Episode 42: Diana Azzam, PhD, Assistant Professor at Florida International University, Co-Founder at First Ascent Biomedical

Episode 42 July 16, 2024 00:36:14
Episode 42: Diana Azzam, PhD, Assistant Professor at Florida International University, Co-Founder at First Ascent Biomedical
Few & Far Between
Episode 42: Diana Azzam, PhD, Assistant Professor at Florida International University, Co-Founder at First Ascent Biomedical

Jul 16 2024 | 00:36:14

/

Show Notes

“Our studies are open for those that have exhausted standard of care, and we're showing improved responses, and the data is promising. So, I really hope that as we scale up and generate more data, we can actually move to newly diagnosed patients.” - Diana Azzam, PhD, Assistant Professor at Florida International University, Co-Founder at First Ascent Biomedical

Welcome to the latest episode of Biorasi's Few & Far Between podcast. Join host Chris O'Brien and guest, Diana Azzam, as they discuss technological innovations in guiding individualized patient treatments.

Tune in today! 

View Full Transcript

Episode Transcript

[00:00:14] Speaker A: Welcome to the latest episode of the few and far between podcast. I'm your host, Chris O'Brien. The philosopher Confucius is quoted as saying, choose a job you love and you will never have to work a day in your life. My next guest very much believes in that mantra to love what you do. And and it has truly illuminated her career path with breakthroughs in individualized treatments for cancer patients, glioblastoma biomarkers, and more. Doctor Diana Azam is assistant professor at Florida International University and co founder of First Ascent Biomedical, a biotech research company providing new tools in the fight against cancer. You may have seen Doctor Azam in the news a lot this past April, sharing her success story in functional precision medicine and genomic profiling for for pediatric oncology patients at Niklaus Children's Hospital in Miami. Doctor Azam has graciously joined us on the podcast today and gives us a chance to delve into the details of her work, explaining her methodology in guiding individualized patient treatments from early chemosensitivity assays to AI powered technology innovations. We'll talk about what drives her mission to transform her functional precision medicine research into the future standard of care for oncology testing. And we'll find out what favorite lebanese dish gives her the energy to move forward in clinical research. It was a complete delight to have Doctor Azam on the podcast, and I hope you'll enjoy this new episode. It's a fun one. Okay, let's start the podcast. Doctor Diana Azam, welcome to few in Far between. [00:01:46] Speaker B: Thank you for having me, Chris. It's a pleasure to be here today. [00:01:49] Speaker A: All right, so let's start out with talking a little bit about your background. A little girl in Lebanon, you did not necessarily have a plan that you were going to be at FIU and in Miami and all this stuff. So tell us a little bit about how you got here. [00:02:03] Speaker B: Yeah, I look back at how I got into science. I really remember the day I got into a lab for the first time during high school. It's the first time they get you into this lab. I remember it was all white, and I was like, oh, my God, this is where I can make discoveries. So really early on in high school, but that was in Dubai. So I am Lebanese originally, but I went to school in Dubai. But then when I discovered that I wanted to major in science, I went to Lebanon and I majored in chemistry. And then I also did a graduate degree in biochemistry. And then I was like, okay, I'm just loving this, and I really want to continue my doctoral degree. So that's when I came to the United States and started my PhD in biochemistry and molecular biology. [00:02:43] Speaker A: That's fascinating. Okay, so tell us. I think often people would think. Americans would think that when people leave Lebanon, they tend not to go back. So you left with your family or grew up in Dubai and then came back for university. So what brought you back to Lebanon? [00:02:56] Speaker B: I wanted to be in Lebanon because I actually always wanted to get into the American University of Beirut. They're known to have really strong programs and biomedical programs, and so I wanted to get a good education there, and then because I realized that a career in research is going to be in the United States. So I knew that having my training and getting a degree from the American University of Beirut will give me that opportunity to come to the United States. And I was right. So I came to Miami. I started my PhD, and really, that has just changed my whole career. Right. I got into cancer research early on. During my doctoral studies, I started looking at, you know, the most therapy resistant cells within cancers. They're called cancer stem cells, and started looking at how we could potentially target these therapy resistant cells. And that's what just got me excited to even pick my postdoctoral training. So it's really. It's all about following, you know, your passion and you're curious about, okay, what field in science do I want to enter and how do I learn more about it and how can I contribute? So I realized that I had a lot of experience in studying