that cancer in many cases can spread to many locations,
  • 08:03 --> 08:09and when you think about triggering an inflammatory response in the tumor bed,
  • 08:09 --> 08:12you're really thinking about triggering it,
  • 08:12 --> 08:14not just at one site,
  • 08:14 --> 08:16but at many sites all at once,
  • 08:16 --> 08:18and so finding ways to send drugs
  • 08:18 --> 08:26to all of the different sites that cancer occupies in the body is one of the major challenges to getting this approach to work.
  • 08:26 --> 08:33The other approach then is the one that is the mainstay of immunotherapy,
  • 08:33 --> 08:38which is to quote supercharge the immune system to get rid of the blocks.
  • 08:38 --> 08:43I always think of it like Harry Potters invisibility cloak,
  • 08:43 --> 08:48right. The tumor has kind of made itself invisible to the immune system,
  • 08:48 --> 08:57and it's getting rid of that that cloak and getting the immune system to recognize it and to go after it and two,
  • 08:57 --> 08:59to be quote supercharged now.
  • 08:59 --> 09:09You mentioned two molecules, in particular CTL A4 and PDL1, tell us a little bit about the differences between the two.
  • 09:09 --> 09:14I mean we have drugs that will block either pathway.
  • 09:14 --> 09:17How do you figure out which one to use?
  • 09:17 --> 09:20Tell us more about that interplay.
  • 09:20 --> 09:29I think we still don't fully understand the mechanism of either drug and either pathway.
  • 09:29 --> 09:32And people have done a lot of good work,
  • 09:32 --> 09:36in fact, the Nobel Prize was awarded a few years back for some of that work,
  • 09:36 --> 09:39but I wouldn't say that we completely understand which,
  • 09:39 --> 09:41even sometimes which cells are being targeted,
  • 09:41 --> 09:44but certainly which pathways within the cell are being activated.
  • 09:44 --> 09:47So a lot of how we figured this out has been empirically.
  • 09:47 --> 09:52We've done clinical trials with different drugs or different combinations of drugs,
  • 09:52 --> 09:55and we've seen what's been effective for patients,
  • 09:55 --> 09:59and the hope is going forward that as we learn more about the immune system,
  • 09:59 --> 10:04and as we learn more about the tumor that we will be able to do better
  • 10:04 --> 10:09and even predict the next set of these drugs that could be usefully combined.
  • 10:09 --> 10:14So tell us more about the differences between CTLA for an PDL1.
  • 10:14 --> 10:24I get the fact that we've discovered these kind of fortuitously and empirically and have just made drugs that affect each of these pathways,
  • 10:24 --> 10:26and seeing that they work.
  • 10:26 --> 10:29But we must know more about these actual molecules.
  • 10:29 --> 10:33Yeah, they both play an inhibitory role in T cells.
  • 10:33 --> 10:37I think it's broadly thought
  • 10:37 --> 10:42that one of them plays more of a role in T cells initially getting primed against the tumor,
  • 10:42 --> 10:52but maybe plays more of a role in lymph nodes then generating the T cells that are capable of responding whereas the other one,
  • 10:52 --> 11:01that speedy one may play more of a role in activating the T cells that are already primed against the tumor that already have the capacity to attack the tumor.
  • 11:01 --> 11:08And I'm going to steer clear of the term of exhaustion because there are a lot of debates about whether T cells
  • 11:08 --> 11:10are actually exhausted or not,
  • 11:10 --> 11:20but there's this idea that T cells can, after seeing a lot of tumor antigen stop responding very well that they can become dysfunctional and so one
  • 11:20 --> 11:23of the things that PDL1 blockade does,
  • 11:27 --> 11:31is to make the T cells that have become dysfunctional more functional.
  • 11:31 --> 11:35And so if these two pathways then are complementary,
  • 11:35 --> 11:42one being more so for priming T cells and one being more so for T cells that are already primed,
  • 11:42 --> 11:53has there been any work looking into either concurrent therapy or sequential therapy of different immuno therapies that might work better than either in isolation?
