Tag Archive for: EGFR

Advanced Non-Melanoma Skin Cancer Treatment | Partnering With Your Team on Care Decisions

Advanced Non-Melanoma Skin Cancer Treatment | Partnering With Your Team on Care Decisions from Patient Empowerment Network on Vimeo.

 When making advanced non-melanoma skin cancer care and treatment decisions, what factors help determine the best approach for YOUR disease? Dr. Soo Park reviews current treatment options, emerging research, and shares advice for partnering with your healthcare team.

Dr. Soo Park is a Medical Oncologist at Moores Cancer Center at UC San Diego Health. Learn more about this expert.

 

Related Resources:

An Expert’s Perspective on Advanced Non-Melanoma Skin Cancer ResearchAn Expert’s Perspective on Advanced Non-Melanoma Skin Cancer Research What Do You Need to Know About Advanced Non-Melanoma Skin CancerWhat Do You Need to Know About Advanced Non-Melanoma Skin Cancer? What Is Non-Melanoma Skin Cancer?


Transcript:

Katherine:

Hello and welcome. I’m your host, Katherine Banwell. In today’s program, we’ll be discussing advanced non-melanoma skin cancer, what it is, how it’s treated, and you’ll learn tools for advocating for yourself.   

This program is part of the Patient Empowerment Network toolkit series, which was created with the goal of helping patients learn more about their cancer and empower them to play a proactive role in their care. Before we meet our guest, let’s review a few important details. The reminder email you received about this program contains a link to program materials. If you haven’t already, click that link to access a guide to help you follow along during the webinar.  

At the end of this program, you’ll receive a link to a program survey. Please take a moment to provide feedback about your experience today, in order to help us plan future webinars. And finally, before we get into the discussion, please remember that this program is not a substitute for seeking medical advice. Please refer to your healthcare team about what might be best for you.  

Well, let’s meet our guest. Joining us is Dr. Park. Welcome. Would you please introduce yourself?  

Dr. Park:

Great, thanks. So, my name is Sue Park, and I’m an associate professor of medicine at the University of California here in San Diego, and I focus on cutaneous oncology. So, I see patients with all types of skin cancers. And in particular, I focus on non-melanoma, because I think most patients realize that the other skin cancer that is widely known is melanoma.   

Katherine:

Okay, that’s great. Thank you so much for dividing that up, and thanks for taking the time to join us today.  

Dr. Park:

Yeah, of course.  

Katherine:

So, Dr. Park, non-melanoma skin cancer is a group of cancers. Would you define it for us?  

Dr. Park:

Yeah, so, non-melanoma skin cancers is just basically a broad blanket term for any skin cancer that is not a melanoma. And so, that’s things like basal cell skin cancer, squamous cell skin cancer, Merkel cell skin cancer. So, anything that’s not considered a melanoma. A melanoma is another type of skin cancer, but it develops from a different type of skin cell.  

Katherine:

Okay, that’s good to know. So, what are the most common types of non-melanoma skin cancer? I think you’ve just mentioned that, but maybe you could mention them again, and maybe define each one.  

Dr. Park:

Yeah, so, there are lots of different types of non-melanoma skin cancers, but the two most prevalent ones are basal cell skin cancer and squamous cell skin cancer, and they actually both come from the same cell of origin in the skin. but there are actually two different types of skin cancers because ultimately that one cell, develops into another cell.  

And so, for basal cell skin cancer, the cell that it comes from is a basal cell, and that’s why it’s called basal cell skin cancer. And that is very different than squamous cell skin cancer. So, as the name says, squamous cell skin cancer actually comes from a squamous cell, and these are both cells that are in your skin, and both of these cancers are mainly driven by the sun exposure.  

But even though they’re both non-melanoma skin cancers and they’re the most common, with basal cell being the number one most common skin cancer actually in the world, I think a lot of people don’t recognize that, because we don’t capture the occurrences of basal cell skin cancer very well, because it’s so common. But those are the two main types of skin cancers that I think a lot of treatments are focused on right now.   

Katherine:

And are these cells – can they develop into cancer anywhere on the body, on the skin of the body?  

Dr. Park:

Yeah, so, anywhere that you have any type of cutaneous skin, these cells reside, and typically they happen in areas where the skin is exposed to the sun. So, for example, they often happen on the head and neck, because we just get lots of sun exposure there.  

Katherine:

Yeah, yeah, okay. So, who typically diagnoses this stage of skin cancer? Is it a dermatologist, or somebody else?  

Dr. Park:

So, that really depends on the stage of the skin cancer. So, when I say stage, I mean how advanced is it. For an early stage skin cancer, those are typically really small. And oftentimes, patients will have a few of these; and some patients have a lot of these, maybe on their face, their neck, their scalp, across their hands and arms. And typically, they notice a small lesion that won’t go away or is getting a little bit irritated.  

And so, they see the dermatologist first. So, the dermatologist is often the first person that sees patients whenever the patient has noticed like a small skin abnormality that’s not getting better. But sometimes, they also see patients that do not see the dermatologist first.  

They actually either see a medical oncologist like myself, or a head and neck surgeon who I work closely with, because some patients have a tumor or a cancer that’s really large, and it’s too large to the point where a dermatologist is not able to offer them anything. And so, if the tumor is really large, that’s a later stage cancer. So, it’s not as stage.  