these, really, therapy resistant cells within cancers, and we put chemo radiation on these cells, and nothing kills them. So I thought to myself, like, how can I find drugs to target these cells? So I started looking for a postdoctoral training in drug discovery labs because I realized, okay, maybe if I start learning on how to do drug discovery and high throughput drug testing, I'll be able to identify novel drugs that would target these therapy resistant cells. Luckily, then, at the University of Miami, there was a group from Scripps, Florida, that just joined, and all they do is drug discovery. So I was like, that's where I'm gonna be. And they didn't work on cancer. They were working on other diseases. And I started my training there, learned all about, you know, high throughput drug testing and drug discovery. And that's when I also realized that there's so many state of the art technologies available and there's a lot of discoveries available and that we make as scientists in the lab, but very few of them are actually available for cancer patients. [00:04:59] Speaker A: And, yeah, I love that. [00:05:00] Speaker B: Yeah, I was like, shocked by that, and I realized, okay, we need to get. Let's take a step back. And before I start identifying drugs for the most therapy resistant cells, let me start looking into how to provide this technology just to bring it a little closer to the clinic or a little closer to cancer patients. And that's how, when I started my own lab at FIU, that was the first thing I wanted to do, is how can we bring these technologies to patients? And that's when I started this clinical trial on implementing functional precision medicine approaches to cancer patients. [00:05:33] Speaker A: Fantastic. Okay, so let's. Let's define a term now. So, functional precision medicine, will you tell us what that means and how it's different from precision medicine as it's sort of more generally applied? [00:05:42] Speaker B: Yes, precision medicine today, it's a common word. Everybody's familiar with this. It really. The idea behind precision medicine is identifying the right drug for the right patient at the right time. But what we use today, and commonly used approaches today, is looking at the DNA of the tumor and trying to look for, like, mutations or changes in the DNA that will enable us to identify this targeted drug that will work for the patient. And that's what we call genomics precision medicine. So, really looking into mutations in the DNA and identifying targeted drugs, and that has helped a lot of patients. So there's a lot of benefit when we identify that mutation and find the right drug for that patient. But what happens when we don't find a mutation in the DNA, or when we find so many different changes? And that's what functional precision medicine brings in. So there's this additional component to genomics, precision medicine, where we add a drug testing assay, or a drug sensitivity assay, and basically we test hundreds of FDA approved drugs directly on the patient's own living cancer cells, and then we identify what works and what doesn't for each patient. [00:06:46] Speaker A: So this is an amazing, an amazing way of thinking about this. Like most really compelling ideas, it feels like it's, in hindsight, like, how are we not doing this years and years ago? But, so if I summarize and play back what you said, if there's a monogenic solution, if there's a single gene that we think we can target, great, then that is traditional, if I can say that in quotes, precision medicine. But the model that you've pioneered is this idea of testing a huge battery of FDA approved drugs against a cancer to see what is the most effective. So we almost, we don't predetermine what the outcome is going to be it's just very driven by the results. Is that right? [00:07:21] Speaker B: Absolutely. And it's the integration of both that genomics with that functional drug, drug testing component, basically is what functional precision medicine is about. You mentioned something very important I want to touch a little bit on that is, you know, this is a simple idea, right? I mean, adding drugs on tumor cells is, it's a very simple idea that everybody's thought about maybe 20 years ago, and they were actually called chemosensitivity assays 20 years ago, but those were very primitive then. And what happened is, because I just mentioned to you, there's huge advancements in technologies that enable us to test and dispense drugs in very small volumes, like in minutes, and also the way we grow tumor cells in the lab. We've done so much progress in those two different, in that area that today our assays are not the same as when they were 20 years ago. [00:08:13] Speaker A: Fascinating. [00:08:14] Speaker B: And we will talk about this later, because a lot of, you know, there's some skepticism when people, you know, our doctors hear about these assets because in their mind, you know, we knew about this and these never work. But we want to tell them that you need to look at the data today and look at the technologies we have today before you make that, before. [00:08:32] Speaker A: You make that conclusion, before you dismiss this. Yeah, yeah, I totally agree. So. And, Diana, is it about scale? Is it the ability to get a lot of