  • 11:53 --> 11:56There has, and in Melanoma combining two drugs,
  • 11:56 --> 11:58one that targets CTL A4.
  • 11:58 --> 12:07and one that targets PDL1 seems to be better than using either drug alone and potentially better than using them both in sequence,
  • 12:07 --> 12:17although the latter is a less clear conclusion.
  • 12:17 --> 12:27And one of the exciting things in this field has been seeing the slew of approvals for immuno therapies in different cancer types in Melanoma.
  • 12:27 --> 12:39Certainly it's become standard of care in the frontline for most patients and it's being explored in basically every stage of care of the disease other than for disease that
  • 12:39 --> 12:45can just be removed and surgically cut out in the early stages and really beyond Melanoma,
  • 12:45 --> 12:53it spread throughout many many solid tumor types and it's being tried in almost any tumor type you can think of.
  • 12:53 --> 13:00And so two questions. First question is one of the things you mentioned earlier as being one of the
  • 13:00 --> 13:08downfalls of some therapies is that it can't always get to all of the cells where the tumors may be hiding.
  • 13:08 --> 13:16Does immunotherapy have that problem in terms of getting to the T cells and activating them or supercharging them?
  • 13:16 --> 13:21Or is that concept, this may not work if there's a tumor,
  • 13:21 --> 13:23for example in the brain?
  • 13:23 --> 13:27Because this drug can't cross the blood brain barrier?
  • 13:27 --> 13:31Or does it affect T cells wherever they are?
  • 13:31 --> 13:38We know that we can get effects certainly in the brain.
  • 13:38 --> 13:51So you can see effects of these drugs in what are thought of usually as sites of the body that are hard to get to or immune privilege sites but
  • 13:51 --> 13:53I guess what I don't know for sure,
  • 13:53 --> 13:58it's hard to say is whether there is a problem activating immune cells somewhere in the body.
  • 13:58 --> 14:07That is to say, whether we're getting these drugs as effectively as possible to all the immune cells that might be able to be mobilize against the tumor.
  • 14:07 --> 14:14We're going to learn a lot more about Melanoma immunotherapy right after we take a short break for a medical minute.
  • 14:14 --> 14:22Please stay tuned to learn more about this research with my guest doctor Jeffrey Ishizuka. Support for Yale Cancer Answers comes from AstraZeneca.
  • 14:22 --> 14:28Providing important treatment options for patients with different types of lung,
  • 14:28 --> 14:31bladder, ovarian, breast, and blood cancers.
  • 14:31 --> 14:35More information at astrazeneca-us.com.
  • 14:35 --> 14:38This is a medical minute about breast cancer,
  • 14:38 --> 14:41the most common cancer in women. In Connecticut alone,
  • 14:41 --> 14:46approximately 3000 women will be diagnosed with breast cancer this year,
  • 14:46 --> 14:48but thanks to earlier detection,
  • 14:48 --> 14:51noninvasive treatments, an novel therapies,
  • 14:51 --> 14:56there are more options for patients to fight breast cancer than ever before.
  • 14:56 --> 15:04Women should schedule a baseline mammogram beginning at age 40 or earlier if they have risk factors associated with breast cancer.
  • 15:04 --> 15:10Digital breast tomosynthesis or 3D mammography is transforming breast screening by significantly
  • 15:10 --> 15:17reducing unnecessary procedures while picking up more cancers and eliminating some of the fear and anxiety
  • 15:17 --> 15:22many women experience. More information is available at yalecancercenter.org.
  • 15:22 --> 15:26You're listening to Connecticut public radio.
  • 15:26 --> 15:29Welcome back to Yale Cancer Answers.
  • 15:29 --> 15:35This is doctor Anees Chagpar andnI'm joined tonight by my guest doctor Jeffrey Ishizuka.
  • 15:35 --> 15:41We're talking about Melanoma research and in particular we're talking about immunotherapy.
