>> Sandra Charles: Good morning. And welcome to this our third Cancer Moonshot Program. I tell you, I said, "How am I going to warm the [inaudible]," because I thought you're going to come in here, you know, some people come running on the stage to the music. I won't do any of that. But I do pledge to you that when you leave here, you will be much more informed, better informed and feel so edified, as well as fulfilled that you now understand why all of this focus on research, why is it that we're spending so many dollars in trying to do these scientific programs and research. And the great thing is, this being the third program, we did the first one explaining all about Cancer Moonshot, because it was actually fabulous the way it was timed, because our program occurred the day after Congress passed the Cures Act, which actually is going to be funding this and many other avenues of research. So it was very timely. And then last year we did the Cancer Genomics Program showing you how far we had come with that research in terms of precision medicine and moving towards personalized medicine. And so today we thought we must keep this going. And so we're doing translational medicine. Sounds nice and lofty, right? And it's pretty much it's been [inaudible] to bench-to-bedside, basically. And so it's the why of all the research. Why are we doing this? Because at the end of the day we could have a lot of papers and a lot of findings, but unless we can translate that into something usable, where we can help patients, then it does not quite meet its goal. And the legacy of the Apollo landing back in 1969 was the coining of the term "moonshot." To speak to something that is so audacious that it would aim to do something that seemed virtually impossible and beyond the realm of all possibility. And so that alone gives us a sense of hope. Not trying to create vain hope, but trying to have people to understand that all this complex science can be distilled into something very useable. And that is why in our program today we have not only clinicians, but also our experts, scientists and researchers, and our patient advocates. So I think you're in for a real treat. And I would like to ask my colleague, Dr. Mary Mazanec, the director of the Congressional Research Services to come and welcome you to this program, and prepare you to hear our speakers. So thank you so very much. [ Applause ] >> Mary Mazanec: Thank you, Dr. Charles. So I am -- I think I just broke the mic here. What a way to start. I'm very excited to be here with you today. And it is my pleasure to welcome all of our guests here to the Library of Congress. As Dr. Charles noted, this is the third in a series of discussions centered on the Cancer Moonshot Initiative. The Cancer Moonshot Initiative was initially authorized in the 2016 Cures Act. And the ultimate goal of this critical initiative is really to identify and develop new therapeutics to diagnose and treat cancer. Now, despite all the advances in medical science, cancer remains one of those diseases that still poses a major health threat to so many Americans. I think it's fairly accurate to say that cancer affects virtually everybody. Many of you probably know someone whose life was forever changed when they received a diagnosis of cancer, or you, yourself, may have had your life turned inside out and upside down when you received a cancer diagnosis. As some of you know, I'm a physician, I'm a pulmonologist, and before I came to D.C. when I was seeing patients, I took care of many patients who had lung cancer. And lung cancer back then was pretty much a diagnosis that ultimately resulted in death. But I saw firsthand the pain and suffering that my patients went through after the diagnosis through the various courses of therapy. As some of you probably appreciate, cancer is the second leading cause of death, both in the United States and worldwide. And then, although scientific breakthroughs, some of which you have witnessed, I witnessed over the last several decades, have actually improved the diagnosis and the survival rates of individuals with cancer. There's so much more to be done. And we're not there yet in declaring victory over all types of cancer. So recognizing this, President Obama, basically, tasked the former vice president, Joe Biden, who had just lost his son to brain cancer, to lead a new national initiative termed the Cancer Moonshot. Congress has been on board from the very beginning, and has led the charge towards conquering cancer by consistently working in a bipartisan manner. This is one of the areas where I think there is bipartisan agreement. And they have grappled with numerous policy issues in the area of science, technology and medical research. But, as we all know, advancements are costly. There's a price tag associated with medical research. And Congress has been aggressively funding research in the areas of medical science that are needed to precipitate clinical progress. So, just for example, in fiscal year 2019, the appropriations included 5.74 billion to the National Cancer Institute, which was a $79 million increase over the previous fiscal year to fund research. As Dr. Charles said, this is the third in the series of seminars that she has organized focused on the Cancer Moonshot Initiative. The first one was done shortly after the bill was passed, and brought together scientists, healthcare providers and patients in a dialogue surrounding this major initiative. The library hosted its second seminar in October 2017. And I had the opportunity to participate in that one also. The topic of that one was cancer genomics. Cancer genomics dictate how a tumor behaves, and whether or not it responds to treatment. As many of you realize, the mapping of the human genome was a major step forward in biomedical research, and it really opened the door to a whole new area of research and approach to treating many diseases, including cancer. It also heralded the dawning of personalized or precision medicine, where treatments are actually targeted or individualized to a given patient. So recent genetic breakthroughs have actually expanded our understanding of cancer. And this has resulted in new ways to diagnose cancer and, hopefully, pick it up early on in the course of the disease when it's still very treatable and curable. And it has also precipitated this paradigm shift away from traditional therapeutics to more newer approaches, many of which use one's own immune system to target cancer cells. Now, today we're going to hear from a very distinguished panel of experts, and from the individuals that are patient advocates who have actually experienced cancer themselves, to probe more deeply into this area of research, which is, basically, based on a translational medicine approach. Now, as Dr. Charles explained, translational medicine seeks to improve the health and wellbeing of patients by, basically, connecting the dots between what's happening in the laboratory at the bench with basic research to the clinical practice of medicine. Now, in a former life before I came to Washington, I actually spent quite a few hours at the bench doing research. And my hope was that at some point what I was doing would have an impact on patient care, and would improve the lives of individuals. So this whole area of translational medicine is a major interest of mine that I've followed over the years here in Washington. As I said, it's been described as a bench-to-bedside approach where discoveries in the laboratory are leveraged and "translated" or developed into new therapies. But in order for this to happen, you have to the scientists actually doing the work, speaking to, collaborating and sharing information with clinicians and medical care providers who are actually taking care of patients. And this is what the Cancer Moonshot is intended to do. And I'm going to quote Vice President Joe Biden who explained the purpose of the Cancer Moonshot Initiative with these words. "The Cancer Moonshot Initiative is to better understand and breakdown the silos and stovepipes that prevent the sharing of information and impedes advances in cancer research and treatment, while building a focused and coordinated effort at home and abroad." So, in this sense, translational medicine is a tool. It's an approach that is central to the success of this major initiative. It has both the power and the potential to be the force that drives us towards, ultimately, crossing the goal line in finding a cure for cancer. The National Cancer Institute has a lead and a central role in this effort. They actually have a program called the Transitional Research Program at NCI. And this program sponsors grants in research excellence that actually tries to promote the collaboration between clinicians and basic scientists to discover new approaches to attack diseases such as cancer. So I think I've spoken enough, or I've taken up too much of the time already, because I really want to hear from our distinguished panel of experts. They are going to share their various perspectives with us. And we are joined today by Dr. John Tsang from the National Institutes of Health, who is an expert on human immunology and systems biology. We also have Professor Elizabeth Platz from John Hopkins University, the School of Public Health, who is a leading cancer epidemiologist. We also are joined by Dr. Susan Bates from Columbia University Medical School, who is a clinician and a cancer epidemiologist. And we have two patient advocates, Ms. Juanita Lyle and John Bauer, who are also cancer survivors themselves. So I'm going to turn the program over to them. And, again, welcome. [ Applause ] >> Tomoko Steen: Welcome, everyone. This event was co-sponsored with the Science, Technology and Business Division. You can see this, we organized [inaudible] lectures. And you can see videocasting, this page. And also we had two previous Cancer Moonshot videos are there. And also we invited several Nobel laureate to talk about translational medicine. And that video is there as well. So I hope you enjoy it. Okay. I can't fix this. Okay. I'm Tomoko Steen at the Science, Technology and Business Division. It's my pleasure to introduce our speakers. And the first speaker is Dr. John Tsang. And he is at the National Institute of Allergy and Infectious Diseases. He leads a project, and is also co-director of the NIH Center for Human Immunology. And Dr. Tsang has training in computer engineering and computer science, and also a biophysics Ph.D. from Harvard. So he has received many prizes from the National Institute of Health. And one of the highlights of his program is OMiCC, and he's going to talk about that in his talk. [ Inaudible Speaker ] Thank you. [ Applause ] >> John Tsang: Good morning. Good morning. Thanks for having me here. It's an honor to speak. So, as Dr. Steen mentioned, I work on the human immune system. So today I'm going to tell you a bit of my perspective and highlighting the feel of immunotherapy. And so the title of my talk is "Towards personalized immunotherapies: Predicting and understanding the immune responsiveness of individuals." So I don't think I need to convince you that the immune system is very important. And it's been implicating a large number of diseases. So, therefore, understanding and knowing how it works is very important. So, aside from infection and autoimmunity, [inaudible] you may of heard of, the immune system has been implicated in, for example, metabolic issues, for example, obesity, diabetes, also neurological disorders. So you name any disease, you can always find a connection to the immune system. And, of course, cancer, being such a devastating disease, it's a huge connection to the immune system. So you may have heard that most recently the Nobel Prize in medicine was awarded to two immunologists for their discovery of cancer therapies by inhibition of negative immune regulation. Right? So today I'm going to give you an overview of the science behind this, and also that sort of leads into my perspective on how we need to move towards a more personalized approach to immunotherapy. So the immune system, broadly speaking, it looks for, basically, dangerous signs and signals and attacks. So here's a video showing if you induce damage at a tissue site, you can see immediately the red, those are neutrophils, a type of immune cell, they would immediately sense that damage, and come and swarm that site, and then try to repair the damage. Right? So that's one example. And then the green ones, they are another immune cell type called macrophages. So there they're interacting with the neutrophils in this case to perform the repair. So, in general, there's a type of cell called T cells. It's a type of immune cells that they are the soldiers, basically. You can think of the soldiers of the immune system. They can remember and also recognize harmful things such as pathogens and tumors. So these T cells would look for things that are harmful in your body. They circulate around and patrol. Right? And then they scan for cells, and then they try to distinguish between normal and abnormal cells. So these abnormal cells, typically, sometimes they raise a flag, and they will just display certain patterns that the immune system can sort of tell, "Oh, this is likely some cell that's either infected, or it has some aberration inside the cell." And then once it's detected that potentially harmful cell, the T cells would try to destroy those cells, basically. Right? So here's a very basic picture on how T cells work. On the left is what we call a cytotoxic T cell. So these are the cells that can kill these harmful or dangerous cells. And on the right is a tumor cell or a viral infected cell, for example. So these are dangerous to our body. So these cells, they can present or they can show up on their surface something called an antigen. So that's, basically, a signal for the immune system. The antigen is something that the immune system can recognize as "Oh, this is foreign," for example, to the body, or "This is something that the body shouldn't have." Therefore, the cytotoxic T cells would recognize that, and then start to interact with these cells, and, eventually, trying to destroy it. So that's the very basics of how cytotoxic T cells work. Right? So how does the immune system find and kill tumor cells? Right? So the very first step, it's the tumor cells, they have to display these signs and antigens. Right? So that's the very first step. Basically, this is going from -- the tumor cells would die, for example, and then they would release these kinds of antigens. So that would allow the immune system to start to pick up these signs, and start presenting them to the T cells that I was talking about. So now these T cells, they're, like, in training, basically. So they can be trained now to recognize these antigens or signals from these tumor cells. So once these cells are trained, the step three, it's called priming and activation. So what the the green guy, right, this green guy is, basically, kind of like a messenger. They pick up these signals from these dead cells, and then they start to go and talk to their colleagues, in this case, the T cells. Right? So the T cells are the soldiers. So then these soldiers are now trained by the green guy now. Right? So now they become activated soldiers. They now know what to look for, because the green guys convey the information