different approved drugs tested against this tumor, this cancer? Is that sort of the magic difference from the past? [00:08:46] Speaker B: Yes, it's, first of all, we've had so many drugs approved by the FDA in the last two decades. We didn't have that number of drugs that we have today. So that obviously gave a better opportunity for us to test hundreds of drugs. I mean, we have at least 220 drugs approved by the FDA for oncology. [00:09:04] Speaker A: Amazing. [00:09:04] Speaker B: And then you have over a thousand drugs approved by the FDA for other diseases. And there's so much evidence that many drugs that are, that are non cancer drugs also may have potential repurposing in cancer. So, yes, it's really the idea of we have all these drugs available, and we have the technology to test these drugs, so we can always provide options for patients that have exhausted standard of care. [00:09:30] Speaker A: Incredible. There was an article recently about a patient in the Miami Herald. It was a story of this first trial. I think it was your first trial. You're deploying this, is that right? [00:09:39] Speaker B: Yes. [00:09:39] Speaker A: Can you tell that story? [00:09:40] Speaker B: Yeah, obviously, for me, that story is, is very unique, because as a researcher. Right. You don't get to meet the participants in your study because the samples come in de identified you, of course. [00:09:53] Speaker A: Yeah. [00:09:53] Speaker B: Especially in the study that I had ongoing with Nicholas Children's Hospital. But by chance, I was presenting at one of the local live, like Bella symposiums about my work. And then after I presented, you know, my data, a beautiful woman who was, you know, was attending the conference because also this symposium invited, you know, patients and their families, which is also very unique. So we had researchers, doctors, patients and their families in that conference. So after I presented, she came up to me and she told me, doctor Azzam, I think my son got into your study and he's doing very well. And I asked her, so tell me about your son. Like, the diagnosis, what happened? And she started talking about Logan. And I was like, oh, my God, he's patient number 13. And why did I know? And I'll tell you that. That's what brings me to the story and why Logan was particular. So Logan had relapsed leukemia and he had gone through a bone marrow transplant already. He did chemotherapy and gone through a bone marrow transplant, which is the way you treat leukemias, but he relapsed, the cancer came back. And when he got into my study, because my study is only for relapse refractory cancer patients, we got his bone marrow sample. We tested hundreds of FDA approved drugs. And what we found with Logan is that two drugs were as effective as three drugs. So, you know, the doctor wanted to give him three drugs. And then when we did the testing, we found that we could get rid of one of the drugs that is actually toxic to the heart. [00:11:17] Speaker A: Wow. [00:11:17] Speaker B: And when we did that, he achieved remission, like complete response in 30 days. [00:11:22] Speaker A: Incredible. [00:11:23] Speaker B: Faster than 150 days in his previous regimen. So we got rid of one drug. We killed his leukemia cells within a month, way faster than his previous regimen, and then he underwent a second bone marrow transplant. Wait, there's one more thing that was very interesting about Logan's cells, is that they doubled with steroids. We add steroids in our library because they're also used to treat cancers in many different indicators. But somehow his cells, his particular cells just doubled in number with steroids. Within a few days, you can see the curve. Instead of killing the cells, steroids are usually given to suppress the immune system, especially when they want to give him another bone marrow transplant. So that information was very important because if the steroids are doubling his cancer cells and we're adding a chemotherapy, it's just counterproductive. So they withdrew the steroids we gave him two drugs, and then he achieved complete response, and he got a second bone marrow transplant. And, you know, Logan today has been cancer free for over two years. I mean, double the previous regimen. And that's huge, Chris, because, you know, when cancer returns, it's more difficult to treat. [00:12:29] Speaker A: Yes. [00:12:29] Speaker B: That's why when you're seeing these responses, you know, it's very promising. [00:12:33] Speaker A: Diana, why? This just brings me to a slightly different but related question. Why do you focus on refractory and recurring cancers? Why is that the area of focus? [00:12:42] Speaker B: That's a very good question. Because if you tell me today, can, when we start guiding individualized treatments at diagnosis, I would be like, I wish. That's really my goal. I would really want to see this being utilized in newly diagnosed patients because, you know, as cancer comes back, we mentioned, you know, it becomes more difficult, and then the patient goes through a lot of, you know, toxic. There's a lot of toxic effects of trial and error of some. Some drugs that sometimes are unnecessary and don't work. But we do. We can't do this right now because we can't shift from standard of care. I mean, physicians are