  • 15:41 --> 15:47Jeff, right before the break we were talking a little bit about immunotherapy in terms of,
  • 15:47 --> 15:51really getting the immune system to attack cancer cells,
  • 15:51 --> 15:55which it may not recognize because as you put it,
  • 15:55 --> 15:58these cancer cells come from normal cells and that
  • 15:58 --> 16:07may not be as foreign looking to the immune system to really trigger it and we talked a little bit about two separate pathways,
  • 16:07 --> 16:11CTL A4 and PDL1 and the fact that we now have drugs,
  • 16:11 --> 16:20this explosion of drugs in immunotherapy targeting these two pathways and how this really has become the mainstay of therapy,
  • 16:20 --> 16:23particularly for cancers like Melanoma.
  • 16:23 --> 16:27I had a few questions to kind of follow up on that.
  • 16:27 --> 16:32The first is, tell us a little bit about the side effects.
  • 16:32 --> 16:35We think about
  • 16:35 --> 16:39chemotherapy, and you know, traditionally,
  • 16:39 --> 16:47chemotherapy was therapy that kills off cancer cells and was really thought to be therapy that switches off
  • 16:47 --> 16:55rapidly dividing cells and so people ended up losing hair and maybe getting sick because it effects your GI lining,
  • 16:55 --> 16:57which are rapidly turning over cells.
  • 16:57 --> 17:01Do you get the same kind of thing in immunotherapy,
  • 17:01 --> 17:05or are there other side effects that are the results of
  • 17:05 --> 17:11kind of supercharging this immune system and getting the immune system to attack healthy cells?
  • 17:11 --> 17:13So I think that's it exactly.
  • 17:13 --> 17:20Many of the side effects that you get from immunotherapy are actually side effects of supercharging the immune system,
  • 17:20 --> 17:24so the immune system can accidentally attack different areas of the body.
  • 17:24 --> 17:27Some of the things we see are inflammation in the lungs,
  • 17:27 --> 17:31inflammation in the GI system we see inflammation of the endocrine system,
  • 17:31 --> 17:34and when we first started seeing these side effects,
  • 17:34 --> 17:37there wasn't a good sense of how you treat them,
  • 17:37 --> 17:39how you manage them, or even how you monitor.
  • 17:39 --> 17:41We didn't really know what to look for.
  • 17:41 --> 17:45But I will say that as experience with these agents has progressed,
  • 17:45 --> 17:49we've gotten better at detecting these side effects as they occur,
  • 17:49 --> 17:56and managing them, usually using immunosuppressives and one of the questions that comes up when you start saying,
  • 17:56 --> 18:02well, you're using drugs to charge the immune system and at the same time to shut down the immune system,
  • 18:02 --> 18:05is that going to be is going to be bad for the patients.
  • 18:05 --> 18:11Are they going to have that outcomes and the data isn't really completely mature on this yet,
  • 18:11 --> 18:14but it certainly appears from the early data that
  • 18:14 --> 18:22you can safely give these immunosuppressives and that you don't at least don't clearly make their responses against the cancer
  • 18:22 --> 18:26worse.
  • 18:26 --> 18:32That's really interesting. Why would that be the case? I can imagine that when we think about people who are immunosuppressed,
  • 18:32 --> 18:37people who for example have HIV or other things that turn off their immune system,
  • 18:37 --> 18:40they are more at risk of developing cancer,
  • 18:40 --> 18:45and I guess for the same reason that you talked about before the break,
  • 18:45 --> 18:47which is your immune system,
  • 18:47 --> 18:55unbeknownst to you, might be getting rid of little cancers that you don't know you have because it recognizes them and it gets rid of them,
  • 18:55 --> 19:01and so if you are immuno compromised you're at increased risk of getting cancer,
  • 19:01 --> 19:04and that's the whole point of
  • 19:04 --> 19:10supercharging the immune system to get rid of these cancers.
  • 19:10 --> 19:21Why is it that giving people an immunosuppresant at the same time as an immuno supercharger doesn't seem to affect the cancer in a bad way?
  • 19:21 --> 19:25A couple of potential thoughts here.