Katherine:

Yeah. Now, this may seem like a silly question, but don’t the cells sometimes show up at like a mole, so you wouldn’t even know you had it?  

Dr. Park:

Yeah. So, that’s more common for melanoma. It can show up as a mole, but you’re exactly right. For both non-melanoma skin cancers like basal cell and squamous cell skin cancer, it can look just like a little lesion that you might have thought you hit yourself on like the door handle, and it’s not getting better.  

Some of these cancers actually appear, and then they go away on their own, and then they come back. And so, sometimes it’s really hard to see that it is a skin cancer, it was a skin cancer. But I think what’s really important is that it’s a lesion that’s either getting worse, or it’s not going away, so you should really tell your doctor about.  

Katherine:

Yeah. We know that there’s a multidisciplinary team involved with advanced non-melanoma skin cancer care. Who are the members on the team?  

Dr. Park:

So, I think now, because we have so many more treatments to offer patients, if I’m specifically talking about a non-melanoma skin cancer on the head and neck, which is where it most commonly occurs, because of sun exposure, I think the multidisciplinary team is really important. That typically consists of a medical oncologist like myself, typically a head-and-neck surgeon, and sometimes a plastic surgeon, as well, depending on what we come up with as a treatment plan.  

It also often involves a radiation oncologist. So, that’s a special type of doctor that gives radiation treatment. We always want to keep the dermatologist involved, as well, because most of these patients do have a primary dermatologist that’s been really involved in their care.  

Katherine:

What about other people on the healthcare team, like that are in a supportive form on the team? 

Dr. Park:

Yeah, that’s just as important, because a lot of these patients may have barriers to care, or they may have social factors that affect their ability to get the care that they deserve, and that they need. 

So, we work really closely with social workers, with nurse navigators, even speech therapists. Sometimes if the cancer is involving some really close to your lip or mouth, sometimes that can be really difficult. We work a lot with audiologists, as well. So, we have a really great multidisciplinary team.  

Katherine:

So, once a patient has been diagnosed, what are the tests that help understand more about the patient’s individual disease?  

Dr. Park:

So, it’s always important to get a biopsy, so then we can tell which type of non-melanoma skin cancer it is. 

And that’s when we look at your cancer under the microscope, and a special doctor called a pathologist. And actually, they’re also really important as part of our multidisciplinary team. They look at the tumor under the microscope, and they help us decide and tell us which type of non-melanoma skin cancer it is. 

But aside from that, I think imaging is really important. So, that are things like CT scans, MRI scans. Sometimes we have to also recommend a PET scan, which is another type of special scan. And these images are really to help us look deeper into the structure of your body, because I can only see so much from the outside.  

And they can really help us tell how deep is the cancer; is the cancer around any critical structures? Is it anywhere else in the body? Because if we find cancer far away from where it originally occurred, that may tell us that the cancer is a later stage.  

Katherine:

So, let’s just go with a scenario. Somebody comes in to you, and they have a lesion on their cheek, for instance.   

Would you do a whole body MRI or a CT scan to see if that…once you’ve done a biopsy, you find that it’s cancerous. Would you do a whole body MRI, or a scan of some sort, to see if the cancer was anywhere else?  

Dr. Park:

So, we typically don’t, because we know the patterns that – for instance, like you mentioned, like a skin cancer in your cheek can go to. And so, non-melanoma skin cancers on the face or anywhere in the body, they typically like to go to the lymph nodes that drain that area. And so, if you have a lesion on your face, that’s typically your neck.   

And so, we’ll do a good exam of your face, your neck, but we will also get imaging of those areas. So, we typically get an imaging focused on the head and neck. If we find something abnormal there, then that may tell us we need additional imaging in the other parts of the body. But more often than not, we don’t start with a whole body scan.  

Katherine:

Okay. What questions should patients ask about their test results?  

Dr. Park:

So, I think patients should definitely ask, “What type of skin cancer do I have? How did it arise? Where all in my body is the skin cancer? What does my blood work look like?” And I think patients should also be aware that for many years now, we send tumor samples for something called molecular sequencing, and that just tells us different types of mutations that may be in your tumor. And that’s really important, because there are some drugs we have now that are only for patients that have specific mutations in their tumor.  

And so, if you are one of those patients that has a specific mutation, that opens the door to another type of therapy for you. And, you know, that’s something that’s now recommended, actually, by a lot of cancer societies, to really send your tumor for some type of molecular sequencing, so we can level the playing field for all patients, and offer them the full range of treatments that we have.  

Katherine:

Yeah. What are the common mutations?  

Dr. Park:

So, for basal cell skin cancer, almost all basal cell skin cancers are driven by abnormality in a certain pathway called the hedgehog pathway. Yeah, I’m – 

Katherine:

Interesting. Why? 

Dr. Park:

It was named, I think, by someone. All these names are people by someone that discovered it, and they get the rights to name the pathway. But for a basal cell, it’s the hedgehog pathway. And so, in the hedgehog pathway, there are certain types of mutations specifically associated with that pathway. And one of them, among these mutations, we look for drugs that can inhibit this pathway. So, there are drugs that specifically target the hedgehog pathway.  