always going to first treat using standard protocols. Right. So that's why our studies are open for those that have exhausted standard of care, and we're showing, you know, improved responses, and the data is promising. So I really hope that as we scale up and generate more data, we can actually move to newly diagnosed patients. [00:13:33] Speaker A: Got it. So your vision for the future would be that this has become standard of care, that we would do this kind of a test. We would try a bunch of. We throw the kitchen sink at a tumor and then see what is most effective and not going to the patient until the oncologist is armed with that information. Is that how you think about it? Exactly, yeah. [00:13:51] Speaker B: Yeah, I think about it. Especially in cancers that are rare as well. Many of these cancers don't even have the approved options. And, you know, like osteosarcomas. I mean, osteosarcomas, they're brutal, and they, you know, it always, it's like one line of treatment, and then they amputate, and it's, you know, those are the types of cancers. I believe we can potentially start implementing functional precision medicine early on, and hopefully that will really improve outcomes for these types of cancers. [00:14:17] Speaker A: What has been the reaction from the oncology community to the initial results? [00:14:22] Speaker B: Chris, I've had a lot of oncologists encouraged by the results, especially that we got it to be published in Nature Medicine, and we were on the COVID And the coverage that came out and all the press that came out with the study is very encouraging. But we did mention in our study that there is physician hesitancy in using these tools and technologies to better, you know, inform their decisions or, you know, for their treatment decisions for their patients. And I'm hoping that, you know, as we continue to generate data and as we continue to publish in the field of functional precision medicine, I'm hoping that more and more physicians and oncologists will start adopting these approaches. I I just want to always, like, I want to make sure that. And that's what I say when every time I speak with an oncologist or clinician, and I say, what you known about these chemosensitivity assays 20 years ago is completely different than the essays we have today. So I encourage you to look at the data, reevaluate all the clinical data that's coming out, find a functional precision medicine researcher, collaborate with that researcher, and start using these tools, because this is more data for you to make an informed decision. You don't have to follow the data, but you will see with time that this data is very helpful for you, especially when you are in a situation where you want to select the next line of treatment and. And you're out of options. [00:15:40] Speaker A: Diana, if an oncologist were listening and they wanted to explore this, would you say this is more of a process change or more of a technology change? In other words, do they have access to the sophisticated tools that are needed to run 180 drugs against a blood sample? [00:15:53] Speaker B: So it's a process and technology change, and not everybody has access to it. So obviously, you'll see that. I mean, in my lab, I received their expensive equipment. I received $2 million from the state of Florida to buy the robotic instrumentation that are, you know, like I mentioned, dispense drugs through sound, and it can do this quickly. It's incredible. They are. They're expensive, resource intensive, but there are, you know, multiple labs now within academic institutions or within, you know, startups that are. That have this infrastructure and just look for them and start collaborating with them to be able to take advantage of this type of technology. And I'm hoping, Chris, that as we continue right now, the equipment is expensive, but it enables us to reduce the cost of the task. [00:16:37] Speaker A: Yeah. [00:16:37] Speaker B: So, remember genomics? 510 years ago, sequencing the DNA was expensive, but now it's cheap. So I'm hoping that, you know, with time and five years, the cost of drug testing, hundreds and thousands of drugs is not going to be. It's going to be cheaper than how much it costs today. [00:16:53] Speaker A: Fascinating. So if you sort of look forward with your crystal ball, like, what do you think? And is it five years away? Is it ten years away? What would you guess in terms of this being a five years? Yeah, that's great. [00:17:04] Speaker B: I. Yeah, I'm on the board of the Society for Functional Precision Medicine. It's a relatively new society, but the mission of that society is to bring these functional assays into the clinic. And, you know, we have different assays and different approaches, but very similar in concept. Right. Testing drugs on tumor cells, whether they're organoids or 2d or, I mean, you have different microchips. There's so many different methodologies now being developed. But I really believe that based on my experience on the board, we've seen a huge interest in scientists and oncologists in this field in particular, where we have more members. Now we're working on bringing in different entities. We're talking to pharma, we're talking to patient advocates. And I do believe that in five years, it's going to be an