The Peter Attia Drive - #177 - Steven Rosenberg, M.D., Ph.D.: The development of cancer immunotherapy and its promise for treating advanced cancers

Episode Date: September 27, 2021

Steve Rosenberg is the Chief of Surgery at the National Cancer Institute, a position he has held continuously for the past 47 years. Steve is a pioneer in the field of immunotherapies for cancer and a... recipient of nearly every major award in science. In this episode, Steve discusses his inspiration for devoting his career to cancer research and describes his keen observation of two cases of spontaneous cancer remission, driving him to learn how to harness the immune system to treat cancer. Steve’s personal story essentially serves as a roadmap for the field of immunotherapy, from the very non-specific therapies such as interleukin-2, the discovery of tumor-infiltrating lymphocytes, checkpoint inhibitors, CAR T-cells, and adoptive cell therapy. Perhaps most importantly, Steve expresses his optimism for what lies ahead, especially in the face of some of the more recent discoveries with respect to tumor antigenicity. Finally, Steve discusses the human side of cancer which helps him to never lose sight of why he chose to become a physician. We discuss: Steve’s childhood and inspiration to become a physician and medical researcher [3:15]; Patients that influenced Steve’s thinking about cancer and altered the course of his career [13:15]; The discovery of antigen presentation, Steve’s first job, and why he knew he wanted to study cancer [19:30]; Cancer treatment in the early 1970’s and Steve’s intuition to utilize lymphocytes [26:45]; Cancer cells versus non-cancer cells, and why metastatic cancer is so deadly [31:45]; The problem with chemotherapy and promise of immunotherapy [38:30]; How the immune system works and why it seems to allow cancer to proliferate [43:15]; Steve discovers how to use interleukin-2 to mediate cancer regression [52:00]; The immunogenic nature of certain cancers and the role of mutations in cancer [1:03:45]; The improbable story of how CAR T cell therapy was developed [1:16:30]; The discovery of tumor infiltrating lymphocytes (TIL) and engineering of T cells to recognize specific antigens [1:28:00]; Steve’s experience treating President Ronald Reagan’s colon cancer [1:36:00]; Why Steve has turned down many tempting job offers to focus on his research at the National Cancer Institute [1:41:00]; The role of checkpoint inhibitors in cancer therapy and the promise of adoptive cell therapy [1:43:00]; Optimism for using immunotherapy to cure all cancers [1:48:00]; The human side of cancer and the important lessons Peter learned from working with Steve [1:52:15]; and More Learn more: https://peterattiamd.com/ Show notes page for this episode: https://peterattiamd.com/StevenRosenberg  Subscribe to receive exclusive subscriber-only content: https://peterattiamd.com/subscribe/ Sign up to receive Peter's email newsletter: https://peterattiamd.com/newsletter/ Connect with Peter on Facebook | Twitter | Instagram.  

Transcript
Discussion (0)
Starting point is 00:00:00 Hey everyone, welcome to the Drive Podcast. I'm your host, Peter Atia. This podcast, my website, and my weekly newsletter all focus on the goal of translating the science of longevity into something accessible for everyone. Our goal is to provide the best content in health and wellness, full stop, and we've assembled a great team of analysts to make this happen. If you enjoy this podcast,
Starting point is 00:00:30 we've created a membership program that brings you far more in-depth content if you wanna take your knowledge of the space to the next level. At the end of this episode, I'll explain what those benefits are, or if you wanna learn more now, head over to peteratiaMD.com forward slash subscribe.
Starting point is 00:00:46 Now, without further delay, here's today's episode. I guess this week is Dr. Steve Rosenberg. Steve is the chief of the surgery branch of the National Cancer Institute, a position he has held continuously for the past 47 years. Some of you may have heard of me talk about Steve Rosenberg in the past. He is unquestionably the most important mentor I've ever had. Steve has received numerous awards, in fact, really too many to name. So it's probably easier to explain it this way. He has received essentially every major award in science except for the Nobel Prize. He is one of the
Starting point is 00:01:23 pioneers in the field of immunotherapy, dating back to his early work in the 1970s and 80s with the discovery of interleukin 2 and its effect on lymphocytes in mediating cancer, regression, and patients with metastatic cancer. In this episode, we talk about his entire history from childhood, basically, until now. talk about his entire history from childhood, basically, until now. And it serves as effectively a roadmap for the field of immunotherapy from the very non-specific therapies, such as interleukin 2 into the discovery of tumor infiltrating lymphocytes, up into checkpoint inhibitors, which many people have heard of in the last decade, CAR T cells and adoptive cell therapy.
Starting point is 00:02:06 We talk about all of these things in detail, and perhaps most importantly, we talk about his optimism for what lies ahead, especially in the face of some of the more recent discoveries with respect to tumor antigenicity. We also talk about the human side of cancer, which I think when you listen to this, just completely comes through him in every way, shape, or form. He has really never lost sight of why he chose to become a physician, basically at the age of six years old, and why he was so drawn to cancer. One other point I'll make, if you find this episode particularly interesting, I would highly
Starting point is 00:02:43 recommend going back and reading the book, the Transformed Cell, which Steve wrote with John Barry about 30 years ago. It's one of the best books I've ever read on the process of scientific discovery and the journey and the ebbs and flows of failures and successes. It's really an amazing book for anyone who wants to understand how biomedical research is conducted. So without further delay, please enjoy my conversation with Dr. Steve Rosenberg. Dr. Rosenberg, thank you so much for making time. I know how busy you are. I know as much as anybody how busy you are because I've sat next to you and watched how hard
Starting point is 00:03:21 you work and tirelessly you work. So it really means a lot that you would make any amount of time to sit and talk about what we're going to talk about today. I almost don't know where to begin, but I can't help but want to begin kind of chronologically with your life story because you're probably one of the most focused people I've ever met, if not the most focused person I've ever met. And that focus seems to have started at a very young age. Let's talk a little bit about your childhood. You grew up in the Bronx if I'm not mistaken, correct? That's correct. I was born in the Bronx.
Starting point is 00:03:50 If I recall, we're coming up to your 81st birthday. So you were born, I think it's August 1st, 1940. Yeah. So what are your earliest memories of childhood as they pertain both to your love of science and perhaps more importantly your obsession with cancer? So up until about the age of five or six, I wanted to be a cowboy. I have an older brother and we would talk about going out west together and riding on horses and doing all kinds of exciting things.
Starting point is 00:04:23 But the first things I remember, other than wanting to be a cowboy, occurred when I was about five or six years old. And I've given a lot of thought to how that came to pass. When I was about five or six living at home, right at the end of the Second World War, when all of the remarkable tragedies of the Holocaust. So it came home as my parents got one postcard after another. I remember this
Starting point is 00:04:57 and they are suffering as they got word of relatives that died in the death camps during the war. of relatives that died in the death camps during the war. And I remember being so horrified by that in terms of how evil people could be towards one another. And somewhere around that time, I developed an almost spiritual desire to become a doctor, to do research and make progress in helping people, in alleviating suffering rather than causing suffering. And that persisted as I began to keep scrapbooks about anything I could find about medicine or research. And I think it was in response to the horrors of that particular time that inspired me
Starting point is 00:05:54 to not only become a doctor, but to become a doctor who not only helped alleviate suffering now, but alleviate potential suffering in the future by doing research. And I stuck with that right through my education. Now, you did very well in high school, because, or at least you, we've never spoken about it, but I can only assume you did because you were accepted to the best medical school, and not only that, but you did the combined bachelor's MD degree, which I assume would have been very difficult to go straight into medical school from high school, but you somehow managed to get into the six year program. And what, so just talking about Hopkins for a moment, what was the impression that that place left on you
Starting point is 00:06:37 in what would have been, I guess, the late 50s and early 60s? So I went into this six year program. It was three years of college and three years of medical school knowing that I would want to get further education that I would take additional time. And I knew from the very beginning that I would go on to get a PhD in one of the sciences. It turned out to be biophysics. And Hopkins was a very nurturing environment with respect to respect to that. As soon as I got to Hopkins, I started working in a biology
Starting point is 00:07:09 laboratory in the afternoon and evenings doing some very simple projects in the biology lab. But I knew from the very beginning that I wasn't just going to try to practice today's science, practice today's medicine, but rather try to create the medicine of tomorrow. And that's stuck with me for these last 60 or 70 years or so. Who was the chief of surgery when you were there? Was Blaelok still the chief of surgery? Yes, and it was, the medical school was not necessarily eternally nurturing environment.
Starting point is 00:07:45 You were thrown in there and at rounds, they seem to be a spirit of calling people to task in front of others. It wasn't the way that I thought education should happen, but there were brilliant people around. And that's what I think is the most probably one of the single most important components of a good education and that is being surrounded by people who know a lot, know more than you can inspire you and that kind of person was at Hopkins.
Starting point is 00:08:18 Now I remember when I was in medical school when I was spending time with you and we got talking one night and you explained that the reason you chose to do your PhD at Harvard in biophysics, and this is as close to my remember as a direct quote as possible is, you never wanted to be intimidated by a differential equation, which presumably was a bit of a shortcut for you wanted the biggest or the broadest education possible. But what led to that choice to, I mean, at that point in time, for example, did you know you had an interest in immunology? Have that peaked your curiosity yet? Or were you still thinking just more broadly? I was somehow interested in the mysteries of cancer through high school, biology, and my college classes. I got a PhD because I wanted more formal learning. I never wanted to be intimidated by what I did not know. I
Starting point is 00:09:14 wanted to be able to grasp any area of science and use it to answer questions. And maybe differential equations were in the source of that. I ended up doing a lot of math in graduate school, but it was more than that. I wanted to have the feeling that I had a good enough broad background in the sciences, because when I got that PhD in biophysics, I was doing physical chemistry, quantum mechanics, thermodynamics. It was a lot of non-biology and biophysics. I wanted to have a background in the sciences such that if I encountered a problem, I could
Starting point is 00:09:54 get a good book, read some papers, and understand it. And it was that base of knowledge that I tried to acquire. So when you were doing your PhD, had you already applied to residency or did you take time out between medical school and the application to residency? So I went immediately into the surgical residency at the Peter Brown program hospital
Starting point is 00:10:17 right out of medical school and then took off four years to get a PhD in biophysics. So it was a year of internship, took off four years, and then went back to the residency. And then came down to the NIH for several years to join the immunology branch here. This was during the Vietnam War before going back to actually finish the residency in 1974. Now, was for any more sort of the most significant figure in your life at that point in time as a mentor? Frannie Moore was the chief of surgery
Starting point is 00:10:51 and an incredibly smart person. There are an awful lot of smart people in that educational system, but Frannie stood out in that discussing a problem, discussing a patient. It wouldn't be at all surprising from to come up with an idea or an outlook or a perspective on a problem that most people had not considered. So in that sense, he was an important figure.
Starting point is 00:11:18 He was not so much loved as respected, which is a feature that I think can be very inspiring to young people. Was it unusual for someone to leave their internship or leave after their internship and go into a PhD program at that time? Well, it was, as a matter of fact, and the Brigham and Cardi to take off a year or sometimes two years in the middle of the residency program, generally after two or three years. But I knew at that point I was just itching to learn more. I was just not satisfied after medical school, the college and medical school, that I really knew
Starting point is 00:11:54 enough to do creditable, meaningful, impactful research. And so again, this was a real program. For two years, I did nothing but study. Nothing but take classes and study and learn. And then the latter two years, I spent in the research laboratory doing basically physical chemistry and protein chemistry research of cell membranes.