They’re called hedgehog inhibitors, and they’re oral medications or pills that you can take every day. And those are for patients with basal cell skin cancer, because the basal cell skin cancer came about because the hedgehog pathway is not normal. But for squamous cell skin cancer, squamous cell skin cancer often has a lot of mutations. And unfortunately, they’re the type of mutations that we actually don’t have a drug for at this moment. But one unique thing about squamous cell skin cancer is that it has so many mutations.  

And so, that means that it has a better chance of responding to a different type of treatment. It’s an IV treatment known as immunotherapy. And so, that’s something that’s relatively recent, I think, in the past five years now. We’ve started using immunotherapy for patients that have squamous cells skin cancer, and it’s worked remarkably well.  

Katherine:

How do these cancers typically progress? What are the stages?  

Dr. Park:

Yeah, so, if it’s just a really small cancer like that’s on your face, it’s typically an early stage or a stage I. And I’m specifically talking about squamous cell skin cancer, because actually for basal cell, we don’t have any formal staging for basal cell.  

Katherine:

Why is that? 

Dr. Park:

It just wasn’t included in the staging systems. So, for basal cell, there’s no formal staging criteria, but we’ll stage it as early stage based on what we think, as a clinician, when we see you; or if we get imaging and we see that it’s spread to other areas, it may be later stage. But for squamous cell skin cancer, it’s earlier stage depending on the size. Sometimes when we get a biopsy, and in the biopsy, if we find high risk things in the biopsy, that may actually put you at a higher stage, even if the cancer is somewhat small.  

So, that could be like a stage III. But if at any point we find on imaging that the cancer has spread elsewhere – so, like you have a cancer that has spread to your liver, or to your lungs, or to the bones in your body, that’s a stage IV. 

Katherine:

Okay. And when is the cancer considered advanced? 

Dr. Park:

I think the cancer would be considered advanced if it’s not something that a surgeon can simply just remove. So, the dermatologist cannot just do a standard Mohs surgery, or the head-and-neck surgeon cannot just do a standard surgery, because advanced means that the cancer is either pretty deep, pretty large; or the surgeon can do surgery, but that means that the surgery would be very disfiguring. Sometimes these lesions can be really big on the face. 

And sure, the surgeon could do the surgery, but if we have to take part of your eye, or part of your nose or your ear, and you have to have major reconstruction, that’s considered probably more of an advanced tumor.   

Katherine:

Okay. As I mentioned, in this webinar, we’re focusing on advanced cancer. So, what is the typical treatment path for someone who’s been diagnosed at this stage of disease, at the advanced stage?  

Dr. Park:

Yeah, so, before – and I really love this question, because in the past, we did not have that much to offer patients except surgery, and then they would get a very extensive surgery. They would get reconstruction. But sometimes it’s hard to get reconstruction after a really major surgery, because you have to heal, and you have to get better. And then, after the surgery, you would typically get radiation to try to prevent the cancer from coming back. But nowadays, we have immune therapy.  

So, immune therapy is a certain type of IV medicine that’s not chemotherapy that works really well for squamous cell skin cancer. And so, nowadays, we can actually give this to you before surgery. So, we can give you a couple of doses of this IV immune therapy medicine before surgery, and really shrink your tumor quite dramatically.  

And then, that makes the surgery a lot easier, smaller. And then, sometimes after we do the surgery, and then we look at what the surgeon has taken out under the microscope, we can’t see any tumor left. And that’s really amazing, because then sometimes we don’t even need to do radiation. So, not only did we make your tumor a lot smaller, sometimes we completely made it go away.   

And then, if that happens, sometimes we don’t even need to do radiation. So, it really helps the patient. And I think this is really important, because this is somewhat newer data, and I still see patients that get referred to me for just surgery. 

But I think a lot of head and neck surgeons are now aware of this data. And so, this is something that’s, I think, becoming more common.  

Katherine:

What about targeted therapies?  

Dr. Park:

So, targeted therapies are, I think, mainly used in basal cell skin cancer. So, targeted therapies are typically oral medications or pills. They’re called targeted, because they’re used in cancers that have a specific target. So, for example, the basal cell skin cancer, the target is the hedgehog pathway, because the hedgehog pathway is abnormal. And so, these pills, they specifically target the hedgehog pathway. But for squamous cell skin cancer, we don’t have any true targeted therapies.  

Katherine:

As patients are reviewing their options with their doctor, what questions should they be asking about their care plan? 

Dr. Park:

I think all patients should be asking, what the goal of the treatment is. They should be asking, especially if they’re being offered any type of treatment, what are the side effects? What can I expect from this, in terms of how much better will it make me? They should really ask about how often the treatments are given, because some patients have transportation issues or financial barriers, and we want to know about that, so we can help them.   

Patients should also ask about any necessary blood work that is needed. They should ask what can they do in the future to prevent a similar type of cancer happening, and just make sure that they’re talking to their families, because I think social support is really important.  

Katherine:

Yeah. I think it’s important for patients to ask how the cancer is going to impact their lives overall, really.  

Dr. Park:

Yeah, exactly, because it will affect every single aspect of your life: your social life, your family life, your mental health, your physical well-being. And so, it’s really important to know and work with your doctor on what you think you can expect now, and also in the future.  

Katherine:

Yeah. Well, how do test results impact treatment options, then? 

