>> Sandra Charles: Good morning, and welcome to today's cancer symposium. Each time we try to expand a little bit, bringing in more employees and, of course, with that this technology and some of the attendant issues. So, thank you for your forbearance, and I look forward to us having a very informative and very, what should I say, well-rounded panel. We have people from our Culpeper [assumed spelling] campus in Virginia joining us, and I welcome those as well. I'm Dr. Sandra Charles, the library's physician and chief of the Health Services Division, and over the years, it's been my pleasure to work with Dr. Tomoko Steen of Science, Technology, and Business and [inaudible] Lyle [assumed spelling], one of our stalwart employee advocates to bring you these cancer symposia. This is just a brief PowerPoint showing what, over the past four years, what we have done in terms of addressing the issue of cancer, as we try to keep our employees informed, as well as hopeful and knowing all that's coming down the pike. So initially, we started out like doing sort of like in September, we do prostate cancer, in October we do breast cancer. And then we picked up on the -- in 2016, the Moonshot Initiative, where the whole idea was to start to get some of the, I call them, low-hanging fruit in terms of what kind of progress being made in cancer to get the word out to people. And we had on that date, 2016, members of the Blue Ribbon Panel that Moonshot Initiative, and serendipitously, the Cures Act was also signed right about that time as well. So, then we went on in 2017 to talk about genomics and unlocking the secrets of cancer through genomics and had a very stellar lineup, followed in 2018, by our program, talking about translational medicine, basically bench to bedside, where we had our panelists talking about bringing the fruit of the research to the bedside to the treatment modalities. And so, here it is we are today with four also stellar panelists, talking about those treatments and some of the progress that's been made technologically, and I won't delay much, because we -- there's a lot for them to do, and I don't want to be the one to hold you back. So, I would like to welcome them here and, Tomoko, would you like to introduce our panelists, so that we can get going? But I hope everyone has a program, and we want to leave time for you to ask questions of these guest speakers. So, I take my leave. >> Tomoko Steen: Thank you so much for coming. I'm Tomoko Steen from Science and Technology and Business Division. Instead of spending each biography -- speaker's biography, we have in this program -- so let me quickly introduce our panel. First speaker is Professor Robert Clarke. He is a breast cancer expert from Georgetown Medical School, and the second speaker is Michael Dean. Dr. Michael Dean is a senior investigator, and he's a geneticist, looking at the genetic side of the cancer and at the National Institutes of Health. He's now expanded the two -- he's going to talk about lung cancer but expanded to different types of cancer. So, if you have any questions, you can ask. Next speaker is Dr. Jonathan Lischalk, I'm sorry, my pronunciation. He is [inaudible] proton therapy, one of the few in the country, and we have a proton therapy facility at the Georgetown, and if you have any questions, you know, further questions, you can ask him, and the last speaker is from National Cancer Institute, Dr. Soumen Roy, and he is working on the side effect and [inaudible] microbiome for how the impact of the different type of cancer treatment affect the patient's toxicity and so on. So, first speaker, Dr. Clarke, please. It should be on the desktop [inaudible]. >> Robert Clarke: Good morning. It's a great pleasure to be here. It's nice to have folks in the room and to those that are listening or watching online, also, welcome. This is October, it is Breast Cancer Awareness Month. So, I'm going to talk about breast cancer. This will be a fairly straightforward presentation, not too much research. I'm going to tell you a little bit about what cancer is, what breast cancer is, and how we treat most patients. So, you'll get a sense of the disease from soup to nuts, and you'll get research pieces, I think, from some of the other presenters. So, I'm going to keep mine as straightforward as I can. So hopefully, it will set the scene for many of you, whether the talks are on breast cancer or not. So, cancer has been around for a very long time. The cancers that we know have been around for a long time are those of the tissues that managed to make it into fossils. Those in soft tissues have long since disappeared, but there's clear evidence of cancer in species as early as dinosaurs. In fact, probably older, it's very likely that what we think of as breast cancer arose, as mammals evolved. It's the same basic tissue performing the same function. So, the earliest mammal like creatures came around 200 million years ago. So, there's nothing new in the sense about breast cancer. It's been around 200 million years. We haven't cured it yet, but we're working on it, and you'll get a sense of some of the new advances that have led to changes in clinical practice, as I go through my presentation. So, what is cancer? Fundamentally, it's a loss of control of growth in cells. Most cells in the body don't keep making copies of themselves. There are some that do, because that's their job, but most don't, and some of the ones that get changed, where the DNA gets mutated, gets altered, and that control of growth is lost, leads to proliferation that shouldn't occur, and that causes a tumor to appear. Though not all tumors are spread or are particularly dangerous, some, of course, are. Benign tumors are those that stay locally, they stay put where they arise in the first place. For many of those, they're not life threatening. Some of them clearly are. Those that occur in the brain, for example, may not spread to the rest of the body, but if they keep growing where they're growing and they can't be eliminated, they become very problematic. But there are many benign lesions, and many of the lesions that arise in the breast naturally are benign. They are not going to kill anybody. It's the ones that spread, and it's when they spread that we have difficulty managing the disease. And those are the ones that are referred to as malignant, where the tumor starts in one place, maybe the breast, for example, but it can spread to the liver or the lungs or the skin or the brain. And then we have to use treatments that can go throughout the body, because we don't always know where those little deposits are. We just know that they're -- it's gone beyond the breast. So, benign, you can usually remove them, if you have to, with surgery and radiation, but if they have spread, we have to use different treatments. Now, what causes the damage to DNA that can lead to cancer? Well, every time a cell replicates its DNA, occasionally, it will make mistakes, and while the cell's usually pretty good at finding and repairing those, some are not. Some damages that curse simply naturally, a mistake in the reading and replication can persist, but there are exposures in the way that we live our lives normally that can increase the risk that there will be additional changes in the DNA sequence that are not fixed, and that will transform the cell from a normal cell to a cancer cell. And that loss of growth control will occur, and in some cases, the ability to spread locally and then disseminate throughout the body will eventually arise [inaudible] that tumor progresses and becomes more aggressive. The sorts of things that we think of as causing cancer include chemicals, which are probably amongst the most common, although we don't fully understand why most cancers arise, what the chemicals might be. There are some that, of course, we understand quite well. There are carcinogens. That's the type of chemical that causes cancer, this damage to DNA and tobacco. So, smoking, in fact, is the leading avoidable cause of cancer and cancer mortality in the United States. Alcohol can increase the risk heredity, and we'll talk about the BRC mutations in a moment. There's a lot of [inaudible] for associative data that implicates changes, features in the diet, hormones we know are involved in breast and probably prostate and one or two other cancers. Exposure to radiation, and you'll hear a lot more about radiation today as a treatment. There are viruses, HPV, the HPV vaccine was developed in part of Georgetown and at NCI, and that now can prevent an infectious disease piece that can lead to cancer. We treat, as I described earlier, local treatment, cut it out, burn it basically. With the systemic treatments, the treatments that we put into the bloodstream, which can get our -- the drugs anywhere, almost anywhere in the body, there what we're really looking for mostly are ways of poisoning, specifically, cancer cells. And I'll describe some of those for you in a moment in the context of breast cancer, and there are hormone therapies. If hormones increase the risk, then blocking the effects of hormones are effective treatments and, in some cases, effective preventive approaches. That's also true in breast cancer. So, what is breast cancer? Simplistically, it's a tumor that arises in the ducts or the lobules of the breast, and that's sort of captured here. It's a relatively simplistic view, but what happens is, as the cells lose their growth control, they can begin to acquire the ability to spread into the local tissues in the breast. In some cases, they can get into the bloodstream or the lymph stream and begin to spread, and so they can spread or metastasize throughout the body. And not all breast cancers are diagnosed from a lump in the breast. Sometimes it's the metastasis that's found, and when the metastasis is found, it still has features of the breast cancer that it came from, which can allow a pathologist who looks at it to say this isn't in the breast, but it's from a breast cancer, let's go look and see if we can find where the primary is. Quite a few cancers are diagnosed that way, not just in breast. So, what affects the risk of getting breast cancer? I wish we really knew. We have lots of data that show that some exposures can increase the likelihood that breast cancer will arise, but very few of them tell us in whom they will arrive, that a particular woman won't get breast cancer because of an exposure, and the effects of the exposure on increasing risk are really quite small overall with the exception, particularly, of one or two genes that we know are inherited. And you may or may not be able to read it on this slide, but most women who are diagnosed with breast cancer have none of the established risk factors. So, anyone who gets a diagnosis of breast cancer, it is not your fault. So, the one or two genes that we know that significantly increase the risk of breast cancer, that are inherited, are called the BRCA genes, and there's BRCA1 and BRCA2. And the reason that they increase the risk of breast and, as it happens, of ovarian cancer and one or two others is, because what those proteins normally do is they help correct mistakes in DNA or damage to DNA. And when there's an inherited mutation in those genes, the protein that's produced doesn't effectively find or correct those mistakes or that damage to DNA. And that's what increases the risk, and the lifetime risk for BRCA1 or BRCA2 for getting breast or ovarian cancer is very high, which is why once their -- a family is found and all of those who have that mutation are found, it is not unusual for women to opt for having their breasts completely removed and reconstructed and their ovaries removed. Now, there are lots and lots of other things that we think affect the risk of getting breast cancer. Very few of these have an effect that would double or triple the risk to an individual, and that's been a challenge. We can look at large, large numbers of women with breast cancer and look at exposures and find correlations, but correlation is not causation, as we say. And so, we struggle, we struggle to understand fully what it is that causes many women to get breast cancer. That being said, don't smoke, don't drink lots of alcohol, try and keep your weight normal, have a healthy diet and exercise, and you'll probably eliminate the increase in risk for many of the exposures that could affect your risk of breast cancer. And at the same time, you'll probably reduce your risk of cardiovascular disease of many other cancers, perhaps some neurodegenerative diseases. So, it's kind of just concepts. It's almost all we have to offer at the moment. The one preventive drug we have is tamoxifen, but it's usually given only to women who have a high risk of developing breast cancer because of a mix of other features that they're exposed to, which I won't have time to go into. Now, if cancer fundamentally is a disease of damage to DNA, the longer your DNA is around, the more likelihood it will get damaged. And so, it's not surprising that cancer tends to be a disease of older age, that you can get a sense of that from those pink bars, which show you the age of diagnosis for breast cancer in women. And that peak comes around 50 years of age, which just happens to be about when most women go through menopause. So, that tells you something about lifetime exposure to estrogens, that probably is one of the risk factors that you can do absolutely nothing about. Men can get breast cancer too, about 1% largely, because there's a small residual amount of breast epithelial tissue in the male breast. And that can get changed just like it can in women. It's just there's so much less of it that so many fewer men get breast cancer, but for women, it's one in eight. It is the most commonly diagnosed form of cancer in women. It is the second most prevalent form