about these antigens to these soldiers. So now these soldiers are walking through, basically. In this case, going through the bloodstream, searching for cells that are displaying those signals, basically. Right? So then they enter the tumor. They have to cross multiple bridges before they can get into the tumor. You can see these bad guys. Those are the tumor cells, the red guys. Right? So these soldiers are now going in, and then they would recognize them based on the principles I mentioned earlier. And then they can kill these cells, basically. And this is a fairly specific process, as you can imagine, right, because these cells are not just looking for some random cells, because they are instructed by the step one, basically. When antigens are released from these sort of tumor cells dying, or they're not so happy, they are releasing these things into the environment, get picked up, basically. So this is a very fairly specific process. So this is precisely why this holds a lot of promise, because the person's immune system can adapt to the tumor characteristics that's fairly unique to that individual. Right? So that's the key here. So when you think about cancer, it's really not a single disease, even within an individual. So here's an example of sort of a conceptual picture of what really is in a tumor. So going from left to right, it's time. So, initially, you may have some clones of cells that somehow something went wrong inside of them, and they keep dividing. Right? So that's the basis of a tumor. But as it goes through over time, these things start to mutate, meaning they are changing their genetic material and instructions. And then some of the changes would allow them to proliferate or rapidly divide even faster. Right? So now you have different clones, as indicated by the different colors of cells. So over time, you have lots of, basically, different tumors in your body. And so in the case of more classical non-immune therapies, even very specific targeted therapies, they may be targeting a subset of these clones, but as the tumor evolves over time, it's actually very hard for these drugs to kill all the different clones within the tumor. So that's where the immune system is really holding a lot of promise, because T and B cells, they have enormous diversity, so they're the kind of cells I just told you about. Right? They're not static. They're not fixed. They're learning all the time about what's going on. And they have a lot of diversity. So they're like a personal targeted drug factory. So you don't need to instruct it to what to do. If it sees some harmful things in the body, it starts to react to it, and then clone themselves. In a way it's like your anti-tumor clone army. Right? It's like cloning itself making sure that it has enough numbers to destroy the tumors. And it's massive. And this is actually very fascinating biology. If you look into a T cell, also a B cell if it's another type of immune cell, they're the only cell that's in your body where there's parts of the DNA that can be rearranged. So they can rearrange in a fairly random way to generate lots of diversity. So they can, basically, recognize almost anything, be it in the pathogen or in the tumor. Right? So that's what gives them a lot of the ability to recognize all sorts of mutations in your tumors. So, well, is that it? I mean, that's great. We have this system. Right? It could kill these tumors. But there are actually many inhibitory factors that actually make it impossible for the T cell to kill the cancer cells also. And when you really think about it, that's because the immune system also needs to be regulated, right, because that's, for example, why we have autoimmunity. Right? So you cannot just let these T cells to go out there and just replicate indefinitely once they see some antigens, and start to kill lots of cells, because you still need to pull it back also at the same kind. So that's called resolution. So that's why the immune system has evolved ways to regulate these cells at the same time. But those regulations, of course, can be taken advantage by tumors. So the tumors are not stupid. Right? They are pretty smart as well. They also evolve. So that's exactly what Jim Allison and Dr. Hangzhou and others in the field have discovered. It's a way to inhibit these kinds of negative regulation to then allow to, basically, release the brake, to allow the T cells. So in this particular case, on the left here, you can see the T cells that I mentioned in the blue, and then the other one is the tumor cell. So the tumor cell in this case has evolved a way to display a signal on their cell surface to tell the T cell to stop, basically. So the T cell would then go, basically, idle once it sees that signal, because that's the negative regulatory signal. Right? So Jim and Dr. Hangzhou, what they did is they figured out ways to block those signals so that the tumor can no longer instruct the T cell to stop, basically. So, therefore, the T cell can now become more active and kill these tumors. But despite that advance, if you look at the response to immunotherapy, so here's what's called a Kaplan-Meier plot. So on the x it's time over time. So you, basically, enroll a group of patients, give them the therapy, and then on the x it's the percent of survival. So here there are two curves. On the red it's looking at a [inaudible] immunotherapy called anti-CTLA-4. So that's the approach that Dr. Jim Allison discovered. So in this case you can see that over time a lot of the patients actually don't respond, as you can see. And they approach a steady state of about 25% survival, basically. And that seems to be a fairly constant number. So if you now look at these trials, typically, about 25% of the patients actually respond to that therapy. The rest don't respond to that therapy. Yes, by the way, these are metastatic melanoma patients, a fairly aggressive form of cancer. On the blue here, it's showing you some recent advances where they combined multiple immunotherapies. And that has done really well, as you can see here in this particular type of cancer. It gets to about a 60% response rate. Right? But still, a good 35-40% of the patients, they don't respond at all to that therapy. So the question is why. Right? So what factors may predict and, potentially, determine whether a person will respond to that therapy or not? Right? So that's one of the major questions. So there's some known indicators of responsiveness. For example, whether there's any T cells at all in these tumors. Right? So that's a major factor. There are tumors that there are no T cells. Basically, it's like a desert. And then we're still figuring -- I mean, the few are still figuring out why that's the case. And also there may be T cells that they may be blocked from entering into the relevant sites in the tumor. So there are images showing, for example, T cells, basically, they're fairly active, but they're all in the periphery of the tumor. They just couldn't get in. The other factor is whether the tumor is hot or cold. I mean, there may be T cells, and there may be the tumor, they may be interacting, but there may not be enough signals [inaudible] inflammation. So, basically, the immune system relies on inflammatory signals for it to act, basically. So there may not be enough signal to tell the T cells to, "Ha, activate yourself and kill these tumors." Right? So these are all found in different patients. So one patient may not have the T cells. Another may have the T cells, but the T cells may not be able to enter the tumor, or they may all be interacting, mixing up, but there's not enough external signals to instruct the T cells to go in. And the other factor is also, of course, going back to the picture I showed you earlier, are these tumors hiding the fact that they're tumors, or are they actually presenting these antigens, or are they making antigens for the immune system to be trained. Right? So that's another key factor. But there's much remaining to be discovered. There's many different types of tumors, as you can imagine. I just showed you an example on melanoma. And there's many different types of patients, and many different activation thresholds. The immune system, at the end of the day, is very complex. Right? So you may want to now go to your doc and ask, "How's my immune system? Is it good enough to fight off a tumor, for example?" Right? Unfortunately, right now we actually don't have a lot of ways to tell patients how well our immune system is doing. I mean, typically, you can get what's called a complete blood count so you can get the frequency and level of different immune cells circulating in your body, for example. That's quite informative, like, for example, to tell whether you have an infection or not. But it's far from sufficient to give you a sense of how well the immune system is working, or how responsive the immune system would be if it encounters some danger or harmful signal. Right? So that sort of leads to my perspective on the need to measure the immune and then tumor status in the human population. So the immune system is quite unique in the sense that it's a system in your body that is far from completely dictated or specified by genetics alone. So early when one is a baby, a lot of that may be specified by genetics. But actually over time, because of the environment and exposure, there's accumulating evidence suggesting that the state of the immune system is far from just explaining that by genetics alone. So you need to know a lot more about the immune system than just knowing somebody's genome, basically. So, fortunately these days, there's rapid advances in technology. So, for example, you can now go into immune cells, and look at the activity of the genes. You can now do what's called sequencing, which is also used to look at the human genome. But you can now use to look at the immune cell diversity, like different T cells and B cells. And you can also, of course, look at genetic changes in tumors at the same time within that individual. There are a lot of fancy machines that can now really look at the properties of the immune cells in a fairly high throughput manner. Right? So now from a drop of blood from a patient, you can actually generate tens and thousands of data points, potentially informative of our immune system. So this now then becomes more of a data science problem. Right? So you have lots of data points per individual, and you have lots of different individuals with different types of tumors, and different health and statuses, for example. So this is where the machine learning aspect would come in to help us to find markers of responsiveness. Right? So you start off, as I mentioned, you have the immune measurements of an individual in a large population, so these various characteristics, including the ones I mentioned already. And now the question you want to ask is actually what feature out of your tens and thousands of data points may help you predict whether that particular individual, right, will respond well or not, or how they will respond, or how the individual actually will respond to a particular type of immune therapy. So there are rapid advances in [inaudible] of machine learning and computer science, where these techniques are now fairly applicable due to this kind of problem. And so one example, which is coming from work in my lab in the Center for Human Immunology at the NIH, where we're asking a similar question, but in the context of vaccines. And it turns out that when somebody gets vaccinated to, for example, the seasonal flu, our responses can be very different. So some individuals may not respond at all in terms of antibodies, which are the mediators of protection. Some individuals may not have a gigantic response. But what makes one person respond much better compared to another? Right? So we did a study where we kind of took the approach, I told you, we generated lots of data points for each individual, and then asked the question whether we can find some sort of set point or baseline, meaning before the individual even got the vaccine, then we used their immune statuses to predict how well they respond to the vaccine. Right? So on the right here is a cartoon showing you may have hot individuals, they respond well. While the colder ones, they may not respond as much. Right? So what we found is what we call a signature or a predictive signal in these individuals that we were able to replicate, in this case, across different populations of people who got the vaccine, so in this case. So I'm just showing you sort of a summary of the result without going into the details, on why it's sort of like the immune responsiveness score. Right? So that's what you want to compute in a way for your patients, also cancer patients, when you give a [inaudible] immunotherapy, you want to find a way to combine data from multiple modalities, be it cells or genes or gene activities, derive such a score, and that score should allow you to delineate. So in this particular example, I'm showing you the low responders versus the high responders to a vaccine. But you can imagine a good versus a bad responder to immunotherapy as well, right, such that you can now see this had to work, be applied and applicable across different populations. So in this case across different seasons of vaccination. And this is I'm showing you also on the right, it's even extendable to look at different types of vaccines. So in this case, yellow fever is another type of vaccine. Right? So this is sort of an example showing sort of the applicability and potential extensibility of such signatures to be applied to the broad population, but also to different types of interventions. So moving forward, my perspective is that we require two kinds of science to build the kind of framework and to push things forward. One is citizen, and the other one is team science, basically. So as you can imagine to generate the kind of data I mentioned that requires a lot of patient participation, right, to help with the data generation, we need to look at different tumors, we need to look at different personal characteristics, we need to look at different immunotherapies, and we need to look at the very diverse immune system statuses that I mentioned. The other is team science. It's becoming complex, these problems. So it's not simply collecting data from five patients, and look at them in the lab for six months, and then publish a paper. But rather that we need to collect these data, and you need a lot of collaboration among the clinicians and computational biologists, immunologists, software engineers to build a software infrastructure, and also a lot of technology that's needed to advance to make these the sort of measurement technologies I mentioned. Right? They are working well in the lab, but to push those into the clinic, and make them relevant, and make them cost-effective, there's still some way to go. So with that I just want to wrap up, give you sort of a vignette. It's a mix of citizen and team science. So the traditional way that scientists do their science is, typically, we generate a hypothesis, we may say that, "Well, I hypothesized that neutral fields of a particular type of immune cell may be able to kill a particular type of tumor." So that's a hypothesis I just made. It may not be true. So, well, then what I do is I go out and I think about it, and