exponential growth in the field and hopefully more exponential implementation of this approach. [00:17:52] Speaker A: That's incredibly exciting. Incredibly exciting. Hi, this is Chris O'Brien, host of few and far between. We'll be right back with this episode in a moment. I personally want to thank you for listening to our podcast. Now in our fourth season, it continues to be an amazing opportunity to speak with some of the top thought leaders in the clinical trials industry. If you're enjoying this episode, please leave us a review on Apple Podcasts. It really helps people discover the podcast. And don't forget to subscribe to few and far between so that you never miss an episode. One last request. Know someone with a great story. You'd like to hear me interview? Reach out to us at few and far [email protected]. dot thank you. And now back to the podcast. Okay, I want to switch gears slightly. You've also done some work in biomarkers for brain cancer, glioblastoma. Tell us a little bit about that. [00:18:45] Speaker B: Yeah, that's a very interesting and exciting project. In my lab especially. I mean, as you know, glioblastomas are like the most aggressive types of brain tumors. And unfortunately, glioblastoma patients have a very, very low prognosis. Like, there's less than 5% survival. Less than 5% of these patients survive over a year. So I started working on glioblastomas because my lab is close to Dean Tomas Galarte, who is not a cancer researcher, but he's a neurotoxicologist, and he worked on this biomarker of neuroinflammation for so many years. And one day he comes up to me, our labs are next to each other. We have open lab space, which tells you how important it is to collaborate with different people. And he mentioned translocator protein tspo, which is a biomarker of neuroinflammation. And he said, you know, I believe this may be highly expressed in glioblastomas. Why don't you look at it? Yeah. Fast forward a few years. We started looking at this neuroinflammation protein in hundreds of glioblastoma patients, and what we found, and we collaborated with Cleveland Clinic, Ohio, because they have a huge database of glioblastoma patients. What we found is that a change in this protein or a polymorphism is associated with worse outcomes in male glioblastoma patients or in male than and not in women. And we know glioblastoma is higher in men than in women. So now we have this new protein or a biomarker that, you know, is also correlated with sex differences in glioblastoma. And we're right now it's really exciting because we want to understand, like, why, what is the function? Like, if it, it goes well with the fact that there's sex differences in glioblastoma. So we're understanding the function of this polymorphism and what it does and hopefully be able to use it as a prognostic biomarker not only to understand, like, we can use as a prognostic biomarker not only to predict treatment, but also to identify the right treatment for these patients. [00:20:40] Speaker A: Fascinating. And so where are you in the stage of that exploration? [00:20:43] Speaker B: We've generated very interesting data. Now in terms of its function, we're starting to show, well, what we found is that when we knock it down, take it away from a glioblastoma stem cell, which, again I mentioned, these are the most resistant cells within tumors. We reduce the cell's ability to migrate, invade, and be a stem cell. And so it's definitely important for the survival of these glioblastoma stem cells, and then it makes these cells more susceptible to chemotherapy. So we applied for an NCI grant, and hopefully in a week, it will be reviewed, and hopefully we'll get funding from the NCI to continue to understand and the function of the polymorphism and how TSPO could also be playing a role in the progression of glioblastomas. [00:21:26] Speaker A: Fascinating. Okay, so that's exciting. Exciting project number two. And I think you've also got some interesting work in adult sarcoma with the Cleveland clinic of Florida. Is that right? [00:21:35] Speaker B: Yes. So I just like you. We talked about Diana. [00:21:38] Speaker A: Do you sleep or do you just do? Sleeping. Not a thing for you. [00:21:42] Speaker B: I love what I do, Chris. You love what you do? I love being in the lab. It's been an incredibly busy, busy, busy few years to set up all these projects. But that's why I tell my students, you have to love what you do. And, you know, many of these students come in, they want to rotate in the lab because they don't know if they want to get into medical school or they want to be a scientist. And I said, you know what? It doesn't matter what you decide to do, but I will give you two, three months for you to really know what it's like to come into the lab every day and do basic science research. And if you come back after three months and say, doctor Azzam, thank you so much for the opportunity, but I really want to be a doctor. You really make me so excited because you know what it's like to be a researcher and you picked to be a physician and work with physicians, so you have to love what you do. [00:22:29] Speaker A: I love that. I want to just sort of emphasize that that's, I think it's wonderful advice to young people. Don't commit to graduate school or a