  • 19:25 --> 19:32The first one is that I want to be careful we don't know for sure that it doesn't affect the
  • 19:32 --> 19:34response to therapy in a negative way.
  • 19:34 --> 19:37I think what we can say is that
  • 19:37 --> 19:44at first blush, patients who needed immunosuppressives because they had these bad immune effects and got them didn't do obviously worse,
  • 19:44 --> 19:46at least in the early studies
  • 19:46 --> 19:55then patients who didn't need them in the first place and that actually could be a kind of selection bias issue where the patients who needed the immunosuppressives
  • 19:55 --> 19:58actually were having the strongest immune responses to begin with,
  • 19:58 --> 20:08and so I think we have to do some careful experiments in a controlled setting to see whether it was really true that the immunosuppressives weren't having any effect there.
  • 20:08 --> 20:14And I think that's probably the main thing that I would think about for that issue.
  • 20:14 --> 20:20The other question that I have is, these autoimmune side effects,
  • 20:20 --> 20:26the side effects of people's immune system now attacking their own normal cells,
  • 20:26 --> 20:28are those permanent? Are
  • 20:28 --> 20:37they forever or are they short lived? I mean when you get chemotherapy and you lose your hair,
  • 20:37 --> 20:39your hair will grow back.
  • 20:39 --> 20:44Is it the same with immunotherapy that this is a short term thing?
  • 20:44 --> 20:47Or when your immune system attacks your lungs,
  • 20:47 --> 20:50now you've got pulmonary fibrosis forever?
  • 20:50 --> 20:55I think it depends on the type of immune side effect that we're talking about.
  • 20:55 --> 20:57I think many of them,
  • 20:57 --> 20:59if they're controlled with immunosuppressives,
  • 20:59 --> 21:04and if you take the patient off of the immunotherapy, will actually go away.
  • 21:04 --> 21:06So we see this in a lot of cases,
  • 21:06 --> 21:09inflammation in the colon, or inflammation in the lungs.
  • 21:09 --> 21:16I think the case in which this isn't necessarily true is when the immune system attacks the cell type
  • 21:16 --> 21:26that produces hormones in the body and destroys all of that cell type because in that case you may not really know what's going on until the cell type
  • 21:26 --> 21:30is gone, and after that there's really no bringing it back.
  • 21:30 --> 21:37So in most cases we actually have been able to give hormone replacement.
  • 21:37 --> 21:40It's extremely bad if it's not detected,
  • 21:40 --> 21:45but in a lot of cases it can be solved by giving a pill a day.
  • 21:45 --> 21:47When you talk about hormones,
  • 21:47 --> 21:54are you talking about thyroid are you talking about ovaries, what hormones are we talking about?
  • 21:54 --> 21:57Yeah, so thyroid is one that you certainly see,
  • 21:57 --> 22:05but you see actually a number of other hormones that are produced in the brain that can also be altered,
  • 22:05 --> 22:07and these can be be more rare,
  • 22:07 --> 22:10but can be pretty dramatic if you see them.
  • 22:10 --> 22:14So given the side effects of immunotherapy,
  • 22:14 --> 22:17is immunotherapy really better than classic chemotherapy?
  • 22:17 --> 22:24You had mentioned that immunotherapy has now become standard of care for Melanoma.
  • 22:24 --> 22:26Is it better than what we used to do?
  • 22:28 --> 22:31We used to give chemotherapy for Melanoma,
  • 22:31 --> 22:32right?
  • 22:32 --> 22:36And Melanoma is not particularly responsive to chemotherapy,
  • 22:36 --> 22:41and I think what excited everyone in the field and it's given us all
  • 22:41 --> 22:44a lot of excitement and a lot of hope is not even that
  • 22:44 --> 22:48everyone responds to these immunotherapy's because they don't,
  • 22:48 --> 22:52not enough patients do, and that's something we don't really understand.
  • 22:52 --> 22:54We're trying to understand it in the lab,
  • 22:54 --> 22:59but it's that some of the patients who respond seem to just keep responding,
  • 22:59 --> 23:07and some of them respond so well and for so long that we've actually started to believe that we can take the patients off of the drugs,
  • 23:07 --> 23:10the immunotherapy drugs, and that the cancer won't return.