then I design an experiment, and see if I can start slowly making progress to either prove or disprove the hypothesis I just made. Right? And then you generate data to test the hypothesis. This goes in cycles. So that's how science is, typically, done. So what I would like to propose is a slightly revised paradigm, given the kind of data that we have already made available now in the public domain actually. So NIH sponsors a lot of large-scale data generation projects that some of these data you can download them actually. So instead of jumping right in to design a new experiment, you could think about generate your hypothesis, and utilize these existing data as much as possible, right, to explore and then refine your hypothesis before you go again, and go back to the traditional paradigm with designing your experiment, then go back and do this again. But to enable this, you need tools. So that's where this sort of evolved out of our project in the lab, because we have a mix of computational biologists in the lab, but also bench experimental biologists. The bench folks, they are always envied of the computational folks who can just kind of wrangle the data off the web, and start generating some new hypotheses. So they started asking me, "Can we do better? Can we have some tools that allow us to do this faster?" So we ended up having something internal development, and then eventually we decided this is useful for the general public and for the scientific community, so we released it. So this provides readily usable, in this case, gene expression data. It's a type of genetic data that's fairly widely available. And there's lots of them. So you can look at the activity of genes, and different cells and different tissues and different people. And you don't have to do any programming or wrangle with the data in this case. It's all done for you already. So what you can do is, by point-and-click, you can say, "I want to compare, let's say, melanoma patients, the blood gene expression profile of those patients, versus healthy people. I also want to look at lung cancer patients. I want to look at breast cancer patients. And now I also want to combine them all together, and ask, 'Are there any common things that I can find across all of those different datasets?'" So that you can all do within this platform. And the other thing that it allows you to do is what I call citizen science. It allows you to share all the metadata and the annotations. All of those are done in what I call a structured way, meaning they're not just free text, they're structured in a way that's reusable. Right? So now others can come in and look at what you have built in terms of, let's say, the one I just mentioned about tumor comparison. So now you want to say, "Well, I want to compare these tumor data to infection." That you can do. I mean, if that's your question. You can now get grab some infection data, and now combine them with the tumor data that's already built by another person, and put them together, and ask, "What are the differences, what are the similarities," basically. So that's the idea. So we actually experimented with this idea to see whether it could work with a group of people with no prior training in computational biology at all. So we, basically, just sent out an email to a mailing list at the NIH, and told them that there's an opportunity for you to learn how to deal with a lot of the large-scale public datasets. In return, can you come here for a day? You can learn about the tool, but at the same time we're going to give you a topic of research, and we'll see what you can do in a day, basically. So we call this the OMiCC Jamboree. So they came. And most of them stayed for eight hours, because we gave free lunch and lots of donuts. So about 29 of them came from 13 different institutions. Right? So we divided them into ten groups, asking them to look at human autoimmunity. Right? So we happened to look at five different autoimmune diseases, in this case, in both human and also mouse models. Right? So in a day they collected over a hundred [inaudible] versus healthy comparisons. And you can ignore some of the details in the table. They gathered more than a thousand, basically, samples and datasets. And then when we sort of quality control them later, look at them, about 75% of them were really well formed. So in a way you gather 29 people for eight hours, so that's like about 230 FTE hours in the day. They got a lot out of it. They felt like they learned quite a bit. At the same time we actually got something out of it, too, because from the research we almost answered a question that generated quite a bit of hypotheses about these diseases, especially what were common across the five model diseases we looked at. Right? So you're welcome to look at a website. We have a website for this as well. And you can sort of look at the details if you're interested. So I borrowed lots of images on the web to make this presentation. So these are some additional image credits for the pictures I've shown you. And then with that, I would like to thank you for your attention and the folks in my lab. Thank you. [ Applause ] >> Tomoko Steen: We take the questions at the end of the speech. Sorry. Thank you. The next speaker is my dear friend, Dr. Elizabeth Platz. She's a professor at Johns Hopkins Bloomberg School of Public Health. And she has a joint appointment with the Department of Oncology and also Urology at Johns Hopkins Medical School. And she received her doctorate in epidemiology from Harvard. And also has done a postdoctoral research at Harvard as well. That's where I met her. And she has been working on molecular and genetic epidemiology. And she's focused in translational findings to practicing and preventing cancer, especially her area is prostate cancer. [ Applause ] >> Elizabeth Platz: All right. Let me see if I can get the next slide up. That's all right. [inaudible] do it. Okay. So thanks so much for having me here today, Tomoko and your colleagues here at the Library of Congress. So I am a translational researcher. I'm a population scientist and epidemiologist by training. And I spend a lot of my time collaborating with investigators from other disciplines, whether it's on prostate cancer or other types of cancer. So I should indicate I have no disclosures. So today I'm going to talk a little bit about what we've done to discover cancer risk, protective and prognostic factors. So these are all translational opportunities. So all that we do is multidisciplinary. We collaborate, we come together. The areas I'm going to cover today are opportunities for cancer prevention, so drugs used for other indications, and whether or not they're related to prostate cancer risk. So I'll cover a few common drugs for cardiovascular indications. I'll also talk a little bit about periodontal disease and cancer risk. And then, finally, I'll talk about opportunities for improving prostate cancer prognostication by using a biomarker telomere length and variability in telomere length. So here are the disciplines that we tend to collaborate with for our prostate cancer research. You can see there are clinical disciplines, there are basic science disciplines, there are very focused disciplines like pharmacology, and then my discipline, epidemiology, as well as biostatistics. And we all come together to try to solve the problem of prostate cancer. You saw an iterative process like this on one of John's slides. So in our field we do exactly what he said. We test our hypotheses. Our basic scientists are really great at developing, testing and discovering fundamental pathways that are integral to the development and progression of cancer. But the question that they often pose is what's the relevance of what they found for human disease. So they'll come to me and they'll say, "Can you study this discovery within people, within populations, populations of people at risk for cancer, populations of individuals with cancer?" So we then test these hypotheses. And then sometimes we have interesting findings that may be different from what was hypothesized. So the iterative process is then to go back into the laboratory to try to discover mechanisms underlying, and then come forward again. And then eventually, when we have what we think is an answer, and when we can confirm it, not just through basic science and one epidemiologic study, but maybe several epidemiologic studies, we may feel confident that we have an association that may be causal and could be acted on. So here's the first example that I want to share with you. So this was a collaboration across all those disciplines that I showed you including pharmacology. What we wanted to do was something called drug repositioning. We want to be able to identify drugs for other indications that may be helpful for the prevention or treatment of prostate cancer. So I have a couple of my colleagues shown here, Bill Nelson, who's our Cancer Center director, and Vasan Yegnasubramaniun, who's a cancer biologist at the Cancer Center. So, again, the goal was to identify drugs that are used for other indications that may be helpful in the prevention or treatment of prostate cancer. And this approach is called drug repositioning. So what we did was we used a multistage strategy. So we started in the laboratory performing a drug screen. So our institution has a library that has over 3,000 compounds in it, almost 2,000 are FDA-approved drugs. So in the first screen they used two different cell lines, prostate cancer cell lines, and they looked for those drugs that seem to be cytotoxic. That is, they reduce the proliferation of those cells, reduce the multiplication of those cells. And based on that screen, those that showed at least a 50% inhibition of proliferation, they moved on to the secondary screen, which included many more prostate cancer cell lines, both androgen-independent and dependent cell lines. And then based on the top hits, we then identified those that we thought had the greatest cytotoxic potency. That is, the lowest concentration, the greatest inhibition, and also were drugs that were commonly used in the U.S. so that we'd have a chance to be able to study them in the context of a prospective cohort study. And that was stage two. So when we scanned through all the drugs, we noticed that many that came up were antineoplastic. So that is, they're already used for chemotherapy. So those are not the drugs we wanted to be able to study. So then we looked for those that were not known to be chemotherapeutic agents. And actually the top one that popped up was disulfiram, Antabuse. We weren't able to study that one within our prospective cohort study. But the second one that popped up was digoxin. Digoxin is an old drug. It's used to treat congestive heart failure and also certain arrhythmias. Again, it's an old drug. It's not used very much these days. It is a sodium potassium ATPase inhibitor. What we noticed was there was a second drug that popped up that acted on the same pathway. And then among those that were not FDA-approved there was another that ranked fairly high. And so we thought, "We can study digoxin within one of our prospective cohort studies." It was a common enough drug used in older adults, again, for the two indications that I mentioned. We were excited by that. But then, as we looked through the list of the top hits, we also noticed that there was another class of drugs that popped up. And these are the statin drugs. So these are cholesterol-lowering drugs, very commonly prescribed in the U.S. And this made us think that our approach might have some reasonableness, because we had previously studied statin drugs based on their actions for fatal and advanced prostate cancer, and found that men who took these drugs, especially for a longer term had a much lower risk of developing advanced, meaning metastatic prostate cancer, or dying of their prostate cancer. I mean, a substantially lower risk. And this was taking into account other known risk factors for prostate cancer, as well as other cardiovascular and other commonly used medications. So we thought this approach had some legitimacy from a scientific perspective. So then we looked at our top hits, so, again, digoxin and related drugs. We found that men who tended to use these drugs had about a 25% lower risk of developing prostate cancer. And this was true including for more aggressive disease. And then if we looked at the duration of use, men who used the drug longer tended to have a lower risk of prostate cancer, whether that disease was organ-confined, localized to the prostate, or the disease was advanced or lethal. So metastatic or caused death. So we thought this was a very exciting approach. It was two-staged. It was clearly multidisciplinary. We think this approach actually helps reduce false positive hits. So if we had just taken the drug screen and left it at that and picked a few, we wouldn't have known whether or not that was just a false positive. They had more than 3,000 drugs in that screen. But by doing this two-staged we reduced the likelihood of false positive hits. So we thought that was super exciting. This was the cover of the article. And this is to remind me that the drug is derived from the foxglove. So it's a beautiful plant that grows out in the west. Okay? So we think this is a super exciting approach. And then just to tell you, statin drugs continue to hold up. So as our colleagues have looked, as we've looked in our other cohort studies, we continue to find that statin drugs tend to be associated with a lower risk, especially of fatal prostate cancer. So in that cohort that I showed you before, that was the Health Professionals Follow-Up Study. We also continue to see it in the ARIC Cohort, which is a cardiovascular cohort, the Atherosclerosis Risk in Communities Cohort study. And I show the first author's photo here. You can see that she's a young investigator. And so we're very proud of her. This paper just came out this week. And here are the results. So she sees an inverse association for fatal prostate cancer for both users of fewer than ten years, as well as users for more than ten years. And then for total prostate cancer, and this has not been seen before, because most cohorts don't have this much follow-up time to be able to look at very long use, those who used the drug for more than ten years also had an inverse at a lower risk of total prostate cancer. And, again, this is taking into account the number of factors that are correlated with both statin drug use and prostate cancer. So another drug, it's a very commonly used drug, aspirin, for cardio protective reasons, as well as for pain reduction and reduction in inflammation as well. So it's been well recognized for a couple of decades that aspirin use seems to be associated with a slightly reduced risk of developing prostate cancer. But there's not much work on whether it's associated with a lower risk of fatal prostate cancer, or, in men with prostate cancer, whether it's associated with a lower risk of dying of their prostate cancer. So this is work done by another young investigator, Lauren Hurwitz, who's now a cancer prevention fellow at the National Cancer Institute. And what she found was that, indeed, men who are regular aspirin users have a lower risk of developing prostate cancer that ends up killing them. And this was true both in white and black