of cancer mortality in women, and the only one that comes higher is lung cancer. Don't smoke, avoid tobacco products. The risk of breast cancer death, specific death, is higher in African American women than it is in Caucasian or non-Hispanic white women, and we don't really know why that is the case. The outcomes are also worse. So, we have a lot, and you'll see, as I go through quickly, that we have a lot to learn. Now, not all breast cancers are the same. They fall into different subtypes that we tend to define by whether or not they have receptors for hormones or a specific growth factor. The hormones are estrogen and progesterone, and the growth factor is a member of the epidermal growth factor receptor family called HER2. And they look different, they behave differently, and they are treated differently. And the risk of death is different for each type. Now, the frightening statistics here, 268,000 new cases of invasive breast cancer in the United States each year, 42,000 women will lose their lives to this disease every year. That's one death from breast cancer about every 30 minutes, if you average it out, in the US alone. Over half a million women will die from this disease worldwide every year. We have made significant progress. You will see some of that, but you can tell from these statistics, we have a long way to go. Now, 70% of those breast cancers express either the estrogen receptor or the progesterone receptor, and if you look at the graph below, it gives you a sense of how the presence or absence of the estrogen receptor is very important in determining the risk of death. Those that don't have the estrogen receptor or that big bump at the beginning, where the risk of dying within the first three years is very high, but it drops thereafter. For estrogen receptor positive breast cancer, the risk of dying in the first three to five years is relatively low, but it increases every year. And by the time it gets to about three or four years, that risk stays at that level for the rest of a woman's life. And this is one of the challenges that we struggle with in breast cancer, is we don't know when those tumors with estrogen receptors will recur, if they're going to occur, because they can come back 10 or 15 or 20 years later, when a woman has lived for 20 years with no evidence of disease. And when it comes back, it usually comes back as having spread elsewhere, it has metastasized, and it is then very difficult to cure. So, that's a challenge. The good news about that piece of it is that women can live for 10 or 15 or 20 years completely cancer free, apparently cancer free. And if you're diagnosed in your 60s and you were able to live 20 more years without your breast cancer coming back, you would get to see your kids go up and graduate from college and get married and have kids. So, even when we cannot cure all breast cancers, we can give women lives to live, and we hope to be able to continue to do much better. How do we do that? We do it either by cutting it out, burning what's left, and poisoning what we can't see. Surgeries, in the old days, we used to do radical mastectomies. That's very rare now. In fact, we know that even the smallest of those surgeries, in most cases, have exactly the same clinical outcome as removing the entire breast for most women. And the reconstructions that we can do now, after surgery, are absolutely fabulous. So, even if you have to get a breast tumor removed, that doesn't mean that you have to be, in some way, permanently disfigured. We can rebuild. Plastic surgeons are fabulous at this, and that gives -- the bottom picture there gives you some idea of the different approaches that can be taken to rebuild the breast. Now, the other local therapy is radiation. You're going to get more of that in a moment, but this is one of those things where it's just too much of a bad thing can actually kill you and kill the tumor, because radiation exposure can increase the risk of getting cancer. But if you use a focused, high intensity radioactive beam, you can kill cancer, and that's what you see here. And so, surgery and radiation probably cure quite a lot of breast cancers of the early stage breast cancers, particularly if they haven't spread at the time of diagnosis. And what's been changing, and you're going to hear more about this, is a new way of delivering radiotherapy that greatly reduces the toxicity and the damage to normal tissue, surrounding the tumor. Now, for breast cancer, that's important. For example, if the tumor is in the left breast, if you can spare damage to the heart, which just lies just behind it, then you can kill that tumor and eradicate what was left by the surgeon that they couldn't get out and not damage the heart. And you'll hear a lot more about that proton therapy. Chemotherapy is the one most people think of when they think of cancer treatment. It's the one most people probably hope they never have to get, largely because, again, we use drugs to kill the cancer cells, and many of them kill normal cells too. Normal cells that are proliferating get targeted just the same way as the cancer cells, which have lost that growth control. And it's those killing those normal cells that causes much of the side effects that's associated with some of these chemotherapy drugs, but they cure some patients. There is absolutely an overall survival benefit for chemotherapy, surgery, and radiation therapy for women with breast cancer. And the other group of treatments that we have are those that target the estrogen receptor, because we know that the estrogen receptor is a driver of many of those 70% of breast cancers that are diagnosed new every year, and there are two ways we do that. We can give drugs called aromatase inhibitors that stop the button making estrogen, and we can give drugs called antiestrogens that compete with estrogen and stop it from binding to the receptor and turning that receptor on, and those drugs have been around and very effective for a long time. Tamoxifen was first published upon 1972, and we continue to make progress. So, one of the newest treatments that was published literally a few months ago shows that if you add this drug called ribociclib, which is a drug that stops cells from going making copies of themselves. It's -- blocks their growth. If you combine that with an antiestrogen, more women will live longer, and there is an actual overall survival benefit with this particular drug. There's a whole class of them that target this, the CDK46. They're called CDK46 inhibitors. A phase three study published literally a few months ago shows that there's an overall survival benefit when you combine it with an endocrine therapy in the metastatic site. So, while since we've seen something in estrogen receptor positive breast cancer actually improved overall survival, we have lots that improved clinical benefit that keep the disease back for a longer period of time. But this actually improves overall survival. That's one subgroup, that's the 70%. Let's look at the next one, HER2, the growth factor receptor positive one. Here we made dramatic changes also in the last 20 or so years, with antibody therapy and small molecules that target that particular protein. The one that's probably most familiar to people is Herceptin or trastuzumab, which is an antibody-based therapy that targets the receptor, and there's a significant overall survival benefit for women with HER2. Until we had a way of targeting this, the outcome for women who had HER2 positive breast cancer was pretty poor. Now, we have drugs that significantly improve overall survival, and quite recently, we've shown that if you combine these drugs, along with the standard chemotherapy, you can even push that overall survival benefit further up. So again, in the last few years, we've had a dramatic change in overall survival benefit for women with HER2 positive breast cancer. The last group we call triple negative, because they don't have the estrogen receptor, they don't have the progesterone receptor, and they don't have that HER2 protein. This is about 10 to 15% of all breast cancers, and it's the most difficult to manage clinically, and it tends to be more common amongst African American women than amongst other ethnic groups, although it arises -- it can arise in any woman. We don't have a targeted therapy like the estrogen receptor or like HER2 yet for these breast cancers, and they, if you remember that graph that showed that big bump at the beginning, that's where most of those triple negatives are. So, the risk of dying within the first five years of a diagnosis, when you have triple negative breast cancer, is amongst the highest risk of any of the breast cancer subgroups. So, we treat with surgery and radiation and chemotherapy, and there is a survival benefit for those approaches. What has recently come to the fore is immunotherapy. Now, one of the reasons that the outcomes are so difficult to manage for triple negative breast cancer is, because they tend to grow more quickly, and they tend to have more mutations in their DNA. That's a bad thing, but what the immune system often looks for and detects when something goes wrong is when there is proliferation that shouldn't be occurring, when something is growing where it shouldn't, or when it's got a lot of mutated DNA and those proteins are expressed on the cell surface and not recognized by the immune system called the neoantigens, the problem that we face is that the cancer cells turn off the immune system. So, although there's all these signals present, telling the immune system [inaudible] and get us, because we shouldn't be here, when the immune system gets there, it gets shut down. There are lots of lots of ways that cancer cells can do this, and we've only really begun to target a few of these in the last few years. But some of those have had dramatic effects in some patients in other cancers, and we're beginning to see that there may be a way to make these work in this triple negative breast cancer. We probably haven't quite got the right switch turned off to turn the immune system that the cancer cells are turning off to get the immune system back to work for everybody. But it does seem to be doing quite well. So, I probably talked longer than I should have, and I'm going to finish with this slide, because the positive things, the survival rates for breast for invasive breast cancer, which is the most common type of breast cancer that's diagnosed, the average survival rate at five years is 90%. And at 10 years it's over 80%. Those are significant improvements for women overall. The new and -- the local and systemic therapies that we have clearly are improving outcomes and clearly are letting some women live today, who wouldn't be alive if it wasn't for these new treatments. We have lots and lots of new things coming up in clinical trials. We've only scratched the surface of immunotherapy. So, there's a lot we can discover, and we have tools to make these discoveries today that we didn't have 10 years ago that are absolutely amazing. We probably just don't have quite enough [inaudible]. That being said, I think we have made tremendous progress. We're on a couple of paths of discovery that have tremendous potential to lead to new treatments and quite possibly fairly quickly. So, despite the statistics that are quite sobering, there's a very great possibility that we will transform treatment and outcomes for women in the next few years. I'm going to stop there, and I don't know when we're taking questions but-- >> Tomoko Steen: To the end. >> Robert Clarke: To the end. So, thank you for your attention. [ Applause ] >> Tomoko Steen: Thank you so much. It's very informative. So, next speaker is -- is this [inaudible]? That's the one, okay. Next speaker is Dr. Michael Dean. He's a senior investigator from [inaudible] Translational Genomics at the National Institute of Health, and he has been covering a wide variety of cancers, and I think today he's going to talk about [inaudible] cancer. >> Michael Dean: Okay, thank you very much. It's a great honor to be part of this panel, and it's incredible to be in the institution that hosts Thomas Jefferson's library. Rather daunting, I was just over there before I came here, and it's totally humbling. So, I want to take off from where Dr. Clarke started, tell you about some of the new things, new ways we're using genetics and genomics to diagnose cancer. And this is helping us more effectively treat cancer as individual patient individual tumor, and some of the focus will be on lung cancer, but much of this will work across cancer. So, the recent focus on lung cancer is the number one cancer killer, as Dr. Clarke said, more women are diagnosed with breast cancer than any other cancer, more men are diagnosed with prostate cancer from any other cancer. But what kills more men and women is lung cancer, and, like in breast cancer, we're making steady progress. We wish this curve was going up faster. This is the increase in five-year survival, but we're still at only 20 to 30% of people diagnosed today with lung cancer will be alive five years from now. Obviously, we have a long way to go to improve that. So, one of the big problems with cancer, I'll talk about a minute about the cancers that we actively screened for, but most cancer diagnosis are opportunistic. The patient comes in finding a lump or there's some other symptoms that leave the patient or the doctor to look for cancer. This is a problem, because by the time that cancer is big enough that you can feel it or that you have symptoms from a liver cancer or lung cancer, you probably already have advanced disease, which is hard to treat. For leukemia, it's blood testing, staining of blood cells that gives us the diagnosis. For solid tumors, one of the big advances are many, many different types of imaging, where