Starting point is 00:12:17 And Franny was supportive of this. He was to an extent to take that time off after internship required a few meetings and I Ultimately, I remember sending him a note saying look, I'm 23 years old I've just finished the residency. I need to do what I'm excited about and he gave in and I have enormous respect for him for that And he was quite patient because I came back for a year and then I went down to the NIH It was part of the Vietnam draft obligation. And they kept taking me back each time, which was very nice.
Starting point is 00:12:51 But it turns out I set some kind of record at the break. It took me 11 years from the time that I started my internship to the time I actually finished the residency, which was some sort of record at the time. So you would have been old for a chief resident at that time. I guess, yes, I was, what, I was 33, 34 by the time I had my first job. But of course, you made up for it because you sort of progressed so early at the outset. As you know, well, the book that you wrote with John Barry in 1992, The Transformed Cell,
Starting point is 00:13:23 is a book, I may have the record for most times reading it. I may also possess the record for most copies owned, which I know you get a kick out of that. Every time I say something to that effect, you basically say I'm one of the few people who's read it, which I know is not true. But I still remember the first time I read it, and it's a remarkable story.
Starting point is 00:13:40 I suspect many people will go on to read it after this interview because it is, in many ways, one of the best books about science Which I think was your motivation for writing it, but and we'll come back to that But in it you talk about an important moment in your training which occurred in 1968 With a patient who you met in the ER one day Can you can you tell us a little bit about that story and why it altered the course of your career? Well, there were actually two patients
Starting point is 00:14:05 that I saw early on, or was familiar with early on, that influenced my thinking about cancer. The first was a patient I saw when I was a junior resident at the West Roxbury VA Hospital. We rotated through there three or four months at a time during the residency. And it was a 68-year-old fellow who came in complaining of right-up quadrant pain.
Starting point is 00:14:27 It looked like a typical gallbladder attack, and I got pretty excited about it because it might be a patient that I might be able to perform one of the first operations I was allowed to do. And so I looked into his chart and a remarkable story was encountered. I looked into the chart and a remarkable story was encountered. I looked into the chart and it turned out that 11 or 12 years earlier, that patient had
Starting point is 00:14:52 been seen at the West Roxbury VA Hospital. He had had a gastric cancer, a stomach cancer. He had undergone a laparotomy and I looked at the surgeon's note saying that he opened the belly. He saw a tumor that was encompassing about three quarters of the stomach. They were multiple liver metastases, deposits that were biopsy shown to be the gastric cancer that it spread, multiple enlarged, hardened nodes. He took out part of the stomach, I guess, the palliative measure, left the rest of the
Starting point is 00:15:23 disease in place, and the patient recovered, and about a week later went home. Well, as I turned the patient to the chart, the patient comes back three months later, and nobody had expected to see him. And he was doing fine. He was gaining weight six months later. He was back working. And here he was, 12 years later, having lived the past 10 or 11 years completely normally.
Starting point is 00:15:46 And so I took part in removing his gallbladder under supervision, of course, and his belly was completely clean of cancer. There was no evidence of cancer of any kind, and we went back and looked at it as, be sure it was the same patient, re-reviewed the pathology, sure enough it was a cancer that had spontaneously disappeared over time in the absence of any therapy. One of the rarest events in medicine, and that is to have the spontaneous regression of cancer without any treatments being given. Somehow his body had rejected the cancer, and I then did what turned out, of course, to be a very naive experiment,
Starting point is 00:16:27 but I was wondering whether or not this patient who had somehow cured his own cancer could be somehow taken advantage of to treat other patients. And it turned out we had another patient in the hospital with a gastric cancer of veteran who happened to have the same blood type. And so I called up the head of the surgery department, Brownie Wheeler, and said, hey, I want to take a blood transfusion from this patient who spontaneously was cured and give it to
Starting point is 00:16:53 this other patient. And he said, okay, that was the IRB, as existed at that time. And so we actually got blood from this one patient and infused it into the other veteran, but of course it didn't do anything. And the other patient ended up dying of his gastric cancer. But at least planted the seed that in fact maybe there was something in the immune system that caused the rejection of that cancer much if you would a far and transplant. And by these major defense mechanism, of course, against foreign invaders, is the immune system.
Starting point is 00:17:30 It got me thinking about potential immune manipulations. But there was a second patient that also influenced me a great deal. And that was a patient that had been seen about a year before I came to the Brigham as an intern. And this was a patient who had received one of the early kidney transplants that were developed and innovated at the Brigham hospital. He had received a kidney from a young individual who died in a motorcycle accident. And that kidney was transplanted into the recipient and the recipient developed a widespread
Starting point is 00:18:07 renal cell cancer. And it turned out after study that the kidney that had been transplanted inadvertently had contained a renal cancer that then in this other patient under the influence of immunosuppressive medications had spread widely through his body. So in an attempt to control this, the immunosuppressive medications were stopped. Of course, the kidney rejected and had to be removed. But the patient's cancer then went away as well. Because it too was allergenic.
Starting point is 00:18:41 It too came from the genome of the original donor. So what did that teach one? Well, it showed that a large invasive, vascularized cancer could be caused to reject completely by the immune system if you had a strong enough stimulus that could mediate that rejection. And so it was that spontaneous regression. Maybe this demonstration that the immune system, or directly by removing immunosuppressive medications, could result in tumor collapse and regression that tended to put me on the path towards cancer. But I was already pretty much there because of what I had seen as a doctor in cancer patients.
Starting point is 00:19:28 Now, in the late 1960s, what was understood about the human immune system as it pertained to even viruses let alone cancer? I mean, I had MHC class one and class two been identified yet. I don't think they were identified until the mid 70s, right? Class one in the 70s and early 80s, Class II.
Starting point is 00:19:48 But when I started in 1974, the idea of immunotherapy was a dream. There were anecdotes way back to the late 80s of tumors going away when people got an infection. But really nothing stable. There was no ability to measure an immune reaction against any cancer. There was no such thing as a cancer antigen that it had ever been found. There were no manipulations you could give that might work. So it was sort of a dark period when it came to knowledge about the immune system
Starting point is 00:20:20 against cancer. It's a little hard to understand just how frequent immunologic information developed. In the 1957 issue with the Journal of Immunology, the word lymphocyte was not in the index. We did not understand what small lymphocytes did, how they were doing, circulating in the 1950s. And so that information only came to pass in the early 60s when it was clear that you could transfer immunity by transferring lymphocytes in a way that you could not do by transferring blood or serum. And even in experimental animals, there was no manipulation that could cause an existing cancer to disappear. You could immunize a mouse against a tumor by letting it grow and then removing it and cause that mouse to resist
Starting point is 00:21:11 an implantation of the same tumor again. But once the tumor was growing, there was no maneuver that could keep it from growing, no immunologic maneuver that could keep it from growing. So the field was desperately in need of more information. So before we get to how you arrived at the NCI, well, actually, let's talk about that. It's a very unusual first job.
Starting point is 00:21:35 How did it come about? And what did you assume you would do at the completion of this otherwise very long residency? So as you're indicating, I finished my residency June 30th, 1974, and the next day I was appointed chief of surgery at the National Cancer Institute, a position that I still hold. I'm still chief of the surgery branch now, 47 years later, 46 years later. The NIH, when I came here, I knew as a remarkable place, it had resources and a commitment, a mandate to make progress. It's a state-of-the-art hospital that provides outstanding care to patients, but it exists
Starting point is 00:22:22 again, not only to practice the best of today's medicine, but to create a medicine of tomorrow. And that always intrigued me from my first knowledge of the NIH when I came here in the midst of the residency during the Vietnam War. Now you did have an offer to stay at Harvard, correct? Was Dana Farber still in existence at the time? Dana Farber was just being built, the hospital was just being built. Frannie Moore was chief of surgery, offered me a position as the head of surgery in that new
Starting point is 00:22:53 Dana Farber institution. Amel Frye was the director of it and head of medical oncology and he too offered me the position. And I had tentatively accepted it, although we're in the midst of some negotiations about whether there would be individual and independent operating rooms in the hospital and so on. But in the course of that, I heard from one of the division directors here at the NIH, who I'd gotten to know when I was a fellow who came to the Brigham to interview me.
Starting point is 00:23:24 I was wondering whether I would be interested in the position. And so he interviewed me and I was expecting to stay at the Brigham at Harvard, but one day I got a phone call from him saying that the chief of surgery offered ketchup. I decided to retire and the position was open July 1, was I interested. And I knew I was, but I had to call my wife Alice, told her about it. She said, just pack. Let's go. And off we went. Was Franny disappointed?
Starting point is 00:23:53 Oh, it was a, it was a really shocking encounter that I had with him. And that I went in to tell him I decided to go to the NIH. I thought it was a place where I could best utilize my interests and knowledge and he said, no. And I said, look, I've decided to do it. He said, you have to stay here. It's too great an opportunity to turn down. And I said, no, and he wasn't an easy guy to say no to. But I knew I wanted to come back to the NIH. And finally, after almost an hour, I finally said, Dr. Moore, I'm not, I'm going down to Bethesda, Maryland. And I offered my hand to shake his hand,
Starting point is 00:24:32 but as I was gonna leave, and he refused to shake my hand, which was a little shocking, but he got over it, finally. Yeah. I left and we became good friends and he said all kinds of nice things about me when he needed to so
Starting point is 00:24:46 worked out well You see at that point I knew I wanted to study cancer I had already made that absolute absolute commitment and the national cancer Institute seemed like the right way to do it and in some sense It was a logical decision from me given And in some sense, it was a logical decision from me, given my childhood experience that got me interested in medicine and science. Cancer is such a devastating disease. It attacks innocent people through no fault of their own, makes them and their families watch impotently as they progress and then die of cancer as a Holocaust and just seemed like the kind of thing I wanted to study. So it was around this time, I guess, a little bit before this time that Richard Nixon and that administration had declared a war on cancer. I believe it was just a year prior to that. How did that resonate with you? Did you view that with great optimism,
Starting point is 00:25:47 or did you think that it was naive that in a matter of years, cancer would be eradicated in the same way that man had gone to the moon? Well, I had great hopes for making progress, perhaps naively, even at that point, not fully understanding all of the complexities. The National Cancer Act mainly influenced funding outside of the NIH. The NIH was already, I thought, well funded, had a building that had been built in 1953,
Starting point is 00:26:16 one of the largest buildings in this area, dedicated to doing research. It had a hospital beds. And so that National Cancer Act didn't have much impact on the intramural NIH that I could see, but again, remember, I'm a worker B. I became chief of the surgery branch and have never advanced in the hierarchy. I was where I wanted to be, turned down a fair number of physicians, and so when it came to the influence of the National Cancer Act on the country as a whole, I really wasn't involved with that very much at all. I focused on the work that I wanted to do
Starting point is 00:26:53 intramurally at the NIH. So how did you lay out a research agenda when you arrived in 1974? You're now finally able to, not only with the resources of money, but perhaps more importantly, with the resources of time, lay out an agenda for hypotheses that you want to test to build, effectively, a program to systematically narrow down the set of questions. So what was the process by which you went about doing that? So when I came to the NIH, knowing I wanted to study cancer, I started reading everything I possibly could about therapeutic approaches, which at that point were simply surgery, radiation therapy and chemotherapy.
