>> Ashley Cuffia: Thank you for joining today's Library of Congress Cancer Moonshot webinar on Immunotherapy and Beyond. Throughout today's session, we invite you to submit questions for our presenters using the Q&A panel on your screen. We will address these at the dedicated question and answer segment at the end of the webinar. If we're not able to get to your question, or if you have further questions after today's event, please send them to Ask a Librarian on loc.gov. Today's webinar is being recorded and a captioned version will be made available on the Library's website in the coming weeks. We will share this information again before we wrap up today. So with that, to get us started, please welcome the Chief Medical Officer at the Library of Congress. Dr. Sandra Charles. >> Sandra M. Charles: Good afternoon, everyone. It is a pleasure to welcome you to the Library of Congress for this sixth panel discussion in our annual Cancer Moonshot series, co-sponsored by the Library's Health Services Division, and the Science, Technology and Business division. This panel comes at the five-year mark from our inaugural discussion held just after the passage of the groundbreaking bipartisan 21st Century Cures Act proposed by then Vice President Joseph Biden. That act authorized seven years of funding for the Cancer Moonshot Initiative, aiming to improve access to therapies and through collaborative efforts and sharing of data in research and innovation, facilitate progress. Today's discussion will focus on cancer immunotherapy, and bring to us several leading scientists, patient advocate, and clinicians in the field. In the US and globally, cancer is a second leading cause of death. Few people on the seminar can say they have not been touched personally by cancer. Immunotherapy is an exciting and ever-expanding approach to treating cancer and this discovery and development came about rather serendipitously as curious minds were trying to better understand the immune system. We look forward to hearing from our distinguished researchers and clinicians to hear how the attributes of the immune system are being harnessed to provide life-changing therapists. One of our key objectives here at the Library is connecting ordinary people and patients to experts, scientists and clinicians, who can further the understanding of cancer diagnoses, diagnostics and treatment choices. The Library is indeed fortunate to have access to researchers and health experts who are willing to share their knowledge and cutting-edge information with us in order to promote health literacy. I now turn the podium over to my colleague and co-sponsor, Dr. Tomoko Steen to introduce our first distinguished panelist. Dr. Steen. >> Tomoko Y. Steen: Thank you. Actually, next slide, please. So we have five different speakers and they are all top experts in our field, and especially, we are looking at the most recent advancement in immunotherapy, so we are very excited about this. First speaker is Dr. Danielle Carnival from White House. She was the initiator for this important bill, and also we have two original 2016 Cancer Moonshot the Library of Congress Cancer Moonshot speakers, Dr. Weiner, who is the winner from Georgetown. He was one of the speakers and he is going to talk about the basics of the immunotherapy and also as a clinician, he can share lots of information. And we have a Nobel Laureate and the discoverer of this, you know, important therapy, Dr. James Allison from the Houston UT Medical School is joining us, and we also have another expert from the National Cancer Institute, Dr. Giorgio Trinchieri. He's going to talk about some fine tuning of this therapy and how we can make it more effective, you know, even better treatment for this immunotherapy. And then we have a cancer advocate and the original speaker also from 2016, Dr. Sigel, and then Sigal is going to conclude the set of speakers and she is going to share with us, you know, how we just connect this wonderful, you know, new therapy to the patients. So can you send me next slide, please? So first speaker is Dr. Danielle Carnival. She worked from original development of this theory, as I said, working under Dr. Biden, administer Biden, and she was the main, you know, architects of this important bill and but she also worked on not only many years at the White House, but also she worked at the ALS, I Am ALS, organization, you know, looking at patient and doctor and the scientists just reaching among them, so I was told not to give too much details. We have a handout coming out, in a virtual handout, so everyone attended this meeting, you can get the virtual handout, so before further ado, I'd like to give the floor to Dr. Carnival, please. >> Danielle Carnival: Thank you, and thank you so much for that introduction. I will amend just not to offend the people who were the architects of the 21st Century Cures Act, I will take credit for helping to frame the Cancer Moonshot here from the White House, but leave it to those in Congress who worked long before we had started on the Cures Act, but we were really excited that we were able to, you know, be there at the finish line and to make a big impact on that bill for people living with cancer, and I'll talk a little bit about that, but thank you for having me, and thank you to the Library of Congress for bringing together the knowledge and speakers on this agenda this afternoon so we can all learn about the amazing impact that immunotherapy is having in the treatment of some cancers, the difference it is and can make for millions of patients. The framing for today as have been mentioned is cancer moonshot, and I want to spend a few minutes just setting the stage for that conversation. To step back a little further, 50 years ago this month, in December of 1971, Congress passed the National Cancer Act, creating the cancer research system we have today in the United States led by the National Cancer Institute. The researchers and clinicians on this call can tell you more, but we learned a great deal over those 50 years. First, that cancer isn't a single disease, but in effect, we have already identified hundreds of distinct types of cancer, that cancer is influenced by our genes, and usually involves a gene changing or mutating in a way that allows the cell to replicate out of control. Because of this, we now know that we aren't going to develop, identify a cure for all cancer, but we have and will develop effective treatments for many types of cancer. Dr. Allison and the panel will go into that in further detail later. I mention this to provide a glimpse into the history of how we got here today, and to go to five years ago when, as others have started to mention, President Biden when he was Vice President, I lead a national focus to make progress against cancer called the Cancer Moonshot. The goals were to inject urgency to our collective approach to end cancer as we know it, to bring all possible solutions and partners to the table with ideas and action, to provide an avenue to hear and prioritize what patients and caregivers and survivors identify as being the most important, change to change the systems and cultures to culture to meet this moment. It wasn't the situation of 50 years ago. We had learned so much and we needed to build on that knowledge we had gained and to really have a 21st century approach to how we deal with cancer of we had gained all this knowledge over decades of research and treating patients, and all of this to deliver on the hope felt by millions of families facing a cancer diagnosis. We brought a national focus on cancer, you know, gathering together all parts of government to do their part from new studies on cancer disparities, new trial networks to drive drug discovery, and new projects on childhood cancer, to addressing prevention and early detection and making sure everyone has access to the resources they need. As part of the Cancer Moonshot, Democrats and Republicans in Congress came together and passed the 21st Century Cures Act in December of 2016, which delivered $1.8 billion over seven years. This enabled the National Cancer Institute to invest in new priority areas of research that hundreds of researchers, clinicians, patients and other members of the ecology community, identified as having the most promise and it streamlined cancer-related decision making at the FDA. A line can speak to this through the Oncology Center of Excellence so that cancer drugs can be approved faster and patients can have direct input into understanding how the process is moving forward. In addition, as part of the Cancer Moonshot, companies, patient groups, universities and foundations launched more than 70 new programs and collaborations, all to make progress to prevent, detect and treat cancer more effectively. As the oncology community spends this year commemorating the 50th anniversary of the signing of the National Cancer Act, and five years since the signing of the 21st Century Cures Act, it has been a year of reflection in many ways. Cancer mortality rates have been declining over the past 25 years, and the rate of that decline has been accelerating. We have more understanding of the underlying mechanisms of cancer, as I mentioned, and will go into further later, including genetics, cancer genomics and the interaction with the immune system. We have more tools to prevent and detect cancer. We have increased outreach to medically underserved communities, rural, urban, tribal, to address gaps in access to care, and although many factors contribute to this trend, more effective treatments are definitely a driving force in recent years, led by many immunotherapies. However, as we all know, there's so much still to be done. We have remaining issues with how we know cancer today. President Biden has said his strong commitment to making progress towards any cancer as we know it, and to be able to do that, we want to make it very clear how we think we know cancer today. It's a disease for which we don't have enough tools to prevent it and don't effectively use all the ones we already have. As a disease for which we don't routinely detect it early enough when it is most treatable, and often with less invasive treatment. We have inequities in time to diagnosis, access to treatment, trial, care and outcomes based on who you are and where you live. We know more about the genetics of cancer than ever before, and yet we are not fully at acting on familial or hereditary risk, and providing people with the tools they need to live as long and healthy a life as possible. We lack some of the -- we still -- excuse me. We still lack the solutions for some of the toughest, deadliest and rarest cancers, and cancer is a disease for which we don't have the systems to learn from every patient or the system to share data and knowledge widely. The good news is that President Biden knows cancer as well. He knows it personally, and he is taking it on as a national and global leader in making urgent progress for people facing this disease. As he has said, he's committed to do everything in his power to make progress toward ending cancer as we know it. He has already made a down payment on that by proposing strong funding for NIH, the National Institutes of Health, the National Cancer Institute, and in proposing a bold new approach to taking on some of the remaining issues facing biomedical research and those living with a disease like cancer in an ARPA for health, ARPA-H. This administration has supported more people getting quality and affordable health insurance, and but we know we still need to make strides to ensure that everyone in the United States has the best in prevention, early detection, treatments, trials and care through survivorship. We all have a role to play and as we organize the administration's response to this mission, we will need your ideas, your action and your change. One area with some urgency is in cancer screening and early detection. Nine million -- more than 9 million -- we have a deficit of more than 9 million screenings over the course of the last few years driven by the pandemic. We can all do our part to get those screenings back on the books personally and from our seats in