major career choice without having tested it out. And so if you can test it out with a three month experience in your lab or in somebody else's lab, that's so much better than making these decisions kind of in a void because they are not entirely reversible. I mean, you can recover from lots of things, but if you've committed to graduate school and you've taken on debt and you've done these things, it does make that more challenging if you decide that's not the right path for you. Right? [00:22:57] Speaker B: Absolutely. And all our labs are open for students who are interested in exactly that because, you know, even students that love research, they end up deciding they want to be a physician scientist. And that's even better for me. Right. Like you're. You want to be a physician and you're going to be your best. You're going to be the best doctor if you continue to be involved in research. So, absolutely, I encourage students to do this and take that step early on in their careers before making any commitments. [00:23:24] Speaker A: Amazing. [00:23:24] Speaker B: Yeah. So going back to your question about the study with Cleveland Clinic, the study that I have that I started, that we just published on childhood cancers was with Nicholas Children's Hospital. And it was really the question we wanted to answer. Like, the main question we wanted to answer was, is drug testing feasible? Can we generate results quickly that will inform the next line of treatment? Really? That was our first question, and that was the main goal. So we show, yes, it is feasible. We can use drug sensitivity testing results to guide treatments. And when we compared patients that were guided versus not, we found improved outcomes. 83% of patients showed improvement. We have to show the same thing in adult cancers. And that's the type of study that I have open with Cleveland clinic, Florida. We started enrolling patients in November, and it's open to relapse, refractory adult cancers, all types of cancers. And the idea of can we use drug sensitive detesting and genomic profiling to inform the next line of treatment and provide individualized treatment approaches for or options for patients? [00:24:29] Speaker A: And I want to just underline the point there that you said to all kinds of cancers, it's not cancer type specific. [00:24:35] Speaker B: We started with sarcomas, but now we opened it and expanded to all types of cancers. And I'm going to tell you this, Chris, we are starting to do drug testing from fine needle biopsies and core biopsies from patients. [00:24:49] Speaker A: Wow. [00:24:49] Speaker B: That is so important to increase accessibility of this type of approach because, because of the technologies, right. We can dispense nanoliters of drugs on cells. We don't need a lot of nanometers. So instead of us getting only an excise tumor after surgery, we can start off with a fine needle biopsy. We won't test 150 drugs, but we can still test 50 drugs and 50 single end in combination. That is very important in broadening accessibility of functional precision medicine approaches. I'm excited about that. [00:25:21] Speaker A: So what's the status on that trial? Is it fully enrolled? How are you coming? [00:25:25] Speaker B: No, we're only. We just started in November and we have two more years to go. So, you know, if anybody wants any information on this, on this trial, please feel free to reach out to me. And it's open for all types of cancers, but relapse refractory cancers. [00:25:39] Speaker A: Okay, so are there any other fascinating, huge initiatives that you have underway in your lab right now? We've talked about three extraordinary things. [00:25:47] Speaker B: One important, I would say a milestone that I'm trying to achieve right now is getting CLIA certification for this drug sensitivity testing. So what does that mean? It's basically moving it from a research task to a diagnostic test. So we're trying to make this test accessible outside of a research or a clinical trial. And that is, that requires. So basically, it requires us to show that our test is accurate, reproducible. I mean, all the measures of quality, all the quality measures that are needed. That's a huge milestone for us, because once we're cLIA certified, then we can start providing this test outside of a research study. [00:26:27] Speaker A: That's fascinating. And so the process on that, if that happens, does that mean that those tests are reimbursable through insurance companies? [00:26:34] Speaker B: No, Chris. No, I wish. Well, that's so one step at a time. So we are working. There's a lot that we need to do to get this test covered right by shorts. And that's why I started a company. I'm a co founder of this company with other two co founders. And really, our main goal is to try to get the data that we need to show insurance companies that not only we can improve outcomes, but also, hopefully, we can save money, because we are saving, we're getting rid of the trial error that sometimes doctors have to do. And so all these health economic studies are studies that are ongoing right now as part of the company, as a part of go to market. And hopefully those will help us get one of the insurance companies to cover this test so it can be accessible to patients. So we have a