Starting point is 00:27:35 Most used alone. And it was clear to me at that point that although incremental advances had been made over the years, surgery 3,000 years old, radiation therapy began immediately after RENKIN discovered X-rays in 1895, and chemotherapy arose in biological and chemical warfare laboratories here at Fort Dietrich in Dietrich, Maryland, to attempt to develop these agents. And it was in laboratory accidents in 1942 when nitrogen mustard was inadvertently exposed to laboratory technicians and found to develop a lymphopenia throughout their body with their lymph nodes shrinking down
Starting point is 00:28:25 that let a Yale physician to attempt to use nitrogen mustard, now known as Melfalan as a chemotherapy agent, and that was the birth of chemotherapy 1942, and that started chemicals to treat, to search for chemicals to treat cancer, but the advances that were being made were slowly and tiny incremental, I wanted something that big, that would make a big difference. And as I've again, to read about the immune system, how little was known about it,
Starting point is 00:28:56 but with the examples that I had, the intuition that I had developed, which is so important in science, that this might be something valuable that I decided to study immunology. Look, everything I could read about, and it seemed to me that the immune cells that would then be recognized as mediators of organ rejection were the agents that one needed to stimulate and why not use an immune cell as a drug
Starting point is 00:29:28 that has taken advantage of a patient selling immune reactions to try to treat the disease in immunotherapy And I started with them unbelievably naive experiments There was no way at that point to keep lymphocytes alive outside the body talking about 1974 You could take them out and they would die on a day or two. And you had no way to keep them alive. You could mix them with other cells and they would stimulate for a few days, but then they would die after about a week. And yet I was desperate to try to use lymphocytes with immune reactivity to treat patients. activity to treat patients. And so I began implanting human tumors into the mezzantery of mini-pigs.
Starting point is 00:30:11 Good friend of mine, David Sacks here, had developed a mini-pig colony that was partially imbred at MHC Losa. And so I would embed tumor in the mezzantery of these mini-pigs, wait about two weeks, operate and remove the inflamed lymph nodes that were draining that tumor, and gave those lymphocytes to six patients that as we take out their tumor, generate lymphocytes reactive against that tissue,
Starting point is 00:30:39 and then harvest lymphocytes from that pig and administer them intravenously to patients. And of course, nothing happened. But it's just a sign of how desperate I was a scientist from that pig and administered them intravenously to patients and of course nothing happened. But it's just a sign of how desperate I was at that point to have some impact to be doing something. I have over the door of my lab, you probably remember it, when you were in the lab, it said, it's a modification of a Louis Pasteur saying that said,
Starting point is 00:31:04 chance favors the prepared mind and what I added to it was, Chance, favors the prepared mind only if the mind is at work. And so I was trying things, and it was only with the discovery of T.C.L. growth factor in 1976 by Morgan Rossetti and Gallo, that opened the door to be able to manipulate lymphocytes outside the body by putting them in a growth factor called interleukin-2. And that was something I began to study quite intensively to see if one couldn't then grow lymphocytes that had anti-tumor activity and would retain it as they grew.
Starting point is 00:31:42 None of that was known, but those were the first experiments I was doing along those lines. Now, before we go further, I think it's worth making sure people understand some of the semantics, because obviously you and I can take so much of this for granted. But let's start with some basics about cancer. How does one define cancer? What separates a cancer cell from a non-cancer cell? Well, if you look at the broadest properties, there are two properties that separate cancer from other cells in the body. The first is uncontrolled growth. Virtually all of the tissues we have,
Starting point is 00:32:20 the fingernails or eyebrows, or you name it, they'll grow to a given amount and then they'll stop. Well, cancers don't have that signal to stop. They'll keep growing. And the second is it's the only cell that can arise in one part of the body and spread and live and divide and grow in another part of the body. And that's not true of virtually any other kind of cell. So cells with uncontrolled growth that can spread and grow elsewhere are the biologic properties. Now, we can dig down layer by layer by layer and get to the point of, well, what is the white of the normal cell ultimately become a cancer cell. And we now understand that that's due to the accumulation
Starting point is 00:33:05 of mutations in DNA of these cells divide, which explains why it's the common organs of the body, all of which have ducts, lining of those ducts, are constantly turning over. And as that DNA is turning over, mistakes are made called mutations, and it's that accumulation of mutations that results in the cancer itself. So we can take it all the way from the biology of uncontrolled growth, but down to the very molecules that are involved. We can describe it.
Starting point is 00:33:35 It doesn't mean we really understand it all, but we can describe it. And let's also explain to people the difference between the epithelial tumors, the hematologic tumors, and even let's frame it as it was in 1974 in terms of what was a person's odds of surviving. So maybe tell folks what the common epithelial tumors are and explain a little bit about the not we're not going to go into staging in great detail, but what's the difference
Starting point is 00:34:05 between local tumor versus metastatic tumor and what's the impact that has on a person's survival at the time you arrived at NCI? So the human logic cancers, of course, are the blood cancers and they start from progenitors in the hematopoietic system. Because after all the hematopoietic system starts from an individual stem cell that then divides into multiple different characteristics much as we all grow from a single fertilized egg from one cell that makes us what we are. So even back then, and a little more so now, if you developed a cancer of the bloodstream, which were about 10% of all cancer deaths,
Starting point is 00:34:50 so due to those, 90% of cancer deaths so due to the epithelial cancer. So these start in the solid organs of the body. And that go all the way from the rectum up through the GI track, through the stomach, through the esophagus, the pancreas, the G.U. organs, the testis, the ovary, the prostate. All of these solid organs have ducts. And as I've mentioned, it's the epithelial lining of the ducts that are turning over
Starting point is 00:35:17 that become the cancer. In blood cancers, it's the more primordial cells that develop into neutrophils and lymphocytes and other types of cells. So let's talk about the solid tumors, which are 90% of all cancer deaths. The last year in the United States were about 600,000 deaths due to cancer. 550,000 were due to the solid epithelial cancers. If you operate on a patient who develops a cancer to remove that cancer, then well over half the time that patient will be cured that is going to live their normal lifespan. But the less and half of patients that cannot be cured result in this enormous tragedy of 600,000 innocent people dying of cancer every year.
Starting point is 00:36:10 Once the cancer spreads, however, and this, in my view, is a dirtial secret of oncology. And that is that if a cancer spreads from its local site and cannot be surgically removed, then the death rate in that patient is 100%. That is, we have virtually no treatments that can cure systemic treatments that can cure a patient with a metastatic solid cancer,
Starting point is 00:36:41 that is one that has spread to a different site that can't be surgically recected. Now there are a couple of exceptions to that. There are two solid tumor exceptions that have existed for several decades. One is Choreocarsinoma. These are cancers that start in the placenta of pregnant women that then spread and you can have 90% of the lung replaced by that tumor, received methotrexate, a chemotherapy drug and it will all disappear. Still don't understand exactly how. Germ cell tumors in the male, tumors of the testis, like Lansom, Strong, at brain meds and lung meds, no matter how much they've spread, if you give
Starting point is 00:37:20 patients, platinum derived chemotherapy regimens, you can cause complete, durable regression of that metastatic disease. Up until 1985, those were the only cancers that could be cured. We can now add to that list, solid cancer. It's we can now add to that list melanoma and renal cancer because interleukin two
Starting point is 00:37:43 administered to patients back in the mid 80s caused complete regressions of widely metastatic cancer and patients that are still alive today. But that's it. Choreocarsinoma, germ cell tumors, melanoma, renal cancer, other than those for everyone who develops a spread cancer will die of it despite all the best treatments that we spread cancer will die of it despite all the best treatments that we have. Well, you read in the paper that this celebrity has cancer and they're going to fight it and they're going to beat it, but nobody beats it. We're in such desperate need of better treatments for patients with metastatic cancers because we just, we can beat them back a little bit we can improve survival by months
Starting point is 00:38:29 and for some cancers maybe a few years like breast cancer and colon cancer but everybody ultimately will succumb to the disease. And that's what I was actually going to ask you about which is if you think about the past 50 years in cancer and what you just said I really I still starkly remember having those discussions as a medical student with you. And the main point was we've basically just extended median survival of metastatic cancer, but we haven't increased overall survival. And what would be the extent to which even median survival has changed if we are just talking about stage four of the common cancers, breast, colorectal, lung, pancreatic.
Starting point is 00:39:06 How much has the needle been moved with respect to median survival, notwithstanding the fact that overall survival hasn't changed? Well, if you look at current papers and advertisements, most regimens that ultimately get approved by the Food and Drug Administration prolong survival by months. Probably the best example in modern oncology is the treatment of metastatic colorectal cancer. When I started median survival, it would have been maybe eight to ten months. Now it's two and a half years. So there's an example where life has been extended by years. Breast cancer patients can go from one regimen to another.
Starting point is 00:39:50 Each one causing some temporary regression of the cancer or limitation of its growth, but the cancer will ultimately grow and the patient will have to move on to something else. And that's why cancer care is so remarkably expensive because people just move from one treatment that can prolong life by a few weeks, like a lot of live in pancreatic cancer, six-week improvement in survival.
Starting point is 00:40:14 For $40,000, right? For enormous toxicity and huge, huge life-altering expense. The most frequently prescribed drug and oncology today is save aston bevacism ab which can impact on blood vessels and tumors and the trials of that regimen and combination with others will prolong survival in patients with colorectal cancer by about four and a half months. But those are the tiny increments, which can provide substantial, some can provide substantial benefit to patients, but not a curative, and people are always living under the cloud of that cancer that is going to regrow.
Starting point is 00:40:59 So we need something more dramatic than the application of surgery radiation and chemotherapy, barring some enormous advances in those fields. And I think one other point worth making for folks with respect to chemotherapy, I was actually just on the phone yesterday with one of my patients whose life is currently recovering from surgery, from a cancer. And he asked a question about the efficacy of chemotherapy and how good is chemotherapy at killing cancer cells, which I thought was an interesting question. And it led to a discussion where I said the challenge with chemotherapy is not finding
Starting point is 00:41:38 chemotherapy agents that can kill cancer cells. You know, I made a point that he probably had 20 chemicals in his home and in his garage that could kill every cancer cell remaining in his wife. The issue is how can you do that selectively? How could you do that and not at the same time kill the normal cells? And I think therein lies the arbitrage that needs to be exploited with chemotherapy and ultimately what we're going to talk about, which is immunotherapy. But I think that's an important point that many people don't understand, which is how
Starting point is 00:42:09 difficult it is to thread the needle of chemotherapy. It's not the killing of cancer that's hard. It's the killing of cancer and not killing the non-cancer. The point you make is incredibly important, because it's the selective killing of cancer without killing normal cells, which is not the case for virtually any chemotherapy or radiation therapy, even in surgery, you have to remove some normal tissue. And so it's that selectivity against the cancer that's so important.
Starting point is 00:42:38 And in fact, that's almost the perfect explanation for another reason that I think immunotherapy has potential importance because of its immense selectivity and sensitivity of recognition. Can recognize single amino acid changes in a protein and develop an immune response against it, trivial differences that can distinguish normal from tumor, or if you get a viral infection, destroy the virus in the respiratory system without destroying the respiratory epithelium, it's the exquisite sensitivity and specificity of the immune reaction that I think makes it such a seductively interesting approach to trying to develop a new cancer treatments. Let's take a moment and have people get a little bit deeper on how the immune system seductively interesting approach to trying to develop a new cancer treatments.