foundations, companies, health providers and policymakers, we must stand up to make sure that people have the knowledge and access to detect cancer early. President Biden has a bold vision for how to make progress against this disease, progress that can continue to bring hope to every family facing a cancer diagnosis. We will drive a new understanding that we can lead to ways to prevent detecting treat cancer, and we, as I said, must deliver more equitable access to quality care, bring more personalized and less harmful effective treatments. We must take on cancers today for which we don't have an answer, including some childhood cancers, and we must all learn from the perspectives and experiences of patients and caregivers, looking to them as the experts they are. As we look at the five-year anniversary of the signing of the 21st Century Cures Act, I just want to reiterate this administration's commitment to this mission. We will take on each of the ways in which we know cancer today and make concerted progress toward any cancer as we know it together, so thank you for the opportunity. >> Tomoko Y. Steen: Thank you so much, Dr. Carnival. That's very wonderful outlines of some administration's efforts. Next slide, please. So next speaker is Dr. Louis Weiner from Georgetown. He is a Francis L. and Charlotte G. Gragnan -- sorry -- Gragnani Chair, and he's Director of Lombardi Comprehensive Cancer Center, and he's Chair and a professor at the Department of Oncology at the Georgetown Medical School. And he's also director of the MedStar Georgetown Cancer Institute, and he's -- he was, as I mentioned, the first speaker 2016's Cancer Moonshot, and he is really making an effort to, you know, pass on this information to the public, and we can distribute his detailed bio after this event, so before further ado, Dr. Weiner, floor is yours. >> Louis Weiner: Thank you, Dr. Steen. It's a pleasure to be back here again to speak with everybody, to be on a panel with so many friends and colleagues, and to share with you some of the most important foundational elements of what has really become an important way of treating cancer and understanding cancer. So I'm going to be talking about immunity in cancer, and with my good friend, Dr. Allison following, I'm not going to talk about immune checkpoints in any great detail in my presentation, leaving him to share that information with you, but rather to provide you with a broader context so you can understand what the field is really about. Next slide, please. So traditionally, there had been two ways to think about how you can kill a cancer cell. In the 1940s, we began developing chemotherapy drugs that attack what you might think of as the bricks and mortar of cancer cells, such as DNA, the cells' cytoskeleton, the basically the girders that are keeping cells going. In the '90s and early 2000s, we began developing targeted therapies, the so-called smart bombs, the idea being to attack the electrical wiring system of cancer cells, their receptors, their enzymes, and their cell signaling molecules, and the idea was that this was going to be far more selective, and much easier for people to take, because you wouldn't have some of the same challenges associated with nonspecific killing mechanisms. Next. The problem is it's hard to know what target to go after. This is a 20 year old paper from Hanahan and Weinberg, which just takes a very, very simplistic view of the complex signaling that goes on inside of a cell, and these are all things that can be disrupted when people develop cancer, and so it's a legitimate question to ask what target is it that you would want to go after? And how could you be sure if you went after that target, would it be enough to kill the cancer cell? And in fact, we learned over painful experience with targeted therapies, that resistance to the treatment that you're going to use is nearly inevitable, because there are so many escape hatches that cells can use through these various different signaling mechanisms, that it's very difficult to use a single drug that will be able to control the cancer for the natural lifetime of the person receiving the drug. There's occasional exceptions, and they're great when they happen, but unfortunately, resistance is nearly inevitable. Next slide. So if that's not complicated enough, all these different signaling pathways that I was just showing you are really part of a larger ecosystem, and you can see this follow up paper that was written a decade ago by the same authors. They talk about sustaining proliferative signaling, evading growth suppressors, and you can see this whole wheel of fortune around here. Note that in this particular iteration, they finally began to recognize the power and importance of inflammation in the immune system, and I point out here that they talked about avoiding immune destruction, how inflammation of tissues can promote cancers, and finally, that the actual DNA of cancer cells might be unstable in certain ways, and there might be mutations that are driving many of the malignant events that we see. Next slide. Now, let's take a step back a couple of hundred years to the late 18th century, to the beginning of immunotherapy when a Dr. Edward Jenner, a Scottish family practitioner, observed that milkmaids who got a very mild viral disease called cowpox, which is caused by the Vaccinia virus, a commonly used virus in our cancer therapy strategies, did not get the deadly disease smallpox, and so he did something that you couldn't do in the 21st century. Without IRB approval, he conducted a non-randomized, non-controlled clinical trial, and what he did, he actually took the pustules from a milkmaid who had cowpox, and he inoculated himself, and challenged himself to see if he would get smallpox and he didn't. And emboldened by that experience, he began using these cowpox sore contents to basically vaccinate people who did not get smallpox, and that was the birth of the smallpox vaccine, and the idea that the immune system itself might be able to control the outgrowth of certain diseases. Next slide. And this has led to development of many vaccines. I know some of this is controversial for some, but on the left, you can see the impact on cases, and by inference, survival, for polio, measles, mumps, rubella, diphtheria, and tetanus. In each case, immunotherapy was applied in the form of vaccines, and lives were saved and diseases were minimized, and on the right, you see one particular example from the recent SARS-CoV-2 outbreak that we're still experiencing, demonstrating that individuals who do get vaccinated have far less in the way of clinically symptomatic virus infections than do people who were vaccinated with a placebo control. Vaccines save lives. Next. So vaccines usually work in those contexts by eliciting the production of antibodies that can be used to control these various viruses. It doesn't work as well with cancers. Antibodies have been used and they are widely used to treat human cancers, because they're able to be designed now through recombinant antibody engineering techniques to target very specific molecules on the surface of cancer cells, as you can see here, and this is an older slide, and I've stopped trying to update it because the approvals are coming so fast and furious now, that we can't keep track of them all, but about 25 different drugs and antibodies were approved in the last year alone to treat cancer. What you can see here are cancers such as non-Hodgkin's lymphoma, breast cancer, certain kinds of leukemia, colorectal cancer, head and neck cancer, kidney cancer, low grade leukemias, and a variety of breast cancer again. Many different diseases can be treated by unconjugated antibodies. These are just antibodies that attach to the surface target on the cancer cell, and that does something that makes the cancer cell get killed. Next slide. And here, you can actually attach toxic molecules, so-called magic bullets to the antibodies, fulfilling a vision that was articulated first by Paul Ehrlich, a famous physician-scientist in the late 19th century and early 20th century, who hypothesized that you could create toxic molecules that would be attached by things like antibodies to eliminate human disease. And these drugs that I'm showing you on this slide, are actually the embodiment of a century-old dream that a brilliant physician-scientist had. And here you can see that there are drugs that can be conjugated to various toxins, antibodies to treat acute leukemias, lymphomas, and breast cancer, and the breast cancer drug and its derivatives have had very important impacts on the management of patients with advanced metastatic breast cancer. And finally, I told you I wasn't going to talk about Dr. Allison's work, but with all homage to the work that he and many others have done, there's a whole list of checkpoint antibodies that have been developed. These are the ones that target PD-1/PD-L1, and you'll hear a little bit more about the CTLA-4 antibodies shortly. Next slide. So these antibodies, as I just described, they can work in a variety of different ways. They can mess up the signaling, the electrical wiring system of the cancer cells that they're targeting. They can also target the cells for destruction by elements of the body's immune system, so-called antibody-dependent cell-mediated cytotoxicity, or complement-dependent cytotoxicity, or they can simply modulate the immune system so the immune system will know how to attack the cancer cells more effectively, and they can deliver a variety of different kinds of toxic payloads, but antibodies can also be used to attack, not the cancer cells, but the normal cells in the area where the cancers are, in the so called tumor microenvironment, and they can block a whole bunch of different functions in the microenvironment so that when cancer cells corrupt the local area around them in order to support their survival and metastasis and invasion, these things can be selectively interrupted by a variety of different drugs. Next slide. So there's a limit to what these antibodies can do, and in fact, most scientists would agree that the most important immune population of cells that are responsible for the control of cancer are T cells of the immune system, the cell population that is disrupted when people have immunodeficiency, such as those that are caused by HIV. And mutated proteins in the cancer cells caused by the genomic instability that I was talking about earlier, can represent potential tasty tidbits that can be attacked by the body's immune system. They're known as antigens, and on this slide, which now is a an eight year old paper that has really become a classic, what you're seeing here are the numbers of mutations in each different kind of cancer when you sort of catalog them using the Cancer Genome Atlas, a National Cancer Institute-initiated process that allowed us to get an extensive encyclopedia of various cancers and mutations and other abnormalities. And you'll see here that melanoma, on the far right, has the largest number of mutations, so that becomes important when we think about how certain kinds of T cell activating treatments might work. Next slide. So because of these mutations, cancers possess innumerable targets and decoy targets, and have a capacity to survive targeted attacks, and that led to the question, do cancers result because our bodies aren't able to see these things, or are cancers actively thwarting the development of anti-cancer immunity, or is there some kind of combination of the two? Next slide. And in fact, we get a hint from -- if you just look at a cancer under the microscope. It's chaotic. All the normal organization of the organ is destroyed by the cancer. You have disordered blood flow and disordered vascular distribution in the cancer. You have a lot of fibrous tissue, scar tissue, that makes it hard for cells to move around and do what they want to do. And in fact, it's become quite clear that tumors go to great lengths to evade or subvert the immune response, because as they're developing, it's the only thing that is attacking the cancer is the body's immune system, and so therefore, the cancers have to develop evasion strategies and ways to