Dr. Park:

So, there are sometimes when we have a skin cancer that actually happens inside a gland in your face. It’s one of the salivary glands in your face. And we sometimes don’t know if it’s a skin cancer that happened on the outside and that spread to the gland inside your face, or did it actually first just start inside the gland? Because a cancer that just starts inside the gland is not technically a skin cancer. It’s a different type of head-and-neck cancer, and it’s very, very rare, and it’s treated very differently.  

So, nowadays, because we have that molecular testing, like I talked about, I see lots of patients where they have a cancer in their salivary or parotid gland. We don’t know where it came from. And so, we send it for molecular sequencing or molecular testing, and there are certain clues in the molecular testing that can tell us, oh, it probably actually came from a skin cancer.  

You just didn’t know it; or maybe it’s the skin cancer that kind of was there and went away; or maybe it was a skin cancer you had like five years ago, that you didn’t think caused any problems, but it did spread, because knowing where it came from through molecular sequencing, if it’s really hard to find out where, really impacts the treatment I may give you.  

Katherine:

Dr. Park, we’ve been hearing a lot about innovation technology, or we’ve been hearing a lot about innovations in technology. How are these advances improving skin cancer care?  

Dr. Park:

They’re approving care, because we can offer patients more minimal procedures. We can tell them you don’t need this type of other treatment, and you can have the same outcome. So, we can tell you need less treatment, and the outcomes are just as good, because sometimes more treatment is not always better. More treatment sometimes means more toxicity, more time away from family, more time away from home. Advances mean that we can keep you cancer-free for longer.  

Or even if I can’t ever get rid of the cancer, we have drugs that can keep it under control for a long time, and it stays under control, even if I stop the medicine. So, all those are really remarkable things for our patients, that we have options that can help them live healthy, full lives.  

Katherine:

How do you know if a treatment is working? How is a patient’s response monitored?  

Dr. Park:

For skin cancer, that’s pretty easy, and I think that’s one of the most satisfying things, because I can often see the cancer visibly. I don’t always have to rely on a scan, as for some patients for the cancers in their stomach or something like that. So, patients will often see a dramatic reduction in the size of their tumor, sometimes even after the first treatment I give them. And not only can we tell by looking at them; eventually, I will get a scan to compare it to the scan they had it in the first place, and we see that the tumor has gotten a lot smaller.  

Katherine:

That’s good news. What about side effects of these therapies? How are they managed?  

Dr. Park:

Yeah, so, for immunotherapy, there’s one specific side effect that we don’t find with chemotherapy, and that’s really when your body’s own immune system kind of ends up attacking the other parts of your body. And so, it can cause inflammation of other organs. And so, for patients that experience that, it can be very mild, and it can be all the way to very severe, requiring a patient to go to the hospital.  

But in all cases, we just have to tell the immune system to quiet down a bit, because it’s attacking your body. And so, the way we do that is we give the patient steroids. And so, if it’s really mild, maybe you have like a small rash; maybe we can just give you a steroid cream, or maybe we have to give you a steroid pill. But sometimes, if it’s really severe, we have to tell you to go to the hospital so you can get steroids through your IV.  

Katherine:

Okay. Beyond what has been approved to treat advanced non-melanoma skin cancer, where do clinical trials fit in?  

Dr. Park:

Clinical trials are great, because they’re the reason why we have the drugs that we have today that are working so effectively. And so, I’m really fortunate to be at a center where we offer clinical trials for patients.  

Clinical trials always fit in at any point in the journey, as long as they fit your disease. So, most of the clinical trials we have are for patients that have advanced disease, not for early stage, because the early stage patients don’t really need it; because with just simple surgery, they tend to do really well, and their cancer doesn’t really ever come back and cause issues.  

But clinical trials are really important, because they’re the only way we can study a promising treatment option; see how well it works, and if it works really well, then move it further on to hopefully help a lot of other people. And that’s why we have immunotherapy today – is through clinical trials; and immunotherapy is used for lots of cancers. 

Katherine:

It is, yes. Are there barriers to accessing trials? And if so, do you have any recommendations on how to tackle them?  

Dr. Park:

Yeah, there are barriers to trials, and I think the thing that the cancer community is really trying to work on is barriers to accessing trials if you’re from an underrepresented population in medicine. So, based on your socioeconomic status, meaning how much money do you make or what your education level is, what race are you, what ethnicity, what is your background; because we know that there’s a disparity for those individuals. And so, I think really asking your doctor, staying curious; asking them, “Could a clinical trial help me?” 

Even if you have no idea of really what trials there are, or what they really mean, you can just throw that word out; and that then, the doctor knows that you’re potentially interested, and they can actually give you the information and help you learn more about it, even if you don’t know much about it; because I think it’s still quite a black box, and we’re trying to overcome this barrier, but it’s difficult.  

Katherine:

And what about researching clinical trials online? What sites would you recommend?  

Dr. Park:

So, I have to admit that there are multiple sites, but sometimes they are not very patient-friendly. Even whenever I look at them, they are not very friendly. I think the largest repository of clinical trials is the NCI database. It’s just where every clinical trial that’s open has to be registered, and it’s run by the federal government, I believe. And because it’s not super regulated, it’s just the person working on the trial entering information, and they’re typically of a medical background, sometimes the information is not very easy to digest or understand.  

So, I think really just letting your medical team know that you might be interested in a trial, then they can help you find the right trial for you. And if they don’t have one, they can tell you other areas or other centers that might have one. You are more than welcome to go search in Google and all that, but it can be really hard, and then I don’t want that to make the patient feel that this is not for them.  