Starting point is 00:43:25 Let's take a moment and have people get a little bit deeper on how the immune system works. I remember, for me personally in medical school, it was one of the most interesting sets of courses we took where the courses in immunology, in particular how T cells worked was fascinating. It seemed to lend itself to a story almost and with generals and soldiers and all of these things. So, explain to people, let's maybe
Starting point is 00:43:52 start with a virus as the example, because obviously in the era of coronavirus, that's on everybody's mind, and we can talk about how the body defeats a virus, but then pivot to then how, in the case of cancer that exact same immune system can accomplish what you just said. So let's take viral infection as an example, whether it's a common cold or coronavirus, the virus comes into the body and infects the respiratory epithelium in the pharynx and the bronchi and the lung. And as that virus then infects those respiratory epithelium, the virus replicates and the infected cells
Starting point is 00:44:37 then express the viral proteins. The immune system has evolved to detect proteins or other molecules that are not part of the normal self of the body. As the immune system evolves, cells that can recognize foreign invaders get spared, whereas cells that can attack normal tissue get eliminated in the thymus, and so except for autoimmune diseases, we don't have cells that can recognize normal tissues. They've been eliminated in the evolution of our immune system. So you have lymphocytes, B cells that make antibodies, T cells that act directly by interacting with other tissues. And so the immune system, via antibodies or T cells, recognizes viral protein that's now being expressed by the respiratory cell.
Starting point is 00:45:34 The lymphocytes are constantly patrolling the body. Every 14 or 15 seconds, your heart's pumping out these lymphocytes, sort of circulating through the vascular system, sometimes extravacating into tissues coming back into the lymphoid system and returning to the heart via the thoracic duct. Well, when the lymphocyte encounters a foreign antigen to which it can have reactivity, that's not self. And define an antigen for folks to tell people what an antigen is, how long is it, what's
Starting point is 00:46:03 it made of? So an antigen is a molecule in the body that is not normally being expressed in the body by tissues. They're generally proteins, but they can be carbohydrates, and what makes that molecule an antigen is its ability to be recognized by a T lymphocyte or a B lymphocyte, that is a T lymphocyte that can directly recognize an infected cell or a B lymphocyte that can make antibodies against it plasma cells. And so if a molecule is recognized as foreign, the immune system can recognize that that antigen. Well lymphocytes are patrolling the body, They encounter this viral antigen and the respiratory epithelium. They stop at that location and you can visualize this in mouth-hearsal
Starting point is 00:46:49 with something called two-fold time microscopy. They stop at that location and you can see them extravacate into the tissue. When they're there, they then begin to divide. As they divide, the dividing cells can further recognize the viral protein and starts making molecules that can destroy the infected cells but also call other cells into that arena macrophages, neutrophils and so on. And that's what
Starting point is 00:47:15 an immune reaction is. As the antigen is eliminated by these mechanisms, lymphocytes, other cells, there's no reason for those cells to stay around anymore. They're not stimulating. They enter the circulation. But now you have patrolling the body for the rest of your life. Long live lymphocytes that can recognize those foreign molecules.
Starting point is 00:47:36 And that's why when you get immunized against smallpox, you have that immunization for the rest of your life. And hopefully for coronavirus, although we don't know that, we don't know the extent to which those cells survive. Now, at the outset, you said there are two things about cancer that make it different from self. It has these two properties that individually wouldn't be the end of the world, but when you combine them, they're devastating. It's this failure to respond to cell cycle signaling, which results in unregulated growth, and it's this capacity to leave the site of origin and grow in an unregulated manner elsewhere.
Starting point is 00:48:14 And you also mention that this is the result of, although you didn't use the word, somatic mutations, and we can clarify for people, these aren't typically mutations that people are born with, although in diseases like Lynch syndrome, that might be the case, that it leads to that, but these are acquired mutations. So the natural question would be, why is it that a cell that has these acquired mutations that clearly produce a phenotype that is different from self? why wouldn't that be foreign enough for the immune system to act? In other words, why does cancer even exist in the first place? Why doesn't it get squashed out in its infancy? These mutations, these changes in DNA that a random events as the cell is dividing can produce proteins that can be
Starting point is 00:49:07 recognized or other molecules recognized by the immune system and they do it in complex ways by breaking down small molecule peptides and putting it on the cell surface but the immune system can recognize these mutations. And it's only been in the last, I'd say, three or four years that we now recognize these mutations as commonly recognized by the immune system. And about 80% of patients with the common epithelial cancers, it turns, as a result of the research done in recent years, do exist that can recognize the products of the mutations. But the immune system against them is too small, is not vigorous enough, what does that mean? Create enough cells, create receptors that have a high enough validity for recognition
Starting point is 00:50:01 to the tumor. The immune reaction is not very strong, and the growth of the tumor can overcome the small impact that an immune reaction might have in killing some tumor cells. Plus, for a tumor cell to survive and grow, it develops certain properties that can suppress the local immune reaction.
Starting point is 00:50:30 It can make molecules like transforming growth factor beta, TGF beta. It can make cytokines like interleukin 10. It can cause the development of cells lymphocytes that inhibit immune reactions. I mean, virtually every physiologic system in the body has stimulatory elements and inhibitory elements. You have hormones that can increase gastro-secretion, some they can decrease it. You have a sympathetic nervous system, a parasympathetic nervous system. Well, the immune system is the same. It has effective cells that can be very aggressive in recognizing antigens, and it has regulatory T cells that deliberately suppress immune reactions. And that's one of the things that keeps us from developing autoimmune disease. But there are many of these regulatory elements. Recently,
Starting point is 00:51:16 described myeloid derived suppressor cells can suppress immune reactions. And so it's the balance of the aggressive immune reaction against the inhibitory molecules that can prevent that immune reaction that is the holy grail of trying to find effective treatments. And effective treatments come in both directions. Interleukin II stimulates immune reactions. And we now have checkpoint modulators,
Starting point is 00:51:43 like Ibalumimab or PD1 inhibitors that can inhibit these inhibitory factors and thereby stimulate the immune reaction by taking away the breaks on the immune system. So the more we understand, the more we can take advantage of the biology. So let's go back to the first of those because that seems to have been the first big break you got at NCI after Gallows discovery was interleukin 2. So now you had both a cytokine that could allow you to grow lymphocytes in vitro, but also something that could be given to patients in vivo to stimulate the immune system. So how did that sort of propel your work? Well, with the advent of interrelugion two, what had been shown was that there were some bone marrow cells could make a substance which would keep lymphocytes alive outside the body. But the
Starting point is 00:52:38 minute I heard about that, there were a series of questions that arose. Well, if it kept lymphocytes alive, could it keep lymphocytes alive and dividing in a format that enabled them to have all of their immune recognition? That is, as they grew, would they just lose that property? And so we try to demonstrate that by developing cells that could recognize what we call alloe antigens, that is very strong antigens that are present in one person
Starting point is 00:53:08 that inhibit the ability to transplant organs, for example. And so our initial studies were to see whether or not we could develop lymphocytes, grow them in culture, and cause experimental skin grafts in mice to disappear faster. We're not talking about tumor, a Beth normal tissue. And we showed that, in fact, we could grow lymphocytes that retain their function in the laboratory and then retain their function in vivo. Well, with that knowledge, we didn't want to cause skin grafts to disappear more quickly. With that knowledge, we had to try to develop cells that could
Starting point is 00:53:45 react against the cancer. And very early on, when we grew cells in interleukin 2, we found that in fact, they could destroy tissue culture cancer cells, have some impact on normal culture cells as well, just by virtue of exposure to interleukin 2. And we call them lack cells, lymphocaine activated killer cells, and we studied them for three or four years, turned out to be a false alarm because they could impact on tiny little tumors in mice before they became vascularized, but why the time they were vascularized, they would not work in mice at all. But interleukin 2 seemed like a molecule that might be able to stimulate those rare cells in the body that could recognize the cancerous foreign or develop cells in the laboratory that could do that recognition. And that then led us to many years of experiments in the laboratory, but also clinical trials trying to see whether or not either interleukin-2
Starting point is 00:54:45 administration alone, well cells that you could devise in vitro that could recognize the tumor and administer those. And that was a very frustrating time. It wasn't until 1984 that we finally figured out a way to use interleukin-2 to mediate regression. We treated over 70 patients with use interleukin 2 to mediate regression. We treated over 70 patients with either interleukin 2 or cells that we grew and interleukin 2 and administered to patients without seeing a response until we modified the schedule of interleukin 2 administration knowing it's pharmacokinetics that is only has a half life inside the body of about seven minutes. So we had an alter of the schedule. We had to give higher and higher doses, which mediated toxicity.
Starting point is 00:55:33 Until finally a patient that we treated in 1984, where widespread melanoma was administered in a low-con2. It was the first patient, finally, after 70 other patients, to show us a tumor regression. The first time that a deliberate immunologic maneuver could reproducibly cause cancer regression. It was one of the few eureka moments that I've had in doing research, but the realization finally that after all of those patient deaths, due to everybody had advanced cancer, all would go on the diet of their cancer, survived in that patient,
Starting point is 00:56:09 now alive over 35 years later, free of disease. You know, it reminds me a lot of the Thomas Starsel's work in the 1960s with liver transplantation, where the number of patients who died, it was hard to keep track of before finally achieving the technical success that was necessary. Both the perioperative care and the postoperative care and the technical skill necessary, plus the immunosuppressive regimen. All of those four things had to be firing on all cylinders for patients to finally undergo liver transplantation.
Starting point is 00:56:44 to be firing on all cylinders for patients to finally undergo liver transplantation. And this patient in 1984, if I recall, it was the 67th patient treated, meaning 67 consecutive patients died of metastatic cancer, and were unresponsive to interleukin 2. The first question is really just a logistics question. How many different histologies were in that group? How many different types of cancers were you treating at that time? We were treating all cancers, metastatic cancers, with the idea that although they each arose from different organs, had somewhat different properties and methods of spread, they would be commonalities that could be attacked. And so we were treating all kinds of histologies. It was the first patient that we treated with this revised regimen happened to have a melanoma.
Starting point is 00:57:32 The third and fourth patients had renal cancer. And as we continued using Erlukin II, we found that those two histologies, patients with those two histologic types of cancer could respond and ultimately response rates and those two diseases turned out to be about 15 to 20% of patients with about a third of those patients having complete durable, durable regressions.
Starting point is 00:57:55 But it was a little different than the liver transplant situation. Because in that situation, there were technical problems that had to be overcome and it was a genius of Tom Starvel to stick with it and to figure out those technical problems. When it came to immunotherapy for cancer, it was a little different. We didn't know that it would ever work.
Starting point is 00:58:16 We didn't know that there was ever going to be an immune system that could cause a cancer to disappear. In contrast to, if well, you could work the technical problems out of so the vessels together, you could get this thing to work. And so that first patient had an enormous impact on me and on the field because it showed that it was possible. And until you know it's possible, you never know that it's ever going to occur. And so that changed everything because it showed that simply administering this one molecule, a T cell growth factor, could cause a cancer regression in a patient, and that then led us to studies to understand how that was occurring, and that then
Starting point is 00:58:58 led to a lot of different direction, cell transfer, gene modification, and so on. directions, cells, transverging, modification, and so on. How did you keep going in the face of all of those failures up until this patient's miraculous remission in 1984? Because again, if you were working as a surgical oncologist at the time, you would not have been exposed to that death. The surgical oncologists work would have been done after the primary resection. Typically, the medical oncologist would be the one that would be at that patient's bedside as they progress through treatment.
Starting point is 00:59:37 But you were seeing something that you would not have seen had you chose a different arc to your surgical career. And I just, I wonder how you coped with that. What were those drives home, you know, what was it like to be alone in your thoughts? Well, you know, as I look back on it, it seems remarkable that there were so many patients, one after another, everyone died eventually of their cancer because we did not have any impact in the manipulations that we were applying. And, you know, you're a doctor and you know that it's not the patients that do well, that you remember.