defeat the cancer. Next slide, and you can see this in clinical samples. This is I'm not going to go through what the pictures mean here, but this was a study done by a French group about 15 years ago now, where they were able to demonstrate that if you just take a look at the kinds of killer cells of the immune system, the T cells and their various subsets, and you characterize them in different early stage colon cancers, and ask, so who did well, and who didn't do well, more important than the conventional clinical staging that we do, which is based on the size of the tumor, the number of lymph nodes and other factors like that, the immune infiltration of the cancer was the most important predictive variable. The more immune infiltration you had, the better the cancer was going to do, by and large. It was a very powerful independent predictor, and that's really important. Of course, I also will point out that despite this powerful immune reaction, people were still getting these cancers. Next slide. So the challenges are that cancers look like self. Even if you have a few thousand mutations, that's a few thousand mutations out of literally billions of different kinds of molecular targets that might be present in a given cell, so the cancers look like self. So the question becomes, is there a way to effectively and safely break what we call immune tolerance to self? In other words, if you were to attack self, you would actually hurt self, and that would be normal organs and normal tissues, and you don't want to do that. Next slide. And that brings us to something that Dr. Carnival mentioned earlier, which is the National Cancer Act of 1971. This is when the war on cancer was declared by President Richard Nixon, but -- next slide -- we have been at war against cancer throughout human history. This is a photo of a grave that was exhumed from somebody who died in medieval times, who was a male, and it looks like that person had a sarcoma, a bone tumor, probably a young male who died, and the battle was not between -- next slide -- society and cancer, doctors and cancer. The primary combatants in the war on cancer is one person at a time and it's a malignant cell population against the host's own immune system. The host immune system is the dominant active enemy that's faced by a developing cancer, and every successful cancer must solve the challenges of overcoming these defenses. Next. And we can help. As physicians and as scientists, we have developed a suite of tools that allow the body's immune system to overcome some of these defenses that cancers have erected even after they're established, and you see here a list of treatments and diseases. The single asterisk then are those situations where treatment can be curative as a single agent. The double asterisk are where you have a long-term remission, maybe too soon to know if there are cures. And the triple asterisk is when you combine these drugs or these antibody immunotherapies with chemotherapy. And the list of diseases that we can now treat and sometimes cure with immunotherapy is really impressive and getting larger, bladder cancer, kidney cancer, melanoma, a form of skin cancer, lung cancer of some types, Hodgkin's disease, bladder cancer, head and neck cancer, cancers of the thymus, and there are a variety of other strategies that can be used, but you'll note that the checkpoint antibodies, the PD-1 antibodies, and also the anti-CTLA-4 antibodies that are frequently combined with PD-1 antibodies have an expanding role, and it transformed the landscape of how we think about immunotherapy and treating cancer. Next slide. As we think about where we go from here, because only about 15% of the people we treat with these immunotherapies are going to have the kinds of fabulous responses that we all want them to have, and when they happen, they're dramatic, and they're remarkable, and I've had them in my clinic, and it's just like nothing -- it's like nothing you can imagine as a physician to treat somebody and see their cancer melt away, because you basically didn't treat the cancer by trying to kill the cancer cells. You treated the immune system so that the immune system could eliminate the cancer. But as we move forward, we're going to have to think about the resistance phenomenon even here and how we begin developing novel combinations, and when we do that, we have to think about how cancers get -- can overwhelm the body's immune response by out-proliferating it, racing ahead too quickly. That doesn't happen very often, but cancers do know how to hide. The things that are necessary for the immune system to recognize the cancer are frequently eliminated or lost or diminished in cancers that are evading immune recognition and response. Sometimes cancers will subvert the immune response by elaborating molecules that suppress the body's immune functions, so that the immune cells can't do their jobs and kill the cancer. Other times, the immune system will be shielded from access to the cancer because the cancer elaborates molecules that don't necessarily turn the immune system off, but rather, they simply will make it hard for the cells to migrate into the cancer. And finally, when all else fails, the cancer cells will erect a molecular picket fence, if you will, around themselves to protect themselves against the attack by the -- by T cells of the immune system, and those are those immune checkpoint antibodies you'll be hearing about shortly from Dr. Allison. Next. I would be remiss in not recognizing the promise of so called CAR T cell therapy and other cellular therapies, and again, a very complex slide but what I want you to remember from this is that you can take cells out of the body, you can manipulate them genetically so that they will now express the ability to recognize and attack a cancer, and then you can use these cells as drugs essentially, to go in and invade into the cancer, recognize the cancer and destroy it. This is a very powerful new technology, and I think many of us in the field, myself included, believe that cellular therapy represents one of the next great opportunities for improving the ability to control the development or the sustenance of cancers using the tools of the immune system. Next slide. So finally, I used to have this slide titled cancer immunology is the next great frontier of cancer research, and I don't think that's true. It's the current frontier. It is the great frontier of cancer research, both to treat cancer, and maybe as we understand how to do this more safely and in a more targeted way, I think it may be possible to prevent cancers this way, using -- if we can identify people at high risk, and then basically use the immune system as a very specific probe in order to essentially get rid of the pre-malignant clones as they're developing. And with that, I'll stop and I thank you for your attention and look forward to the rest of the presentations. >> Sandra M. Charles: Thank you so much, Dr. Weiner for your clear and thorough description of how immunotherapy has been harnessed to provide a variety of cancer treatments, and how it differs from other modalities. We've always enjoyed your presentations and the hope it provides for so many who have different cancers. And I hope we will have you back again. Next, I would like to present to us our next speaker, Dr. James Allison. Dr. Allison is the Regental Chair and Professor of the Department of Immunology, the Olga Keith Weiss Distinguished University Chair of Cancer Research, Director of the Parker Institute for Cancer Research, and the Executive Director of the immunotherapy platform at MD Anderson Cancer Center. You will get a copy of his bio, but I'd be remiss if I did not at least say he has spent a distinguished career studying the regulation of T cell responses and developing strategies for cancer immunotherapy. He earned the 2018 Nobel Prize in Physiology or Medicine, which he shared with Dr. Tasuku Honjo, for their discovery of cancer therapy by inhibition of negative immune regulation. And one of his most notable discoveries was how the T cell receptor structure and another structure were blocking the activation of the killer T cells, and through their work and their -- in individual labs, but certainly in his lab, he paved the way for the emerging field of immune checkpoint blockade therapy for cancer, and that work led to the development of the drug ipilimumab, which is the first immune checkpoint blockade therapy approved by the FDA and is used among other things in the treatment of melanoma. So without further ado, I will introduce to you, Dr. James Allison. Dr. Allison. >> James Allison: Thank you very much, Dr. Steen, for that kind introduction. Today, I'm going to give you a little bit of a description of how we came up with the idea of blocking immune checkpoints to treat cancer, and then sort of show you what's going on now and show some work that we've been doing at MD Anderson with Dr. Padmanee Sharma to really discover how to extend the therapies to more kinds of cancer and make them more effective. I'll show my -- Go to the next slide, please. These are just disclosures of myself and Dr. Sharma. Next slide. This work-up, it shows mostly done by a number of people over the years but as I said, some of it more recently and I'm going to show us about with Dr. Sharma and fellow Sumit Subudhi at MD Anderson. The next slide just shows the story of what we're talking about. This is -- I've always been interested in T cells since I was in graduate school and they were discovered, particularly in the possibility that we could use them to treat cancer. It was a daunting task because we didn't really understand that much about how -- what regulated T cells at the time. The left panel of his shows the simple idea was that T cells recognized an antigen on an antigen-presenting cell or a tumor cell and then would kill it, but we realized by the -- and we worked out the structure of the T cell receptor in the early '80s, but quickly a number of people realized that it's the T cell receptor signal by itself was not enough to activate a T cell. It needed a second signal called a co-stimulatory signal provided by a molecule called CD-28 that we showed on T cells, when combining with a molecule called B-7 on very specialized antigen presenting cells, would provide all the signals necessary to tell a T cell to expand. To put this in context, you've got an enormous repertoire of T cells, probably in the order of 10 to the 10th, or so, you know, tens of billions, but the problem is that you need an army of them to attack a tumor or attack a virus infection, but you only got, because of this high diversity, you've only got a few of any given clone, and so you have to rapidly expand that clone to get the army generated to go out and, you know, do whatever it has to do to eliminate the virus or kill the cancer cells, and so that practice -- that regulation has to be -- the proliferation has to be tightly regulated, so that's why you have these two signal requirements. Now as it applies to cancer, I'll get to in a moment, but basically when a T cell gets those two signals, it makes a lot of stuff and it makes growth factors such as IL-2 that tell it to proliferate almost as fast as bacteria, so you go from just a few dozen, perhaps, T cells of any given specificity to hundreds of thousands within a matter of days to ward off that problem, but you have to stop that process, and it used to be thought that was just a passive process to the T cells undergoing a process called activation induced cell death, but it sort of ran out of steam, but we discovered a molecule along with that bit of molecule which we previously discovered, along with Jeff Bluestone called CTLA-4, highly analogous to CD28, very similar in structure, is turned on when the T cells get that CD28 signal. A way of looking at it is that the T cell receptor engages antigen along with C28 given a co-stimulatory signal, you get this go program, shown in green. It's started by CD28, but when the [inaudible] that also turns on another program mediated by CTLA-4 that accumulates and turns off that proliferative program and stops those T cells are proliferating, and we showed