we have many different tools that find cancer in different parts of the body. And then the ultimate diagnosis is a biopsy, going in taking a piece of that tumor, putting cells on a microscope slide and looking to see what we have and categorizing it. And a huge advance is the whole area of molecular pathology, like in breast cancer where we look for HER2 and progesterone receptor estrogen receptor, pathologists have multiple different antibodies that can stay in tumors there to classify them. And now, increasingly, we're taking the DNA from the two tumor, sequencing them and finding out exactly what is going on with each individual tumor. And as you'll see, this totally changes our treatment. So, there are cancers that we actively screen for, cervical cancer with HPV testing and pap smears, skin cancer mostly by the individual finding something abnormal and going to their doctor or from a dermatologist. Colon cancer is highly effective to screen by colonoscopy, by finding a blood in the stool, and we can largely prevent those cancers by these screening programs. Then there's a class that includes breast cancer, obviously, we're using image to find cancers earlier, but that is not, obviously, eliminating all cancer. We're beginning to in very high risk patients that are heavy smokers screen to find cancers early. In certain parts of the world, where there's a lot of stomach cancer, they actively screen people in the population by endoscopy to find those tumors. Last is the lowly prostate cancer where we've been doing PSA test, digital rectal exams on millions of men, but the US public health service, when they really look at that data, does that really improve mortality? The answer is not so much, and so we're left, you know, with a very important cancer without a lot of -- without a really good screen to find the cancers early. All the rest of the cancers we don't have an active surveillance problem, so a program. So, we don't have an effective method to screen everybody out there to find cancers early, when we might be able to treat them better, and so this is obviously an area of need. It's an area of lots of research, both by companies and scientists and an area where we need to make a lot of improvements. As you already know, the three main areas of cancer treatment are surgery, chemotherapy, and radiation. When we give that chemotherapy or radiotherapy before surgery, that's called neoadjuvant or after surgery that's called adjuvant. And for every cancer, there's a different program, there's a different system and schedule for what is most effective. And what's important to know, that's all been worked out mostly by trial and error, by clinical trials, and this is where there are two heroes, the clinicians and scientists who carry out those very difficult studies and the patients who volunteer their time, so we can figure out really what is the best way to treat all different types of cancer. Now, to get into more targeted therapy, we need to have a little bit more background. So, one of the things that's been puzzling, we have all these different agents that in the environment that can cause cancer. What's in common? One thing that's in common is almost all these agents cause chronic inflammation or damage to the tissue. And what happens when that occurs over decades is that the normal tissue needs to grow and repair that damage. And exactly as Dr. Clarke says, simply the act of those cells dividing when they didn't need to means mistakes get made. Those errors can accumulate as genetic damage, causing a premalignant lesion which, if that continues, can go on to a malignant lesion. And exactly as Dr. Clarke says, some of these agents actually damaged the DNA on their own. So, from studying these genetic events in cancer cells, we've identified different classes of genes that are mutated in the cancer cell. One group we call oncogenes. An oncogene is essentially an accelerator of the cancer. These are mutations that cause or create an activated gene driving cell growth and survival. So, one example, our growth factor receptors that are normally there on the surface of ourselves that normally are only active when the growth factor binds to the receptor, then they send a signal, and then they're supposed to shut off. What can happen in the tumor cell is that receptor gets mutated, and now it's stuck on. So, this is like the accelerator of a new car. You know, you push it down when you want to go, you let it up when you want to slow down. If it gets stuck, you know, down, then you have a big problem. The good thing about this is that we've come up with ways to begin to try to reverse this or block this from happening. So, many of these oncogenes include growth factor receptors. There are a class of enzymes called kinases. What they do is bind a molecule called ATP and transfer a portion of that to another molecule, typically, another protein. Now, often that's a tyrosine amino acid of another protein. So, these get called tyrosine kinases, and there's a whole class of drugs of tyrosine kinase inhibitors, which you may have heard about. So, researchers begin screening for molecules that would bind the net little pocket where ATP binds and block the molecule off, kind of like sticking a key in the lock and breaking it off, and then, you know, nobody can open the door. So, a cancer where this has worked spectacularly well is a type of leukemia called Chronic Myelogenous Leukemia. This disease is caused by an activated kinase called ABL, A-B-L. It's actually activated through a chromosomal rearrangement, and Dr. Brian Druker [assumed spelling] at Oregon Health & Science developed a molecule now known commercially as Gleevec or imatinib, which is a kinase inhibitor. It binds here in the pocket where normally ATP would bind in the ABL protein and sticks there and prevents ATP from binding and prevents the thing from signaling. Now, back when the first clinical trials were started, chemotherapy was the standard of care for this type of leukemia, and about 50% of patients were alive two years after getting chemotherapy. Interferon came along, that improved things a little, but Gleevec was really a miracle. These patients, almost all of them survived. A few of them couldn't tolerate the drug, a few of them had their tumor develop resistance, but by and large, this stabilized the drug in nearly all of these patients. Now, the treatment doesn't kill the leukemic cells. So, the patient has to keep taking this drug, essentially, throughout their life. I think there are now some patients that have been weaned off the drug, and then the leukemic cells were gone, but the drug has no really terrible side effects. So, you take a pill once or twice a day, and your leukemia is under control. I wish every single kinase we had an inhibitor like this that would have this kind of effect. But one of the things that was truly amazing, and this is where science is really exciting is this same treatment had a big advance for a rare intestinal tumor, gastrointestinal stromal tumor. This is caused by different activating kinase that are called KIT, and it was shown that the same drug Gleevec can inhibit KIT. Now, GI stromal tumors about, you know, three to to 6,000 Americans get this disease every year, equally amongst men and women, about average age of 50. And we don't know any of the risk factors for them, so we have no way to prevent this disease. It just occurs. Now, before Gleevec came along, this was not a very good disease to get. Fifty percent, only 50% of patients were alive nine months after the standard of care at the time, a type of chemotherapy. When those patients were put on Gleevec, getting either one pill a day or two pills a day, you can see they did way, way better. So, 70% of patients were alive 30 months out. Not as good a response as we saw with the leukemia, but still, obviously, way, way better than the standard of care at the time. There are now a whole host -- there's a whole library of these tyrosine kinase inhibitors that are being used on a variety of cancers, and this has been, you know, one of the major advances in therapy over the last decade. One of those areas is in lung cancer. So, one of the growth factor receptors that is often activated in lung cancer is called the EGF receptor, and there are a number of inhibitors specific for that specific receptor. There are other inhibitors that are more broad that affect EGF receptor and a family of related proteins. Once we identify a target that's effective, you know, research biotech pharmaceutical companies are great at coming along with other ideas. And so, there are monoclonal antibodies against the [inaudible] receptor, and those can block the receptor and also be affected. Now, others -- other -- one of the major types of lung cancer is non-small cell lung cancer. Some of those cancers have other kinases activated like ALK or ROS1, and they're targeted drug specific for those. Now, typically, a given lung cancer will typically only have one of these receptors activated, and there's no way to know from looking at the scan or even the pathology slides saying there's malignant cells which one it is. We have to have a molecular test, either to sequence of DNA from the tumor or some other antibody test to figure out whose tumor has EGF receptor, whose tumor has ALK and give them the proper drug. But one of my professors in university is a woman who never smoked, who had lung cancer. Her tumor has one of these receptors activated. She has gone into complete remission, takes a pill twice a day, and I just saw her a few months ago, and she is perfectly healthy so far. Now again, as Dr. Clarke said, if cancer only stopped here at the local malignant, you know, the local growth where it started, we wouldn't have a big problem. The big problem we have with cancer is a lot of most cancers become resistant to therapy and or become metastatic. And so, this is an area where a lot of focus needs to be put on how do we understand these processes, and then how do we use that understanding to try to come up with better treatments? One of the things that we have learned is that although there's genetic damage that we can recognize to get us to this stage, it seems to be mostly nongenetic, but aberrations of gene regulation that get us to these later stages. This is called epigenetic, because it doesn't involve changing the DNA it causes -- it's involved changes in the regulation. Now, you have about this much DNA in every single one of your cells, and how that gets packed in is that DNA gets wrapped around proteins called histones, and those get packed up together in a structure called chromatin, and this is highly regulated. So, in your pancreas cell, your insulin gene may be, you know, unpacked and available, when you need to make insulin. None of the other cells in your body need to make insulin, so that gene is sort of packed away and inactive. What we found is that many tumors actually mutate the genes involved in this kind of regulation, and so we have a whole new class of genes that are altered in cancer that are involved in remodeling that chromatin that are involved in regulating entire programs of gene expression. We're only at the very beginning of understanding what the implications of this are, but we are starting to see therapies that are in this class. But let me back up and sort of explain sort of why this makes sense. So, all animals start off as a fertilized egg, grow into an embryo and a fetus, and then the adult, we have hundreds of different cell types. We have blood cells, neurons, muscle cells, kidney cells, et cetera. All of those cells, with a few minor exceptions, have exactly the same DNA, as did the original fertilized egg. The reason all these cells have different properties is that they're regulating their genes differently. They turn on different sets of genes and turn off other sets of [inaudible]. If we lay that concept onto the cancer cell, yes, we have these genetic events that get us to the original malignant cell, but then what we think may be happening is that the cancer cell can then turn on, if it needs blood supply, because it's got too big and it needs its own blood vessels, it turns on the gene to generate blood vessels. It turns on the gene to migrate to other parts of the body. It can turn on the genes to become resistant to the immune system resistant to therapy, and because a large tumor has millions of cells, if every one of those cells has the possibility to turn on in programs of gene expression and turn on different properties, then you can begin to imagine of why it is such a challenge to completely eliminate cancer, especially once it grows big. I like to think of this as the tumor developing the ability to unlock the genome to make all of the tools that are in the tool chest of our genome available for its use. Now, this may overwhelm you. It overwhelms me. There are days I think this is so incredibly complicated, we'll never be able to figure this out, but then I think of how innovative human beings are, that once they understand exactly how a problem works, they can figure out ways to fix this. So, I'm excited. I think we're finally at the part that we understand how cancer works, and now it's the next phase which is, you know, I won't kid you, it's going to be difficult to figuring out how do we attack this, but we are starting. Here two FDA approved drugs, they're called HDAC inhibitors, histone deacetylase inhibitors, and they work by altering gene expression patterns in tumor cells. So, here's two that have been used on a rare form of lymphoma that affects your skin. Here's a patient before and after treatment with one of these drugs, and you can see things have gotten a lot better. Now, these are early days and, you know, the response rates are, you know, are measured in months or a year or so. But this