Katherine:

Right, right. Are there any recent research highlights that you could share with the audience?  

Dr. Park:

Yeah, so, one thing that just recently came out is that – so, squamous cell skin cancer is actually a lot more common and a lot more aggressive in patients that have an organ transplant. So, I’m talking about patients that have a kidney transplant, or a liver transplant, or heart transplant.  

But the problem is, I can’t really give them immune therapy like I can somebody else that does not have a transplant, because like I said, sometimes one of the side effects of the immune therapy is that it can attack other parts of your body. And so, for patients that have an organ transplant, one of the risks, if I did give them immune therapy, is that it would actually attack their organ that they got from somebody else.  

And except for kidney, because we have dialysis: if I injure that organ, I don’t really have much else, and that does not bode well for the patient. But now, they’re trying to really figure out ways we can actually combine immune therapy with steroids in different doses to really see: can we actually help the patients that have organ transplants? Can we help their skin cancer? But then, can we also not injure their organ? And so, that’s something that a lot of research is being conducted on right now, and it’s really exciting.   

That’s great information. Well, Dr. Park, as we wrap up, what would you like to leave the audience with, in terms of the state of advanced non-melanoma cancer care?  

Dr. Park:

Yeah, I think as a patient, no matter your background, I think it’s just really important to be your own best advocate. And sometimes, that’s easier said than done. Some patients have families that can be advocates for them; but if you are by yourself, you can ask your doctor anything. You have the right to know, because it is your health and your body, and we do want to hear from you, because we do want to work with you to provide the best care that we can for you. 

Katherine:

Dr. Park, thanks so much for joining us today. I really appreciate it.  

Dr. Park:

Thanks so much, Katherine. It was a pleasure. 

Katherine:

And thank you to all of our collaborators. If you’d like to watch this webinar again, there will be a replay available soon. You’ll receive an email when it’s ready, and don’t forget to take the survey immediately following the webinar.  

It will help us as we plan future programs. To learn more about advanced non-melanoma skin cancer, and to access tools to help you become a more proactive patient, visit powerfulpatients.org. I’m Katherine Banwell. Thanks for being with us.  

Advances in Targeted Lung Cancer Treatments | What You Should Know

Advances in Targeted Lung Cancer Treatments | What You Should Know from Patient Empowerment Network on Vimeo.

Dr. Thomas Marron discusses how these therapies work to treat lung cancer, how the presence of certain mutations can impact care and treatment choices, and the research being done on new therapies to target specific lung cancer biomarkers.

Dr. Thomas Marron is Director of the Early Phase Trials Unit at the Tisch Cancer Institute at Mount Sinai Hospital. Dr. Marron is also Professor of Medicine and Professor of Immunology and Immunotherapy at the Icahn School of Medicine at Mount Sinai. Learn more about Dr. Marron.

See More from EVOLVE Lung Cancer

Related Resources:

How Has Lung Cancer Molecular Testing Evolved?

How Has Lung Cancer Molecular Testing Evolved?

Antibody Drug Conjugates for Lung Cancer | Advances in Research

Antibody Drug Conjugates for Lung Cancer | Advances in Research

Expert Perspective | New and Emerging Progress in Lung Cancer Treatment

Expert Perspective | New and Emerging Progress in Lung Cancer Treatment

Transcript:

Katherine Banwell:

Welcome, Dr. Marron. Would you introduce yourself, please? 

Dr. Thomas Marron:

Sure, I’m Tom Marron. I’m the Director of the Early Phase Trials Unit at the Tisch Cancer Institute at Mount Sinai Hospital. I’m a Professor of Medicine and also a Professor of Immunology and Immunotherapy at the Icahn School of Medicine at Mount Sinai. And I’m trained as both an oncologist and an immunologist.  

Katherine Banwell:

Excellent. Thanks for joining us today.  

Dr. Thomas Marron:

Thank you for having me.   

Katherine Banwell:

We know that the presence of certain mutations can affect lung cancer treatment options. Can you share the latest updates in targeted therapies?  

Dr. Thomas Marron:

Sure, so there’s been a lot of developments in targeted therapies as of late.  

Mutations in a patient’s cancer can represent a potential therapeutic target, and we have increasing numbers, every year we have new FDA approvals for typically pills that target very specific mutations and are able to either control cancer or even kill cancer. Additionally, we use DNA sequencing of tumors to identify mutations that could be predictive of a response to certain therapies. So, even though we don’t have a specific drug to target that mutation in their DNA, that change in their DNA that’s making the cancer grow, we do know that patients with certain DNA mutations do better on certain therapies than other therapies.  

And so, we can use mutations specifically to help guide therapy, even if we don’t have a targeted therapy for something like EGFR mutation or a KRAS mutation. And additionally, one of the things that we do as we’re treating patients is, often times we will give a patient with lung cancer a therapy and then their cancer may respond for weeks, months, even years.  

But then it might recur, or it might just start growing if it never went away entirely. And at that time, we’re oftentimes repeating the genetic sequencing, whether doing a biopsy or sometimes we can do what we call a liquid biopsy, which is just taking some blood and looking for some of the DNA from the cancer floating around in the blood.  