that by showing that if you eliminate the gene for CTLA-4, the stop -- the T cells lose that ability to stop their immune responses, and the T cells just keep going and it's lethal. Your immune system is killing you, so that's the role of CTLA-4 is to stop immune responses, and this was shown by [inaudible] as well as ours, so you have to restrict that proliferation eventually, and as part of normal regulation of the immune responses, but what does this have to do with cancers? Well, when we discovered this -- next slide -- and apply this to cancer, because one of the things that we learned very quickly, which was that tumor cells, solid tumor cells are basically invisible to the immune system, because they can't give that second signal through CD28, so they can grow for substantial period of time without being detected, until as shown on the next slide, there is the T cells get big and or can't get enough oxygen or enough nutrients, so they begin to die a kind of death called necrosis, and that causes inflammation. These professional antigen presenting cells, there have to be seven molecules on them, pick up antigens from the tumors, reprocess, then put them on the cell surface, and then that starts the immune response, but as with everything else, that induces CTLA-4r, which can come along then, I show on the next slide, and attenuate or even stop a immune response to a tumor, and so when we worked out all these basic aspects -- and the early work had nothing to do with cancer whatsoever. It had to do with regulation of T cell responses, basic T cell biology that was actually funded by the National Cancer Institute, because the ultimate relevance was, I think, not necessarily obvious, but was at least a possibility. But anyway, this led to this situation where it's kind of a race, you know, if you get enough T cells to kill the tumor before CTLA-4 turns them off, then you win, but if the tumor get out grows them, then the immune response gets shut off and the tumor wins. So we had the idea I show on the next slide, just to interrupt this process, to interrupt CTLA-4 with an antibody that keeps it from binding to the B-7 molecules and let the T cells just keep going long enough to eliminate the tumor and then they stop giving the antibody and everything returns to normal, and this was compelling for a couple of reasons. As Dr. Weiner mentioned, this does not attack the tumor of these antibodies, unlike the ones conceived of by Dr. Ehrlich in the early 1900s. The target here is the immune system itself, not the tumor cell, so we're treating the immune system to allow it to kill cancer. So since you're doing that, the possibility is that this could work for all cancers, because Dr. Peter told you all cancers have mutations in them, and they're going to have foreign things that can be detected by the immune system, and so potentially one drug could be a universal cancer treatment. It doesn't work out that way for several reasons, some of which I'll get to later, but the second compelling reason was that shown on the next slide, this whole process is started by cell death, and so one of the things that you can -- that the immune therapies, the T cells give you, you know, is memory for example. Once you've got T cells, some of them stay around for the rest of your life, and if the tumor comes back, they can easily be reactivated to attack it, and it can also change as the tumors change, and help continuous immunity. And also the immune system does not necessarily attack just that mutation in the target therapies that Dr. Weiner told you about. They cause the tumor, but it can attack the passenger mutations as well, that have nothing to do with the cancer process, but are there in the cell, and if you can hit enough of them, then you can eliminate the tumor despite not doing anything to the causal mechanism, but anyway, you can combine that with anything that kills tumor cells potentially, chemotherapy, radiation, hormone therapy, anti-angiogenesis, and those targeted therapies themselves can also extend the range of targets of vaccines, so it was a nice idea, but would it work? And the next slide shows an example of one of many, many mouse experiments that we did. This just shows a mouse tumor, a transplantable tumor that can be passed from mice to mice that are genetically identical, and it will grow and kill the mice. If we inject antibodies to CD28, you can see the tumor gets bigger, faster, showing there is an attempt by the immune system to get rid of the tumor, but it's just not able to completely eliminate it, but in many animal models, when we just injected antibodies to CTLA-4, the tumor would grow for a while, and then the T cells would reach them to where they could eliminate the tumor, and the tumor would be rejected, and the animals then would reject that tumor for the rest of their lives when we tried to re-challenge them. And, you know, this worked for, we found out later, for -- it worked by itself with tumors that were considered strongly immunogenic, and we know now by sequencing those, those were tumors. Typically that were chemically induced, and had a lot of mutations. There were other tumors that had very few mutations, and were not by themselves immunogenic, and that you could not immunize a mouse, for example, by giving them radiated tumor cells, but we found if we combined anti-CTLA-4 with chemotherapy or a little bit of radiation or with a vaccine, we could even eliminate those kinds of tumors, so we were able to cure almost every single mouse model that we went for in our testing this. So we show on the next slide, after a time, we found a company -- next slide, please. -- that had a mouse, a company called Medarex, that had a mouse that didn't -- that they had eliminated the mouse antibody genes and replaced it with human's, human genes, so we could make a fully human antibody that was immediately ready to go in the clinic and so with Medarex, we made this antibody which has, Dr. Steen told you, is called ipilimumab. They are developed by Bristol-Myers Squibb, which was used initially in many small trials and in many tumor types and was found to cause objective tumors, objective responses in many of these including melanoma, prostate, kidney, bladder, ovarian and lung, and prostate. It was a combination with a vaccine. But there were adverse events. In mice and monkeys, we never saw -- in toxicity studies, we never saw any adverse events other than some deep pigmentation in melanoma models, but when it went into patients, we began to see a lot of adverse events, colitis, hepatitis, a lot of [inaudible]. These are inflammatory responses that are not really autoimmune diseases when looked at carefully. Now they are immune related in that they represent a dysregulation of the immune response, but they could be treated with systemic steroids typically, and they go away, their symptoms go away, and then you can stop giving it, wean the patients off the steroids and they do fine and the tumors still respond, but now we know that hundreds of thousands of people have been treated with this, that are, pretty rare, 1% or so of patients develop type one diabetes, or a T cell mediated problem called myocarditis. It's a very interesting thing that we've developed an animal model for and are studying now and have developed a way, I think, a way of treating it. But overall, it's -- although it has adverse events that have to be paid attention to like any cancer therapy it turns out, they can generally be managed, so the next slide just shows a patient, one of the early patients treated. This was a -- I like to tell the story. This is a personal friend, now. She wasn't then. But anyway, she was 22 years old in 2004, had just graduated from college and was engaged to be married when she was exhausted anyway. She was diagnosed with metastatic melanoma. Most of the tumor metastases were inside her body, in her lungs, as shown here, in the left panel circled. She had 31 lung metastases. She also had liver metastases and she had a brain metastasis. She went on an early trial was treated, by Jed Wolchok at Memorial Sloan Kettering. She failed, actually, two chemotherapies, high dose IL-2, a dendritic cell-based vaccine before she -- and radiation for her brain tumor before she went on ipilimumab. She was hospice-bound at that point in 2004, so and then after the treatment with ipilimumab, her tumors completely disappeared. This shows -- CAT scan shows the metastasis in the lung. The next slide shows that the metastasis there in the lower right that was in her brain, and it also resolved. All of her tumors completely resolved, and she was a complete responder in 2004. The next slide just shows her -- she sent me this photo in 2016 with her two beautiful children, and I actually saw her last, two years ago, and she is still doing fine with two wonderful teenage children now, with no sign of disease, and able to live without looking over our shoulder after a time now, so she's now, you know, close to 14 years -- 16 years actually out, so I show on the next slide. This went to a phase three randomized trial with a overall survival standpoint, which completed in 2010. It was started about four and a half years earlier. Anyway, the lower curve here shows a placebo control, and the situation with melanoma at that time was a median survival of about seven months after diagnosis with fewer than 3% of patients being alive in five years. The top line is the ipilimumab line, and it's moved over several months, which moves the median survival, 50% survival over a few months so that would have been sufficient for FDA approval, because no drug at the time had ever done that in a randomized trial, but as you can see, the other interesting thing is that the survival curve flattens out in about two and a half to three years, and then stays flat for the duration of this treatment. Anyway, this resulted in approval by the FDA in 2011, but as shown in the next slide, by 2015, there was a sufficient number of patients as shown here, that could do a meta-analysis of survival, and it turns out that the overall survival at 10 years was about 20%, a little over 20%, after monotherapy, so which was, you know, a remarkable achievement at the time, although it's only 20%, you know, given the alternative, but it was only 20%, and why isn't it better? But I can give you a number of reasons, a number of mechanistic possibilities, but of course, the more interesting one is that maybe there were multiple checkpoints, and sure enough, we show the next slide, Tasuku Honjo, this was mentioned, in about the time that our phase one data were coming out, he found that, in collaboration with Arlene Sharp and Gordon Freeman at the Dana Farber Cancer Center, showed that a month molecule Tasuku had been studying for years, PD-1, was another one of these checkpoints. It had two ligands, PD-1 and L1 and L2, as shown in the bottom. A difference though between this and CTLA-4 is that one of its ligands, PD-1, can be expressed on tumor cells, and this is different than what goes on in the CTLA-4, because PD-1 is induced in response to gamma interferon, a substance produced by T cells to kill tumor cells, which can kill tumor cells, and so this is an acquired defense mechanism. You know, the specific T cell that can detect antigens on the tumor makes gamma interferon. The tumor responds by up-regulating PDL-1 which turns off the T cell, so this is an active defense mechanism. This mechanism has also been shown to play a role of protecting the fetus from destruction by the maternal T cells, targeting paternally encoded antigens, so this pathway seems to be co-opted by tumors to protect themselves much that same way, so different than CTLA-4. The next slide just shows the result of a phase one trial, just a summary by Suzanne Topalian, where like CTLA-4, which responds to react -- many tumors respond to it, including, again, of course, melanoma, non-small cell lung cancer in this trial, renal cancer, which also responds well to CTLA-4, but colorectal cancer did not respond. Now that's since been shown that there is a subset of colorectal cancer that has hypermutation, has defects in DNA damage repair, so it has a lot of