men in this cohort. We took into account, again, the number of factors are associated with risk of developing prostate cancer, as well as use of aspirin. And you might ask, "Well, what about aspirin use? It's still associated with a reduced risk among statin drug users." As I've just told you, the statin drugs are protective. So, indeed, in fact, even among those who didn't take a statin, there's still an aspirin effect. Now, interestingly enough, NSAIDs that are not aspirin, so other non-steroidal anti-inflammatories such as ibuprofen, were not associated with risk of fatal disease. Okay? And then among men who have prostate cancer, if they took aspirin, they also had a lower risk of dying of their prostate cancer. So we think these are exciting findings. Aspirin is not indicated for cancer prevention except in one situation. And that's actually colorectal cancer in middle-aged individuals who have a slightly higher cardiovascular or higher cardiovascular risk for whom aspirin has already indicated. And the reason why aspirin use at this time is not indicated for cancer in general is because of the side effects of aspirin. So GI bleeds, for example, and also stroke. Okay? So now on to periodontal disease. So why study periodontal disease? Well, it's clearly an inflammatory disease. There's an immune component to it. And so we wanted to know, this is a collaboration with a colleague up at Tufts, she's an expert in periodontal disease and cancer, as well as with dentists. So this was a new collaboration for me. So I'm used to collaborating with basic scientists and urologists and oncologists, and now we've moved to collaborating with dentists. And so we were fortunate, because one of the cohorts that we work in, they conducted a dental examination on all of their participants at one of their visits, visit four. So most cohort studies do not have dental exams as an integral component of the work. So we were super excited to be able to collaborate with the dentists who had performed this work. And so what did we find? We found that individuals who had periodontal disease, especially severe periodontal disease, and edentulism, meaning not having any teeth, which is often caused by severe periodontal disease, had a higher risk of cancer, very modest, but there was a dose-response. And then, in particular, there were clear patterns for colorectal cancer and lung cancer. This was taking into account the number of factors related to periodontal disease and to cancer, including colorectal and lung cancers. So we were excited about these findings until you stop and think, "What is a major risk factor for periodontal disease?" It's smoking. Smoking is also a major risk factor for cancer. So what we did is we didn't just adjust for smoking, we looked in the never-smokers, not the non-smokers, but the never-smokers. And, sure enough, the patterns were still there, not as strong, right, not as clean, but the patterns were still there. So we were very excited about these findings. And at the end, I'll tell you a little bit about what our next steps are. And so the last story I want to tell you about is developing a biomarker for prostate cancer prognosis. So prostate cancer is a very common cancer. It's the number one cancer in U.S. men. It's the number two cause of cancer death in U.S. men. At this time, we have few tools to help us distinguish which men have disease that's so aggressive that it needs to be treated, and which men can have either a less-aggressive treatment plan, or just be watched. Okay? So, typically, what's done is on biopsy the patient, along with his providers, will determine whether or not the morphology of the tissue is such that the man is likely to have a higher risk of progression or not. And it's a value judgment for that man whether he wants to undergo treatment. Now, if it's a super aggressive disease, it's metastasized, that's a different issue. But if it's early, so it's working confined, and the disease doesn't look particularly aggressive under the microscope, there's complexities. The man has to decide with his provider what to do about treatment or non-treatment. It would be much better if we had a tool that could complement, it could help men with their decision-making, and help providers be able to give the best option for management of that patient. And so years ago we had a thought. And this came out of a discovery in my colleagues' lab, Angelo De Marzo and Alan Meeker, Alan is up here, here's Angelo, where they identified that throughout the natural history of prostate cancer there was increased shortening of telomere length. Okay? So the pre-cancer cells even have shorter telomeres than the normal cells. And so they asked the question, "Could information about telomere length," -- and telomeres are the ends of the chromosomes, and when they shorten that can lead to chromosomal instability. And so they asked the question, "Could information about how short the telomeres are in individual cancer cells tell us about prognosis?" And so we came together to do a cohort study. So this is a population science design using real people who have cancer who are participating in a cohort study, the Health Professionals Follow-Up Study. We came together to be able to make measurements, and then relate these measurements to risk of progression to lethal disease, to metastasis and death from prostate cancer. This took a big collaboration. So it wasn't just our team. We collaborated with Harvard, where the Health Professionals Follow-Up Study is located. My colleagues, Alan Meeker and Angelo De Marzo, had developed a technique. This technique allowed us to look at individual cells, so not looking across the tissue as a whole, but to be able to look at individual cancer cells, individual cells that are non-cancer cells. And when I say individual cells, I mean not just the individual cell, but of specific cell types. Which then allowed us to explore in detail which cells are the ones that were providing information about prognosis. And what we found was it wasn't telomere length in the cancer cells that mattered. It was the variability in telomere length from cancer cell to cancer cell that provided information about prognosis, along with how short the telomeres were in the stromal cells adjacent to the tumor cells. And so the stromal cells were primarily fibroblasts and smooth muscle cells. And so what we were able to do was evaluate about 30 to 50 cells per type, using a fish approach. And so these are beautiful fluorescent scans. And you can see the pink dots are the telomeres and the normal tissue. And then you can see in the tumor the pink dots are very faint, in some cases you don't see them at all. So that indicates the telomeres are very short. So here are the original findings that we had that were published a few years ago. And so I'm going to draw your attention to prostate cancer death. So, again, these are all men who have prostate cancer. And what we found was, compared to men who had less variability in telomere length among their cancer cells and had longer telomeres in their stroma, that's the reference group, those men who had shorter telomeres in their stroma and greater cell excelled variability in telomere length in their cancer cells, had the highest risk of progressing to prostate cancer death. And this is after taking into account the known prognostic indicators. Men who had the other two combinations had intermediate risks of progression. We found the same pattern for lethal prostate cancer, but, interestingly, not for biochemical recurrence. A biochemical recurrence is when the prostate cells that have escaped from the prostate before surgery, I should say all these men had a prostatectomy, those cells that escaped in some men later they start to produce PSA, prostate specific antigen. And while there may not be evidence of metastases, this is an early indication that it's possible that this man might eventually develop metastases. But in most cases they don't. And so what we want to be able to do is figure out which men are most likely to actually progress to metastases and death. So we've now repeated this work in a number of additional studies. And we've put all the data together in a form called a meta-analysis. And you can see that the pattern remains the same for prostate cancer deaths and for metastases. And for recurrence, we don't see the same pattern. What's popping out is information about the variability in the cancer cell, but not the stromal cells. So we're super excited about all the studies that I've shown you. These are great examples of collaboration across disciplines to discover cancer risk, protective and prognostic factors. And perhaps the most important part of what I'm going to tell you is what the translational opportunities are, what are the next steps that our teams together are taking to take the results that I just showed you to the next step, so that we can actually get these risk and protective factors and prognostic factors to the population for prevention, or to patients to help with better decision-making. So with respect to statin drugs and periodontal disease, our next steps are additional basic science research. So we have currently funded projects to go back into the laboratory with respect to statins. It's to understand how statin drugs influence a cell that's related to what John talked about, T regulatory cells. T regulatory cells hold back that autoimmune response. So they're immunosuppressive. And we have evidence from our colleague, who's an immunologist, that statin drugs may, in fact, reduce the immunosuppression by those T regulatory cells. So we're going to test with him directly that hypothesis. And then for periodontal disease, with our colleague, Dominique Michaud up at Tufts, we received funding from the American Association for Cancer Research to more directly test whether periodontal pathogens and the immune response are related to lung cancer in particular. And then the next one for translational opportunities, I didn't show you research we've done to look at how common risk factors for cancer development may relate to cancer outcomes in people who have the disease. And, in particular, we've had an interest in obesity in men with prostate cancer, and how that may relate to their prognosis. And so what we've found over several studies is that obese men, and especially men who gain weight from before their diagnosis to after, have a much higher risk of progression. And so my colleague just received funding to conduct an intervention study to take men who have prostate cancer, who have early recurrence, that's biochemical recurrence, and then she's going to randomize them to a lifestyle intervention for weight loss versus standard of care, and see if that affects their outcomes with prostate cancer. So that's developing and testing strategies to intervene to reduce the burden of cancer. Another example is the telomere biomarkers. So what are we doing next? We've developed the biomarker. We've actually optimized it for use. The question is, "Well, what is the use beyond just being able to say to a man, 'Here's what we think your prognosis is.'?" Does it have any role in helping for treatment decision-making. And we actually think that it does. We think the biomarker is telling us something about disease biology. So we've just submitted a grant application to determine whether the telomere biomarker will give us information about which men, who've started to recur, the PSA has gone up again, which men would benefit from simple salvage radiation therapy, and which men would benefit from additionally having anti-androgen therapy on top. And the reason why not all men are given anti-androgen therapy in the first place when they start to recur is because it does have side effects. And so to figure out which men would benefit versus which men would not would be an advance in the care of those men. And then the last one, as I mentioned, we have optimized the telomere biomarker. And we had funding to do that. So thank you for listening. Here are the acknowledgments, the funding and then our colleagues, an example of our joint lab meeting that has our basic scientists, our urologists and our population scientists who come together to study the problem of cancer, and in particular prostate cancer. Thank you. [ Applause ] >> Tomoko Steen: Thank you so much. Thank you so much. So our third speaker is very close to our patients. Actually she's a clinician. She's a [inaudible] clinical investigator, Division of Hematology and Oncology. And she's also director of the Translational Cancer Medicine, and head of the [inaudible] Therapeutic Section at Columbia Medical School. And she was at the National Cancer Institute. And she has done significant discoveries and studies. And her work is epigenetic modification agents, as well as her group received approval by the FDA for [inaudible]. That was a medicine originally developed by Japanese scientists for the antibiotics purpose, but it was too potent. And her group got approval for [inaudible] T cell lymphoma, as well as peripheral T cell lymphoma. And she has been working now at Columbia still a combination of laboratory studies and clinical studies. So she has a goal to translate the ideas from the laboratory to clinical trials. So, Susan, thank you so much. >> Susan Bates: Thank you very much. Thank you. I really enjoyed those last two talks. And I hope we can find the third [inaudible]. [ Inaudible Speakers ] So what I've been doing since I went to Columbia is actually working more on pancreatic cancer. It's a very, very difficult disease. And I have learned an enormous amount about it. And I wanted to just talk to you some about that. That is completely translational, trying to identify strategies to try to bring to patients. Anyone know anyone who's ever had pancreatic cancer? So you've known some people. So it's not a commonly diagnosed disease as others, as you can see here on the slide, where only 3% of patients who are diagnosed with it, of all 1.7 million cancer diagnoses expected this year. But, in fact, it accounts for a larger fraction of patients who die from cancer. And it's on target to become the third leading cause of death from cancer in general. So it is very important that we try to start figuring out how to solve the problems of pancreatic cancer. This is the five-year survival from -- oh, my goodness. Sorry. I don't know how that happened. Oh, my. This is the wrong version. This is completely the wrong version. So I'm going to have to give you a different version. I'm sorry. [inaudible] to do that. I guess we can keep talking. This is totally the wrong version. I apologize. So I had my husband give me some edits, and all he ever edits is font. The font, that's all he ever edits. So the problem with pancreatic cancer -- I'm going to get my computer open and I'll talk at the same time. [ Inaudible Speaker ] So the point about pancreatic cancer is that it's a big problem because of the local disease. Yeah, you can [inaudible]. It has the local disease, it has distant disease, and it is drug resistant. And then we have problems with -- and we have made progress in precision medicine and immunotherapy, but we have not made the kind of progress that we need to make. And that's what we'll be talking about. So the local disease, probably the pancreas, is it's nestled right up under the -- right