  • 23:10 --> 23:15And this is true even in some cases for very aggressive disease.
  • 23:15 --> 23:19And so seeing those effects are the ones that have really made everybody excited.
  • 23:19 --> 23:26And you see that in clinical trials when we study how patients survive on different drugs and
  • 23:26 --> 23:30it was no contest between the immunotherapy's and chemotherapy.
  • 23:30 --> 23:44When we talk about therapy for cancer a lot of times we're talking about personalized medicine and we're talking about how we can figure out what a cancer likes to
  • 23:44 --> 23:48eat, what receptors cancer has,
  • 23:48 --> 23:51what genes are turned on and turned off,
  • 23:51 --> 23:54and then we target our therapy accordingly.
  • 23:54 --> 24:04Talk about immunotherapy. It seems to me like we're talking about a blanket turning on supercharging the immune system,
  • 24:04 --> 24:09is that right, or are there ways where we're actually tailoring this therapy?
  • 24:09 --> 24:20Are we looking at who those people are that are super responsive to immunotherapy versus the people who are not super responsive to immunotherapy?
  • 24:20 --> 24:25And which immunotherapy might work better in particular patients?
  • 24:25 --> 24:28Yeah, so this question is near and dear to my heart.
  • 24:28 --> 24:39We in general don't do a great job of selecting patients to get particular immunotherapy, there is one biomarker which is the expression of PDL1 in the tumor as
  • 24:39 --> 24:43we talked about PDL1 and PD1 is one of these key pathways,
  • 24:43 --> 24:51so if you have PDL1 expressed in the tumor microenvironment either by immune cells in the micro environment or by the tumor,
  • 24:51 --> 24:57we know that you are more likely to have a response to targeting the PD1 PDL1 axis.
  • 24:57 --> 25:07But basically everyone in the field spends a lot of time complaining about this biomarker because we know there are a lot of patients who will have PDL1 expression
  • 25:07 --> 25:10in their tumor who won't respond well to these drugs.
  • 25:10 --> 25:18And conversely, there are a lot of patients who won't have PDL1 expression in the tumor who will still respond to these drugs.
  • 25:18 --> 25:20So what's the point of the biomarker then?
  • 25:20 --> 25:28We know it's better than not using it in terms of you have some predictive value and in some cases you might not even be able to see
  • 25:28 --> 25:32a signal of the drug working in a patient population and unless,
  • 25:32 --> 25:37you used a biomarker, and also it's a stand in because we haven't done a good enough job yet of
  • 25:37 --> 25:40finding better ones.
  • 25:40 --> 25:44So we have this biomarker that if you have it,
  • 25:44 --> 25:49you won't necessarily respond to the immunotherapy, if you don't have it,
  • 25:49 --> 25:52you may still respond to the therapy,
  • 25:52 --> 25:57so either way you're likely going to get immunotherapy if you have Melanoma,
  • 25:57 --> 26:01regardless of whether you have the biomarker or not.
  • 26:01 --> 26:06That's true, and that's where I think we have the potential to do much better,
  • 26:06 --> 26:10particularly as we talked about these two pathways,
  • 26:10 --> 26:19there are a lot of other immuno regulatory pathways that can activate immune cells or can activate the tumor to recruit immune cells.
  • 26:19 --> 26:23And we're still at the beginning of understanding these.
  • 26:23 --> 26:31But as these drugs come out and as they are available we have the potential to start thinking about OK for a given patient.
  • 26:31 --> 26:37How can we assess that patients immune system and how can we understand the tumor,
  • 26:37 --> 26:45the genomics, the genetics of the tumor in such a way that we can find the best combination of drugs to work for that patient.
  • 26:45 --> 26:48That sounds really interesting,
  • 26:48 --> 26:59because that sounds like the stuff that we've been doing for awhile now in terms of cancer and looking at cancers in figuring out which therapy is going to work
  • 26:59 --> 27:02better. What targeted pathways are turned on versus turned off.