those neoantigens that Dr. Weiner talked about, so that subset of colorectal cancer does respond to PD-1, but there were no responders to castrate-resistant prostate cancer, and this is different from CTLA-4, which did respond in some early trials, at least there were some responses to it, and I'll come back to this in a while, because there's certain several lessons that we've learned from studies of prostate cancer. At any event, show the next slide, and these quickly began to be realized that these pathways were indeed not only purely mechanistically independent, but because of that they -- one drug could be used after the other. In other words, patients that failed ipilimumab could go on a PD-1 antibody nivolumab and still have a good chance responding in the reverse which was also true those that were treated with PD-1 could respond, sorry -- fail PD1 could respond or subsequently treated with anti-CTLA-4, so this plus a lot of mechanistic studies that were done could show that they were different, in part, lead to a trial combining these two agents as shown in the next slide. Putting them together, the actual data I show in the next slide is a trial -- sorry, [inaudible] or a little bit. Anyway, this is an experiment that we did in mice very early on showing that these two could be combined. The next slide then finally shows the results of a trial where these were put together. This shows that 52% of patients here we're alive five years after therapy, and in a different -- this a phase three randomized trial, Jed Wolchok recently published an update where the response is six and a half years and another trial is even slightly higher than this, but anyway, over 50%. So the lesson here is that by combining these two checkpoints now with melanoma, this drug -- this disease, which, you know, no drug had ever treated before this and now the majority of patients are alive at six and a half years now, and then there's no reason why they're not going to be alive at 10 or whatever, just as shown within our CTLA-4 alone, so quite an advance, especially with melanoma, as shown in the next slide. This isn't just melanoma though, and this is as Dr. Weiner said, a very classic slide at this point, but here I've just marked it up a little bit. Actually, this is with Bob Schreiber and Tony Schumacher, to show you that in the red box, these are these tumors that have hundreds to thousands of mutations per cell and lots and lots of new antigens, and you notice again, melanoma is at the high end of that, and also other kinds of diseases caused by mutation. Like lung cancer, for example, not caused by UV induced mutations, but by carcinogens in tobacco smoke along with, you know, lung cancer, but also bladder cancer, head and neck cancer, other things like that. Those tumors all respond very well to monotherapy, so either CTLA-4 or anti-PD-1 [inaudible] tested, and now are being tested with their combination. But as you move to the left on this slide, with other kinds of cancers, they have much lower frequencies of mutations, and down here, kidney cancer, although it's at a level where you would not necessarily expect it to respond, responds very well again to monotherapies, either PD-1 and CTLA-4 but even better to the combination, where the response rate is 40% or so, and then when you go to the left, though, you get down to where pancreatic cancer, glioblastoma, and prostate cancer, there in the blue box, which there are occasional responses to, but not regular responses. Again, I'll mention this in a moment, but this is the challenge now is to deal with the rest of these kinds of tumors, but as shown on the next slide, this just shows the approvals by the FDA and different cancer types, and there -- this is only a partial list, as Dr. Weiner said, with the antibodies with the checkpoint antibodies. I got tired about three years ago keeping up but there are 40-something approvals now. You can see here, not just melanoma, where if you're doing ipilimumab was approved in 2011 and CTLA-4 and the Pd-1 antibodies, 2014, and a combination in 2015, but also lung cancer, renal cancer, colorectal, on and on, but the notable or what I should point out, though, where we're at is that it's gradually moving towards frontline use of checkpoints, generally with a combination of anti-CTLA-4 and anti-PD-1, PD-1 which gives you a higher response rate and ipilimumab was thought to provide perhaps better, you know, sustained responses. Together, they work quite well, but not quite as well as in melanoma and kidney cancer, for example. The combination is active, like somewhere between 35 and 40% of patients, so we've got a ways to go in improving the rates of responses there, and so far, there's been no approvals in prostate cancer, although we have seen clinic -- some complete responses within anti-CTLA-4 plus anti-PD-1 and also no responses so far in two very well-known and very lethal tumors, pancreatic cancer and glioblastoma, although there's a lot of effort here, and I think progress is being made, although not much yet. We just got a lot of work to do. So in the next slide that just again, a summary because I don't have time to show you the data, but we did some studies along with Pam Sharma in prostate cancer. Remember that I told you there was no responses with PD-1 and we took biopsies for patients before and after therapy with CTLA-4 monotherapy. There was a phase three trial of anti-CTLA-4 in prostate cancer because there were sufficient numbers of responses to justify it, but the phase three trial failed, and so it led to Bristol-Myers Squibb to completely shut down their prostate cancer program. You know, we got biopsies from a number of patients before and after ipilimumab therapy and what we found was that before therapy, they were as cold in terms of immune cell infiltration as pancreatic cancer, for example, but after ipilimumab, they were just as infiltrated as melanoma. On the other hand, what we found was that those cells in the prostate cancer now expressed a large number of checkpoints including not only PDL-1 which, you know, involved in PD-1 blockade, but also several others. This led us to then look at the combination of drugs because PD-1 antibodies do not drive T cells and prostate cancer, and so we went to Bristol-Myers Squibb and said let's give CTLA-4 to get the T cells in PD-1 to take away that checkpoint, and then we'll see some responses, so Dr. Sharma and Dr. Subudhi led a trial. There were complete responders in that trial, meaning PSA became undetectable. The tumors went away, including primary prostate cancer, but also soft tissue metastases, and there were some patients that are now two, three years out, but bone metastases did not respond. In any event, that trial was being expanded now, and there was a registration trial. There was some toxicity, but the amount of CTLA-4 is being cut back to minimize that, and we'll see if it's going to be effective, but one of the other things that we learned in the course of this study was that with CTLA-4 monotherapy, we could get response to neoantigens in this tumor, even though as I -- as Dr. Weiner just showed, and as I showed you earlier, prostate has very low levels of mutations. In our sequencing, we found most tumors had only a few dozen, a dozen to a few dozen mutations, but we found in a smaller panel of patients that we looked at, one patient and we had 10 mutations. Two of them, those mutations were recognized by T cells and induced after CTLA-4 therapy. Another one that had 12, only 12 mutations had one response, so there is some hope for patients that have low mutational loads that need not be a total reason to not try. So I show on the next slide -- next slide, please. Just to summarize some differences between these two checkpoints. CTLA-4 is hardwired. PD-1 is an induced resistance. CTLA-4 seeks to target C28. PD-1, the T cell receptor pathway. CTLA-4 works during clonal diversity, and because that has been shown to work pretty slowly, but also to move T cells into cold tumors, and to recruit new T cells into tumors and expand the clonal diversity. Whereas PD-1 works very rapidly and only expands the T cells that were already there before treatment, because it works on fully differentiated T cells. Main difference is that adverse events with PD-1 are much less frequent, and adverse events with CTLA-4 much more frequent, but it's found that disease recurrence after PD-1 monotherapy at least, is significant, but it's pretty rare after a response to CTLA-4. At any event, so a very powerful pair checkpoints that can be used, but the story doesn't end there because I showed you. It was on this -- the prostate cancer slide. There was also a molecule called VISTA that we detected there, which is a very powerful inhibitor of T cell responses. It's not ever found on tumors, but it is found on a myeloid cell population that we find after treatment of prostate cancer, but not melanoma, for example. So there's some complexity of these checkpoints, as to the kinds of tumors that they expressed in that needs to be paid attention to if we're going to continue to combine these. At any event, the next slide just shows a partial list of the molecules I talk about. I've already talked about the main two, PD-1 and CTLA-4 which I think are going to be the combination that can be built upon in most therapies coming forward. Antibodies to molecule called TIGIT are looking pretty good in the clinic now as are antibodies to LAG-3, but this other class of molecules that are on just myeloid cells that I've mentioned on the lower right here, VISTA is one of them. Another one that we're looking at now is LILRB4. Again, both inhibitors of T cells that are not expressed on tumors. The mechanisms aren't completely elucidated yet, but there are antibodies to them, and for patients that don't respond, for example, to anti-PD-1 plus anti-CTLA-4, I think in certain kinds of tumors, we're going to be able to add these if we see that they're upregulated in response to the other checkpoints. And then the next slide just shows what I think is going to happen. We're going to continue blending antibodies to both positive and negative checkpoints. OX40 4-1BB are molecules and ICOS molecules that I talk about, that actually increase T cell responses, and those could be combined, but I think, going back to the first part of the talk, we're going to start seeing more combinations with conventional therapies and target therapies, which again, can prime T cell responses by killing tumor cells, and allow us to treat exactly, you know, when we know these tumors are beginning, these antigens are going to be released by tumor cells and prime a response, which should make the therapies much more efficient, so any problems to face, as I told you, we're not that far along. There are many kinds of cancers, a long way to go, but I think we know the basic rules now, and we can get there and show on the next slide is something that I like to show and this is something that as Dr. Weiner told you, beginning with chemotherapy, in the '40s the goal of oncology is largely to take a -- do a clinical trial with a fairly large number of patients, treat them with the drug that you're testing, compare and see if you get an increase in immediate survival. And if you do, that's obviously a good thing, providing a longer life with quality of life, but we know now from ipilimumab, the early studies there shown on the next slide, you can do that, but also get a tail on the curve at about 20% or so in melanoma, but the next -- the final slide is not any data yet. It's an aspirational line showing you what we're trying to do now, and that is to raise that survival tail as high as we can get it, not just the response rate, but the overall response, the durable responses, like we've seen in melanoma that can last for decades can finally be considered cures of cancer, and as I said, we've got a lot of work to do, but we're learning the fundamentals and we've learned the fundamentals. Now we can begin to start kept combining them in more productive ways to try to achieve this goal, so that I hope I haven't gone too much over time, and I'll stop and thank you for inviting me and allowing me