Starting point is 01:00:11 It's the patients that you fail to help, that you remember. And it was just a remarkable number of tragedies, young people dying of cancer, people of all ages. But I had this intuition based on everything I knew about biology and everything I had studied in biophysics. I had an intuition and also influenced by these inklings of the first two patients I mentioned that this would work. You know, I recently saw a quote by Abraham Lincoln that said, success consists of moving from failure to failure
Starting point is 01:00:53 without loss of enthusiasm. And that actually happened to me. I actually just saw that quote a few months ago. But I always felt it was going to work. And it did eventually, and a small number of patients, still a long way to go. But at least now we have effective immunotherapies for a variety of diseases that can be effective.
Starting point is 01:01:18 And when that first patient responded, it all exploded in my brain. It does work. This can work. And we'll figure out now ways to make it better. And I imagine Alice was a big part of that support for you. Again, I know I have the privilege of knowing her and knowing how important she is. Do you think you could have got through this alone without the support of your family? I mean, you had to do this very difficult thing, which was basically have this remarkable obligation
Starting point is 01:01:48 to your family, which every father does. And at the same time, you felt like you were carrying the weight of your world, the weight of the world on your shoulders trying to take care of these patients who were otherwise really left with no hope. Do you see that as sort of a synergy between those two? They were probably, and I'm not exaggerating 40 days in the first 40 years of my work here That I was in town not traveling that I was not in this hospital. I
Starting point is 01:02:20 Would come in every day, of course. I would come in every Saturday. Sundays, I would come in to go over research with some of the fellows. You probably remember that or see some patients. That kind of life requires support of some kind. Then there are not a lot of wives who I think would tolerate that kind of commitment outside, outside the home. And Alice was such a person. Never hassled me about it, always understood that, I remember once she said, look, I know what you're doing is important.
Starting point is 01:02:57 And so what I'm going to do as much as I can is relieve you of the burdens that we commonly face as part of daily living. She handles, I haven't written a check in 20 years. is relieve you of the burdens that we commonly face as part of daily living. So I've I haven't written a check in 20 years. Alice pays our bills and really takes care of so much that enabled me to work at that level. But it was a family thing. I have three daughters who are growing up as all of this was happening. And I remember when my oldest daughter applied to her,
Starting point is 01:03:20 my first daughter applied to college, Beth. She opened her college essay with it, set in somewhat along the lines of, at our dinner table at home, we're much more likely to be talking about cancer and AIDS than the Washington Redskins. And it made me understand how much the kids had been affected by all of this talk of death
Starting point is 01:03:44 and suffering from these diseases? So it takes a family and I doubt I could have done it without that kind of support. So once you identified this patient, patient number 67, did you have an inkling what it was about melanoma and renal cell cancer that made them particularly immunogenic relative to this whole host of other epithelial cancers that were less reactive. The answer is no, but the answer would be yes, 35 years later. But I think now we do understand what's different about melanoma, but we didn't at the time. We were seeing responses to interleukin 2 in patients with melanoma and kidney cancer,
Starting point is 01:04:29 but no other diseases would respond to interleukin 2. And we learned that the hard way treating over 600 patients with interleukin 2 here at the clinical center, it turns out those two diseases were uniquely responsive. And we now know, at least from melanoma, why that's the case. And that is the immune system is recognizing the products of mutations. And melanoma, if one looks at the number
Starting point is 01:04:58 of the frequency of mutations among different cancer types, melanoma has more mutations in any other cancer type with the exception of lung cancer. They're about equivalent, about 400 mutations per tumor as a median. And that's very likely because melanoma induced by a carcinogen, ultraviolet light, lung cancer by the carcinogens, largely cigarette smoke or the environment that leads to an increased number of mutations. And that at least is part of the answer. And that is the more mutations
Starting point is 01:05:30 you have in the cancer, the more likelihood that you'll develop a particular protein, foreign protein that can be recognized by the immune system. Did you ever see patients with Lynch syndrome response? They would typically have many mutations as well, wouldn't they? You're exactly right. So there are some situations like the microsatellite unstable cancers, colon cancer, other kinds of cancer types, Lynch syndrome. But we didn't understand that with mutations that were
Starting point is 01:05:59 involved at that point, and I don't remember ever treating a patient with Lynch syndrome. You're right. They would have a very large number of mutations. So for comparison, if we talk about a standard, you can't even use the word standard. There's no such thing as a standard cancer right now. We know so much about cancer that every cancer is different, but what would be the median number of mutations in a metastatic breast cancer, colon cancer, or pancreatic cancer? You would probably encompass 80% of these common cancers if you considered mutation frequency between 60 to 70 and 150.
Starting point is 01:06:38 That would be the measure. The median would probably be somewhere about 110, but it would vary from cancer type to a cancer type. Some pediatric cancers have very few mutations. Some cancers have more, but about, I'd say the median would be very likely to be about 110. How many of those mutations would be driver mutations? So they are oncogenes, tumor suppressor genes, they are playing a functional role in the cancer, versus what we might call passenger mutations that can still produce antigens. They would still generate a peptide that could be recognized by MHC, but they're not playing a functional role in those two properties of cancer that we spoke about.
Starting point is 01:07:23 So about six years ago, we described an assay that would enable us to actually identify the exact molecular nature of these antigens that are recognized by T cells. And that came from, again, the understanding that it was the products of these unique cancer mutations that were being recognized by the immune system. Well, it turns out that some of these antigens that are recognized by T-cells are recognizing the proteins that derive from driver mutations that is it caused the cancer in the first place. Like P53 present in half of all cancers, but only about 2% of patients develop immune reactions against it.
Starting point is 01:08:08 K-RES, about 90% of pancreatic cancers, will have that as a driver mutation. But what's stunning to me about oncology and the biology of cancer, and that is how few of these shared cancer mutations exist. There's P53, there's K-RAS, to some extent, pick 3CA and breast cancer, maybe B-Raph mutations in melanoma. Other than that, the incidence of driver mutations as a cause of cancer is low single digits. You'd think there would be many mutations that would change the cell so dramatically,
Starting point is 01:08:46 but it turns out it's not only those few driver mutations, but the accumulation of mutations, each with its own property that in and of itself appears unlikely to cause cancer, but in concert with the action of other genes, other mutations does cause a cancer. And we've, you know, more recently shown in a breast cancer patient that we published a few years ago that we could mediate complete, durable regression now over six years later by targeting for what appeared to be random somatic mutations, none of which had a driver function, but in concert caused the cancer and by attacking them, you could cause cancer to disappear. I mean, I still, I'm struggling to understand why it is that a P53 mutation or a K-RAS
Starting point is 01:09:35 mutation is not immunogenic. Is that simply due to the evolution of cancer that because of the ubiquity of these mutations in cancer, cancer has come up with enough evolutionary tricks to evade detection of those mutations. I mean, that's a teleologic question, but I mean, do we have any biologic insight into this? So you have to get a little deeper into the biology. For a mutation gives rise to a molecule, a protein, right?
Starting point is 01:10:06 It's DNA, RNA transcribed, translated into a protein. For that protein, even though it has now a mutation, a string of amino acids that are not seen as normal by the body, that mutation, that abnormal amino acid, called a non-synonymous mutation, is only recognized acid, called a non-synonymous mutation, is only recognized by the immune system. If the molecule in which it occurs is broken down inside the cell into small peptides, that is small sequences of amino acids,
Starting point is 01:10:38 or the tumor cell or the antigen presenting cell takes an antigen, a protein from the outside of the cell into the cell and breaks it into small peptides, strings of amino acids. Well, that has to happen. And then one of those strings of amino acids has to fit onto the surface of the patient's own HLA molecule, that is the kind of molecules that we call transplantation molecules. And so for a mutation to be recognized by the immune system, it has to be broken into these nine to 11 amino acid peptides and fit on that patient's own transplantation molecule.
Starting point is 01:11:20 And that transplantation molecule is highly polymorphic. There are hundreds of them. And so if you had a mutation and your cells made small peptides, but it didn't fit on your HLA molecule, it wouldn't be recognized by the immune system. And so as we've learned more in the last five to six years about what cancer antigens are. It's these mutations that are broken down and put on the surface of a patient's presenting cells or tumor cells.
Starting point is 01:11:54 And that turns out to be between one and a half and two percent of all mutations. And when you look at melanoma, it's 1.3 percent. When you look at the gastrointestinal cancers, 1.6 percent, breast cancer, 2.1 percent of mutations are immunogenic because they happen to fit into that individual patient's HLA molecule. And the most stunning finding of recent years, in my view, in this field, is that virtually every patient recognizes a unique antigen. And so we're in the process of writing a paper now on 195 consecutive patients that we've identified the exact antigenic nature of what the T-cell can recognize,
Starting point is 01:12:46 and it can recognize in about 80% of all histologies. And that turned out to be 363 individual antigens that were recognized in these 195 patients. And no two patients shared the exact same antigen with the exception of two patients that had a K-RES mutation that was recognized on a very rare Class I molecule, CW-802, HLA molecule. So just to make sure I understand that, you're saying that in this series of nearly 200 patients, the first interesting finding is each of them produces at least one
Starting point is 01:13:29 neoantigen that is immunogenic. 80% of patients will antigens, 392 of them were unique, not shared by any patients with the exception of K-RAS. Now, it's not to say that P53 or other driver genes can't recognize it, but they don't naturally recognize you. You might be able to raise those very rare cells, but that's correct. It's good news and it's bad news. Explain why that is to people, because I was just about to say,
Starting point is 01:14:15 that's really good and creates a huge problem for scale. You got it. It's exactly right. It's good news because we finally understand after all of these decades, what a cancer antigen is. Back in 1985, I knew one existed. I didn't know what it was, whether it would be shared, and we spent an enormous amount of time trying to identify shared antigens, especially in melanoma, these melanocytes antigens, shared by normal pigment-producing cells. these melanocytes and it's shared by normal pigment-producing cells. But now that we understand what an antigen is, and we understand that most cancers contain multiple mutations, which give rise to the antigens, well since almost all cancers have mutations,
Starting point is 01:15:01 if we can figure out ways to target these mutations that are foreign, we potentially have a treatment applicable to all cancer histologies. Since almost all cancers have mutations, let's recognize them. And now you don't have to worry whether it's a breast cancer or a colon cancer or a brain tumor, the T cells are there. So the possibilities of developing broadly applicable treatments exist. The bad news is as you point out it's going to have to be as you target these mutations highly individualized treatments unless you can fully unleash the immune system against even the most minor of antigens which we can't do now, checkpoint modulators
Starting point is 01:15:47 have virtually no impact on the overwhelming majority of the solid epithelial cancers. It means that if you're going to stimulate immune reactions via a vaccine or a T cell as a drug, it has to be individualized to target that cancer because that antigen is present only in that patient, but not in any other patient. And that's going to make it very complex to develop.
Starting point is 01:16:14 But when we developed this other form of T-cells, CAR T-cells, a whole other story, people said that it could never be applied. But in fact, if you have something that works and can cure people, the genius of modern industry will figure out ways to make it available. Well, I wanted to go back to 1985 to pick up the story with both Ronald Reagan and Tittle unrelated, but temporarily related. But before we do, because you brought up CAR T cells, let's let's tell the story about the diffuse B cell lymphomas that ultimately led to kite, because it's a great way to, I think most people listening to this will have heard of a CAR T. Let's tell the story about the diffuse B cell lymphomas that ultimately led to kite, because
Starting point is 01:16:45 it's a great way to, I think most people listening to this will have heard of a CAR T. This is an illustrative case to explain how they came about, how they differ, of course, from a regular T cell receptor. And as you said, how industry basically came in to solve a problem that, at the outset, looked pretty daunting. So pick it up wherever it makes sense with respect to. So again, you have to understand the biology. You have to go back to the biology that normal
Starting point is 01:17:10 T cells have receptors that can recognize antigens on the surface of a cancer cell, right? These tiny peptides that are put on the surface. Those alpha beta chains are expressed by a lymphocyte, and that's how the immune system recognizes its antigens. Well, there's a way to create an alternate way for a lymphocyte to recognize an antigen that was created by some scientists at the Weitzmann Institute about 10 or 12 years ago, Zelligesshar and Gideon-Bros. They took advantage of antibody recognition.