And the reason we do that is that if you see a change in the mutations, it might represent either a change in the type of cancer or it might represent what we call an escape mutation, or an escape mechanism where the cancer that had been responding to therapy X is now not responding because it changed its DNA to overcome the therapy you were given. And that might suggest that we try a specific new therapy, or that we just change our approach entirely.  

Katherine Banwell:

You’ve answered my next question to some degree, but I’m going to ask it anyway. How do these therapies work to treat lung cancer? 

Dr. Thomas Marron:

So, cancer is caused by changes in your DNA. So, your DNA is your instruction booklet on how cells should grow and when they should grow. And every cell in your body theoretically has the same DNA, except for, because of a variety of things like smoking or exposure to radon or just living in a large city full of pollution. As we get older, we basically accrue more and more mutations and changes in our DNA, our instruction booklet. And while most of these changes really don’t have any sequela, and they’re not going to affect the ability for the cancer, or for normal cells to grow.  

Sometimes you’ll get a mutation in a very specific gene that’s important for telling cells when to divide and when to grow and when not to grow. And you can think of it as a light switch where the light switch gets stuck in the on position and constantly, cells are growing and growing and growing and that’s when you have cancer. So, when you have these mutations, one of the approaches that we’ve been working on for the last few decades, in particular in the last few years.  

We have lots of these new drugs that target these mutations, and they basically turn that on signal off. So, they disrupt, it’s like turning the light switch off. You’re disrupting the constant grow, grow, grow signal and keeping the cancer from growing. Typically, we think of these targeted therapies that do this, not as cures for cancer, at least when patients have metastatic disease, but they’re very good at controlling cancer. And some of these therapies can work for years, even a decade and control the cancer. But often times, unfortunately cancer always finds a way to outsmart us, even when we’re outsmarting it.  

Katherine Banwell:

Right. Are there new mutations being discovered that can impact the future of small cell lung cancer care? 

Dr. Thomas Marron:

Well, I’m not sure I would say that there’s a lot of new mutations that’ve been discovered, per se. Every time that you come in and get a diagnosis of lung cancer, we typically will take the tissue and like I said, sometimes we’ll take some blood and do a liquid biopsy and look for a slew of different known mutations.   

And typically, we’ll look for anywhere from three to 500 known mutations in the cancer, even though we only have drugs to treat about 10 of those three to 500. The nice thing though is that as we learn more and more and more about these mutations and we study them, we are developing more and more drugs to address specific mutations. So, five years ago we really only had three different mutations that we could target.  

Now, we have around 10 because we have all these new drugs that target very specific mutations whether they be in genes like MET or RET or KRAS or BRAS.  

So, I think that while we aren’t necessarily discovering that many new genes, we’ve been looking at the genetic sequence of cancer and also, just the human genome for 20 to 30 years at this point, we’re discovering lots of new drugs that can target those specific mutations that we know patients have, but that most of the mutations we identify are not necessarily druggable targets.  

Understanding Oncogene-Driven Lung Cancer: Targeted Therapy Advances and Challenges 

Understanding Oncogene-Driven Lung Cancer: Targeted Therapy Advances and Challenges from Patient Empowerment Network on Vimeo.

How do the genetic mutations of EGFR and exon 20 impact lung cancer? Expert Dr. Christina Baik from Fred Hutchinson Cancer Center discusses oncogene-driven lung cancer and how it differs from other lung cancer types.

Download Resource Guide

See More from START HERE Lung Cancer

Related Resources:

Lung Cancer Patient Expert Q&A: Dr. Christina Baik

Lung Cancer Patient Expert Q&A: Dr. Christina Baik 

advice

Newly Diagnosed Non-Small Cell Lung Cancer | Key Advice for Patients

Bridging gap

Advancing Lung Cancer Treatment: Bridging the Gap in Personalized Care

Transcript:

Lisa Hatfield:

What exactly is oncogene-driven lung cancer, and how does it differ from the other types of lung cancer?

Dr. Christina Baik:

So in lung cancer, there are certain lung cancers where the growth of the cancer is dependent on a particular genetic abnormality. So there is one gene that makes that cancer grow. And because of that, there have been treatments that are developed against that particular genetic abnormality.  So, it is referring to lung cancers that have that particular genetic abnormality. A prime example of this is lung cancers that have what we call an EGFR mutation.

That means that there is this gene called EGFR that is abnormal, and that’s making the cancer grow. Now, not everyone has a cancer gene that is driving that cancer. I would say about 30 percent or for 30 percent to 40 percent of patients would have an oncogene-driven cancer for which there may be treatments either as a standard treatment or in clinical trials. But the majority of patients do not have an oncogene, meaning that genetic abnormality where there is a targeted therapy option. So that’s the distinction we make. And I know this term or phrase is used a lot, but that’s what it means. And if you want to know if one has an oncogene-driven lung cancer, you would know based on the genetic test results.

Lisa Hatfield:

Okay. Great. Thank you. And just for clarification too, the genetic mutations are found in the cancer cells, not in their body, the cells? So that’s what the genetic testing is done just on the cancer cells. Is that correct?

Dr. Christina Baik:  

Yes. Yes.

Lisa Hatfield:

Okay, great.

Dr. Christina Baik:

Thank you for clarifying that. That’s a very important distinction.