is the beginning, I think, of developing drugs that we're going to use to change how cancer cells express genes and hopefully be another tool that we have to fight this disease. Dr. Clarke introduced the idea of the immune system. The other tool that we have is the immune system can regulate abnormal cells throughout this entire process, and if we can harness that, we may be able to eliminate cancer cells. So, the proof of concept that this really would work has come from Stephen Rosenberg, right here [inaudible] at the NCI. He has been, for decades, taking, for instance, melanoma tumors from patients, isolating from the patient their very own T cells, identifying those T cells that will react against that patient's tumor growing up in big labs, and I've been to this lab, and I've seen it, it's totally phenomenal, and then infusing those back into the same patient. When it works, it's spectacular. Metastatic tumors all up and down the patient's body disappear and don't come back. Now, he, you know, he announced that [inaudible], you know, I think to do this on 10 or 20 patients a month, it's not something that we can do across millions of patients across the country, but it shows the power of what the power of the immune system if we can properly harness this, we can get rid of cancer. Here's one of those patients, you know, of they were completely filled with tumor before treatment. Here's a few weeks after treatment, those tumors are mostly gone. Here's a few years after treatment, and the tumors still have not come back, but I mean, what we obviously want is some way to do this across many patients, and we know that our cells have mechanisms to suppress T lymphocytes from killing our cells. We have to very carefully -- the body has that very carefully control the system not to develop autoimmune disease. You don't want your lymphocytes killing your own cells, and tumors have been able to use those same pathways to activate what are called immune checkpoints to survive. So, tumor cells are turning on many of the same inhibitors of the immune system, but once we've recognized that, we've come up with ways to fight this. So, here's a T lymphocyte that's recognizing a tumor as foreign. It's all primed to kill it, but the tumor cell has turned on this other protein, PD-L1, which binds to PD-1 of a T cell and keeps it from being activated, keeps it from killing the tumor cell. Well, we can come in with antibodies or therapeutics to either bind up that PD-L1, so it can't bind to the T cell's PD-1 or bind a PD-1 and a T cell, so it can't bind to PD-L1 of the tumor cell. And when that works and the T cell is now activated and can go back to what it wanted to do and kill the tumor cell. Here's one of these early trials with an inhibitor, PD-1 and lung cancer. All these patients in blue have had their tumor shrink. These patients in gray have had their tumor stay the same size, and these patients in red, their tumor progressed. And you can see their survival, all the patients in blue were alive 12 months later, most of the patients in this gray area were also alive 12 months later. These therapies, these checkpoint inhibitors have had many very impressive effects on cancers. They typically tend to be in cancer with lots of mutations, and there are lots of immune cells out there that can recognize that as for. So, lung cancer, melanoma, but also bladder, stomach cancer, there's about 10 or 12 cancers now where there's an FDA approved drug from one of these classes that is working. And now every pharmaceutical company in the world has a drug part of this class, and they're all trying to figure out where's the best place to use this. We first started using this on patients with very advanced disease, who, you know, who were not responding to any available therapy. Now, we're going to start using this more and more in earlier stage disease and figuring out, again, the schedule, when is the best time to give the immune therapy. Maybe it's best to give that first, activate the immune system, and then use the other therapies we have. That all has to be worked out one by one, through the different cancer types. So, moving forward, we have a number of really tremendous challenges. We're starting to come to the idea of thinking of the tumor as a multicellular 3-dimensional entity, more like an organ than just a group of cells that are all the same. And so, that changed our perspective on how -- what we need to understand and what we need to go forward. We need to understand how tumors are changing their gene expression, how this occurs over the evolution of the cancer, and how might we be able to intervene with that to shut off the tumor cell or get it to differentiate and die like it is programmed to do that. We need to understand much better the immune response of the host, how the tumors -- how the patients that didn't respond to checkpoint inhibitors, what have, you know, what's different about those patients and the ones that responded, and can we use this to develop more tools, using the immune system to [inaudible]? And all this knowledge, obviously, we want to use to better diagnose and treat the cancers that we failed to prevent. Now, because we're in a library, I have to give you some resources and also some homework. Cancer.gov is the National Cancer Institute's website. Cancer.org is the American Cancer Society's website. Both are completely outstanding resources, if you want any information about cancer in general, any specific tumor. Clinicaltrials.gov is a website where you can search for any clinical trial in the country or actually in the world, if you want information. Patients treated in Bethesda at the National Cancer Institute, if they're accepted in a clinical trial, all the care and travel costs are free. So, if you have friends or relatives anywhere in the country that might qualify for a trial, you can make this available. There are a number of popular books out there. "The Emperor of All Maladies", I think, you know, has been super, super popular, and it's very, very good, and Dr. Clarke spoke of Herceptin. This is an outstanding book. It really gives you the entire story of how hard it was to develop Herceptin of the heroics of the doctors who developed this, who set up the first trial of the first patients who'd volunteered, people at Genentech who fought to keep that program alive, even AIDS patients who told breast cancer patients you've got to go out and protest, you've got to go out and fight for this drug, so that it gets in there. This is an excellent book on immune therapy that that came out fairly recently, that tells the story of how it happens. This is not a cancer book. This is Richard Preston's latest book about Ebola, but it's a phenomenal story of how public health science medicine came together to prevent what was a terrible outbreak but which could have been a disaster for the entire world. And thank you very much, and if you don't know what -- if something funny -- mole looks funny on your body, if you don't know what you should be looking for, here are those pictures. Thank you. [ Applause ] >> Tomoko Steen: This was a great example of translational medicine [inaudible] genetics to [inaudible]. So, next speaker is Jonathan Lischalk. Dr. Lischalk is a clinical instructor at the Department of Radiation Medicine at the Georgetown University Hospital, and you are going to hear about proton therapy now. >> Jonathan Lischalk: So, thank you for the introduction. My name's Jonathan Lischalk. I'm a radiation oncologist at Georgetown, and we just had two really great lectures to give us a background of what we're dealing with here in oncology. I'm going to be focusing on a specific type of cancer treatment, and it's radiation, and within radiation, we basically primarily utilize X-rays to treat cancer. That's usually what we're talking about, but an exciting, not necessarily new, but type of treatment that we have available now [inaudible] is called proton therapy. And that's something that I use a lot of Georgetown now to treat patients. So, this is a outline of the talk. I'm going to start by discussing some of the history. I think it would surprise a lot of people that this type of treatment goes back many, many years, almost all the way back to the discovery of X-ray. We'll talk about the physics of proton therapy. I'm going to try not to bore you, but this is really where the bread and butter kind of differentiates X-ray-based treatment from proton therapy. We'll see a little bit of the modern evolution towards proton systems and centers that we see today. We'll talk a bit about why this is an important type of treatment relative to X-rays, when we would it use it [inaudible] why it may be beneficial, and then finally, our experience haven't been open for over a year now at Georgetown [inaudible] treat patients. So, starting with the history of proton therapy. To give you a bit of context, the X-ray was discovered in the 1890s in Germany. So, only two or three decades later, Ernest Lawrence was a scientist out on the West Coast, invented the cyclotron. And the cyclotron is essentially a machine that accelerates a particle, and shortly thereafter, Lawrence Berkeley Labs built one of these machines. Now, the first clinical kind of idea for using this for cancer treatment came out of this paper here by Robert Wilson in 1946. And about three years later, at Harvard, which is where Dr. Wilson was at, commissioned the first cyclotron. Now, the first patients were treated in 1954 and then in 1961 at Harvard, and for decades and decades, there were only two facilities in the United States, one on the West Coast, one on the East Coast, that were able to treat these patients. In 1988, FDA approved proton therapy as a form of radiation to treat different types of cancers, and about a year later [inaudible] California [inaudible] open the first hospital-based system. Prior to that, these systems had been more closely aligned with the Physics Department of Harvard and [inaudible] out of Berkeley Labs. But this is the first hospital-based center. So, you can see, for many decades, this was a type of technology that was just not available widely. These are some interesting photos. This is the first cyclotron that was built by Ernest Lawrence out there in California Lawrence Berkeley Labs. And about a decade later, the Harvard cyclotron was commissioned and then taken over to the campus at Cambridge. So, what about the physics of proton therapy? In radiation oncology, we primarily treat patients with X-rays, and if we remember high school physics, the proton is part of the nucleus. It's in there with the -- it's in there with the neutron, and then electrons are running around, but a hydrogen with an electron removed is what we're using when we're using proton therapy. And there's a lot of different machines that accelerate this particle, and the reason why you need to accelerate it is compared to an X-ray, which is essentially massless, a proton does have a mass. And so, you need really big heavy equipment to be able to accelerate that proton, and this on the right side here is an example of one of those types of machines. It's a cyclotron where you basically have two magnets that are alternating and charged and kind of accelerating an electron that's placed right at the center of those two magnets. And then as they speed up and speed up, you get to a point where this proton is now of an energy that's high enough to be useful clinically. This is another form of an accelerated -- this is a synchrocyclotron. It's one of the more modern ways to accelerate a proton, but essentially, these are accomplishing the same thing, where they're accelerating this proton, this particle to be able to penetrate into a patient in through the skin and enter into where the tumor is and then deliver the dose to kill those tumor cells. Now, the energy of a proton is very important, because it dictates the depth to which that it can reach. So, if you have a tumor that's very deep in the pelvis or deep in the lungs, you need to have very high energy proton to penetrate through that patient's tissue and ultimately reach that tumor. So, you can see here on the X axis, the energy of a given proton dictates how deep it can penetrate relative to water, which is oftentimes will be used to measure a tissue equivalency. So, to get up that to that energy, you did need these huge machines to accelerate that particle and use this technology to treat patients with ligands. Now, this is probably one of the most important slides in my talk, and it shows the comparative dose distribution for the most common type of radiation treatment that we have, which is the X-ray here in the blue line relative to the proton here, the green line. And what we're seeing here on the X axis is depth. So again, depth and tissue, you can think about this as particle traversing through somebody's body and getting into the tumor. And then on the Y axis, you can see the dose distribution. Now, if you looked at an X-ray, the X-ray comes in, and its dose maximum is relatively superficial, and then it continues through a patient's body and exits out the other side. It's like why we use chest X-rays to evaluate patients that have [inaudible] problems, we have CT stands, but when we use these therapeutically, the X-rays do get to the tumor, but then they continue past that target and outside. The difference with the proton is that it can come in, oftentimes with a lower energy, and because this is almost like throwing a bowling ball through a room, it doesn't slow down until it gets to the range of the proton. And once it gets to its range, again, dictated by the energy of that proton, it starts to very rapidly deposit its dose. You can kind of think about this area as the location of the tumor in a patient's body, and it rapidly deposits its dose, and