up under the liver -- It's going to be the last [inaudible]. Right up under the liver, where the bile drains from the liver, part of the digestive process, into the small intestine. And the pancreas sits here, right nestled in behind the stomach. And then when you get a cancer there, it's actually sometimes diagnosed very early, unlike other cancers in the body. It can be diagnosed when it's one or two centimeters, because it is in such a vital location. But that doesn't seem to make a big difference in what happens, because the recurrence rates are very, very high. The recurrence rates -- [ Inaudible Speakers ] Recurrence rates are very high. And the reason they are high is that it is a distant disease from the very beginning. So while people think, "Oh, this is a two-centimeter pancreatic cancer, I should be able to get that out and cure the patient," in fact, what happens is that the tumor cells have already escaped into lymph nodes, this is a drawing of a lymph node, escaped into the bloodstream and traveled to the liver, and set up what we call micrometastatic disease in the liver and in the lung. And, in fact, 75% of patients are not candidates for surgery, because it's already invaded into those local structures, because it's right next to everything that matters in your digestion, and also because it very soon, very rapidly travels into the bloodstream. So 75% don't get surgery. And I tell this to patients all the time, "You're in the lucky 25% of those who do get to go to surgery." And then I try to gently tell them, "But there's a 75% chance that you're going to die of this disease anyway." So that makes that 8% 5-year survival a reality. It really is a death sentence. This is a very difficult disease. So it's drug sensitive, but we do have drugs for it, like we do with other cancers. Which for many years, when I was in medical school, there were no drugs. No one thought that pancreatic cancer would respond to anything. But now we have found, eventually, that there are a couple of treatment regimens. And these are a bunch of different patients, where we have response of the tumor, it shrinks in size, and then it grows back. Response, grows back, response, grows back, response, grows back. So you can see that the majority of the time it grows back by about -- it really starts turning around by 100 to 200 days. So that means three to six months. So by the time I'm kind of relaxing and I think I've got this patient on autopilot, they're getting their chemotherapy, the scan comes back and tells me I've got disease progression. And I have to start all over. And that is the life of the patient with pancreatic cancer. Once you start down that road, it's a never-ceasing source of the cancer's going to respond to treatment, but it's going to come back, until eventually we run out of treatment. So there it took actually research. The concept of how difficult it is to do clinical research sometimes amazes me. And I've been doing it for all these years. But I sat down once and I added up all the patients enrolled in the clinical trials that it took to find that one drug combination called for Folfirinox was better than Gemcitabine, and a two-drug combination, Paclitaxel with Gemcitabine, is better than Gemcitabine alone. So these are our two leading drug combinations. So better than a single agent. It took, basically, 40 clinical trials and 14,000 patients to get to the point of proving that, because over and over again we tried and we failed, we tried and we failed. And we, finally, got two regimens to try to treat these patients with. [ Inaudible Speakers ] My computer's so full. And this is just the benefit that we got out of these two, that in terms of survival. That's how we measure overall survival and how we measure a benefit in pancreatic cancer. We don't have to measure how good does this drug work, how to shrink the tumor. We just have to measure did it make you live longer or not. And that's a devastating comment in itself. The FDA has spent enormous effort trying to accept other endpoints for other diseases, tumor shrinkage, a disappearance of tumor, various things. And pancreatic cancer is one thing. It doesn't make you live longer. And so these are the two chemotherapies. And this is for people who couldn't go to surgery where the disease was spread outside the pancreas. So in those patients, it made you definitely increase survival by four months, live from, basically, seven to 11 months. And this is one of the reasons, it's not the only reason, but you heard about lymphocytes and lymphocytes being in cancer cells. So this is one example of a pancreatic cancer where there are plenty of lymphocytes or immune cells, those T cells, and up here is the cancer. But what you can see is that there's a lot of fibrous material. It's actually called hyaluronic acid. Dermatologists use it to make your cheeks puffier when you see people that have been to dermatologists. And it actually prevents those -- we think prevents those lymphocytes and from getting into the tumor cells. So the question is, "Can we do better with precision medicine?" And that's where -- okay -- [ Inaudible Speakers ] Well, we'll see. [ Inaudible Speaker ] I'll just talk when I -- I have such lovely, better slides than this. It might be that I sent you two versions. I probably send you the correct one the first time. So I don't know if you have that on your flash drive. [ Inaudible Speakers ] Yeah, yeah. Did you save both versions I sent you this morning? [ Inaudible Speaker ] Because maybe I sent you -- it was very -- [ Inaudible Speaker ] They were identical? Okay. Anyway, so what we're going to be talking about now is precision medicine, and how that relates to pancreatic cancer. So what we've done in oncology is, as a result of the Cancer Moonshot, as a result of a lot of fabulous progress that you've heard about already, we've learned to figure out specific changes in patients' cancers that lead to help drive that cancer, and that we can identify a drug that will target that cancer, and have succeeded. So that's called targeted therapy. That's how we've come to call it. Precision medicine and targeted therapy are, basically, the same thing. And a lot of, for example, the American Cancer Society in trying to explain what is targeted therapy, they say these drugs target certain parts of cancer cells that make them different from other cells, or they may target other cells that help cancer cells grow. But it's a specific target, as opposed to our traditional chemotherapy, which -- [ Inaudible Speakers ] So that's different from traditional chemotherapy, which binds DNA, binds tubulin, binds something in the cancer cell, and the cancer cell has to be more vulnerable to being killed. This is something that makes the cancer a cancer. And that's the reason why we call it targeted therapy. So it is clearly a validated treatment in some tumors. This is just an example that I got from a website where they're showing if we take a number of patients who have, for example, lung cancer, we know that some are going to have what we call an EGF receptor mutation, some are going to have what we call an ALK mutation, different changes in cancers that lead them to be different, so they're alterations that lead them to be susceptible to different treatments. So the key here, though, is that this person has to be identified as having had the EGF receptor mutation, and this person has to be identified as having an ALK mutation. So we have clear examples where it's become a real cancer medicine by doing that. We know that in chronic myelogenous leukemia we can treat patients with Imatinib that inhibits this protein. And patients now are not even dying from the disease. It's turned it completely into a chronic disease. We know that in breast cancer patients with HER2-positive, that's a mutation or an amplification of HER2, they have very good, very long treatment responses with an antibody against that. Erlotinib and Gefitinib. And I'm going to show you more about that. And then a melanoma mutation against BRAF that I'm sure you've seen in the news. So here's the mutation. EGF receptor signals cancer cell proliferation, cancer cell survival. It makes the cancer grow. It's a ghost signal for the cancer as a whole. And that EGF receptor mutation is the one that we can specifically target with EGF receptor inhibitors, Erlotinib, Gefitinib, drugs that are FDA-approved. These make people live longer. It converts their disease, at least for a time, into a responsive disease. And then other cancer drivers have been eventually recognized. Targeted therapy for all of these has been developed -- for most of these has been developed. Not for all of them. But they are effective. They're not curative. But it has been a watershed for lung cancer. It completely changed it from the kind of disease we talked about earlier where no benefit to a disease where we have drugs that target it that hold the disease at bay. So if we don't get to the end of it, we'll be okay. You think you've got it loaded? Okay. [inaudible], but let's wait and see. Maybe I somehow just got the whole thing tangled up. So what people have done then, and now this is where personalized medicine, basically, went out into the community, before we were completely ready for it to be there, but it went because every patient wanted to know what was in their cancer genome, what caused my cancer, what is my cancer. So many different institutions have developed their own 500 cancer gene panel. There's agreement that about 400 genes are cancer genes. And the people took it on their own to develop their own panel for whole genome sequencing. Columbia has its own. Memorial Sloan Kettering has its own. University of California San Francisco has its own. FoundationOne is the most widely used. And so these actually cost an enormous amount of money. And patients want to know, and they hope that that will give them information that will lead them to a treatment. Now, what comes back on these cancer panels -- [ Inaudible Speakers ] Okay. So can we switch to the -- can we switch over to the other [inaudible] that she saved? I'm sorry. I know this is taking [inaudible]. [ Inaudible Speakers ] I'm sorry. And that's the last time I let my husband work on my talk. [inaudible] a big [inaudible] like that [inaudible]. He's also a physician. So what those 500 gene panels do is they are looking for all different kinds of mutations. They're looking for many more things than just individual mutations. But the thing that we look the most for is where, at the DNA level, you have one base change that leads to a change, potentially. It can be [inaudible] in the RNA, and leads to a change in the amino acid. Those are called missense. It doesn't, we think, really hurt if it changes a base that doesn't change the amino acid. Sometimes it causes a change that leads to the protein not being synthesized, and the loss of a protein. So these things can all be related to cancer. And that's what we get back from our 500 gene panel. So this is a patient of mine with pancreatic cancer who we ran the Columbia panel on. This is the kind of result we get back. Four hundred and sixty-seven cancer genes. We have mutations in KRAS, TP53, SMARCA4, CDKN2A. So a whole bunch of genes. Some I don't even know what they are. I don't know what all these genes are. And that's what your doctor is getting back when you sent your thing to FoundationOne, he's getting back a report just like this with all kinds of genes that you don't actually know what it has. And so you get the report back, and they classify these variants, and they say, "Well, some are unknown mutation," like the EGFR mutation that I showed you. And there's an FDA-approved drug for it. And if you have lung cancer, it's an FDA-approved indication. So in that situation, bingo, you're settled. But many of them are it's an EGFR mutation. There's an FDA-approved drug, but it's a different tumor type. Is that the same thing? We actually don't know. And sometimes it comes back a known mutation, but there's no FDA-approved drug. And sometimes, as with many of these, I have no idea what that gene is, or what the mutation is doing there. So the problem with that is that it's a $5,000 test. And we're, basically, running $5,000 worth of experiments on lots of patients without actually knowing the outcome. So the precision medicine strategy is to take a newly diagnosed cancer, and find out which of these categories it really fits in. Is it a known mutation in a known tumor type with an approved drug? Great. Or is it a known mutation in a different tumor type, in which case the doctor has to work hard to get the drug and off-label use, but you can do that. I've done that. And I actually did it for that patient. And I picked this one because there's a breast cancer drug called palbociclib. Now, I could have picked this one, but there's a drug that's in clinical trials that might work against that. That would be a lot harder for me to get that than an FDA-approved drug. But you see I have no data for either choice. And then there can be a completely unknown mutation. So I've already alluded to some of the problems with where we are with precision medicine. It sounds great. We all want it. We all absolutely want it. Every day when I see patients, I want to order the tests for the patients. But the truth is knowing the mutation, it may have a different role in the tumor I'm looking at. Many mutations can be found, not just one. We don't have drugs for most mutations. And when we direct a patient toward a targeted mutation, we direct that patient away from the proven therapy. It's new. It's expensive. And patients and insurers pay precious dollars for what's effectively an experiment. Now here's where I'm going to show you how it's an experiment. So BRAF in metastatic melanoma, a fabulous discovery, that most melanomas have BRAF. It's driving the cancer to grow. And when we treat with a drug called vemurafenib, or BRAF inhibitor, it turns off the signaling. It went from bright on this glucose scan, all these are spots that patient had all over his body, and now they resolved. It doesn't work forever, but it works. And this is the whole patient. This is the way we show patient responses, called a waterfall plot, where it's the percent shrinkage. So patients didn't have 100% shrinkage, typically. Some did. But many had a fraction of that percentage. I mean, this is a fabulous waterfall plot. And it made people live longer. Those who got the drug lived longer than those who didn't. So then they said, "Well, this mutation shows up in lots of other cancers. We're going to practice precision medicine on those other non-melanoma cancers." So the biggest group that also has this BRAF has colorectal cancer. And, in fact, there were six patients that had shrinkage. Nothing like you saw in the waterfall plot before. This looks totally different. And, in fact, they all returned to baseline by one month. It wasn't by 11 or 12 months. So this did not succeed at all. This was a total failure. And, in fact, a number of people said they took a broader approach and they said, "I'm going to do the FoundationOne in a variety of patients, and then give them all the best drug I can think of with the FoundationOne precision medicine strategy. And I'm going to see if they live longer." And so they collected all those patients. And, really, there's