long way to go, but we're trying to, you know, overcome one, one barrier after the other. [00:27:24] Speaker A: Well, you're certainly making a lot of progress. It sounds like terrific progress on the actual science. And now there's a lot of work that is education, that is persuasion, and that is evangelizing for this. Is that kind of how you think about this path, or is there still a lot of hard science to be done on this as well? [00:27:40] Speaker B: The science is always going to be done because, you know, you know how we are as scientists. We want to make it faster. Like now, we give back results in a week. I want to give back results in two days. So we're always going to continue to optimize this test. We want to be able to test immuno oncology drugs. Right now, we're testing everything immuno oncology, but we're starting to include immuno oncology drugs because our patient derived cultures, we found that they have 10% of immune cells within the cultures. So now we're starting to optimize those tests to include immuno oncology. So there's always going to be more science done. But on the clinical side, on the clinical efficacy side, I believe the data is promising. I believe that we're going to continue to show, hopefully we're going to continue to show that this approach improves outcomes. Hopefully we'll move to newly diagnosed. So that's all great. I want to expand the clinical program as much as I can, but you want to bring this to market. It's a totally, totally different journey. And I believe that when it becomes accessible to patients and we start showing, you know, we start all these health economic studies, we integrate it with AI, hopefully, you know, that will bring it to market easier or faster. Hopefully. [00:28:48] Speaker A: That's fantastic. Okay. Sort of a, I guess a big picture question. Obviously, new drugs for a new treatment in oncology. Huge area of focus. Big area of focus for us at biorosity and for the industry as a whole. When you think about your research, if we sort of fast forward five years, a hunch, an opinion, I hate to ask a scientist for a hunch, but do you have a hunch or an opinion about how much of the improvements we're going to see are going to be from using already approved drugs more broadly, and how much of it do you think might come from new drugs, or maybe a better way to say it is? How much gain do you think there is to be had by testing lots and lots of drugs against individual cancers and creating these personalized plans? [00:29:27] Speaker B: The answer would be both. I think that based on what I've seen, there is use of many existing drugs in different cancer types. I mean, you know, if an osteosarcoma patient is responding to idirubicin and an allergy medication, I mean, that's incredible, right? [00:29:40] Speaker A: Yeah. [00:29:41] Speaker B: We're repurposing these drugs on an individual basis. So even for pharmas, you know, I think that many of their drugs could potentially be used in many different indications, and so that data is important for them. I think the fact that we can test hundreds of drugs, and if we include drugs that are new drugs that they've developed or, you know, drugs in their pipeline, in our library, we will be able to probably provide new information for them about their drugs and new indications. So I think that as we generate more data and we'll get more pharmas involved and we will have the opportunity to do both, use existing drugs, but also find novel indications for new drugs. [00:30:20] Speaker A: Makes a lot of sense. Dana, are you making an ethical argument here? I think about the example you gave of the patient who was going to be prescribed three drugs and ended up being prescribed two or would have been prescribed steroids, which were counter indicated. So is there an ethical argument that we should be doing this at scale for as many patients as possible? Is that part of the story that's. [00:30:42] Speaker B: Going to be part of our process of getting FDA approval? I mean, if we're providing safer options for patients that are just as effective, I think we should be doing that. Obviously, you need to choke the idea of the more the better when it comes to children. I mean, this notion that the more drugs you give, the better you can kill the cancer cells is not true. And that's what we're showing with this test is like when we have four drugs versus three drugs versus two versus one. You know, is one as effective as three? That would be great because we can just get rid of, you know, the toxic effects of these drugs. [00:31:15] Speaker A: And of course, it's also. It is also a savings for whoever's paying for it. So there are economic arguments, but I think it sounds like you have both a strong economic and sort of ethical arguments for. And then effective, I guess just general effectiveness. A lot of wins in your sales right now. [00:31:29] Speaker B: I hope so, Chris. It's just a matter of convincing the right people. [00:31:35] Speaker A: Well, you're a perfect spokesperson for this, so. Okay, a couple of last questions as we close out here. We've talked a little bit about where cancer treatment is likely to go over the next few years. Will you talk a little bit about AI? It is top of mind for everyone these days. And, you know, tell