  • 27:02 --> 27:09Should you be using, you know an anti HER-2 agent in somebody who's got a HER-2-positive breast cancer?
  • 27:09 --> 27:12Or should you be targeting KRAS in lung cancer?
  • 27:12 --> 27:16Sounds like you're moving in the same direction in Melanoma.
  • 27:16 --> 27:19But looking at it from an immune perspective,
  • 27:19 --> 27:23and I should say this is mostly on the research side,
  • 27:23 --> 27:29right now we're trying to understand the flavors of inflammation in the tumor microenvironment.
  • 27:29 --> 27:35The composition of the immune cells that are there and why they're there,
  • 27:35 --> 27:44and then once we understand that, we're simultaneously starting to look at OK if we take pieces of the tumor and study them in tissue culture,
  • 27:44 --> 27:46if we study them in a dish and treat them
  • 27:46 --> 27:52with different immunotherapy drugs, can we see patterns of response from some patients but not from others?
  • 27:52 --> 27:55Those are things that we're working on here,
  • 27:55 --> 28:01and others are working on as well that we think could lead to the development of better biomarkers.
  • 28:01 --> 28:03That's one kind of major approach.
  • 28:03 --> 28:09Another one is focusing on the technologies that have emerged to sequence patient genomes.
  • 28:09 --> 28:11The immune cells from patient genomes.
  • 28:11 --> 28:15We do technologies now to look at individual cells in sequence.
  • 28:15 --> 28:17Everything that that cell is expressing.
  • 28:17 --> 28:24Basically everything it's doing and we can do that for a bunch of cells in the micro environment all at once,
  • 28:24 --> 28:33and the thought is that we may find particular genetic lesions in the tumor that lead to a better response to immunotherapy A versus B.
  • 28:33 --> 28:40We may find particular features of the immune system that interact with the tumor as well that predict that,
  • 28:40 --> 28:47and so I think that in the next 5 or 10 years we're likely to see progress in this direction.
  • 28:47 --> 28:50Whether that will translate affectively into
  • 28:50 --> 28:53guiding precise therapy choice for a patients
  • 28:53 --> 28:59Melanoma, I'm not sure.
  • 28:59 --> 29:01When you talk about,
  • 29:01 --> 29:09essentially taking tumors and looking at the micro environment and seeing the composition of these cancer cells,
  • 29:09 --> 29:13and what kinds of immune therapy they may benefit from,
  • 29:13 --> 29:16you can also look at the immune system and see,
  • 29:16 --> 29:23maybe my immune system is different from your immune system in terms of attacking a particular cell.
  • 29:23 --> 29:26Is that on the right track?
  • 29:26 --> 29:28It's exactly on the right track,
  • 29:28 --> 29:36and you know, even taking a step back when we first started to see that these therapies could work for patients,
  • 29:36 --> 29:38people started to ask,
  • 29:38 --> 29:41why do they work for some patients but not for others?
  • 29:41 --> 29:51And we started to look inside patient tumors and one of the things that was clear is that some patients have a lot of attacking immune cells
  • 29:51 --> 29:54even prior to immunotherapy and others don't.
  • 29:54 --> 29:56And just unpacking that basic
  • 29:56 --> 30:00observation is something we're still doing,
  • 30:00 --> 30:09but as I was mentioning, as we start to understand it as we start to understand the chemical signals that the tumor in the immune system makes.
  • 30:09 --> 30:15It's giving us a lot of inputs to try to determine which drugs could be affective in each case,
  • 30:15 --> 30:18and what the basic flavors of immune micro environment are.
  • 30:18 --> 30:25Doctor Jeffrey Ishizuka is an Assistant Professor of Medical Oncology at the Yale School of Medicine.
  • 30:25 --> 30:35If you have questions, the address is canceranswers@yale.edu and past editions of the program are available in audio and written form at Yalecancercenter.org.
  • 30:35 --> 30:39We hope you'll join us next week to learn more about the fight against cancer
  • 30:39 --> 30:43here on Connecticut Public Radio.