to participate in this conversation about progress and moonshots, so thank you very much. >> Tomoko Y. Steen: Ah, thank you so much, Dr. Allison. It is really a very informative presentation, and it's great to know that, you know, continuously improving, and we have the combination with targeted treatment, it's available, and that brings to a wonderful, you know, introduction to next speaker, Dr. Giorgio Trinchieri. He is Chief at the Laboratory of Integrated Cancer Immunology, and he is the head of Cancer Immunology Section in the Center for Cancer Research, and he is at NIH, NCI, and he's also the Distinguished NIH Investigator, and now he's working on fine tuning and, you know, how we use this technology therapy to even better level and so that's Dr. Trinchieri is going to share with us, so before further ado, the floor is yours. >> Giogio Trinchieri: Thank you, Dr. Steen. Thank you for the introduction. If I can have the first slide, can you show the slide from the beginning? >> Tomoko Y. Steen: Yes, yes. >> Giogio Trinchieri: This is -- there is animation. It's not showing the animation. >> Tomoko Y. Steen: Yeah, it's perfect. >> Giogio Trinchieri: Okay, it is supposed to be shown with animation, show only the center of box. >> Tomoko Y. Steen: Oh, animation. I'm sorry. >> Giogio Trinchieri: Yes. >> Ashley Cuffia: The animation is not coming up, Doctor. >> Giogio Trinchieri: Okay, can I show my slide myself? >> Ashley Cuffia: Sure. Give me one second. >> Tomoko Y. Steen: So then, meantime, I like to tell audience, please put in your question. It's bottom center part. It's the Q&A, so some people were putting in the chat, but please put in the Q&A, and you can write in any time so we'd like to have the questions. >> Giogio Trinchieri: Can you see my slide? >> Ashley Cuffia: Yes. >> Giogio Trinchieri: Yeah, perfect. Thank you very much. I'm sorry for this inconvenience. So I said it was -- >> Tomoko Y. Steen: Sorry, it's in like a speaker's mode, so you can switch. >> Giogio Trinchieri: Okay, sorry. Okay. It's fine now? >> Tomoko Y. Steen: Yes. >> Giogio Trinchieri: Okay, perfect. Apologize again. So it's been discussed by Dr. Allison and Dr. Weiner, even with a great result with immunotherapy and immuno-checkpoint blockade, still unfortunately, not all patient respond to immunotherapy, so the question really come out, can we predict and improve the response? And we know that the tumor cells was discussed. The presence of a new antigen mutation is a major role as a tumor increasing factor, but there are also tumor experiencing factors, the cell, they are present in the tumor microenvironment, circulating factor like different inflammatory factor present in the blood, but also [inaudible] present for example, the number of granulocytes neutrophils in the blood. There is a correlation of the response with adverse effect. Adverse effects are always a problem in the therapy, but also are associate normally with a good response, and then in many different host factor, metabolic factor, what is I want to discuss today is to generate quite a lot of interest is the role of the intestinal gap for commensal microbiota means the bacteria and other organisms or microorganisms that are present in our gut that affect our physiology and pathology, and it's been shown to have an effect on cancer immunotherapy. Now, what are these commensal microbiota? We know that we have evolved with microbial partners. It means our body is formed by our own human cells, together with these microorganisms, bacteria, but also virus, protozoa, fungi, and they are present in all the [inaudible] surface that divide our body from the outside world, and particularly are very abundant in our lower intestinal tract, lower small intestine and in our colon. And what is very important is that the commensal microbiota is known now to be indispensable for health and survival of the metaorganism, our organism with our bacteria, and the crosstalk between our cells, the host cells, and the microbiota regulate many physiological function that include many different one, but particularly metabolism, inflammation and immunity, and in this way, also think cancer, may cause cancer, may favor the cancer progression, but they may also regulate cancer there. And our group and others about eight years ago has shown that the gut microbiota is require and determine the efficacy of cancer therapy by modulating the anti-tumor immune response and so naturally doing that by educating one type of cells in the tumor microenvironment, the myeloid cells and in this way regulate immunity and the ability of the immune cell to regulate the tumor growth. We originally described that for a new type of immunotherapy based on CPG oligonucleotide, as well as chemotherapy based on platin compound, but then many other group and other paper has been described the role of this gut microbiota in regulating cancer therapy, and a lot of interest was more recently, when immune-checkpoint inhibitor therapy has been shown to be regulated by the presence and composition of the gut microbiota. Now well just summarize in this slide. There are several studies been published, but they are mostly based on animal model, on mouse model. Therefore, really the major interest in this type of research was about three years ago and several papers starting come out, three in the journal Science showing that the in a clinical trial, a patient treated with immune-checkpoint inhibitor, particularly anti-PD-1 and anti-PDL-1, the gut microbiota controlled the response, and here we have, for example, two studies of [inaudible] in Chicago on melanoma patient and [inaudible] Paris on lung and renal cancer. Now, this patient clearly established with many different type of data and also correlate in animal model that the composition of gut microbiota is important for the effectiveness on the PD-1 cancer therapy.. What there was a little bit disappointing that even it's on this paper describe bacterial species that were associated with a good anti-PD-1 response. The species they described are shown here in red, were nearly different in each study. There were almost no communality, no overlap between the bacteria that were identified before the study. So that put the faith in a little bit disarray for a couple of years, but I think things are really now start changing and I think we can start seeing some common result among the different study, and really start to understand a bit better how the bacteria and which bacteria are regulate the cancer immunotherapy. I want to show you a series of data has been published by many different groups. This is one example that showed that if you're disrupt in the patient the composition of the gut microbiota by using antibiotics, you can see the patient that use antibiotics had much worse survival than the patient did not use antibiotics. Antibiotic disrupt the bacteria present in the gut and make the patient less responsive to therapy, but also other pharmacologic agent that can change the competition bacteria, for example, the acid-reducing drugs, the proton pump inhibitor, you can see the patient use proton pump inhibitors. They respond less well, and if they use antibiotics and proton pump inhibitors, they really respond very poorly to the anti-PD-1 treatment. Also, the different the data in different study, within different study, at least, were quite strong to show there are certain bacteria. This is our data in melanoma patients in Pittsburgh. We show on the top three different bacteria that were associated with a good response to anti-PD-1, and in red, you see the response and survival of the patient that have either of these three bacteria, in blue, the one with low levels, so you can see here it is drastic and significant different response in the patient. On the bottom are three bacteria. They are associated with poor response to anti-PD-1. You see [inaudible] the patient that have high abundance of these three bacteria have a very poor response, very poor survival after anti-PD-1 treatment, but if you put all the result together, these are five different study in melanoma and the patient here, each one, the microbiota, the bacteria present in the gut of the patient is shown by a red dot. In blue, the one respond in red, the one do not respond. You can see that the patient that responded different from the patient do not respond, and if you combine in a meta-analysis all this together, even if its study really describe somewhat different bacterial species, you can see that if you look this family of lots of spirochete, [inaudible], three family of bacteria, most of the bacteria that are associated with a positive response belong to these three families. Wherever the bacteria they belong to Bacteroidetes, many of them shown here by the -- shown to be associated with a bad response, so it did find some commonality and probably some share mechanism in the result has been present in the different cohort. And more than that, even if cohort, like you show five different cohort here in five different city, even if they -- each cohort was very different, and the patient were very different in the composition of their microbiota, if you get the information of the composition microbiota and a response to anti-PD-1, derive from four of the cohort. You can test this information on the cohort that was not used in this analysis, and you can predict the response to anti-PD-1. That was done for all five different cohort. This is [inaudible]. This actually show that in [inaudible] we can -- we have an accuracy about 80%, so it's not perfect, but just by knowing the composition microbiota of the patient with an 80% accuracy, we can predict whether they will respond or will not respond to anti-PD-1. The other thing that was discussed in detail by that [inaudible] was that when you get a good response to anti-PD-1, often it is associated with immune related adverse effect, and this slide on the left, you know, just showing the many different type of adverse effect, collateral effect that you get with anti-PD-1 treatment, and the study we did in Pittsburgh, as many other study before us, it showed that the patient that have any type of immune related adverse effect [inaudible] had a better survival of the patient without immune related adverse effects show here in blue. And if we look the microbiota, the bacteria they are in the gut of the patient with or without adverse effect, we see that they are different, in red, the patient with adverse effect, in green here, the patient without adverse effect. There are bacteria significant different in this two group, but we'll also show that was not unique type of bacteria. There are different type of bacteria doing different things and using different type of adverse effect, and in particular, we found that the Lachnospiraceae family were associated with various immune related adverse effect, and also like we show in the totality of the patient, were associated with improved survival. However, there was other patient that had high number of Streptococci in our gut, and these patient also had adverse effect. They were slightly different the one [inaudible] Lachnospiraceae, particularly they have very often after a [inaudible] and arthritis, but they had a much worse survival, and these patients Streptococcus species were actually most of them were using a proton pump inhibitor, the acid-reduce drugs, and that's because they use the proton pump inhibitor to actually increase the abundance of streptococcus species in the gut to the patient, and this is actually associated with both side effect, but in this case, to a worse survival, worse response, poor response to anti-PD-1. The other thing that I mentioned that if you have systemic inflammation as show by high neutrophils in the blood, particularly high neutrophil PFS ratio. This is associated with a poor response to anti-PD-1 and again, it's been shown by many study, but what we found that the neutrophil to lymphocyte ratio also distinguish two group with a patient with a different composition of the gut bacteria, the fecal microbiome, and when we look which bacteria were associated with this systemic inflammation and show by a number of neutrophils in the