Starting point is 01:17:42 Now, antibody recognition is very different than that of T cell recognition Antibodies recognize the three-dimensional structure of a molecule on the surface of a cancer cell or of any cell Not a processed peptide brought to the surface, but an actual molecule on the surface and that antibody like a lock in a key will latch on to that antigen and recognize it. Well, T cells can't do that, but by creating a chimeric T cell, you put antibody recognition domains into a lymphocyte that converts the lymphocyte from its normal recognition from its own receptor into the recognition of this antibody that you've put into the lymphocyte.
Starting point is 01:18:28 And that expands the number of molecules that can be recognized by T cells. And so it's this chimeric antigen receptor, which is part receptor, normal receptor, but with antibodies attached to it that enables the lymphocyte to recognize now molecules that it never was able to recognize in the course of evolution based on antibody recognition.
Starting point is 01:18:53 And it turns out that there are very few molecules on the cell surface, very few, like I could name the few I know of in the fingers on one hand, that are unique to a cancer that are not on normal cells. And we learn the hard way that the immune system will destroy a normal cell just as quickly as it will a cancer cell and we've mediated cancer deaths by targeting antigens that are present even in very low levels on normal cells. Well, it turns out as a molecule on B lymphocytes called CD19. We don't exactly know what its function is, but when B cells turn into lymphomas and
Starting point is 01:19:37 leukemias, they continue to express CD19. And so with this understanding, and as soon as I heard about these chimeric T cells, I invited Zelle Geshar to come to a sabbatical in my lab, he came the next day, the next year, and spent three years working in the surgery branch. And we worked out ways to use car T cells to attack cancers, but we can never get them to disappear because the molecules that we were giving could not be used in large enough for numbers because of their ability to recognize normal. Well, CD19 expressed by leukemias and lymphomas, they developed from normal B cells, express CD19. And we developed a technique to introduce these anti-CD-19 car molecules,
Starting point is 01:20:27 chimeric antigen receptors, into T cells, to create a car T cell, that when we administered the patients would kill every cell in the body that expressed CD-19. So all the normal B cells were eliminated. But so too were lymphomas and leukemias. And that became the first actual cell in gene therapy ever approved by the FDA. So how did that happen?
Starting point is 01:20:53 Well, we studied the ability of these anti-CD-19 car T cells to kill cells and experimented animals and they did, they wiped out normal B cells, but you can live without normal B cells. Because you can give antibodies by giving antibody infusions. We used these CAR T cells to treat the first patient ever to receive a CAR T cell. This was in 2009. This was a patient that had a lymphoma that is spread throughout his chest had been through four different chemotherapy regimens that had enormous chelogram tumor burdens in his body. We treated with CAR T cells
Starting point is 01:21:32 that could recognize a CD-19 molecule on the surface of normal B cells and tumor. And all this tumor disappeared. And he's, well, we treated him in 2009. He's 12 years later and completely diseased free. We published a series of those over the course of the next two years. And we had seven or eight patients
Starting point is 01:21:55 who had a complete disappearance of all of their lymphoma, diffuse large B cell lymphomas, the most aggressive form of, and lethal form of lymphomas that people develop. And they develop complete regressions that were ongoing for at least seemed like several of several years. Well, once we did this, and incidentally all the patients' normal B cells disappeared, but again, you can live without B cells.
Starting point is 01:22:22 And so in 2011, two years later, after we had published several of these papers, and a year after we published that where Carl Junet University of Pennsylvania used these CD-19 CAR T cells to treat leukemia patients. And so two years after our description of multiple lymphoma patients undergoing a complete regression, I was contacted by a former fellow named Ari Beldira who had worked in my lab 25 years earlier. He was just finished his urology training, became a professor of urology at UCLA. And he, we had become friends. And he came to see me in 2011 saying,
Starting point is 01:23:00 hey, you're treating patients with these T cells, these chimeric T cells, these car T cells, and successfully treating lymphoma patients. I want to commercialize this. I want to start a company to do this. And we had had several companies come through, like Johnson and Johnson, brought in 12 people, examined everything we had done, said,
Starting point is 01:23:22 hey, if we have a lymphoma, we'll come back and get treated by you, but we don't see how we can make any money doing this at Johnson and Johnson. Well Ari Bellingham had a different vision. He said we can figure out a way to make this available and in 2012 we formed what's called a creative cooperative research and development agreement that enabled us in the lab to work with this company, this biotech company, started kite farmer. They were able to give us funds to help support the research. And so we just started that in 2012. We signed the creative. We worked together. We treated over 50 patients, showed that this could happen. And over half of patients will undergo durable regressions. He then did a multi-institutional study, and Novartis was doing this in leukemia patients
Starting point is 01:24:09 almost simultaneously. His multi-institutional study reproduced a results exactly about a 70 percent objective response rate with 50 percent durable, complete regressions. And in 2017, five years after Kite started working with us on this, Kite was sold to Gilead for $11.9 billion. That all happened in the course of those five years, and that treatment is now available, thanks to Kite and the Vartis available for use in patients in the United States and Europe and parts of Asia, that can effectively treat these B.C.L. lymphomas and leukemias.
Starting point is 01:24:53 So, it's really a pretty incredible story that evolves so rapidly. Yeah, it is. I mean, do you think that CAR T cells can have efficacy against non-hematologic cancers? Right now the answer is no We have no way to use them against solid cancers again because the solid cancers to be treated with a car T Cell you have to have a molecule on the cell surface that's unique to cancer We originally didn't fully realize quite how sensitive they could be, and when we targeted molecules that were on normal cells, patients died, devastating events in the development of the treatment.
Starting point is 01:25:36 But monocle antibodies were first described about 45 years ago, and no one has found unique monoclonal antibodies against molecules uniquely on cancer cells and not normal cells. And that's what you need to make a chimeric T cell receptor. You need an antibody that you can put into a lymphocyte that has specificity. And antibodies just have not evolved to recognize individual cell surface molecules on cancer which are shared by normal cells, whereas conventional T cells do because internal proteins then get digested and brought on to the surface. So right now, there's very little prospect for CAR T cells being useful for the treatment
Starting point is 01:26:22 of solid tumors, but that's not to say that some brilliant ideas will come forth in the years to come that will make them available. Right now, they're not useful. Now, what about for organs that are not essential? So, where you could wave the need to recognize or differentiate between cancer and non-cancer. So, for example, breast or even pancreas. I mean, if a person had metastatic pancreatic cancer and you were willing to completely lose both normal and non-normal pancreatic cells and render that person a type one diabetic, it would still be worth it. So, are there any antigens that are present on exclusively pancreas or exclusively breast or colon? Obviously, this wouldn't work for liver and lung, but is that a slightly easier problem
Starting point is 01:27:06 to solve? Or is it just as hard? Well, it's just as hard because for the past 45 years, some of the best immunologists in the world have tried to develop these monoclonal antibodies that can uniquely recognize cancer and they have not found any. Either because they haven't done it right, or they just aren't these molecules on the cell surface. And even very recently last several months, you probably heard about this T-immunity, these two deaths for patients that were targeting what was thought to be a prostate-specific molecule, BSMA,
Starting point is 01:27:36 but it was present on normal cells, and that can result in death of patients. And so, yes, if you could find the molecule unique to prostate cancer, breast cancer, that is expendable organs, you could develop more effective cell-based therapies against them. But right now, none of those molecules have been identified. So, let's now go back in time to post the IL-2 insight. You have this other amazing realization, which is there are certain types of lymphocytes that manage to track to tumors, these T cells that infiltrate the tumor, and they're called
Starting point is 01:28:18 TIL. How did you come to understand these and understand the efficiency with which they could identify tumors? So things seem to move very slowly, although we had an explosion of ideas, but looking back on it when it comes to scientific advance, it actually moved along pretty quickly. Because IL-2 as a T cell growth factor was mediating reproducible regressions, it seemed reasonable that is being mediated by the ability of interleukin 2 to stimulate lymphocytes in vivo. And so in melanoma patients,
Starting point is 01:28:53 we looked for T cells that could recognize the tumor deposit itself. We didn't know what it was recognizing. And what better place intuitively to look for a cell doing battle against the cancer than within the cancer stroma itself? And so we grew those cells, one out of peripheral blood, but we also grew cells invading into tumor, called tumor infiltrating lymphocytes, or till cells. We grew them in vitro, and in animal, and then very quickly in human experiments, grew those lymphocytes to large numbers in vitro and administered them to patients with metastatic melanoma. And now instead of the 15% response rate that we got from giving interleukin to alone,
Starting point is 01:29:41 by giving lymphocytes that we grew and interleukin to these till cells We got response rates 30 35% in melanoma patients and so it represented a substantial improvement But they were pretty short durations they were real, but they did not appear to be durable But it was the first demonstration that lymphocyte transfer as a sole modality could cause tumor regression in patients with melanoma. And so some lesser extent kidney cancer was mediated by lymphocyte. So that intuition then became a reality of a biologic finding. And that is lymphocytes were the cause biologic finding and that is lymphocytes were the cause of These regressions or could be the cause of regressions and then things moved along fairly
Starting point is 01:30:34 Well slowly hand quickly depending on your point of view It immediately became a Parent that if we had these lymphocytes naturally maybe we could genetically modify them to be more potent That is they were making factors that drew other cells in. Well, let's introduce that gene into them, but no one had ever introduced a gene into human cells. And so, I teamed up with two scientists at the NIH, French Anderson and Mike Blaise, who were trying to develop gene therapies to replace a denison deaminase deficiencies,
Starting point is 01:31:07 a lethal deficiency, and young children to see if they couldn't introduce those genes, but of course, no genes had ever been introduced into people. We decided to see if we could break the ice about putting foreign genes into humans, by putting a marker gene into the lymphocytes we were administering to patients.
Starting point is 01:31:28 We picked the gene, a bacterial gene called Neomycin Fast-Fatraspharase, inserted that gene into a patient's normal lymphocytes, and our plan was to administer them so we could track where these lymphocytes were going inside the body because they would have this unique bacterial gene that were being expressed. And so we proposed that to investigation review boards at that point the government had established what's called the Recommonant DNA Advisor, he committed the rack adequately named to add a review any clinical proposals. We went through, we tabulated at 117 different review groups having the old back and forth as they made changes until the rack finally voted was a painful time 13 to 4 to allow us to do it but the director of the NIH
Starting point is 01:32:16 James Weingarden insisted that before we would start tampering with the human genome we needed unanimous consent back and forth making changes. Finally, there was a vote of the RAC 13 to nothing to zero with one abstention, which was unanimous. And so we got permission to proceed with the clinical trial, biotechnology activists, and filed lawsuits against the NIH saying, we shouldn't be tampering with the human genome. It was immoral, it was ungodly,
Starting point is 01:32:44 but we finally got permission to do it and inserted these lymphocytes that were genetically modified with this bacterial gene that did enable us to track the cells inside the body. When we did biopsies, it was a paper we published in the English Journal of Medicine. And that then led to the gene modifications of lymphocytes that we attempted to use to improve them.