then it stops, and it goes no further. So, you have this area of no radiation exposure. So, you're effectively able to decrease the amount of radiation that you're using to kill the tumor, while still killing the tumor, by 50 to 60%. It's almost like if you were giving somebody chemotherapy for leukemia, and then all the sudden you could drop that dose by 50%, and then you could achieve the same outcome. And so, this is why it's a really exciting form of technology in our field. So, what were we using this for the first several decades in treating patients? Well, you can imagine that 50, 60 years ago, the technology was really not there. We didn't have CT scans or MRIs, computers, or planning software. So, we were very much limited in the types of cancers that we could treat. One of the most common things that was being treated in -- at Harvard and Berkeley Labs were tumors within the brain, so central nervous system tumors. And by 1975, they'd actually treated a very -- one of these rarer tumors called a pituitary adenoma at Harvard almost up to 1,000 patients. And so, these were tumors that were easier to treat, because it was simpler to set a patient up and make sure that they're not moving, because it's easy to see on an X-ray, the skull anatomy. The tissues were not changing, there was no motion like breathing. If you're trying to treat a lung tumor, that tumor can move, as a patient's breathing and try to manage a proton beam with a moving target is very challenging. So, in the early era of proton therapy, these were limited to only a few sites of cancer that we could treat. So, what about modern proton systems? We've come a long way since the era of just having two facilities, one on the West Coast and one of the East Coast. And really, if you look at the evolution of centers, after Loma Linda opened in 1990, you didn't really have another one open up until MGH opened theirs at Boston in 2001. A couple more centers coming online of the subsequent 10 years, but really just a handful of centers across the country compared to today, where we're seeing almost over 30 operational proton centers. So, this is a form of technology that has become more ubiquitous, and that's an improved patient access, which is very, very important. When you're getting treated with radiation, oftentimes you have to come in for a week, two weeks, six weeks, nine weeks of treatment, Monday through Friday and to try to have a facility that's very, very far away from somebody's home is extremely challenging. So, this has changed our ability to offer this type of treatment to patients across the country. One of the other important aspects to all early proton centers was that they were very, very, very big. This is actually an interesting example of the Heidelberg Ion Therapy Center where I worked that has a facility, and this is a man to give you kind of context of the size of this facility. Now, these things were three, four, five stories big. This blue thing here is called the gantry. What the gantry is, it's basically a rotating machine that aligns that proton beam up, so that it can come in from different angles to enter a patient for a given tumor. The geometry [inaudible] into the tissue and how the patient is lined up is very, very important. Now, this facility actually treats patients with carbon as well, and that's why this gantry is so large, but these are huge facilities, 100,000 square feet facilities that you need oftentimes over $250 million dollars to invest in these types of facilities. And this is kind of thet raditional model of a proton therapy center. To give you an idea of what this looks like, schematically, that accelerator that I mentioned at the beginning of the talk, you'd have one of those kind of -- like maybe two, three rooms away from where a patient was actually getting treated. This accelerator would then accelerate a proton into what's called a beamline. So, it gets to that useful speed, it's pulled out and extracted, and then there's potentially one, two, three, four, five other rooms or gantries similar to the photo that I just showed here. This is a gantry, where you could treat patients. Now, these aren't autonomously functioning rooms. They actually are feeding off of the same accelerator. So, you can't treat all these patients at the same time, but these facility's really big, and it was really challenging a medical center to want to invest in what is -- would easily be the most expensive piece of medical equipment, over $200 and $250 million dollars oftentimes. You could imagine the amount of energy that would be required to maintain a facility like that. So, these -- this conventional model of proton therapy center, I think, was really challenging to became ubiquitous throughout the US. They were so expensive to invest, and now this technology has improved, and costs have come down. We've been able to invest in these from a lot of NCI designated cancer centers can now [inaudible] open these centers up. So, we're in Washington DC right now, obviously, we're limited in space both up and down, we have limits in where we can build. And out where we work in Georgetown, there's even less space. So, one of the things that we wanted to do when we invested in proton therapy was have that center on our campus, so that we could [inaudible] treat a lot of patients that get chemotherapy at the same time. So, working with our medical oncology colleagues, who are directly on site or surgical oncology colleagues, who are on site, rather than have a facility 10, 15 miles away, where patients are being seen at the same time. This is something that we wanted to focus on, so that could be a kind of all-inclusive care and not require that massive upfront investment cost and also be a more of a patient centered form of treatment. So, our facility is actually right on the campus, right within the Lombardi Cancer Center itself. So, patients come in, they get the radiation treatment and they're getting chemotherapy at the same time. They go up one floor, and they're already in that [inaudible] in the infusion center. So, I showed you a photo of the accelerator that the kind of the graphic of an accelerator, but this was a picture of one of the early accelerators. It was 250 tons, a really large piece of equipment, and as time has gone on, those accelerators have become more and more advanced, smaller and smaller. And it's allowed us to minimize the size of those facilities as well to the point where we're now with a facility over at Georgetown it's only three stories high. You can see that a patient brought into this room here, and I'll have another couple of photos to show you what it looks like in this room. And that's the three-story facility above and below, because the gantry is actually rotating right here, and the accelerator is placed onto the gantry. So, that's why we're able to kind of fit this piece of equipment right into the Lombardi Cancer Center and treat patients right there in our own department. So, if you were to walk into the treatment room and take a look at the equipment, this is basically what you would be seeing. This photo is basically this portion of the room right here. So, if your patient is getting treated with proton therapy, you would walk in, you would lie down on this table. And this is what we call a treatment couch. We have various types of equipment to kind of immobilize the patient, so that they're not moving around. You can imagine we want to be on the order of a couple of millimeters of accuracy here. So, if we're treating a head, neck, or brain tumor, we'll have to create a patient specific mask, and we'll put that onto this table, so the patient is kind of fixed in place. Now, this is what we call a robotic couch top. It's kind of cool. It basically has six degrees of motion adjustments, so that we can get that patient lined up once they're on the table into the position and adjust on the order of a couple of degrees or a couple of millimeters with this robotic couch top. Now, the location where the proton is actually coming out is right here. This is what we call the treatment head, and what's cool about our facility is we, right here, there you can't see it, but there's little leaves that come in and out and shape that proton beam, as it's entering the patient, so that the lateral aspect of the radiation field can be very sharply, I mean, kind of personalized to the patient's anatomy and the tumor. Now, depending on the location, whether you need to treat a patient from the front or treat the patient from the back, this gantry is going to be rotating about 190 degrees. When that happens, this thing right here basically goes above or below the treatment level. Now, once the patient's set up, we'll oftentimes get a CT scan, and this looks at 3-dimensional anatomy. So, if we're treating a lung tumor, we get the CT scan, we line it up, we make sure that the breathing is within a certain degree that we've already kind of compensated for, that there's no other surprises before we treat the patient, so a lot of image guidance in terms of setting the patient up and making sure that we're hitting the right spot. This is a different view, kind of this is from the treatment head. You can see the gantry here, again, the patient couch top. Back here is where we have the radiation therapists and physicians. This is where they're getting images to set the patient up, and this gives you an idea of what goes on in the background here. These are X-rays that are obtained for patients being treated. We line these X-rays up with the radiation plan that we've developed prior to the patient getting treated to make sure that we're in the right spot. This is, for example, a CNS tumor here, and they're getting their -- the bones and their head lined up to the X-rays that are being [inaudible] with this X-ray panel that kind of comes out before the patient's treated, and this is one of those masks that I mentioned before. So, that's kind of the evolution of the systems and the facilities that we've treated. How does this type of treatment relate to our bread and butter X-ray type of therapy? Well, the basic idea goes back to that first physics slide, which is the proton comes in, it gets to the target, and then it stops and goes no further, and that's basically called the lack of an exit dose for a proton. I think this is best illustrated here. This is a type of tumor that pediatric patients get, a medulloblastoma, where you have to treat the whole brain spine. Now, with X-ray type of therapy, which is shown here in the middle, you can see that to be able to do this, because you don't have a Bragg peak or that lack of exit dose, you're radiating in all of this tissue, lungs, heart, GI tract, all of this tissue is receiving radiation. You can see the excess radiation here when you compare it to a proton plan that is achieving the same type of cancer control or cancer outcomes, but sparing all of these anterior structures, and what that means is that you're reducing the risks of side effect profile risks down the line for patients that are young. Young patients with these types of cancers, they're at much higher risk of developing maybe a tumor from radiation. Radiation can cure cancer, but there's a risk that you can cause cancer. So, to reduce that risk, you want to get radiation dose down to zero for all of these structures. You really just want to target where the malignancy is. So, we have a lot of publications over the last several decades that have demonstrated improvements in the dose, improvements looking at how when we create these plans, we're using software to kind of create a radiation plan that's specific to a given patient's tumor. And when we compare the proton plans to photon plans or X-ray plans head to head, we have a lot of data that supports improvements in the dose. The challenging part is trying to improve the clinical outcomes also will improve with reductions in dose to these normal tissues. What's interesting is that there's certain areas in oncology where we've already decided that protons are better, and we don't need to use a randomized trial to prove that, and one of those sites is pediatric patients. I think if you had a child and you saw these two dose distributions, you wouldn't need to put them on a randomized control trial. You would say there's no equipoise for that. We don't want to radiate these normal organs, and we've done that in radiation oncology for pediatric patients. Now, in adult tumors, it's a little bit more challenging, especially depending on the site of the tumor and the age of the patient. So, what we're trying to figure out now is what is the clinical -- the real clinical benefit in what we see from a dose symmetric improvement here. I'll give you some other pictures. This is a pituitary adenoma, and what I'm showing you here as proton therapy is advanced, we've seen the dose in the surrounding normal structures. And let me kind of give you some landmarks here. This is the tumor here in the red, and all this dose out here is stuff that really doesn't need to be there, okay? But depending on the radiation technique that you're using, you have to get the dose in somehow. Now, this is an X-ray-based plan, and you can see that there's different beams that are coming in and converging on this area. But there are all these normal structures here, including brain temporal lobes, the optic nerves, that are receiving radiation and if we can do a better job of reducing that dose of radiation in those normal structures, maybe we can reduce the side effects as well. And I think as you compare the evolution of proton therapy, this is an older form of proton therapy, and then a newer form of proton therapy and really the newest form here, and I'm not going to get into the details of the physics on how this works. But you can see this -- the surrounding