very little difference between those curves. It has a ratio of .88 is, basically, saying almost no difference. So precision medicine, we have to think about -- we cannot think of that right now as the answer to everything. What we have to do is keep looking for actual targets in tumors that are drivers in most tumors. So this is pancreatic cancer. And this is a sequencing of about 400 patients. And these are a variety of genes that were found to be in one or two percent of those patients. But there is a gene called KRAS. Very similar to that EGF receptor. It drives growth, drives size, drives metabolism. It's found in pancreatic cancer almost always at the very beginning. It's the first gene that changes. So it is a key gene. And the NCI actually has set up something called the RAS Initiative to try to find a drug for this. But we don't yet have one. Now, RAS signals a lot of things. It signals through ERK, which is in that BRAF. But it signals down to a protein called MYC. MYC is highly expressed in pancreatic cancer. And when it is expressed at very high levels, you actually have a lot more patients dying. This is a survival curve. This is more survival, this is less survival. So patients with pancreatic cancer have less survival when you have active signaling through MYC. So one of the things my laboratory now is working on is a compound that will turn off MYC in pancreatic cancer. And you can see here, this is a western blot of protein expression. You can see very high MYC expression, and then you see how it's turned off. Very high and how it's turned off. It also kills those cells, at least some of them. Some are not very sensitive. This is a killing curve of pancreatic cancer cells in the laboratory. And we start out at 100%. And at least two of the cell lines that we study are very simple. But we're stuck in this research because we need $50,000 to make this drug. And we don't have it. And we keep putting in grants over and over again. And we still aren't getting a grant back to finish this research. So if any of you have any influence over directing dollars toward pancreatic cancer research, think about that. So my patients always come back and they say, "Okay, what about immunotherapy? I want a treatment. I want immunotherapy." And you saw this a little bit earlier. The way I think about this -- see here's the big tumor cell, the little bitty T cells, which is amazing. All these drawings, the T cell is no match for the cancer cell in terms of size. There's this handshake. I call it a handshake that is, basically, this tumor telling this T cell not to be active. You saw the inflammation that we talked about earlier that you need to have. Well, when we add those new drugs that are FDA-approved, you block that handshake, and then you get T cell activation. And the tumor dies. So the T cell feeds things into this tumor cell that make it die. And so we want to block the handshake that turns off so that the T cells can attack the tumor cell. Well, this is what -- I got this off of Google and then I couldn't refine it, but someone had drawn it showing that immunotherapy -- it's a great time in oncology because of immunotherapy. You can see that they've drawn this 100% survival curve, dropping [inaudible]. So 80% of patients are living because of immunotherapy. Whereas without it, patients die. Whereas this is so much better than traditional cancer treatment. Right? How many believe this? I've got some land in the swamp to sell you. So, okay, there are patients who've done amazingly well with immunotherapy. But it is not 80% of patients. The reality is far closer to this than it is to this. So that's one problem we, who've developed small molecules have, "Well, your grant doesn't even have immunotherapy in it. You just want to develop a small molecule." So the reality is that immunotherapy is not helping. Only in melanoma is it able to even approach the majority of patients. Every other disease, it is much closer to this reality. But I'm not trying to be negative. There are lots of other handshakes that need to be studied. And all the things that you talked about -- Dr. Tsang talked about in terms of why people respond, why they don't. But too much in -- to my mind, there's too much energy currently invested in immunotherapy, and not enough back in small molecules, back making the precision medicine dream a reality. There are still other oncogenic drivers that need drugs. And that's the work I think is so important. So we have 361 clinical trials, including ten that are in exactly the same category of the exact same handshake that we already have very good drugs FDA-approved for. So we need new immunotherapy, different immunotherapy. And we need more small molecules to be developed. So just to sum up, pancreatic cancer's on track to become the third leading cause of death from cancer. Surgery's not enough. Responsiveness to chemotherapy, yes, it's there, but it rapidly develops resistance. Precision medicine and immunotherapy are works in progress. And research funding is critical to make progress in this disease. So to sum up, I have a lot of people that have helped me with all the work that we do. A lot of people at Columbia. Just been blessed with many, many collaborators over the years. Thank you. [ Applause ] I'm sorry for the confusion. I don't know those two slides got in there. I'm completely baffled. All right. And that slowed us down, too. I apologize for that. [ Inaudible Speakers ] >> Juanita Lyle: Good afternoon. Welcome back to our third Cancer Moonshot Program. It gives me great pleasure to be here in the midst of progress that I had hoped to see and continue. I have surpassed the time of being a recipient of such a wonderful good news time with the Cancer Moonshot Initiative. I came from times that things were still in the research process and sheer experimentation. Now everything to be here -- to be now progress with this Cancer Moonshot. Now it's today it's more about the treatment of the patient than the treatment of the patient with the disease. So I welcome aboard today John Bauer, [inaudible] club of the big little c. I told John that the reason why I called it the big c is because it becomes the little c as we go through the process of beating the disease itself. Meaning sometimes it looks too big to defeat, but when you do it, it becomes something little to kick to the curb. John Bauer has walked the journey of the c. He started working in the Library in 2010. He started his journey in 2014 with thyroid, head and neck cancer. Fortunately, they were treatable and curable. He, like I, did all the right things with food and fitness. He is a father and husband with three young children. I, of course, was a mother and wife in 1976 with a six-year-old daughter and an eight-year-old son. But I look at John today, and John is [inaudible], John, welcome to thriving in survivorship. So John it's you're moment to share now. [ Applause ] [ Inaudible Speakers ] >> John Bauer: So I can't offer any cures for cancer. But there's no -- I know nothing about telomeres or [inaudible] or any of that. But I do offer a perspective from a patient. And, hopefully, offer a better engagement on physicians' part and researchers' part with patients. And also to offer patients a better ability to cope with the situation at hand, which can be devastating, of course. So I just borrowed the title page here from -- [ Inaudible Speakers ] Got it. Thank you. Okay. I'm an IT guy. I don't do too many presentations. So, anyway, that slide's just to start it up. I didn't know -- the guidance was to convey my own personal experience, but also to emphasize the engagements with medical personnel along the way. So I thought how do I do this in an engaging and thoughtful way? Cancer is kind of in the same category as 9/11 or the Holocaust. And it's just, you don't joke about it. It's just too dreadful and it's pernicious. And as affected as people are by it, it certainly doesn't lend itself to standup comedy. But, nevertheless, I did try to put together something that was entertaining and thoughtful. So here we go. Here's my dad and I in the early '60s. And then my dad was swept up in 1969 in the Vietnam War, and he was exposed to Agent Orange. Myself, I was doing okay until 1985. When Chernobyl hit, I was studying German in a small German village. And Chernobyl happened. I knew it happened. I didn't know exactly how serious it was. Nobody really did at the time. They just picked up some sensors in [inaudible] had picked it up. And the cloud was rolling across Eastern Europe. And it was really unsure how serious it was. The German politicians were assuring everybody [foreign language], don't worry about anything. Everybody stay put. How are you going to evacuate millions of people anyway? So I thought, "Okay, I'm going to sit this one out." But then I saw my classmates coming back up from Switzerland from hitchhiking, [foreign language], which is okay to do in Switzerland. And half their face was completely burned, just burned to a crisp. And the other half was completely unaffected. So I knew something -- that was the side that was pointing where the cloud was rolling in. So I thought, "Okay, it's time say auf wiedersehen and jump on the plane." So my Iceland air flight, my chief Iceland air flight I had, I wasn't due to return until months later after the study concluded. I found myself up in Iceland. I thought, "Oh, great. Up in Iceland, I'm home free." The cloud's not going to make it up here, certainly. But then when I discovered later that Iceland took a really big hit, of course, I was outdoors admiring waterfalls and doing all the Icelandic things. So if I had to take a guess of what -- have the thyroid cancer [inaudible]. I tried to get iodine. Of course, nobody spoke English, and there wasn't any iodine on hand. What they had went to Icelanders. I couldn't get iodine. Okay. So in any event -- because I know iodine, of course, soaks up the radiation, it flushes it out without it affecting. Okay. Fast forward to 2004. My dad, by the way, his cancer started in the early '90s. And his second wife was a Thai doctorate who had professional commitments back home. So he moved with her back to Bangkok. And I get this call, it was actually May 2004, late May. My twins were born by in vitro fertilization. That's a different story. August 2004 I get the dreaded call that every son or daughter dreads. I'm in the shower and getting ready for work on a Monday morning. And my wife comes running in, "John, come quick. Your dad's in trouble," et cetera. So I hop on the next plane to Bangkok. I was by myself at the Faculty of Medicine at Ramathibodi Hospital, where my dad had formed friendships with the research staff there. And he would grade their papers -- he would edit their research papers in return for them keeping him alive. And it was a good exchange, a good arrangement. They keep him on the latest drug, and until that drug stopped working. Then another one would come along. And they managed to prolong his life for ten years because they caught -- back in America they caught his condition late, so it had progressed. And here they are here, the physicians. They used Mr. Robert. They addressed you by your first name. This is the ICU chart. He was in ICU when I arrived. This was taken later so I'm smiling. And, of course, everybody in Thailand smiles. So she's the ICU nurse, the hardest job in the entire world. And I went back to thank her afterwards. So this is a bit of a staged photo. This didn't happen as I arrived. It wasn't [inaudible] arrival. The IXU chief at one point, as my dad had a tube down his throat, and he had a needle in his neck because they would have to transfer [inaudible] every couple of days. And they were just, basically, artificially keeping him alive. And the ICU chief -- this went on for several days. And his heart was strong. And I, you know, "What do I do? I never -- in a new situation -- you had to know." The ICU chief pulls me aside, and he laid down the law with his, read me the riot act. And he said, "There comes a point at which we're no longer extending life, that were prolonging death." And I said, "Right." So I took a walk around Bangkok with his doctors. And I said, "What do I do?" And they said, "Well, do what we all do, and just make the hard decision." And I said, "Okay, when would you like to do this?" She said, "Whenever you like." And I said, "Well, how about tonight?" "Yeah, okay, great." So they put out the word. All his friends, every friend he had in the world at that point, gathered in the ICU room. And a fellow came in with a syringe, and I held his hand as he passed. And I had a certain amount of engagement with him when his eyes were still open. And I was able to communicate everything I needed to say. But it was certainly a one-sided conversation with a tube down his throat, there was no argument or anything involved. But, anyway, so here's the temple. I'm going to blast through these quite quickly just to kind of show you what it involved. And on this thought everybody says, "Well, we're praying for you." Regardless of religious belief, it's more of a coping mechanism on their part, really, because what can people say? I mean, this person has cancer, for heaven's sake. You don't want to say, "You're going to be fine," or, "Well, you shouldn't have this to drink. You shouldn't of drank Gatorade." I don't know. But, really, the discomfort was in everybody being required to confront their own mortality. And I think it's really more for them. I said, "Oh, thank you, thank you. I'm not a religious person. [inaudible] a spiritual person. Thank you, thank you." But just to hedge my bet I did have prayer groups going in all major world religions pulling for me there. And, ultimately, it worked. So, again, here's back in my day. This is [inaudible] momento where we flashed back and flashed forward [inaudible]. You never really know where you are in the presentation at the time. But I managed to get the point across at the end. So here's a [inaudible]. I had it [inaudible], sorry, picture of him. I think it was midsummer heat, Bangkok, Thailand. Full uniform, he's got a cigarette hanging out of his mouth. He's doing his work. Look, I don't get it. His job is to cremate. So they arrange the bones in a certain order, sprinkle some nice roses and pebbles on them. And then they box it up. And it wasn't all bad. This was back when I ate meat. This is a fish. Well, it was a fish. And it was just delicious. Anyway, fast forward to 2014, and I'm diagnosed. And I said [inaudible] first commenter was, "John, we're just in a state of disbelief with your list of health hang ups. You're the last person that this should happen to." I agree, because since college days ended, since I met my wife and got serious about life, I tend to follow a fairly healthy diet regime. I certainly never smoked. Drinking was a thing of the past, et cetera. So, anyway, I find myself several surgeries, 27 lymph nodes, 36 radiation treatments and various chemotherapy appointments later, I find myself in this treatment program here. So here are the kids. And it was another everybody says is, "Oh, you're so brave." And I'm thinking, "Right. [inaudible] my choices are exactly?" So kind of as a patient, you're kind of along for the ride. And I was really actually measuring up the pine box, because I personally didn't expect to survive. I