Starting point is 01:33:04 We put in the gene for interleukin 2. That didn't improve them because we couldn't regulate it, but it just started our endeavors to genetically modify cells that finally came to their fruition in the CAR T cells by inserting the genes that would insert these new receptors that could recognize molecules on lymphomas and leukemias. So that started us on the track of trying to improve the cells. And then there were a variety of advances. We learned that you had to first wipe out all of these inhibitory T cells, regulatory
Starting point is 01:33:37 cells, before you gave the administered cells. And that then jumped the response rates up to 55% in melanoma patients with about 25% being durable, complete remissions. We then started developing ways to use T-cells to do cancer treatment and 2013 finally realizing that it was a unique mutations. We developed techniques that enabled us to develop T-cells specifically targeting mutations and published the first paper on that in 2014. It was a patient with a biodecance or a collageocarsinoma that was widespread in the lungs and liver. We gave her bulk TIL till cells did not work. We gave
Starting point is 01:34:25 cells that were uniquely directed against her mutations and she's undergone a dramatic regression, it's now disease-free. But those were till that were not genetically modified? Those are the nuts. So our work went in two directions, genetically modifying till or figuring out ways to use natural till and these were natural till that were selected for mutation reactivity and given to a patient whose natural immune system was temporarily eliminated and she's living disease free now eight years later all of her liver and lung disease is gone and we subsequently now have published on these T-cells that recognize unique mutations in their ability to cause regression and cervical cancer
Starting point is 01:35:12 induced by human papilloma virus, by colon cancer, that patient recognized K-RES, breast cancer, recognized four random somatic mutations. We're now struggling to figure out ways to more efficiently target the products of unique mutations that cause the cancer. So if I run it, that the Achilles heel of the cancer is gonna be the very abnormalities
Starting point is 01:35:37 that cause it in the first place. And so that brings us up to 2021. Can we now take advantage of all this new biologic information about the role of mutations and T cells that target them about the ability to genetically modify cells and large numbers using retroviruses? Can we take advantage of that technology, that biologic information to develop more effective immunotherapies in the years to come? And that's what we're working on today.
Starting point is 01:36:05 That's what we're working on in the lab as we speak. Well, I wanna go back a little bit and talk about a few other things, both for posterity, I suppose, and also because I think there were some other things we've glossed over quickly. But in 1985, you had this opportunity to operate on the then president of the United States
Starting point is 01:36:22 who had developed colon cancer. Why is it that you were a part of the team that would take care of the president? Why is it that the chair of the National Cancer Institute would be involved in the president's care? Is that something that's sort of mandated at the federal level? No, it's part of the aberrancy involved in treating high government officials. It turns out that there is a set of modules in Walter Reed or Bethesda Naval Hospital that are set aside for the treatment of the president.
Starting point is 01:36:56 It's an isolated set of rooms. I learned this as it happened. I didn't know it ahead of time, where the kind of equipment and availability of technologies was available so the president could actually run the country from his hospital bed. But it turned out that it was the physicians in this particular case at Bethesda Naval Hospital and they have marvelous doctors there that would be treating the president. And it turned out there that would be treating the president and it turned out that the chief of surgery at Bethesda Naval Hospital had just rotated off an aircraft carrier to
Starting point is 01:37:31 be the chief of surgery at Bethesda Naval. It would change very commonly as officers in the Navy had different assignments and he happened to be an expert in vascular surgery, not oncology. And he was the one responsible for calling the shots about the patient's cancer. And it turned out he was an excellent doctor and excellent vascular surgeon, but not an oncologist. Never really was operating on cancer patients in any volumes. And so they needed an expert in oncology to take part. And just out of the blue on one
Starting point is 01:38:07 Friday evening, I got a call saying, well, you come over to Bethes and Naval Hospital, we have a patient we need your help with. And it turned out to be President Reagan. And it was simply because I was nearby, I had previously gotten a security clearance because I had tentatively been assigned to be part of a medical team that would take care of high government officials in the case of calamitous nuclear emergencies. So it was because the vascular surgeon was in charge and I was across the street that I got called and took part in that surgery. And if I recall that the press conference following the surgery, you explained point blank that
Starting point is 01:38:47 the president had colorectal cancer. And if I recall, Nancy Reagan wasn't too pleased about that, right? No, and I'm not telling stories out of school here about a patient because it was public information. And he later wrote a memoir that describes some of this. But Nancy Reagan, when we had to have a press conference and they were a little concerned about having a vascular surgeon hand with the questions and I had been in as an operating surgeon as part of the operating team.
Starting point is 01:39:19 She before I went on to hold a press conference, I remember standing backstage, and she said, you cannot use the word cancer in describing this because if foreign officials know, think the patient has cancer, the president has cancer, they won't pay any attention to him anymore, thinking he would not be around. And I said, I'm sorry, I can't do that.
Starting point is 01:39:43 If I have to go out, I have to just tell the truth. And it was Don Regan, who was the chief of staff, who finally talked her off that ledge and said, look, we've just got to let him do what he wants to do. So we went out and the surgeon who led the discussion just basically read off the path report. It was an ad in the carcinoma, a disproportionate of the colon and so on,
Starting point is 01:40:08 and nobody understood what he said. And so they asked me to explain it. So I said the president has cancer, which got me into all kinds of trouble. I later learned that when Vince Davida, who is the director of the NCI, resigned to become chief at Memorial Sloan Kettering, I was on a short list to become the director of the NCI, not a position I would have thought of accepting.
Starting point is 01:40:35 I was told at that point when I actually wrote this book in 1992 that my name got taken off the list as someone who would become the director of the NCI. I was very upset that I used a word has cancer and not had cancer. That is past tense. But we got over it and the president did very well and recovered and never recovered from his early colon cancer. You mentioned it in passing earlier that although you've been in the post year and now for 47 years, along the way you've had a few job offers that have tempted you. I'm sure you've
Starting point is 01:41:13 had many job offers. What are some of the other ones that tempted you at least where you thought you could even do better work or continue your work, because obviously you're so mission focused, it would be a special opportunity that I would imagine would get you to leave NCI, but what were some of those other opportunities that you even contemplated? There were only three that I looked at once. When was here at Georgetown, because of a relationship I had with the surgeon
Starting point is 01:41:42 in terms of collaborating things and it was sort of a favorite to him. To look at the job, I was also invited to look at the job as chief of surgery at Hopkins. And ultimately I was told, God, boil down to John Cameron and me and the shortlist, but I went back a second time but refused to go back a third time because I knew I would not go to Johns Hopkins and leave the NIH. I was also asked to look at the job at the Brigham. I'm already bread and a friend of mine, and I were again being pursued to accept that position. But again, I backed out.
Starting point is 01:42:19 I knew that I didn't want to take an administrative job. I wanted to be in the lab. I wanted to be mentoring fellows. I wanted to be trying to make progress. I didn't want to guide other people making it. I wanted to be there. I wanted to be doing it. I wanted to be guiding it because I thought I could do it well. So I actually refused any of those offers. I never looked at another job. I actually refused any of those offers. I never looked at another job and actually turned down opportunities to advance in the hierarchy here at the NCI because I wanted to remain at the level that I'm at. The control of resources that enabled me to pursue the kinds of research that I thought needed to be done in an environment of enormous resources.
Starting point is 01:43:06 So let's talk a little bit about checkpoint inhibitors. They've come up now a couple of times in this discussion. You've mentioned anti-CTLA for an anti-PD one. Certainly my time at the NCI, my second stint, I got me, got me very familiar with anti-CTLA for and I was an exciting time. And of course, James Allison would go on to receive the Nobel Prize a couple of years ago for his work in the discovery of this. Maybe go back and explain how that system works, how the removal of breaks works.
Starting point is 01:43:40 And of course, as part of the undercurrent of this, it only works if there's a tumor antigen to be recognized. In other words, taking the brakes off when there's no stimuli doesn't do anything. But how does that system work and how is it a two-edge sword? So again, there are stimulants and there are inhibitors, virtually every physiologic system that we have. And one of the inhibitors are molecules on the cell surface, on the surface of a lymphocyte that when engaged by a receptor will inhibit a lymphocyte from developing an immune reaction. And surprisingly, there are two molecules
Starting point is 01:44:25 that have been found on the cell surface. Now, many more that when targeted by an antibody, will not kill the cell, but actually turn off the brakes that are keeping that cell's activity from exhibiting itself. So it's releasing the brakes, and it turns out to have a very important function in the body because there are some cells that can react against normal tissues
Starting point is 01:44:53 that do not react because they're being inhibited by these brakes. And when you release those brakes, now the T-cell can be very active. And it turns out that cancer has manipulated those and by taking the breaks off, you can attack certain cancers and explains why melanoma is one of the
Starting point is 01:45:15 more common cancers to be attacked because it has so many antigens, so many mutations. And it was a startling discovery that simply attacking a molecule, single molecule on the cell surface could take the brakes off a lymphocyte and let it attack cancer. And when it comes to melanoma, kidney cancer, cancers that have large numbers of mutations
Starting point is 01:45:37 because they have mismatched repair gene mutations, Lynch syndrome. The MSI, the microsatellite and stable tumors, they can very strongly react against cancer, but the common epithelial cancers that result in 90% of deaths in patients have very little reactivity against the checkpoint modulator. So, although they can be life-saving and very likely, although it's been too soon, curable for some cancer patients, the overwhelming majority of cancer patients just do not respond to taking off the brakes, because when you take off the brakes, there's not a strong enough reaction to take advantage of. But hopefully, combinations of treatments, using checkpoint modulators will be more effective in the future,
Starting point is 01:46:25 but it was a major step forward and the beauty of it easy to apply because all that required was the injection of an antibody. So when you think about these amazing conceptual advances in the field, the ability of CAR T cells to recognize CD19 on B cells and eradicate any lymphoma originating from that lineage. The checkpoint modulators, NTCTLA4, NTPD1, and the durable effect that they can have on patients in whom you have enough mutagenic burden that relieving the checkpoint is enough to initiate it. Obviously, what you've alluded to earlier with IL-2, high dose IL-2, I don't want it at all sound disparaging, but just for the lack of, let's just call that the low hanging fruit of immunotherapy, which is, of course, completely ridiculous, given that that's 50 years of work
Starting point is 01:47:25 and countless lives. But for the sake of being cheeky, the low hanging fruit of immunotherapy are those pillars. Do you believe that the final frontier to go from where we are in 2021, until the point where all of those, let's just call the 550,000 patients with solid organ metastatic cancer, who have neoantigens or 80% of them have neoantigens that are unique to them, but not occurring in high enough frequencies that they will respond to a checkpoint inhibitor in isolation and or in combination with cytokines. Do you believe that there is a path for these people to be cured using adoptive cell therapy, either genetically or naturally occurring in some sort of a customized format? Do you think that that is the path forward from here?