blood, we found that the majority of the bacteria they were using these increased neutrophils number on the blood, where endotoxin produce gram-negative bacteria, and in a series of study in human and the mouse and in the fecal transplant and other clinical study, we could show that the gram-negative bacteria in the gut by producing the toxic LPS, inducing inflammation in the intestine with production of inflammatory mediator, but also attraction in the guard of inflammatory cell like neutrophils and macrophage. These are also were seen in the liver where we have neutrophils infiltration, and that favor grow tumor and metastasis in the liver, at least in the mouse, but in human we could show that was [inaudible] in the blood as I mentioned to high neutrophil/lymphocyte ratio and presence of soluble factor like the cytokine IL-8, and this reflect a poor response to immune-checkpoint blockers at the tumor level and also the presence of this cytokine IL-8 we see in the blood and then intestine also being produced in the tumor, tumor and patient I mentioned that have a poor response to immune-checkpoint blockers. So why we see this discrepancy different study? Well, it's a study the gut bacteria is very complicated because the gut bacteria may change with time and may change with geography and so on, and what we know is that the composition of the gut bacteria is really mostly regulated, involved by contact with the mother then in the first few year of age by interaction with the environment, then become relatively stable, during adult life, but can still be changed by diet and lifestyle. So what is really very important is where you're born, where you live, the geographical organizational lifestyle, diet and aging is very important, but also use a food supplement like probiotics. The use antibiotics infection that can change the composition of the gut microbiota. So when you look at a different study, you can immediately know and I already mentioned some of the city, they really their patient populations come from different geographical place, and that's obviously associated with different lifestyle, and different type of food, and these are all things that can have an effect on the type of bacteria that you have in your gut, so we look at that specifically. We look in the United States. We have the ability, because of the American Gut Project has analyzed more than 20,000 donor in the US to show that the [inaudible] U.S. is really clustered, divide in different group that have characteristic type of bacteria in their gut, and if you look this different classes, different types of microbiota, and you look different bugs, different bacteria, you can see that some bacteria present some cluster in red, not in other and for example, what surely on top is bacteria, that are good for anti-PD-1 response, the one on top of the one they are bad, so you can imagine that this different type of gut bacteria [inaudible] gut bacteria may associate already with good or bad response to anti-PD-1, and what we will show that if we get each one of this type of cluster of composition of gut bacteria, and we look where they come United States, you see that some are present in different populated state, but this one, for example, is present only in the northeast, this one only in Southern California, so they have different geographical localization, and not surprisingly, when we look different melanoma patient cohort, you can see these are patient. They're not only normal donor, you can see they are different, the different patient study in different place, they have different composition microbiota, and if you look their presence of the microbiota type, they are good or bad for anti-PD-1, they're really different in its different cohort, and that's probably one of the reason explain the different result, the original has been described by the different study. So if the microbiome is important, the question always immediately come, can we target the microbiota? Can we change the microbiota, and we can do that in a way to improve therapy response? This was another slide showing now at the beginning, when we really had no idea what was going on. We knew that there was this association. We said the microbiota with good response or bad response to anti-PD-1. We didn't know exactly which bacteria were good, and what the mechanism was. We know a bit more now but immediately, everybody always was interested we can change this balance, this between different bacteria and make the patient better response to therapy. Now what there are many studies that are ongoing, that they're trying to use single bacteria, or consortia bacteria probiotic, and see whether we can change the microbiota composition. This is always difficult, because as I mentioned, we don't know exactly which bacteria are important, how they work, so we really have no result even different study are ongoing. We have no result from this study yet, but where we have a result and we have been directly involved in the study is two type approach to change the microbiota. One is a fecal microbiota transplant from one patient to another patient or change or locally change in the diet that the patient are intaking during the before during the therapy, so the fecal microbiota transplant is based on the idea that the patient did to respond to anti-PD-1, either they failed completely respond to PD-1 and together we work out the bar and since They're all in Pittsburgh, we decide based on their qualities in this two group of patients, the different microbiota, the responders, a good one, the non-responders the bad one, see if we could transfer the fecal microbiota from the patient that responded what's supposed to be good microbiota implant in the unresponsive patient and see if we can change the response to anti-PD-1 and indeed we -- it was a small trial with the 15 patient. Another group [inaudible] but what we found that by doing that we can transform 40% of patients that were completely unresponsive to PD-1 before the fecal transplant in response to PD-1 and the data showing here the size of the tumor in red are nine patient that fail the even after the fecal transplant, but in [inaudible] the six patients there respond and they either have a complete respond, part response or stable disease that let you say that all the six patient now are stable disease for more than two years after the fecal transplant and the continuation of the anti-PD-1. The other data that I mentioned is diet. This is a study in collaboration with the group Jennifer [inaudible] Anderson, and this is not an intervention study, but just to analyze what the patient that were treated melanoma patient were treated anti-PD-1 how they responded to anti-PD-1 and look what they were eating, and what it came up that if the patient were eating more than 20 grams per day of fiber, so in high-fiber diet, about two-third of the patient [inaudible] of a patient that respond to anti-PD-1, but if the low fiber-diet, only about 1/3 of them respond to anti-PD-1, and you can show the [inaudible] America for survival here. You can see the patient with high fiber is a much higher survivor than the patient with low-fiber diet and the study in human [inaudible] study in the mouse were able to show the high-fiber diet really change the gut bacteria in a way that the ratio between bacteria that favorable to anti-PD-1 response to [inaudible] one is changed by the high-fiber diet, so what I've been trying to tell you is that our gut bacteria really very much depend on the way we interact with the microenvironment. Is very important where we are born, where we live, what we eat, what is our lifestyle, and all of these affect the microbiome, and based on this emerging data show that they may have a major effect in modulating the response to cancer therapy with different mechanisms and some of the data on fecal transplant diet, and hopefully some of the ongoing clinical trial with single bacteria or consortia bacteria are start to suggesting the microbiota could be targeted through cancer immunotherapy and this could be a future way in a relatively non-invasive way to change the ability of the patient to respond to therapy. Just like briefly to thank my laboratory group for the data I presented, the great collaboration of the patient and the clinician and the Pittsburgh Medical School, particularly the group at [inaudible] the group of Jennifer [inaudible] Anderson and obviously all the great code that we have NCI for doing this type of work. Thank you very much. >> Tomoko Y. Steen: Well, thank you so much and, you know, this has really proved that the gut microbiome is really an important part of the immune system and especially for this treatment. If you can put in the questions, please put in Q&A and it's the middle center part. So next speaker is Dr. Sigal. Actually, can you put back to slide? >> Ashley Cuffia: One minute. >> Tomoko Y. Steen: Yup. There some people putting a question in the chat, but please put in Q&A, and if we can't answer on time, we will answer to you or send you the email answer after the event. So our last speaker is Dr. Ellen Sigal, and she was speaker in the 2016 Cancer Moonshot panel. It was in person, and unfortunately, this time we had to do webinar. Hopefully we can do in person next year. So she's a patient advocate, the founder and the chairperson of Friends of Cancer Research. And she has been working on this reaching patients and clinicians and scientists for quite many years, and she'll give background work, and we will have future of plans as well. So floor is yours, Dr. Sigal. Thank you for coming. >> I believe you need to start Dr. Sigal's video. >> Tomoko Y. Steen: Yup. >> Ellen Sigal: Good afternoon. It's quite a pleasure to be here, and I'm really overwhelmed by the extraordinary panel and the opportunity to discuss cancer, and certainly how it impacts the patient, and thank you for being at the Library Congress again. Last time I was there in person, and I'm hoping next time we can, and I'm going to make it a little shorter, because I know there's time for Q&A. Friends of Cancer Research is in existence now 25 years, and it started as a result of the 25th anniversary of the National Cancer Act, and now we're at 50 years, so it's quite a lot has been accomplished. We've done a lot and we have a lot of -- a lot more to do, so I'm going to talk about some of the major accomplishments from the 21st Century Cures Act, and the impact the legislation passed and many of you worked on it. And Danielle Carnival, you were extraordinary, and what was extraordinary about the moonshot is that you actually listened to the patient and the advocates and what they thought was really important, and the fact has had a profound difference. The legislation has really enhanced biomedical research and development of safe and effective new products. One of the key provisions is strengthening the FDA and streamlining clinical trials. Clinical trials are the backbone of science. It's how we learn. It's how we really get to better treatments, but they have to be accessible, and they have to be meaningful, and we have to get to the patients where they live, and we have to make sure that underserved and patients who normally do get treated in a community get access to these to these trials and Cures went a long way in streamlining these clinical trials. One of the other things it did is modernize the regulatory process, the OSE, the cancer, the creation of the Oncology Center of Excellence at the FDA's enhanced patient centered regulatory decision making through innovation and collaboration. It also organized all of the different divisions, so they all work together which is really important. It created a collaborative scientific environment in advance of the development of regulation of oncology patients, for patients, and it also put patients at the center of drug development. We are doing all of this for science, but we are doing this so we can benefit patients and patients are really the north star of what we're doing. The provision created new steps that operationalizing corporations direct patient feedback and assisted advocacy organizations and medical research FDA for FDA and industry