was, like, they say prepare for the worst and hope for the best. So there I was. And it's a bit like they did [inaudible] in the foxhole, the battlefield, when they hear the charge, and you can't see in the foxhole. You've got to advance. So this is a [inaudible]. So they locked and bolted me in in one these [inaudible] my little custom-made headset here where they bolted you in. And, of course, just what I needed after Chernobyl was more radiation. But, anyway, [inaudible]. And these are the fellows that helped. There I am after it. You see the sunken eyes and just thin as a bone. I mean, just absolutely -- just absolutely -- and the kids they [inaudible] the best they could. [inaudible] the Legos. It was actually [inaudible] where they took me away and where they put out the call for prayers, because John may not make it. There was blood on the wall where I'd walked into it, and I just had no memory of it. And I just -- it was [inaudible]. I just want to [inaudible] say cancer affects not just the individual, but instead the whole family. And it's the cancer patients that have a support structure in place. But the caregivers don't. So it really affects the family, really most of the family. The patient, himself or herself, is really just kind of along for the ride. And maybe I'll survive this and maybe I won't. But let's give it my best shot. You see, here's my youngest son. He's about 11, ten or 11 at the time. And he's giving me the feeding tube there. And here's my little guy. Evidently any fertility issues were overcome in 2010, the same week I joined the Library, coincidentally, it was a good week. When he was born on my 48th birthday. So, anyway, the kids would take [inaudible]. They wouldn't do this now, because the twins are 14 [inaudible]. But back in the day they would keep me warm and whatnot, because I was suffering temperature issues. This [inaudible] certificate. I barely squeaked through college, freshman-level chemistry. So I'm the least qualified person to grace the stage today. But, obviously, my talents lie in other areas. But of all these certificates and awards and diplomas and whatnot that are on my wall of shame, this is probably my hardest earned, was this. And it talks about a hero at the bottom. It says here [inaudible] it says, "Here was a person that takes personal risks through their own life to save others." And that never really occurred. I was just kind of in it for what I could get out of it, basically, surviving. Here's a little guy ringing the gong in the radiation oncology waiting room at the end of treatment. Okay. Now, 2015, this is another very important point. Recovery is way harder than treatment. Again, in treatment you're just along for the ride. You do what you're told. And you take what's given you. But recovery, you're, basically, on your own. That's when you have -- you're suffering this chemo brain, and you're sitting in meetings at work, and you just can't string two thoughts together. And it's like you can't formulate sentences. And it's just life is very difficult. And you're still smelling burning flesh from the radiation. And it's just getting these chemicals out of the body, because I really, like I say, [inaudible] vegetarian, you're drinking filtered water and never drinking bottled water. There's some hang ups a mile long. But the point is that I tried to run a clean ship, and suddenly, as the chemotherapist told me, he said, "You've been, basically, poisoned with the nastiest stuff known to man to get rid of this." This was all for a good cause. Here's the [inaudible]. This is, again, a staged shot after the fact. And the radiation oncologist said something to me at one point in the treatment, "You have to prepare yourself for a life after cancer." I just thought that's a radical thought. I'm prepared to die here, and you're talking about life after cancer. I can't see beyond the next appointment. So that was just a radical thought. And I took it upon myself to really make those extra 100 days or 200 days or whatever, providing you don't have a tube down the throat, providing they're mobile and [inaudible] and whatnot, it's worth it, so they can wring that extra little bit out of life. I don't mean to go all Joseph Campbell on you, the hero's journey sort of thing, but, really, that's what it amounts to. So, basically, I sprinkled this. I thought, "What do I do to keep [inaudible]. Photographs, sprinkle it with metaphors, various quotes and whatnot. Get my point across and it will all come together. Great." So here we are, me and the twins. The little guy he said he had to stay home with Mama because I knew from visiting [inaudible] before that he would not be able to navigate squat toilets. Here's the daughter busting a move here, she's a little yoga [inaudible]. There she is at the Taj Mahal. Little fellow. Okay. These guys, [inaudible]. Basically, they don't have any family or any job, and they just, basically, embark on this internal quest in life. And they don't have any sort of home. They're accounted for as homeless people. But they dress up in really bright colors, a lot of makeup, and they they smoke a lot of dope. I mean, a lot of dope. And they're constantly stoned. It's like Elon Musk says, "That's why they call it a stone, because you sit there like a stone." Okay. So, anyway, the oncologist's initial consult -- okay, now, they've already exhausted conversationally the introduction and the how do you do's and the qualification. Okay. Then the possibility of genetic treatment has been set aside, because that's not going to work [inaudible] conventional [inaudible] getting it. He shows me a laminated flow chart showing cancer type and stage, easily accepted surgery, radiology and chemotherapy protocol. So I thought, "Okay, right, we're going to do this the old way. We're going to cut it out. We're going to burn it out. And we're going to poison it. Right?" And I'm thinking, "Okay." So he shows me on this chart, and it was very comforting actually, you are here. You can expect trouble swallowing and talking here elsewhere on the chart. Since we can't operate during treatment, you'll need a feeding tube starting here. And I thought, "Oh, my God, you're not going to be able to swallow your food." So okay, great. I said, "I see." And I just -- I don't know, it seems like it's all in the news, this medical marijuana, miracle [inaudible]. I live in Virginia. It's going to be last state in the union that [inaudible] marijuana. But I said, "What about medical marijuana?" And he says, "Look, I don't want to be known as the doctor who prescribes pot." So it's all about reputation. And I get it. "And the last thing I need is a paranoid patient calling the office." And I tried to make the case here. I argued, "Look, I'm already losing weight. I understand Drabinol, I've done all my research, which every doctor hates probably. It stimulates the appetite. Because already my pants were falling off. So I had to drill extra holes in my belt. My co-workers said, "Hmm, you might want to get that checked out." Anyway, he said, "Rest assured, you'll get whatever you need to get you through this." So I found that very comforting. So here we are, morning sunrise on the Ganges, goats, interacting with the folks from [inaudible]. This was done on a budget, I want to emphasize, a backpacker's budget. We only flew to where there weren't roads, or where there were bandits, or 1,000-drop off cliffs or something. So mostly by train. Okay. Here we go. This was another quote, excuse me, "We're all dying. Cancer patients just have more information." And there may be some comedy in that, a little dark humor, I know, but it's true. You've got all the information in the world. You've got every test in the world coming at you and saying, "You can expect to live this long. This percentage, you've got all these stats coming at you." It's like, "Okay. I can -- ." It's the first time in your life, and your only time probably ever, you'll have that much insight into your own mortality. And here's some healing bowls where they're dinging in the sides. They call her the camel whisperer. This camel had some issues from being mistreated in the past. She was calming it down. The castle on the right was 3,500 years old when Alexander the Great unsuccessfully attacked it. Just to give you perspective timewise here. We're just here for a short journey. And whatever we can do to make that journey more worthwhile or extend the journey, make a positive impact on others, I think that's the name of the game. Okay. Food. I'm in the ENT's office mid-treatment, I'm emaciated, a total stick figure, can't consume enough to sustain weight. She receives the nurses records, she's reviewing them and showing substantial weight loss plus the current medications I'm on. First question, "All right. Who prescribed the marijuana?" I said, "Well, Dr. so and so, but he asked me not to broadcast that fact. I insisted on it. It was my idea. I was feeling that an appetite stimulant might help." She's a short-statured woman, but 100% no nonsense. She looks me in the eye, points directly at me, she's up like this. She says, "I want to see proteins, I want to see carbohydrates, I want to see fats." So she was trying to, in her way, fatten me up. [inaudible] stage. We didn't -- yeah. Okay. Okay, now, my thoughts on exploitation of elephants for tourist purposes have evolved since this. I wouldn't do it again. But they do, in fact, like being covered with chalk, and European girls rubbing their noses and whatnot. It's calming for them. Here are the kids at the end of it all. Okay. It's 2017, it's the little guy's turn. We go to Iceland. My first trip back to Iceland since the Chernobyl days. Blue Lagoon. Yes, that military cargo plane that crashed on a black sand beach in 1973. One of my yoga instructors said to me that you enter life taking a breath, and you leave life breathing, or breath out. And so everything in between is really a gift. Here's the little guy with the waterfall. [inaudible] there's [inaudible]. Okay. Thank you very much. Let's just end on a quote here. Do not regret growing older. It's a privilege denied to many. And thank you for extending my life and helping me out. I appreciate it. [ Applause ] >> Sandra Charles: Our panelists are making their way up to the podium. And John will join them. [ Inaudible Speakers ] So we've had quite a morning. And I think I delivered on my pledge. You've been stimulated, informed, entertained. And, really, it's been revealed to you just what an integral part each of the people represented here play in this whole journey, this whole Moonshot. This is trajectory that we are taking to overcome cancer. Time for questions and answers. I've heard so many things. And John sort of brings it home, because John and Juanita as patient advocates represent the why in all of this. Why are Dr. Tsang, Dr. Platz and Dr. Bates doing what they're doing? And these are the why. And so having heard that, and having heard the full plethora of things going on, anyone with a burning question before I pose one to the panel? Questions? Right here. Go ahead. Sure, please. [ Inaudible Speaker ] >> Elizabeth Platz: That's a great question. Is this on? Can you hear me? Okay. Terrific. If we think about ecology, and think about survival of a species, if you have all of the animals being exactly the same from a genetic perspective, and there's something catastrophic that happens, all of the animals could die. But if there's more variability in the characteristics of those animals, some may die, some may live. We actually think it's the same thing in the cancer cells. If they have different telomere lengths, they may have other changes that are correlated, and maybe some of those cells are more likely to live, which is a bad thing in the case of cancer, versus those that are more likely to die. And so we think it's related to that. And I should, I guess, be clear when I talked about telomere shortening and chromosomal instability, why is chromosomal instability important in the context of cancer? Well, when you have chromosomal instability, there's also higher chances of there being crossing over, exchanges of pieces of DNA, which then increases the risk of cancer becoming even more aggressive. So we think it's probably a marker. It may be involved in the cancer process, but, certainly, it's marking just more aggressive subsets of cells. Now, the stromal cell, a shorter telomere length, that's an interesting question, too, because it turns out that that relationship was independent of the cancer cell relationship. Together they give us information, even more information than either one alone. But when we look separately at the variability in length in the cancer cells and at the length in the stromal cells, they both provided independent information. So what is a hallmark of an aging cell that maybe a senescence? Shorter telomeres. Well, what is it about senescence stromal cells, for example, senescence fibroblasts that may influence cancer progression? Well, it turns out some of those fibroblasts, when they're senescent, they change what they elaborate, they change what they secrete. And in some cases they start secreting cytokines, which ordinarily immune cells would produce, but we're talking about fibroblasts now. And they might also secrete survival factors. And so if you have short telomeres in the stroma that could be related to senescence and this phenotype called the senescence associated secretory phenotype, which then acts on those cancer cells to make them even more aggressive. So it's a great question that you asked. Yeah. Thank you. >> Sandra Charles: And that sort of is underscored by a quote attributed to William Osler saying, "Worry not for the individuality of patients. Medicine is merely a science and not an art." And that is, an art. I saw another question back here. Barbara? [ Inaudible Speaker ] >> Elizabeth Platz: I'll take this one, too, if that's all right. So for many years we had a field called nutritional epidemiology, where we tried to identify individual foods, individual nutrients, and sometimes food patterns, that are associated either with an increased risk of cancer, or, more importantly, protection against the development of cancer. And I think we've come to a point where we think about more a general pattern of lifestyle as being potentially beneficial, not just for protection against cancer, but protection against other chronic diseases, like cardiovascular disease. So the general strategy that is probably appropriate for everyone is don't smoke. I know that's not food, but that's the number one cause of cancer. Don't smoke. The second one is avoid weight gain. And if you've already gained weight, lose weight. So many cancers are related to obesity. And, certainly, there's now emerging evidence that those who have cancer and are gaining weight during that timeframe -- now I know, John, you were telling us about massive weight loss that you experienced, but for some cancers that is not the typical situation. And so those who gain weight just because of dietary factors, have a higher risk of later recurring, even after they apparently were cured following treatment. So don't smoke, avoid weight gain, lose weight if you've gained too much weight. And then eat a well-balanced diet. And so the old story of everything in moderation is probably the right answer. Big trials that have tried