halo of radiation, reducing down as you kind of advance even within proton technology. Now, depending on the site that you're treating with radiation, you can see various clinical benefits and using another brain tumor as an example, say, this is a low grade glioma or a tumor of the primary nervous system in an adult. You can see that, as you compare an X-ray-based plan to a proton-based plan, there's a lot of reduction in normal dose, and we know that brain tissue is very sensitive to doses of radiation, all the way down to very low [inaudible] doses of radiation. Patients can have neurocognitive effects, you know, it can almost cause a type of dementia to occur earlier in patients. You can have endocrinopathies when you radiate somebody's pituitary access, hearing loss, optic problems, cataracts, secondary malignancy. So, the results of having this excess dose can lead to this. Now, moving on to other sites for lung cancer, which we heard of, number one killer in the US, locally advanced lung cancer in particular. We know that radiation dose for patients that have lung cancer is very important to consider when you're thinking about the dose to the lungs and the dose to the heart. These are patients that might have been smoking for many, many years, and they don't have the same tolerance of side effects as younger patients do. So, this is an -- kind of a cool plan that I wanted to show here for a patient that had a large, a locally advanced lung cancer, because these two beams are coming in here from the back and then one slightly from the front, this is a side view of the heart, and the tumors over here. And what's kind of amazing in a patient that has lung cancer and a lot of cardiac disease and lung disease, this is able to come and stop and go no further and reduce the risk of any cardiac side effects for this type of patient. Similarly seen in a different type of thoracic tumor, thymic cancer, which is a rare tumor at the front of the chest, you can see the same idea here, except kind of flipped, two beams coming in from the front stopping, going no further and then blocking all this heart out here, reducing the risk of a patient's risk of cardiac toxicity down the line. Same thing can be seen for other sites like lymphoma. This is a younger patient with lymphoma that I treated, and, in this case, any tissue exposure for a young patient is going to reduce the risk of a secondary cancer from radiation 10, 20, 30, 40 years down the line. And then more complicated cases where, say, this -- this is an instance where a patient had a lung cancer before they got radiation and chemotherapy to cure it, but then it came back. A lot of the times, they wouldn't have any options for curative treatment, but we have the ability to get radiation dose and treat a tumor that's recurred already in an irradiated field, which is a very dangerous situation and do this while staring all these normal structures including the spinal cord. So, this is a form of technology that is really exciting to have as a complimentary form of treatment in our department. So, the -- I think the question, which is a very complicated question to answer, and I have trouble answering this on a day to day basis, and sometimes I actually just have to create plans, both X-ray-based plans and proton plans for a given case to figure out which is better in which situation. Now, this was published in the JCO about 10 years ago, and these are just kind of generalized bullet points when you would consider protons being better than X-rays. There are some cases where X-rays caused a lot of problems, that cause a lot of side effects, and we want to reduce those side effects, and this is one way to do it. When the normal tissues are so sensitive to X-rays that you want to reduce any radiation dose, this is -- a good example's radiating somebody's brain or treating a pediatric patient, protons might be a better idea. When you need to increase the radiation dose, because a tumor is just so aggressive that it doesn't respond to standard doses of radiation, sometimes you can achieve this with this type of technology, [inaudible] radiation cases which I already mentioned and then also pediatrics, which I think is the most commonly used site. So, just briefly, in the last few minutes, this has been our experience at the Proton Center at Georgetown over the last year and a half. We've been using this technology to treat almost every site in the body, and so it's been a very heterogeneous group of patients. I think if you look back in the 1950s and 60s and 70s, when this technology was only available in a couple places in the US, where they were really just treating a couple of sites, this is where we're going with this type of technology to be able to have advanced treatment planning and delivery techniques that we can treat, really any cancer anywhere in the body, brain cancers, gastrointestinal tumors, thoracic tumors, genital urinary really across [inaudible] including breast cancer here, really across the board. And this is what we're hoping to offer to our cancer population. So, I'll conclude by saying this is a exciting type of radiation treatment that's actually been around for a very long time, but is now becoming more available for patients, both in the United States and abroad. Our advancements in technology and the cost and our ability to integrate this into an existing cancer center have allowed a lot more centers to adopt this type of technology. I think this is a great complimentary form of radiation to have at a radiation oncology center. I don't think this is going to completely replace X-rays. And, in fact, there's a lot of cases where I prefer to use X-rays, but this is an excellent option for a lot of clinical situations, pediatrics, of course, taking the highest priority. And then we have a lot of ongoing trials, looking at proton therapy versus X-ray therapy head to head which is better and a lot of future challenges as well in terms of treatment planning, managing motion, as I'd mentioned earlier is a big challenge. And, of course, we always have challenges with insurances, as physicians in oncology. Insurance coverage, I think will be an issue in the future as well. So, thank you for your time. [ Applause ] >> Tomoko Steen: Very interesting. Next speaker is going to talk about gut microbiome. We have two lectures already we sponsored for the microbiome, especially gut microbiome, from the supervisor actually, who was Dr. Roy [inaudible]. >> Soumen Roy: Yeah. Cancer, yes. >> Tomoko Steen: Is going to talk about side effect of cancer therapy using gut microbiome. >> Soumen Roy: Thank you so much. So, good afternoon, and I would like to really thank Dr. Steen for inviting me, Library of Congress to sharing my research. And before me, the other three panelists, Dr. Clarke, Dr. Dean, and Dr. Lischalk [inaudible] giving a wonderful foundation and information, different kinds of cancer and the therapy options. So, that made my [inaudible] much easier to give you the other aspect of the cancer therapy that how, you know, you can impact the quality of life of cancer patients. So, one of the thing in the cancer survivors and the quality of life by the development of different cutting edge therapy options, the percentage of cancer survivors has increased really, really high in the last couple of years. And especially, you know, the statistics number like yesterday like based on January 2019, it is estimated that there's like 16.9 million cancer survivors in the United States from the different cancer therapy [inaudible]. And also, it is estimated that by 2019, 29% increasing [inaudible] should become like 21.7 million cancer survivors. So, there are different therapy options, as the previous speakers mentioned that chemotherapy, radiation therapy, and also immunotherapy, and all of this therapy has severe side effects, not all, but most of them. And those side effects cause, like, nausea, vomiting, and gut toxicity, and also it causes nerve toxicity and anorexia, which is like [inaudible] which they lose the interest of eating food, they lose weight [inaudible] a lot of toxicity during the therapy. Once the patients are cured, they can get on to other kind of toxicity. So, today I'll focus one of them, so in terms of side effects, and that is communication. So, communication is the key component of life, and we all communicate by talking, by writing letters, and discussing and sharing our stories. And one of the interesting thing I came across recently from one of the articles in [inaudible] of society that before the fire was, you know, invented the primitive people, like, they didn't know what to do in the evening. And once the fire was there, they just started [inaudible] sharing a story, and communication was -- became one of the biggest thing and born and forming the social structure. And if you look someone who's communications and the mode of communication that impacts the quality of life, and one of the communication -- the mode of communication is hearing, and then hearing loss in nearly 750 million adults worldwide, and those are experiencing hearing loss. And one of the biggest inducer of this hearing loss is one of the drug called cisplatin. So, cisplatin was discovered by Barnett Rosenberg in 1965, and it was approved by FDA in 1978. It is in the market and the clinic, lasting more than four decades, still like the efficacy, and the side effects are, like, really [inaudible]. So, cisplatin is to treat like lung cancer, testicular cancer, bladder cancer, solid tumors, and many other head and neck cancers. And you can see, like, it goes, and it leads to many side effects. One of the biggest side effects is that hearing loss. So, I started working at NIH and the National Institute on Deafness and other communication disorders before joining [inaudible] I studied how chemotherapy can cause hearing loss and what could be the protective mechanism, and others are like nerve toxicity, and patients experiencing, like, heavy toxicity in the kidney and a lot of time due to kidney failure the therapy has to be stopped, although the efficacy is great. And others are like [inaudible] neuropathy, so the -- although there are some patients, those who are treated with cisplatin, even if the cancer is cured, after many years they feel the pain in the nerve. So, there are, like, a lot of research is going on to understand what is the cause of these side effects and how to treat that. So, I asked questions, like, in my previous institute like how -- what is the mechanism of [inaudible] causing the hearing loss? So, you can see the sound travels through the outer ear, and it vibrates this bonds, and then this physical -- the sound energy goes through this tunnel, and this is the inner ear. And this then converted to electromechanical -- electromagnetic energy and goes to the brain and then [inaudible] here. So, if you do a cross section in this -- the inner ear called cochlea, and you see this kind of structure, and when -- once you give -- and this is the structure, when you see the inner ear, is it technologic -- the architectural [inaudible], and it has three rows of outer hair cells, and one of those [inaudible] hair cells will [inaudible] I'm talking with you this [inaudible] membrane is vibrating, and this, an electromagnetical energy goes in -- go -- is going to your brain, and you can hear it. So, if you give cisplatin -- so after cisplatin administration [inaudible] given systemic -- thorugh systemic infusion, and it goes to the ear, and there's a portion of the cochlea called stria vascularis [inaudible] enters in this tunnel, and it kills those hair cells. Just to show you one of the figure how it looks like. So, this is the cochlear in mice, and these are the three rows of outer hair cells [inaudible], and you see how the hair cells are missing. And nearly 50 to 80% of the cancer patients, those who are treated with cisplatin become death permanently. And although you might think that, you know, there are so many other options now, why cisplatin, you know, cisplatin is one of the cheapest drug in the market, and it's so effective. New -- recent therapies are coming up -- they're also taking to combine cisplatin along with the immunotherapy, so that they have less toxicity, while still long [inaudible] long way to go. Okay, so what could be the protective strategies? So, what happens is, in case of hearing loss, once the [inaudible] is in the system, so either you could do repair those outer hair cells, those are missing by [inaudible] by targeted therapy, and those are like -- none of [inaudible] therapy can do [inaudible]. I will give the cisplatin inside another [inaudible] specifically to the tumor, so that you can relieve the system [inaudible]. But so far there is no drug in the clinic which can cure [inaudible]. And another possibility for would be intervention. So, how that would be, you know, reduced. So, I was thinking like, you know, once you persist [inaudible] in the patients, there's like a window, the hearing loss doesn't happen immediately. It takes some time, like, you know, sometimes it's one week to a month to start losing high-frequency hearing loss. So, I was thinking, is there any human [inaudible] that could be responsible for causing this hearing loss? So, in case of [inaudible], you see, [inaudible] deficient and it should kill the tumor and goes to the tumor and then tumor killing. And in some cases, it can cause resistance, and -- but what happens is, you know, earlier Doctor Clarke mentioned that it not only does kill the cancer cells, it also kill the healthy tissue. And it can cause [inaudible], in some cases -- also in the ideal cases should clear of the unnecessary drug and reduce the toxicity. Then after giving [inaudible] in the system, it cause [inaudible] toxicity, and it impairs the barrier of the gut