Starting point is 01:48:20 My intuition is very strong that the answer to that is yes. For a variety of reasons One we know it can work from multiple tumor types and as I've mentioned we've We've described it and published treatment of liver tumors mild that cancer is breast cancer colon cancer cervical cancer We have responses in ovarian cancer that we've published. And so it's no longer a question of can it work in these other cancers. The answer is yes, it can work. And it's a world of difference. Like before I'll, too, we never knew that immunotherapy would work. But once
Starting point is 01:48:56 it did, we knew the immune system could do it. Now we know that antigens recognized by T-cells are present on 80% of the common cancers. And if you can develop unique reactivities, lymphocytes, select reactivities against them, and administer them, they can cause those regressions. And in fact, now, because we know the exact T-cell receptor sequences that we've cloned and isolated, it's almost an engineering problem because since we know the receptors, we've now isolated libraries of receptors against P53, K-RAS, that we can now use to genetically modify lymphocytes to turn a normal lymphocyte into a lymphocyte that can attack the cancer. And we have our first example of that now that we've
Starting point is 01:49:46 submitted for publication targeting P53 by genetically altering a lymphocyte by giving it a receptor that could recognize some of these driver antigens. So we know it can work. And tell you the truth, I finally feel like I have the hang of this kind of research. And that by sufficient work, creativity, this is going to be a problem that is solvable. We know the antigens of there, we know the t-cells of there, it should work. And it can work, and I believe it will work as the year has gone. work and it can work and I believe it will work as the year has gone on and it's 100% of what I'm working on today and that is how to utilize these unique mutational reactivities to cause the solid common epithelial cancers that result in 90% of all cancer deaths, how to get them to respond to immunotherapy. I mean of all the Eureka moments in your career of which you've had several,
Starting point is 01:50:48 this one seems to be the most promising, which of course, maybe they always seem that way when you're glowing in them. But do you see it that way that every one of these milestones that came before this was vital, but this has the greatest potential, this recognition that virtually every solid tumor out there has novel peptides that can be recognized by a patient's own immune system. Right. Now, realize how current this is. We published a lot of these individual cases that can respond. We published the first 40 or so colorectal cancer showing that mutations were all unique.
Starting point is 01:51:29 But we haven't published much of what I've told you. So for example, none of the breast cancer work has been published. We now have looked at these 195 individual cancers and found, regardless of histology, they're there. So it's very recent. This realization that mutations are the antigens and now that T cells can recognize these mutations. It's really a new world. But that happens every time you make an advance, you find the immune
Starting point is 01:51:59 system can be stimulated. Okay, well, let's get to work. I'm figuring out how, you know, cells can do it. Let's figure out how and science works that way by incremental advances. We know this can work and I have every confidence that Scientists around the world will figure out ways to make it work That kind of reminds me of some of the important lessons that those of us who've been privileged enough to work alongside you have learned along the way. You never sort of pounded the table to make these lessons, but it was it was abundantly clear. And one of them
Starting point is 01:52:35 was no secrecy. There was never any secrecy. I remember working on my experiments and before data were published, people from other labs. I don't mean other labs at NIH. I mean other labs in different parts of the country. You would encourage me to reach out to them and share my results with them, even running the risk that they would quote unquote scoop me. But none of that mattered to you. Your goal was, what is the fastest path to the accumulation of collective knowledge in the field? Am I accurately representing that? Is that the, I don't think I'm overstating that? No, I've always been horrified by the secrecy that exists in medicine.
Starting point is 01:53:17 And it's an ongoing problem. The need for biotech companies, pharmacologic pharmaceutical companies to protect their intellectual property, given current patent laws prevents them, will often prevent them from sharing information from sharing reagents, and this is holding back progress. If we could somehow overcome this secrecy that results from either people's own personal jealousies about wanting to be the one who does it, or intellectual property that companies have to protect to preserve themselves and raise funds to continue to do their work, we need kinds of regulations that will bring lawyers and doctors together to figure
Starting point is 01:54:05 out ways to prevent that kind of secrecy from being a part of modern science. It's not like we're trying to create a better air conditioner, we're trying to save the life of another human being. And I think when you take care of cancer patients, it puts a lot of things in perspective. And I think when you take care of cancer patients, it puts a lot of things in perspective. And the idea of having a policy or a rule that you live by that inhibits your ability to help people who could potentially be helped is that parent to me. I wrote as you know a perspective in New England Journal of Medicine trying to change things, but they haven't changed. Every bit is common today as they wear back then. As you know, the first thing a fellow here is in my lab when they start the first day is look, anything you know, you share. Any experimental result you have, you can tell somebody, any experiment you plan to do
Starting point is 01:55:00 tomorrow, you can tell somebody about. Our goal is to help people that are involved in the suffering of cancer, and there's no excuse for not doing everything you can to try to help, and that means sharing what you know. On the other side of your office, right, if one side of your office is the lab, the other side of your office is the clinic, The other lesson I think that you've infused into the literally hundreds, you might even be it into the thousands now of people who have come through and trained with you. Basically, this idea that one never retreats from the bedside, one of the things that struck me actually, especially in medical school when I was there,
Starting point is 01:55:42 because I spent the entire time on the clinic, I lived in the clinical ward, he may recall, I had rented an apartment in Bethesda, which I never went to except on Sundays to get new clothes, but I slept in. I slept the day. Yes. You're quite a legend at the NIH,
Starting point is 01:55:59 with respect to that incidentally. I mean, people thought I never left, but you never left. So it was really quite an experience to see you in operation there. But the thing that was hard to believe and hard to process until you experienced it was nine out of ten people that walked in the door died. You know, eight or nine out of 10 of the people who walk because I think most people don't maybe understand because it's implicit, but it should be stated, the patients who are coming to NIH have progressed through every standard therapy. People aren't
Starting point is 01:56:37 coming here for whom there are standard options elsewhere. By definition, these are patients who have the most advanced, the most aggressive, the most recalcitrant cancers imaginable. These are people who would probably be expected to live no more than six months without a miracle. And those, they come to the NCI for those hail marries. And if 20% of them are saved, that's remarkable, but it means 80% of them don't. And I think what I remember most was the way in which you described taking care of those 80% and fighting the urge to retreat from them because of the failure we saw in ourselves. How did you develop that? I mean, I assume it came naturally to you, but how deliberate has it been in how you teach those of us that came through?
Starting point is 01:57:36 You know, I have enormous respect for practicing non-collegists who face this every day in their practice. And it's difficult if you point out, especially when you get treatments that not only don't work, but actually cause some harm, which has happened in the course of the development of these treatments as we try to figure out exactly what we were doing in the right ways and the right ways to do it. But I always had the feeling that I was working and working hard to try to improve the situation, to try to somehow repair this Holocaust. And that kept me going. I don't know what it must be like to be taken care of cancer patients knowing
Starting point is 01:58:26 you have limited tools at your disposal that are not good enough. And yet that's all you have. And that's what you do day in and day out. I mean, that, that must be even more trying. And so I think one of the things that keeps me going at least is the fact that I'm doing everything I possibly can within reason to improve the situation. Without that I think it would be much, much more difficult. Yeah, there are so many patients that I still remember from more than 20 years ago, who were those ones that didn't make it. And I can't imagine how haunting it is for you sometimes because I can see their faces,
Starting point is 01:59:15 I remember their names, I remember their voices, and I remember their stories. You know, the newlywed girl who came to clinic one day,. I mean, she had literally been married for maybe three months, a beautiful 25-year-old woman with metastatic melanoma, and she was not one of the survivors. A young man whose name and face, I remember every detail of single guy, metastatic melanoma, and what was most tragic in his case was, I remember everybody kind of abandoned him at the end of his life
Starting point is 01:59:46 and I've said this before I I feel like cancer takes families that are close and brings them closer and takes families that are fractured and fractures them more and I got the sense that his was fractured to begin with and He was maybe 26 years old So I certainly understand the motivation. There's no lack of motivation for how you do what you old. So, I certainly understand the motivation. There's no lack of motivation for how you do what you do. I guess I'm amazed that you talked about how the immune system has the stimulatory and the inhibitory components. Well, it's similar that taking care of patients like this, there's the motivation that comes
Starting point is 02:00:22 from it to do more, but at some level there's the depression of the death toll. I'm not sure everybody could do that. You seem to have found that balance. Well, you know, when I lie awake at night, it's not the successes that I think about. It's the tragedies, patients that you're remembering even now that I'm sure are impacting on you and gets even worse because there are patients that we've killed by doing the wrong thing to them, not understanding some of the underlying biology. That's the hardest thing to deal with. But again, given the fact that I'm doing
Starting point is 02:01:11 everything that I can reasonably do to help them, it certainly uses the burdensome. And if you know being a doctor, what an unbelievable privilege it is to have the opportunity to help people like that, given the skills that you've developed. One of the first lines of the prayer of my monodies goes, you have been given the wisdom to alleviate the suffering of your brothers. And that's true in medicine because you know, we spend a lot of time just learning how to help people. It's a unique opportunity in the world and in life in general. So there are the satisfaction that you're trying hard, even if most often, things don't
Starting point is 02:02:02 work out. But they're clearly asleepless nights involved in all of that process. The good news is you have wonderful longevity in your family. So despite being 81, I have every confidence that you're gonna be doing this for many more years. I know you don't like golf enough
Starting point is 02:02:20 to hang up what you're doing for the golf course. You know, I found, I pulled this off the wall today. I wasn't sure if you still recognize these guys here. This is us from, I think, I think this is 16 years ago. We both look quite a bit younger, and this is the only picture of me in my office is this picture. And it speaks to what an influence you've been in my life. I think the list of people who have had a greater impact on the course of my life than you is somewhere between zero and epsilon. It's a decidedly small list. So I feel forever in your debt. And though I have not been able to follow in your footsteps,
Starting point is 02:03:07 your impact is greater than you could ever recognize. Well, thank you for saying that. That means a lot to me knowing that someone of your incredible intellect and perseverance feels that way. So thank you. I know again what a sacrifice it was for you to take time today to speak. And I know that every minute you spent talking with me was literally a moment you were not working on this problem. So I'm beyond grateful and I know that the people watching this or listening to this are equally grateful. So thank you so much Dr. Rosenberg for everything. Well, you're very well. Thank you for listening to this week's episode of The Drive.
Starting point is 02:03:50 If you're interested in diving deeper into any topics we discuss, we've created a membership program that allows us to bring you more in-depth exclusive content without relying on paid ads. It's our goal to ensure members get back much more than the price of the subscription. After that end, membership benefits include a bunch of things. One, totally kick ass comprehensive podcast show notes that detail every topic paper, person, thing we discuss on each episode.
Starting point is 02:04:15 The word on the street is, nobody's show notes rival these. Monthly AMA episodes are asking me anything episodes, hearing these episodes completely. Access to our private podcast feed that allows you to hear everything without having to listen to spills like this. The qualities which are a super short podcast that we release every Tuesday through Friday, highlighting the best questions topics and tactics discussed on previous episodes of the drive. This is a great way to catch up on previous episodes without having to go back and necessarily listen to everyone. Steep discounts on products that I
Starting point is 02:04:49 believe in but for which I'm not getting paid to endorse. And a whole bunch of other benefits that we continue to trickle in as time goes on. If you want to learn more and access these member-only benefits, you can head over to peteratiamd.com forward slash subscribe. You can find me on Twitter, Instagram, Facebook, all with the ID, Peter, a T.A.M.D. You can also leave us a review on Apple Podcasts or whatever podcast player you listen on. This podcast is for general informational purposes only. It does not constitute the practice of medicine, nursing, or other professional healthcare services, including the giving of medical advice.
Starting point is 02:05:25 No doctor-patient relationship is formed. The use of this information and the materials linked to this podcast is at the user's own risk. The content on this podcast is not intended to be a substitute for professional medical advice, diagnosis, or treatment. Users should not disregard or delay in obtaining medical advice from any medical condition they have, and they should seek the assistance of their healthcare professionals for any such conditions. Finally, I take conflicts of interest very seriously. For all of my
Starting point is 02:05:56 disclosures in the companies I invest in or advise, please visit peteratiamd.com forward slash about where I keep an up-to-date and active list of such companies. you

There aren't comments yet for this episode. Click on any sentence in the transcript to leave a comment.