to test individual supplemental nutrients, so selenium, vitamin E, have failed. Right. They haven't protected against cancer. Except in those who are truly deficient. So looking in subgroups, every now and then there are patterns that are found. But most people living in the U.S. are replete, they're not deficient. So making sure that your diet is very well balanced, eating fruits, eating vegetables. You know, the logic of eating colorful fruits and vegetables so you get a wide array of nutrients. Not overdoing meat. And, John, you said you're a vegetarian. There are many other sources of protein that are non-animal-based. And that's absolutely appropriate, too. Avoid eating, basically, empty calories. Right? You don't want to gain weight, so you want to make sure you eat foods that are providing nutrients, not just sugars. >> Susan Bates: I saw something recently, though, that said that "junk food," processed food was not good for you. >> Elizabeth Platz: So IARC, the International Agency for Research on Cancer, has concluded that a certain type of processed food, which is processed meat and red meat, is supported as a risk factor for colorectal cancer. So it's not to say that there aren't some specific factors that are agreed to, but, in general, we just want to make sure that we minimize the chance that we develop cancer, these sort of bigger picture strategies are appropriate. And, by the way, processed meat, do you know what processed meat includes? >> Sandra Charles: Bacon. >> Elizabeth Platz: What's that? >> Sandra Charles: Bacon. >> Elizabeth Platz: Bacon. Bacon, yeah. Lunchmeat. Right? Nitrates, nitrites. And then also there's some laboratory-based evidence, not so much human evidence, that meats cooked at high temperature may -- well, actually they do produce mutagens, so that's clear that they're mutagenic. But it's unclear if the amount that we typically eat of grilled food, meats I mean, cooked at high temperature, whether those are cancer risks or not. >> Juanita Lyle: From the patient's standpoint, John and I both shared the fact that I didn't smoke, I didn't do any of the things to get cancer. I had a very good diet, did all the right things, but I got cancer anyway. But one of the things that, because I'm now, from the first time that I had cancer, 42 years ago, stage 3 breast cancer, one of the things that my doctor told me is the reason why my survivorship is such after having cancer four times was because I had a good diet, I did fitness, I did all sorts of things. So diet does help. Even though you might get cancer, it does help your survivorship. >> Sandra Charles: Okay. Well, thank you. [ Inaudible Speaker ] >> Elizabeth Platz: Yeah, that is a good question. So that is an emerging area of research. So trying to understand how the microbiome influences cancer development, and then, given cancer, how that might influence progression in long-term survival. So probably the most studied location of the microbiome for men and women together is the gut microbiome. So, again, it's an emerging area. We don't have consensus yet. Typically, what's been looked at is the variety of bacteria that are present, as well as sort of the general amount of each type, or the proportions present. We don't have final details, like I said, yet for the gut microbiome and colorectal cancer. There's some, of course, information on the cervix. Right? We understand HPV, but not bacteria. Right? So a virus, we understand that relationship very clearly. But are there other bacteria that may be present in the [inaudible] that relate to risk? That's an area of study. For men, understanding the bacterial environment that may influence prostate cancer risk. That's an area of active study. But I'd say at this time we don't have clear evidence that the microbiome, in general, is protective or a cause of cancer. There's great variability in moment to moment in one's microbiome. And, typically, the kinds of studies that we do that are in large cohort-type studies, where you might have 50,000, 100,000 people in it, we, typically, only have the money to measure at one point in time to collect the right specimens and measure once. And, really, what you'd want is long-term exposure, not just in adulthood, but perhaps earlier in life. >> Sandra Charles: And talking about translational medicine, a lot of people have translated that information into overindulging in prebiotics and probiotics, and a number of different microbiome containing foods. So everything, again, in moderation. And we have to be sure that we aren't overdoing it just because we hear one story. >> Elizabeth Platz: That's right. >> Sandra Charles: Any other questions from the audience? [ Inaudible Speaker ] Please. [ Inaudible Speaker ] >> John Tsang: Thank you. Yeah, that's another good question. So I actually visited Watson a few years ago, and they showed me sort of, at that time, the most recent progress. And at the time, my understanding was that they, basically, ingested, right, and digested I suppose, too, knowledge-wise, a lot of the information in clinicaltrials.gov. So a lot of, basically, textural, natural language-based information sources. And then they tried to develop tools to help oncologists to make sense of the, for example, mutational profiles that you saw from Susan, because often they would get, basically, gigantic amounts of information for each patient, especially those who went through a lot of sequencing studies and other types of OMiCC's analysis. So the idea is to integrate the existing knowledge with this individualized information, basically. And I think it made some good progress at the time, especially in terms of reducing -- sort of shrink the space through which the oncologist had to look through in terms of the information. Right? But I think the major challenge remains, which is that a lot of those gene mutations, and also expression profiles -- and we don't have much information out there yet. So drawing on existing databases and existing publications on what these complex profiles could mean, it's through a daunting challenge, basically. And especially in terms of combining them, because you may look at one mutation and look at a couple of mutations, and you can say, "Well, this looks like it may predict this." But together, how they all give you information on how the patient will do in terms of therapy and response to therapies, it's still very difficult, basically. So I think we need a lot more data, in a way, to start to discover some of the associations, starting with associations, and then eventually getting to, hopefully, causation as well. Yeah. >> Susan Bates: And I would just add one thing to that. That was a very good answer actually for that. The problem with the mutation profiles is that it's quite variable. One cancer type will have anywhere from 20 mutations per tumor, to another cancer type may have 300 or even 3,000. So when you have that, you have to say which of these are relevant. And you really have a difficult time figuring that out. The pancreatic cancer example I gave you where we have a founder mutation from the very beginning, that's highly likely to be important. But it doesn't mean that when we have a drug for it that that will work, because by then you've accumulated other changes in the cancer. Every single time you make a discovery, you have to validate it in the clinic with a drug. And so it becomes a very time and long process. You can make a prediction, but then you have to be able to target it to make a difference. So that's why it should be a Moonshot. That's a great way to think about precision medicine, including immunology. >> Sandra Charles: And I wanted to direct a question to you, Dr. Bates, because pancreatic cancer, though the numbers are swollen in terms of incidence, it is a growing incidence of pancreatic cancer. And maybe, particularly for our audience, the patient advocates, we're getting feedback from patients who are survivors. How can we help in terms of improving the database that you have to do some of the research that you're doing with pancreatic cancer? What things can help and maybe stimulate funding of that $50,000? >> Susan Bates: Exactly. That's a really good question. I mean, I think it's important for patients of pancreatic cancer to try to go on clinical trials. That's been something of a challenge, because many of the -- I showed you two lines of therapy. What we don't have is a third line of therapy. So patients will be still fit enough for a new treatment at the end of the two lines of treatment, and we don't have one. So that's the point in which we need more and different clinical trials. But it's difficult to get patients onto clinical trials. There's a movement to try to improve the eligibility criteria, to decrease how tight it is. >> Sandra Charles: Right. >> Susan Bates: Many patients have problems with liver test abnormalities, or they were getting a treatment just last week, and don't really want to wait six weeks to go on the next one. So there needs to be more work done to make it easier to get cancer patients of all types, but, in particular, pancreatic cancer on clinical trials. You need to be able to pick up the phone, and have them on a trial the next week. That's a huge unmet need. >> Sandra Charles: Is there any ongoing work at any level to enhance that? >> Susan Bates: To? >> Sandra Charles: To get patients into these clinical trials? >> Susan Bates: Yeah. I mean, well, there are clinical trials everywhere. >> Sandra Charles: But in terms of bringing them in, and -- >> Susan Bates: Yeah. >> Sandra Charles: Informing the community at large? >> Susan Bates: I mean, NCI has developed a lot of platforms where you can search for clinical trials, clinicaltrials.gov -- >> Sandra Charles: Right. >> Susan Bates: Has gotten very good. And I actually tell patients to start looking at it, start lining up what we're going to do next. That would really be the best thing. I think that information is out there. It just requires some energy to come up with it. >> Sandra Charles: Yes. I've been struck by just reading about translation medicine. And it struck me that what you said about immunotherapy, because, of course, that is a thing now. Immunotherapy's supposed to do it all for almost anything. >> Susan Bates: Incorrect. >> Sandra Charles: And clearly that's not always the case. And one of the things I was reading also was from, I think it was the Microbiology Society, which said we're doing all of this with translational medicine, but we cannot forget basic science. We cannot forget those foundational pieces that lead you to do the research. And so that sort of resonated when you made your statement about dealing with the small molecules and not just with immunotherapy. >> Susan Bates: Yeah, that is a flaw of medicine is that we tend to -- when something works, it's not medicine. It has to do with profits and it has to do with where people -- investors don't want to invest in failure. So they will invest in companies that are more likely to make the second of the drug that already worked, than they are to invest in something that's completely unknown potential. And if there were one thing we could change about the research paradigm, it is so much driven by investors and by pharmaceutical companies. So that's why I showed you that list of ten different [inaudible] -- >> Sandra Charles: The same -- >> Susan Bates: Inhibitors. >> Sandra Charles: Area. >> Susan Bates: We do not need that. We have, I think, four [inaudible] FDA-approved, maybe five. So somehow that paradigm needs to change. But that's virtually impossible to keep people from investing in what was already proven, already proven therapies. >> Sandra Charles: So for any young investigators and researchers in the audience, you heard it. Go ahead, please. [ Inaudible Speaker ] Please. [ Inaudible Speaker ] >> Susan Bates: You mean radiation therapy? >> Elizabeth Platz: [inaudible] radiation. Yeah. Do you want to -- sorry. You go ahead. >> Susan Bates: No, I mean [inaudible]. >> Elizabeth Platz: We'll both do it. So I'm an epidemiologist. So I'm not an oncologist. I'm not a urologist. So it depends on the patient's situation. So it may depend on how many cores were positive. So, typically, 12 cores are taken. If only one core is positive and only a tiny percentage of that core is cancer, and the morphology, typically called the Gleason score, if the Gleason score is low and the man's older, he and his provider may decide to do something called active surveillance. If the man is young, maybe he's 50 years old, he and the provider might make a different decision. That man may not want to undergo active surveillance. That man may want to have a prostatectomy, with the idea that the surgeries done with curative intent. If more cores are positive and a greater proportion of the core is positive, and the Gleason sum is higher, which is a poorer prognostic, especially if that man is an older adult and he has many comorbidities, that man and the provider may together discuss radiation therapy. So it highly depends on the clinical situation of the patient, the man's own values, and what he cares most about for his other aspects of his life. So it's not is it prostatectomy or is it radiation therapy. There's far more that goes into the conversation and decision. >> Susan Bates: And there are clinical trials currently. They're open where you can enroll and be randomized, one versus the other, or you can choose and then the data is collected versus patients show undergo surgery. >> Sandra Charles: Okay. Please. >> John Bauer: Is there international sharing of information with other countries who may have a different funding paradigm for research that you can build on? >> Susan Bates: There's a lot of international sharing [inaudible] a lot of other countries, because they have a more centralized healthcare system, actually do a better job than we do of developing clinical paradigms that ought to be followed for treatment. >> Sandra Charles: Well, one last question and we're at the end of our program. >> Juanita Lyle: Now, I would just like to say about clinical trials, the good of clinical trials, because I was placed in a clinical trial back in 1977. And there were 814 of us women who had breast cancer. And there is probably less than two-thirds of those women who are now living. And in that clinical trial, one of the things that made me go and do the clinical trial was simply because you're followed so closely. And a lot of people had the fear of going into a clinical trial, but it's a bigger benefit to going into the clinical trial rather than just going as a patient, taking all of the chemotherapy and doing radiation, because it's a powerful situation where you can actually talk to your doctor on a level that's totally different, I think, than if you're not in a clinical trial, because everything is being tracked so closely. So I would say for anybody here who is in that situation, clinical trial is a real benefit to the patient. >> Tomoko Steen: We get a lot of questions about the clinical trial possibilities by cancer patients at the Library of Congress. And often the question comes to me. So over 30 years I was dealing with cancer patients as a clinical pharmacologist. And I didn't think this progress is coming. And I'm so excited about the new change and the future progress. Thank you so much for coming. [ Applause ]