lining, and there's a lot of toxicity happens. And then patient experience diarrhea, and I'm losing off electrolytes. And another toxicity is [inaudible] as I mentioned, and then cachexia, which is like really the muscle wasting. Patient become really thin and [inaudible] toxicity. So, why [inaudible]? So, one of the first site of injection of [inaudible] compared to like a lot of microbiota around like 2.5 pound and this is recently considered as another [inaudible], because they regulate pretty much everything now. And so, the first question that I ask, does [inaudible] regulate [inaudible] in the cochlea? So, to understand that then I move from the Hearing Institute to the NCI [phonetic]. And in 2013, a very [inaudible] from my current supervisor [inaudible] promised that came out that commercial bacteria is gut [inaudible]. They control cancer response therapy by modular [inaudible] or microelement. And at that time, I was working with [inaudible] and then I thought to talk to him and then continue this research to see if we can modulate gut microbiology to reduce the toxicity of this [inaudible] drug. So, what are the microbiota? So, [inaudible] with microbiota partners, and, you know, we are composed of specific bacterial [inaudible], cortisol, fungi, [inaudible], and [inaudible], and it actually -- microbiota not only is in the gut, but it also -- it's kind of covered in the entire external surface of our body. And it regulates the different barrier surfaces in the gut, and there is numerous -- more than the human cells and the DNAs called microbiome. And they come to nearly 100 time more genes than the human genes. So, what are the microbiota? So, they're the key orchestrator. So, in a recent bit of news, a paper [inaudible] microbiota nearly regulates everything, every disease. And so, one thing is like in the hearing, it's nothing has done yet, so I'm kind of investigating now. So, one of the thing that regulates microbiota, the age, and mode of delivery. Like how the babies are delivered or that they're from the C-section or from the normal delivery. And there's some studies showing that the babies -- those who are delivered through C-section could be -- more tend to [inaudible] chances of there. But there's no like [inaudible] data applicable, but a lot of studies are going on on that direction and then also, of course, genetics. Also, recollect the composition of gut microbiota, and what else? So, the geographical location and the lifestyle, mutation has a huge impact on the composition of gut microbiota kind of food that we eat, like high fat or the vegetarian or the Mediterranean food, the hygiene, and also like the probiotics. There are a lot of -- the counter in the pharmacies, you can buy probiotics, and really, we do not have much information how these probiotics can interact with our immune system or with the other microbiota. So, the caution has to be taken, but it will take [inaudible] probiotics. And also, antibiotics; it impacts heavily the composition of gut microbiota, especially in the developing countries. So, the antibiotics you can buy in the clinic without a prescription of the doctor, and then there's no regulation on that [inaudible] infections and disperses and different disease also change the composition of gut microbiota. And that can imbalance the composition of microbiota, and it can, you know, promote [inaudible] cancer and other diseases. So, in that paper what the [inaudible] soon, before I joined the lab, that they introduced, you know, the tumor into mice. And then the treatment -- the mice with the platinum compound. There are two kinds of drug, oxaliplatin and cisplatin, and in another set of experiment, they treat the mice with three different antibiotics, which is neomycin, vancomycin, and [inaudible]. So, and the composition of three antibiotics, they remove ground positive bacteria, ground negative, and anaerobic bacteria from our gut. And also, they use jumper mice. So, what has happened is -- and [inaudible] can explain shortly this graph, so the green line here -- so this is the tumor volume. So, higher the graph goes up, the bigger the tumor. So, when the mice are not treated with any therapy and when the mice are drinking normal water, and only the antibiotics, these two group, they are -- the tumors are growing really high. And then once the -- and this is the normal tumor treated with oxaliplatin, the tumors are cleared. But when these mice are treated with antibiotics, you see the tumors are growing, so it's really bad [inaudible] the impact of the therapy. And it also -- the similar case in case of cisplatin is you want the mice treated with antibiotics, the tumor is growing, and the therapeutic efficacy is gone. So, that really -- it issued a new line in the cancer and gut microbiota research. So, then I started asking question that whether gut microbiota can regulate the toxicity. And it can seem recently [inaudible], so recently like [inaudible] radiation therapy and the cisplatin, oxaliplatin, and [inaudible] and different [inaudible] for [inaudible] PDL1, they all is regulated by microbiota. And [inaudible] discussing the [inaudible], so what I told you so far that chemotherapy kills the tumor, and also at the same time it causes system and toxicity. And we know now that gut microbiota also regulate cancer initiation and progression, and it also regulates chemotherapy. However, very little is known how gut microbiota can regulate system and toxicity. And that's what my question was, so then we asked the question whether the alteration of gut microbiota can regulate [inaudible] side effect. So, we proposed this experimental plan, so we had four experimental group, control where the mice are not treated and cisplatin we'll give the mice the drug. And you treat the mice with antibiotics, so the antibiotics is not here given as a therapy or something. We are just creating experimental model where you can remove the bacteria and see the impact of the drug and either germ-free mice. And in the fourth group we used the drug and the antibiotics. So, this is the experimental setup, so we gave the mice three weeks of antibiotics. And then after three weeks of antibiotics we have done sequencing. We know that this different class of microbiota is gone, and then we introduce [inaudible] to a really heavy dose of platinum. We used that dose to create toxicity and see if that can anything in terms of reduction of toxicity. And then we did a readout and every day, like what the weight and collection of different tissue for other experiments. And for germ-free mice, I just put this picture just to give you the background that this is really intricate experiments. [Inaudible] an animal has not a single bacteria, and this really tricky kind of facility that took a lot of time. I took this picture from the [inaudible] we have two [inaudible] facility where our dedicated staff members, they maintain those mice, and then [inaudible] this experiment inside this isolator and then see what happens. So, the first question with this cisplatin hinders [inaudible] loss regulated by microbiota? So, here you can see that normal cisplatin in the blue line when you give this high dose of platinum from day zero, on day four, these mice lost nearly 20% of their bodyweight. Once you remove or modulate the microbiota with antibiotics, the mice are protected. And this is across male, female of both [inaudible] at resonating male and female, some differences. We did this experiment more than seven-eight time; we always see the same thing. And then we did that experiment also in [inaudible] and see also germ-free mice is protected compared to the conventional mice. So, take this point so we know that if you deplete gut microbiota, it has [inaudible] the mouse bodyweight. But one can ask, how do you know this is really microbiota affect? This could be that these mice ate less food or have different kind of metabolism. So, we -- this [inaudible] activate those, but anyway, so we did microbiota reconstitution experiment. So, far I've shown you [inaudible] that you have conventional mice, normal mice. You give platinum; this causes toxicity. You have germ-free mice. You give platinum; it is protected. So, now what happened? You take the normal mice, and you collect the sickle content, which is the, let's say the fecal sample. And then you give the sickle sample through fecal microbiota transfer in these germ-free mice, and then you wait for three weeks and this process called reconstitution. And then you give cisplatin and see what happens, and these mice become not like weighted mice. So, you see that normal mice, this purple, one, this is the conventional mice, and that -- we made it conventional mice, they all lost bodyweight, like same as conventional means. Which means once you convert the germ-free mice with the normal -- the microbiota, those germ-free mice lose protection. But here in this line, you can see the germ-free mice did protect. So, this really shots that gut microbiota modulate the toxicity [inaudible]. And we have done many other different experiments to understand what is the mechanism of this toxicity and what the -- and the reason that the data indicates the gut microbiota, it regulates the transporters in the kidney, and it clears the drug from the system circulation. And this is one of the pictures showing that the in the kidney cross section, you can see the cisplatin causes a lot of [inaudible] more [inaudible] signal is causing -- showing that [inaudible] diversion. Here you can see that once the mice are treated with antibiotics, it is -- the damage is really, really less or more like negligible. And this is the graph; you can see that the treatment with antibiotics in the mice reduces the toxicity by platinum, and this could be really helpful if we understand the mechanism could be used in the clinic as well. Where after giving the treatment to the patients, after like -- the cisplatin has very short half-life, and after that the unnecessary platinum, which is bound with different protein in the system, you can clear and reduce the toxicity. So, what is the impact of cisplatin on gut microbiota? So, you can see, another test we have done to see what the drug are doing in the composition of microbiota, you can see the [inaudible] we have done this experiment with fifty mice. And after four days, they lose 20% of bodyweight, and within this group, within these 45 mice, you can see this is only creating two clusters. And one of the cluster, this one, we focused on this, and this lost significant amount of bodyweight compared to this cluster. So, we focused on this, and we saw they have specific microbiota presence in this. And another striking thing is, this is one of the [inaudible] sequencing of gut microbiota. And these grey lines are the lactose, and this -- these top bars, these are the mice from this group. They are losing lactose [inaudible], so this is one of the examples that I'm showing. So, seems like the toxicity's associated with losing of [inaudible] species from the gut composition. So, then we wanted -- we asked a question, can specific microbiota modulate and also like promote protection. So, if you can bring back those lactose [inaudible] would it protect against platinum? So, then we took three different lactose [inaudible] species, and then we created a synthetic consortium. The reasoning I'm doing that is if you take the total [inaudible] content because like millions of bacteria, you don't know what these are, and then it's really difficult to understand the mechanism. So, we took -- defined -- and these microbiota came from human, and then we -- it is well defined. And then we added this lactose [inaudible] species with this one, because if you give a single [inaudible] to the mice, these bacteria not survive in the [inaudible] conditions. We have to give [inaudible] ecosystem. And then we looked for the toxicity. So, in this graph I'll explain, so this one, and this is the conventional mice, this is the grey color one here, lost 20% of bodyweight. And this is microbiota consortium plus cisplatin, and this experiment was done in germ-free mice. So, we gave the microbiota consortium to the germ-free mice, and then we gave cisplatin. The protection is again lost, and you see here, once the microbiota consortium was mixed with the [inaudible] species, the defined one, and this is a single isolate, and that is protected significantly conflict with the conventional one. And this is the germ-free mice again showing protection against cisplatin. So, this [inaudible] case that if -- you can give -- treat the mice with a specific bacteria, and that can protect the mice against the cisplatin [inaudible] toxicity. So, the -- going back to the question, you know, does microbiota regulate platinum clearance from the cochlea? So, we have done from many different organ, and this is the cochlea data. You can see on day four cisplatin has really cleared from the cochlea. And there's a paper [inaudible] from my previous lab showing that once platinum enters the cochlea, they retain their whole life permanently, and they don't leave from the cochlea. And that might be one of the reason that it's causing the produce of hearing lost over the years. And this data shows that if you modulate microbiota, that can really clear platinum from the system and circulation. And we have also done experiment in germ-free mice [inaudible] data and also a different specific microbiota. So, ongoing study, what I'm doing now is, you know, I don't give much background here, but this cochlea is thought to be [inaudible] organ, and this is very, you know [inaudible] one of the most difficult organ to reach. Any organ you can think of in the body that you can have biopsies, but the cochlea is so inside the bone and this is one of the hardest bone in the body. You cannot get