Lisa Hatfield:

Yeah. Thank you. So that leads right into the next question, and it’s kind of a lengthy question. And this person is asking, “Dr. Baik, you have done considerable research around EGFR exon 20 insertion mutations in non-small cell lung cancer, considering their association with poor survival outcomes, what are the survival implications of having EGFR exon 20 insertion mutations compared to other types of EGFR mutations?”

Dr. Christina Baik:

Now this gets a bit complicated, but not all EGFR lung cancers are the same. And there are patients who have what we call EGFR mutation that is a classical mutation. And I throw out that term, because that’s how it’s written on the Internet and a lot of papers. And then, so that’s one group, and the other group are patients who have this exon 20 insertion mutation. And the reason these are separated is because the treatments that work very well in the classical mutations do not work very well in this particular exon 20 mutation.

So when we look at all patients with EGFR mutation, it is true that the prognosis is poor in exon 20 patients just because there are no great targeted therapy options. That said, I am very hopeful that this is changing. There are a number of targeted therapies for exon 20 that are in trials, and I think these are going to be FDA-approved in the future, not too far off in the future, I believe. So I think the survival implications will start to hopefully equalize amongst all the EGFR-mutated lung cancer patients.


Share Your Feedback

Create your own user feedback survey

Lung Cancer Patient Expert Q&A: Dr. Christina Baik

Lung Cancer Patient Expert Q&A: Dr. Christina Baik from Patient Empowerment Network on Vimeo.

In this START HERE lung cancer webinar, Dr. Christina Baik, Associate Professor of Medicine at the University of Washington and Fred Hutchinson Cancer Center, provides valuable insights into priorities for newly diagnosed patients. Dr. Baik also discusses essential tools for managing disease progression and recurrence.

Watch the program and download the guide for tips on creating an actionable pathway to optimize lung cancer care for yourself or your loved one.

Download Resource Guide

See More from START HERE Lung Cancer

Related Resources:

Bridging gap

Advancing Lung Cancer Treatment: Bridging the Gap in Personalized Care

advice

Newly Diagnosed Non-Small Cell Lung Cancer | Key Advice for Patients

DNA

Understanding Oncogene-Driven Lung Cancer: Targeted Therapy Advances and Challenges

Transcript:

Lisa Hatfield:

Hello and welcome. My name is Lisa Hatfield, your host for this Patient Empowerment Network Start Here program where we bridge the expert and patient voice to enable you and me to feel comfortable asking questions of our healthcare teams. The world is complicated, but understanding your lung cancer doesn’t have to be. The goal of this program is to create actionable pathways for getting the most out of lung cancer treatment and survivorship.

Today, I’m delighted and excited to welcome Dr. Christina Baik, a distinguished expert in the field of lung cancer. Dr. Baik serves as an associate professor in the Clinical Research Division at Fred Hutch. She also holds the position of clinical research director of thoracic head and neck oncology at University of Washington. Additionally, she has an associate professor, or she is an associate professor in the Division of Hematology and Oncology at the University of Washington School of Medicine. We’re so happy she’s joining us today. It’s a pleasure to have you with us, Dr. Baik.

Dr. Christina Baik:

Thank you for having me.

Lisa Hatfield:

Okay. Before we dive into today’s discussion, please take a moment to download the program Resource Guide using the QR code. This guide contains pertinent information to guide you both before and after the program. In this program, we’ll provide you with a comprehensive update on the latest lung cancer news and its implications for you and your family.

Following that, we’ll launch into questions we have received from you. So let’s start here. Dr. Baik, can you speak to the latest news and priorities for the treatment of non-small cell lung cancer? And what are the notable advancements in understanding resistance mechanisms or novel therapeutic targets?

Dr. Christina Baik:

So it’s a good time to be a lung cancer doc, I would say, just because there’s so much in advances. We’re seeing different treatments be FDA-approved every other year, if not every year. So it’s really good to have all these options to offer our patients. Now the priority, however, is that not everyone is benefiting in an equal way from all these advances. And really the research priority, including my own personal research, is to really understand why some patients are benefiting and why some are not.

So, for example, in the immunotherapy world, which is a big advance we’ve had in lung cancer in the last 10 years, we know that some patients respond very well, some do not. Yet we give the same sort of treatment to patients. So one thing to understand is who are…and one thing I would say is we don’t personalize immunotherapies for our patients.

So one of the research priorities is to really understand where the different subgroups of patients who are going to benefit from this one treatment type…one type of immunotherapy treatment versus the other. So I would say that’s a big priority for me as well as for the field and all the researchers so that we’re giving the right treatment to the right patient. Now, there have been advances, I would say, in this theme in those patients who are able to receive a targeted therapy. So that is a type of treatment that we give to target the genetic abnormalities that exist in a particular patient’s tumor.

And these treatments work very well. But at some point, it stops working. But nowadays, there are certain sorts of resistance mechanisms as we call it. These are changes that occur in the tumor when a targeted therapy stops working. And we’re starting to understand better in terms of reasons for that and actually develop treatment options for those mechanisms of resistance. So I think we are starting to understand better, and I think we’re going to get there in terms of personalizing immunotherapy. But there’s still a lot of work to be done.

Lisa Hatfield:

Great, thank you. So just a follow-up question to that, when a patient is diagnosed with non-small cell lung cancer,is genetic testing always done on the tumor, or do patients know what their mutations are right upon diagnosis if testing is done?