us if you think it is overrated or underrated. Tell us a little bit about how you're using it and what the applications for AI might be in your world in the near future. [00:31:57] Speaker B: So will AI replace a human being? No, I don't believe that. Okay. But can AI help us revolutionize, you know, cancer treatment? Yes. So I'll give you an example. Think about this, right, from every cancer patient, we do DNA sequencing. We look at, we do rna sequencing, transcriptomics. We measure response to hundreds of drugs on each patient. All that data, right, integrated with AI. As you build that data set, what happens? Right? So the AI will slowly start to recognize patterns and start to identify, right. Biomarkers with drugs. As we built this data set. I believe in five years, when we have, you know, we've done this on, let's say, hundreds and thousands of patients, maybe in five years, the AI would predict what drugs work for each patient without having to do a biopsy. Think about that. So that's the power of AI. And this is why I'm starting to use AI really early on. I'm collaborating with Doctor Noah Barlow, who's, you know, has developed this machine learning approach where he can take all this data and put it in this engine. And as we generate and as we increase the data set, I believe in five years we'll start predicting what works for each individual patient, maybe without even having to do a biopsy. [00:33:09] Speaker A: Incredibly exciting. I think one of the things that we've heard from several people who are sitting on the cutting edge of AI right now is that the time horizons that people think in are shorter than the time horizons that normal people think in. So we're in the steep part of the change curve right now. And so when you ask people to sort of predict out to the future, they're sort of saying two years from now, four years from now, five years from now, they're not talking about 15 years from now, because we don't really know. I think it becomes hard to say anything really interesting when you go out that far in time. So that is incredibly, incredibly exciting. All right, final question. I have to ask this in my mother's honor. What's your favorite lebanese dish to cook? I will say my mother's gibbee is still something that I dream about. [00:33:50] Speaker B: Oh, my God. That's my favorite. Wow. I did. That is my favorite. Chris, I thought it was yours as well. Rokimbe. I mean, come on. So I know. I'm sure you've gone through this, right? You're telling your friends to try Rokibe, and they're looking at you like, what is that? It's raw lamb, minced lamb with special spices. And you only can have it with onion and olive oil and mint. And everybody looks at me like, there's no way you're kidding me. But when they try it, they love it. [00:34:26] Speaker A: They're converted, right? Yeah. It's easy to understand why hummus went global before Kibbe did. But I'm with you. I'm a kid fan. [00:34:34] Speaker B: Oh, I love to hear that. My daughter, she's 13, but, like, a few years ago, she would go to a friend's house. I can't make it. So I'm gonna tell you this. I'm not a good cook. I'm sure you've heard that I'm not a good cook. But she knows that I'm not gonna make her kibbeh. But she goes to my friend, and she sleeps over. And in the morning, my friend asks her, what do you want to eat, Leah? And she looks at her can you make me kibbe? [00:35:03] Speaker A: A true lebanese child asking for Gibbee for breakfast. That's great. Well, Doctor Diana Azam, it's a pleasure to talk to you. This is incredibly exciting research that you're doing. Thank you for spending some time with us today on few and far between. [00:35:16] Speaker B: Thank you, Chris. I appreciate you having me. Thank you so much. Was fun. [00:35:24] Speaker A: Thank you for listening to the latest episode of few and far between. Our podcast is now available on Apple Podcasts and other major streaming services. Please take a moment and leave us a user review and rating today. It really helps people discover the podcast and we read all the comments. Those comments help us to make few and far between better and better. Also, be sure to subscribe to few and far between so that you don't miss a single episode. Got an idea for a future episode? Email us at few and far [email protected] or contact us on our [email protected] dot. I'm your host, Chris O'Brien. See you next time.

Other Episodes

Episode 11

September 28, 2021 00:34:38
Episode Cover

Episode 11: Rebecca Stewart and Nicola Miller from Rare Revolution Magazine

How important is transparency to the rare disease community? Today's full episode of Few & Far Between features Biorasi's Lindsey Rios, with guests Rebecca...

Listen

Episode 4

June 23, 2021 00:38:12
Episode Cover

Episode 4: Marianne Clancy, Executive Director at Cure HHT

On today’s podcast, host Wayne Bowden, Vice President of Partnerships and Strategy at Biorasi, talks with Marianne Clancy, Executive Director at Cure HHT, about...

Listen

Episode 2

May 26, 2021 00:39:31
Episode Cover

Episode 2: Julie Walters, Founder and Executive Director of Xperiome/Raremark

On today’s podcast, host Wayne Bowden, Vice President of Partnerships and Strategy at Biorasi, will be talking with Julie Walters, Founder and Executive Director...

Listen