any biopsy; you cannot reach there without surgery. So, this is one of the difficult -- the problem that we have, and another thing, we do not know which kind of cell entered into this [inaudible] space and whether they have any specific, you know, immune cells. There is a lot of studies showing this [inaudible] by [inaudible] that immune cells are present in the cochlea. So, what I have done in this project, I have done single-cell [inaudible] and high dimensional [inaudible] technology to characterize the immune cells within the cochlea and quantify the amount of cisplatin in the single cell. Then I can now know from the single-cell data that which cell is modulating this inflammation in the cochlea. And the idea is to understand better mechanism into that, and from that, [inaudible] to understand and characterize the immunity of the cochlea. So, in summary and conclusion, but I've shown you that, you know, once cisplatin is in the system and circulation, if you can deplete microbiota by antibiotics, it can reduce cachexia. It can reduce DNA damage, and also it is [inaudible] toxicity and it meant [inaudible] integrity. So, you can think that, okay, in the beginning how can you give, you know, an antibiotics to the patients? And that's a very critical question, and so we are trying to find out a balance. And I think we understand better now that there's a balance. Some of the microbiota is responsible for causing toxicity and some are for efficacy. So, if you can maintain that balance, then we can, one hand, kill the tumor; at the same time, we can maintain the toxicity. And another is happening is, you know, after the [inaudible] toxicity, we have disperses in the microbiota. And that recruits many different kind of immune cells that maintain the -- at the [inaudible] site and leads to toxicity. So, this one, if we can regulate disperses of microbiota by giving probiotics or like removing specific class of bacteria, then we can clear the platinum from human tissue and also reduction of long-term toxicity. And ultimately, which will enhance the quality of cancer patients. And there's a similar kind of study also [inaudible] as well and where we show that microbiota also regulates [inaudible] toxicity. So, with that, I would like to thank all my lab members [inaudible] and [inaudible] team and the funding from [inaudible] agency I had from the [inaudible] foundation. Thank you so much. [ Applause ] >> Tomoko Steen: Thank you very much. So, we have a few minutes to ask questions to the speakers. Anybody? Yeah? [ Inaudible ] Could you repeat the question? >> Michael Dean: So, the question was what are immunomodulatory agents in antiangiogenic [inaudible]? I'll answer the antiangiogenics, so as a tumor gets bigger, it needs to develop its own blood supply. And so, some tumors actually secrete factors to essentially recruit blood vessels into the tumors, so the tumor can get nutrients. And so, the that antiangiogenic factor's the idea, well, if we can suppress that, starve the tumor for blood supply, then the tumor would die. And it's one of those things that worked fabulously in mice. When we went to apply it in humans, you know, we are starting to see some applications, so that is a new line of therapy. >> Robert Clarke: I can try and take the immune one, though it's not my field. Basically, the idea is to try and stop the cancer cells from turning off the immune system. So, when you have an infection, for example, the immune system gets turned on. The t-cells, they come in. They get activated. They remove the infectious disease, and then they get turned off, because you don't want them attacking the normal cells. What happens when they come into a tumor cell is they just turn off. And they do this by signaling between the cancer cells and the immune cells, and there are receptors and ligand that turn off the activation of the immune cells. So, what the drugs that we currently have do is they block that signaling that turns off the immune system and then allows it to become activated and stay activated and then kill the cancer cells. There are lots of different ways that the immune system can get regulated in that way. I mean, we don't have drugs that target all of that communication [inaudible] communication, the question really. But more drugs and more classes of drugs are coming through very, very quickly now as we understand in much greater detail how that communication between tumor cells and immune cells in the tumor microenvironment occur. [ Inaudible ] >> The barrier around it is cancer cell that controls remote-- [ Inaudible ] At least in the beginning, the cancer cells are encapsulated and before it ruptures and spread the [inaudible]. So, the question is whether this cancer cell can be controlled by the growth and [inaudible] by some-- [ Inaudible ] >> Robert Clarke: So-- >> Tomoko Steen: Could you repeat the question, sir? >> Robert Clarke: I will try. So, I think you're asking, in part, how does the body respond to the tumor, or does it wall it off? And is there a way to improve communication or alter the communication amongst the cells in a way that will cause the cancer cells to die? So, yes you do see an attempt to wall off the tumor in some cases where you see what we would call a desmoplastic response. And you see cells coming in and they try to change the tissue dynamics between the cancer and the normal cells, but generally it is not successful. I'm not sure how well we've been able to manage that piece as a target. The other thing though is that cells do communicate with each other, and there are cell-cell contacts, there are gap junctional communications, for example. And there's pretty good evidence that in some cases the normal cells, if they conform those with cancer cells, can slow them down and stop them. And cancer cells have fewer of these gap junctional communications than normal cells do. And there are drugs that can, in experimental systems, break that cell-cell communication, but I don't think any of them made it into the clinic yet. But it's certainly an area of active research. I don't know if anyone else wants to take that one. >> Tomoko Steen: Any other questions? >> Yeah, I got a question here on this [inaudible] therapy. [Inaudible] therapy's good in terms of-- [ Inaudible ] From the surrounding molecule, and then it becomes reactive. But-- [ Inaudible ] >> Jonathan Lischalk: So, the question was, how does -- how do antioxidants play as a role in decreasing the side effects of radiation? Well, the inherent radiobiology of what we're trying to accomplish with whether it's X-rays or protons is DNA damage. And the DNA damage, if you can accumulate that in a tumor cell, tumors are just not -- tumor cells are not able to fix themselves. They have a lot of mutations that basically accumulate over a time or are inherent in the cell, for instance, like the VRCA1 mutation we talked about earlier. And they're not able to repair DNA damage, and so much of the damage that we cause with radiation results in the -- us removing that cancer cell's ability to divide, which is an inherent problem with cancer cells. And we do that by damaging the DNA, and when they try to divide again, they can't, and they die. And so, part of the way that oxygen plays a role in that is that when a -- whether it's an X-ray or a proton damages DNA, if the oxygen is around, that damage is then fixed. We call it the fixed body oxygen, so that it's still there so that when that cells try to go on and divide again, it's not able to. And so, it's something that historically we try to avoid antioxidants during radiation therapy to allow radiation to be more effective. Now, as the proton's traversing through its length, it's not really -- you know, it's getting to the point where it's depositing [inaudible], its range. And so, earlier to that, the dose is much smaller, and the damage is a lot less. But, you know, it really depends on the location of the tumor and the type of tumor. >> Tomoko Steen: Any other questions? Can I ask a question, actually? I was told that proton therapy is not good for the medicalized cancer. Is that-- >> Jonathan Lischalk: So, you know, radiation is radiation, whether it's X-rays or protons, it can kill cancer cells, whether they're [inaudible] or primary tumors. Now, the question is, what are you trying to accomplish? And in certain situations, when a patient has a metastatic disease, at that point it's stage four. And really, theoretically, those cancer cells could be anywhere in the body, and so the type of therapy that you would want to choose is something that can go throughout the body, and unfortunately, we can't just irradiate everything in the body. It would be just too toxic. And so, you know, proton therapy could be used for certain situations where drug therapy is so powerful that maybe there's one focus left that isn't responding to the drugs as well. So, I wouldn't say that it's a very common approach, but it's certain feasible to do. And it would -- you know, that those cancer cells would respond to the radiation. >> Tomoko Steen: Another question is-- [ Inaudible ] >> Jonathan Lischalk: Well, you know, the radiobiology of proton therapy is challenging, because we've studied X-ray radiobiology for many years. And proton therapy's interaction with matter is thought to be maybe 10% more efficacious relative to X-ray. So, when I'm doing a radiation plan with proton therapy, we're kind of giving it a 10% increase in its damage effect. Now, whether that graph that I showed you early on where the proton comes in a lower energy and then deposits and then stops, the relative biological effect may be variable along that path. And right now, it's an active area in investigation to understand if we're on a different part of that proton path, what -- how is it interacting differently? So, it's a -- it's something that I think in the next 10 to twenty years we'll be able to take advantage of. >> Tomoko Steen: Thank you. Another question to Doctor Roy, the [inaudible] actually, working [inaudible] question about diet in the cancer and microbiota relationship. Do you have any comments on that? >> Soumen Roy: Yeah, like a lot of new research has been showing that microbiota, like diet [inaudible] the microbiota. And, you know, there are recent studies that show and that may [inaudible], and also like in general, you know, what we eat, it becomes [inaudible] beside microbiota, which is very important to eat [inaudible] to see like what the body needs. And then -- you know, because -- and then go for it in terms of regulating. And one study show [inaudible] that the diet also regulates the hearing loss, so one of the [inaudible] study. >> Tomoko Steen: I see. Very interesting. Any other questions? Yeah? [ Inaudible ] >> Soumen Roy: So, thank you so much for asking this question. This is wonderful question. So, the question is whether fasting can impact the efficacy of toxicity of the therapy. So, in general, like, you know, there are a lot of recent terms. It's called like fasting, intermittent fasting, so in the current era like there's a tendency that we eat in every four hours, five hours. And before the body could clean up or utilize the entire food, [inaudible] we eat again. So, the body never get rest. So, if someone is fasting, or like it's called like intermittent fasting, if there's a gap between one meal to other meal, at least eight hours, that body gains enough time to detoxify and utilize all the food. And there's a study shows couple of years ago from MIT that if the mice they are fasting, they have more intergenerational stem cells, and they are have better varied integrity in the gut lining. So, once the therapy's given, they are like, you know, causing less toxicity, and they are more resistant to the toxicity. So, the fasting clearly helps a lot in terms of therapy and any other aspect of lifestyles. >> Tomoko Steen: Very interesting. >> Michael Dean: Another thing you may have heard of, some of the growth factors that are important in cancer or insulin or insulin-related compounds. And so, there's some new therapies coming online targeting that system, and maybe they'll work better in patients where we can lower their insulin levels during cancer treatments. And that's also something we've looked at. [ Inaudible ] >> Soumen Roy: You know, like there is to say-- >> Tomoko Steen: Could you repeat the question. >> Soumen Roy: The question is like whether the -- after the treatment, in case of best cancer patients, whether it is becoming more relevant compared to like cardiotoxicity, right? That's what you mentioned. Yeah, they're like lot of -- not a lot of clinical data available like, you know, worldwide which you can make a conclusion out of it, but I'm sure like the doctors can give you more -- better answer. But in terms of like hearing loss, there's a huge heterogeneity of different treatment and also like the record of preexposure of different antibiotics and drugs that could also impact the progressive hearing loss. It's always better to, you know, like check about the hearing sensitivity testing, but [inaudible]. >> Tomoko Steen: One last question. [ Inaudible ] >> Michael Dean: I don't know a lot of good data. Typically, you know, marijuana smokers don't smoke nearly the quantity that tobacco smokers do. I think it's no question that breathing any kind of smoke into your lung is not terrific for your health. But I don't know of specific studies showing a clear increase between marijuana smoking and lung cancer, no. >> Tomoko Steen: Anybody? No? Before I close, I like to thank my colleague, Ashley [inaudible] put the books together. So, if anybody's interested, and also she put together the reference. This is the Library of Congress, so please pick it up, and then thank you so much for the other speakers, wonderful panel, and very informative. I hope--