to align drug development programs with the important information provided by patients. It also went into rare diseases for limit -- where we have really no treatments, and now the FDA is empowered to offer experimental drug approval for narrow patient populations, expanding options for patients without established alternatives. That is incredibly important. In some cases, we have wonderful treatments. In other areas, we really are -- really need to enhance rare cancers and really deal with these cancers where we have no good answers. One of the other things that the moon shot did in 21st Century Cures, it advanced recruitment of best and brightest at the FDA. We have to attract the best people and the brightest people to come and do public service both at the FDA, the NIH and in government, and as the responsibilities grew, we were able to streamline the process and make career enhancement more important for people to join these really important efforts, and the other thing is that we built a framework to better involve and incorporate patients in drug development. You don't bring patients in at the end, you bring them in the beginning. You try to find out trials that are important to them. Are they interested in these trials. Would they go on them? And did they enhance the knowledge? And are they accessible to these patients? This is really incredibly important. Patients, it puts patients at the center of drug development, and patients first should always be our mantra, and I want to thank you and look forward to our discussions. >> Tomoko Y. Steen: Well, thank you so much, Dr. Sigel. Okay. We have a limited time because of the beginning of the technical issue, but Ashley, can you read some of the questions? >> Ashley Cuffia: Yes, we do have some questions. First, is there a way to tell a cancer to grow in reverse like an implosion? >> Tomoko Y. Steen: That's a interesting question. Maybe Dr. Weiner can answer. >> Louis Weiner: Okay, then. So I think what you're talking about here is not necessarily immunotherapy, but rather targeted therapy. You'll recall my slide I showed the smart bomb that was the targeted therapy. That's essentially what you're trying to do there. You're essentially shutting down the systems that the cancer cells need in order to be able to survive, and in doing so, those cancer cells find no alternative, but to go ahead and die, and so they melt down, and that's one of the mechanisms that an implosion, as you suggest, is a well-known biology mechanism called apoptosis, or programmed cell death, and there's been an enormous amount of work over the past quarter century in developing therapies that can cause targeted -- you know, this kind of cell death, and so while I'm not sure it fits neatly into the immunotherapy checkbox, it certainly is another approach that's generated a lot of interest. >> Tomoko Y. Steen: Thank you, very much. Ashley, next question, >> Ashley Cuffia: What might be the potential for this therapy in treating sarcoidosis which is difficult to diagnose and treat? >> Tomoko Y. Steen: Maybe Dr. Allison, would you kindly answer this, or Dr. Weiner. >> James Allison: Weiner would probably be a better one to answer that one. I don't know of any myself. >> Louis Weiner: No. I was hoping you're going to take this one, Jim. So you know, these are different difficult challenges because sarcoidosis isn't necessarily associated with mutations or altered cell states of the type that can be targeted by the body's immune system. So while I think it's certainly possible to manipulate the biology that might lead to sarcoidosis by going after some of the underlying molecular mechanisms, I don't know that the principles of cancer immunotherapy are going to be highly relevant here. Although some others on the panel may have other thoughts. >> Tomoko Y. Steen: Yeah, that's great. Next one, please. >> Ashley Cuffia: Okay. It seems that we are finding cancers earlier. Is this being taken into account when people claim that five-year survival has increased? >> Tomoko Y. Steen: This can be again, Dr. Weiner or -- >> Louis Weiner: So there's good news on all fronts in terms of the improving survival rates from cancer. There's no question that earlier detection and the development of more effective early therapies have combined to put a dent in the deaths caused by cancer, but it's equally true that improved therapies have made an important contribution as well, and from 1990, or there abouts to the current time, there's been a dramatic decrease in the death rates from cancer. I'll also point out that reduced smoking has had a lot to do with this as well, because the burden of lung cancer had been quite enormous in our country. And I'll also mention, and this is really a shout out to Dr. Allison and colleagues, the advent of these novel immunotherapy strategies, the checkpoint antibodies, has not even yet begun to show the impact that they're going to have on death rates from cancer, because certain cancers like melanoma, as he pointed out, that was a disease when it spread, there was maybe a seven month average survival, and now more than half the people are living out their normal lifespans, it seems or close to that. And so there's going to be an indelible impact on survival as these treatments for melanoma but also for even more common diseases like lung cancer become more and more well integrated into our therapeutic armamentarium, so I think the outlook is good for continued drops in cancer mortality, assuming we can get back ahead of cancer with respect to getting people to remember to get their screening done, as Dr. Carnival mentioned. >> Tomoko Y. Steen: Yeah, yeah. Great. Dr. Allision, would you like to add something? >> James Allison: Dr. Weiner did an excellent job of answering the question. There was a study by the American Cancer Society, and I think they put it out that melanoma, the detection rate is actually increasing while the death rate is going down. So it's just a matter of numbers right now, but it'll be a few years before we see the full impact, but that doesn't mean -- we still need to worry a lot about prevention. >> Tomoko Y. Steen: Yes, yes. That's true, that's true. >> James Allison: [inaudible] been there too. >> Tomoko Y. Steen: Yeah. Dr. Trinchieri, maybe diet issue as well, so do you have something you could add? >> Giogio Trinchieri: Well, I mean, diet is clearly important for improving some response, and diet is clear -- and diet and gene and many other things, is clearly important to change the immune system and the interaction immune system with cancer is not always good. I mean, some cancer are going up because we live in different way, and we have different lifestyle, and cancer, and but I think once we have cancer, when we treat the patient, possibly, you know, change of life. Another way is to I mean, at the end of the day is the body defending itself against the cancer, the immune checkpoint inhibitor and approach of that is helping the body to do that, and it's possible that the diet and the composition of microbiota may also help in this interaction. >> Tomoko Y. Steen: Yeah. Japanese has a traditional [inaudible] diet for cancer, so. Next question, one more question. >> Ashley Cuffia: Okay. Ten years from now, what do you think the headline regarding cancer research will be? >> Tomoko Y. Steen: That's for everyone, please. Maybe start with Dr. Carnival? Is she out there? >> Danielle Carnival: Yeah, just coming back on video. Yeah. I think for me, I'll let the experts get into the specifics of where they think we will see advancements, but I think it's not only the kind of new treatments and new understanding of cancer, which I hope will be part of that headline, but that we've really reached everyone in this country with what we do now. I think the disparities around some of the prevention and early detection and even treatment access are some of the biggest barriers we face and 10 years from now hope, even sooner than that, we've made a lot of progress there. >> Tomoko Y. Steen: Thank you. Let's go with Dr. Allison. Would you -- >> James Allison: I think that what we're going to see is a more integrated approach to cancer therapy. I think immunotherapy is not going to be all of it. It's going to be part of it, I think. We're going to have to learn how to put multiple components together. Right now we're doing, you know, doublets. I think we're going to start mixing three, four, you know, different immunological reagents, but also, as I said, taking -- using radiation and chemotherapy as vaccines, basically, to start an immune response and things like that. I think it's going to be just combinations and recommendations for diet, and the disparities to is a very, really important issue to getting [inaudible] to everybody. >> Tomoko Y. Steen: Wonderful. Dr. Trinchieri, would you like to add something? >> Giogio Trinchieri: Yes. I mean, if you look back 10 years, and we had tried 10 years ago to predict what happening, immunotherapy, we are never being able to predict it. What we could predict is that by studying the immune response in cancer, by studying basic immunology, and that's applied to all other areas of science, metabolism, and so on, you really end up to identify a new way that you couldn't predict it, so one page is not to disregard basic science and where that's bring us, but the other things that what was just mentioned, is that it's important to cure cancer, but it's even more important to prevent cancer, and even more important to address all the disparity that are in different part of our population in being affected by cancer. We've been talking about cancer prevention for a long time. We are really not doing very much yet, so I hope that would we be talk more in the next 10 years. >> Tomoko Y. Steen: Yeah. Very important. Thank you. Dr. Sigal, would you like to add something from patient? >> Ellen Sigal: Yes, I think early detection is coming, and that's going to make a huge difference. We now have multi-panel cancer tests. We can detect earlier. We need to go still earlier, and then we need to know how to intervene, and we need to get to all patients, and we need to get to underserved. We have to get access, and we have to get better clinical trials. We're doing much better, but we still have to make them accessible and meaningful to patients, so I'm very optimistic, but we have to prevent, and if we can't prevent, we have to detect early. >> Tomoko Y. Steen: So your most important ambassador to the patient in term these wonderful research is going on, so Dr. Weiner, would you like to add a few words, or -- >> Louis Weiner: Yeah, so I'm going to just think about the headline. I think the headline is going to be continued investment in cancer research improves cancer survival rates to 80% -- >> Tomoko Y. Steen: Oh, wonderful. >> Louis Weiner: -- and I think that's achievable. I really do. And I think that that is only going to happen if all the things that my co-panelists described actually happens, you know? We have to make scientific advances. We have to deal with the science of disparities. We have to deal with the science of prevention and early detection and improve therapy, and it's only by, you know, putting all that together, we're going to be successful, but I think we can get -- we're at around 70% now. I think we can get 80% [inaudible]. >> Tomoko Y. Steen: That's wonderful. On that note, next slide, please. Yeah, so I'm sorry, we couldn't answer every questions, but we will be happy to reach out to all these speakers and answer to your question, so please send in your question. Make sure to add December 6th webinar so we can prioritize your question. Next slide, please. And this video is going to be available on the webinar under the Library of Congress in few weeks, and I hope, you know, you can use even classrooms and spread the word. It's important for everyone to know about, you know, how cancer research is going on in the [inaudible] for the patients. So thank you so much for all the speakers, and this was a wonderful webinar because of the wonderful presentations and hope we can do it again next year, and even sooner, so thank you so much for the speakers and also audience.