Dr. Christina Baik:

So, as a rule of thumb, they should, all patients should be tested, and there are exceptions. So, for example, in lung cancer, there’s the type that we call small cell lung cancer, and there’s non-small cell lung cancer. So we often, we usually do not do genetic testing on small cell lung cancer, because often these tumors do not have a genetic abnormality that for which we can actually give treatment for.

But for non-small cell patients, I would say, if most, my personal opinion is that everybody should be tested with the genetic test and really advocate for that. You know, there are certain types of non-small cell lung cancer where there are genetic targets that are rare, however, you don’t know unless you test. So I would say yes to that question of testing for genetic abnormalities.

Lisa Hatfield:

Okay, thank you. So can you speak to the priorities for newly diagnosed patients, particularly populations who may have poor outcomes?

Dr. Christina Baik:

So, I think there are priorities when it comes to research, and then there are priorities for individual patients, right? So from a research standpoint, as I mentioned before, I think really the priorities, the priority is to develop strategies so that we’re truly personalizing treatment for each patient, and we’re not giving this kind of generic treatment for a bulk of the patients. So from a research standpoint, really understanding the biology, understanding what works for what patient, I think that’s extremely important.

On the individual patient level, we sort of alluded to this earlier, but really knowing the cancer we’re dealing with is extremely important. Know your cancer stage, ask what your cancer stage is, know the type of lung cancer that you have. So I will say as of now, there are, I can think of 12 or 13 different types of lung cancer that I want to make sure I know that patients, you know, what their subtype is.

So know your subtype of lung cancer. Ask those questions. If the knowledge is not known, if they say, “You know your stage is not very clear, your subtype is not clear,” then ask why that is, what type of additional testing that needs to be done. So I think those are the type of questions that each patient and their family member should really ask. And in terms of the poor outcome question, I think the first thing I would say is if a doctor tells you, you belong to a group of patients who are going to have a poor prognosis, then ask why that is, right? And understand the reasons for that.

And if that’s, once you understand, I think I’m a big proponent of getting second opinions, because a lot of these treatments and there’s a lot of medical judgment involved when we recommend treatments, and you just want to get a different perspective with the same type or set of information. So really being an advocate for yourself, I think that’s extremely important when you’re first diagnosed.

Lisa Hatfield:

Great, thank you. You mentioned two things I also feel strongly about, I don’t have lung cancer, I have a different type of cancer, but you said that patients and family members can ask questions.  Having an advocate with you at all times, if that’s possible, a family member, a friend going with you, I think is super important. 

And also getting a second consult to understand your diagnosis better. I appreciate you saying that, because some of us are a little bit reluctant to do that, maybe afraid of offending our doctors. So, I appreciate that as a patient myself, so thank you. Okay. So talking about disease progression and recurrence, particularly for metastatic non-small cell lung cancer, what should patients know?

Dr. Christina Baik:

Okay. So when a cancer initially responds to a treatment and it stops responding, there can be many reasons for that. So the first question to really think about is is there another test we can do to identify the reason for the progression? And can we personalize a treatment according to that resistance pattern or the change that occurs in the tumor? This is more relevant to patients who get a targeted therapy, but I think it’s a good sort of rule of thumb in terms of asking your doctor why that is, and is there more testing that’s required. And the second I would say is once the cancer progresses after the initial treatment, then, unfortunately, in lung cancer the treatment options are much more limited, and the effectiveness is very limited as well.

So, it’s really at that juncture to really seek out clinical trials. There are many trials that are out there. So really working with your doctor in identifying these trials. If there is an academic center that’s close to you, at least inquiring about that. In lung cancer, fortunately, there are many wonderful advocacy groups and these advocacy groups can be great resources in finding out about clinical trials and where to seek out opinions. So, I think it does require some homework at the time of progression but really seek those out.

Lisa Hatfield:

Okay. Thank you. Now, if a patient does have an interest in a clinical trial, say maybe they have, their cancer has progressed, would they seek out that trial through the academic center itself? If, say they live in a rural area and they don’t have access, would they contact the academic center itself, or would they seek out a specialist like you first to ask about those clinical trials?

Dr. Christina Baik:

So they sort of come together in a way, because a lot of the specialists are in academic centers. So I think there are two ways to go about it. One is to meet with the specialist who can give you kind of the landscape of where things are and what might be appropriate. So, that’s one way to do it.

The other way to do it is if there’s a particular clinical trial that you’re really interested in based on discussions with other patients or through advocacy groups, if there are particular clinical trials, usually the contact information is listed on the clinicaltrials.gov website, and the contact number is usually for the research team who can give you more information about that particular trial.

Lisa Hatfield:

Okay. That’s very helpful, thank you. And thank you for this overview. I just want to recap a couple of points that you made that’s really important for patients to know. You had mentioned knowing their type, their subtype of lung cancer, knowing their stage, and knowing their mutations and having an advocate. I think those are all really great tips that you gave. So thank you for that overview. It’s that time now where we answer questions that we’ve received from you. 

Please remember, this is not a substitute for your medical care. Always consult with your own medical team, and, Dr. Baik, we have some questions here that people have sent in from patients. So I’ll just jump right into the first question. “What exactly is oncogene-driven lung cancer, and how does it differ from the other types of lung cancer?”

Dr. Christina Baik:

So in lung cancer, there are certain lung cancers where the growth of the cancer is dependent on a particular genetic abnormality. So there is one gene that makes that cancer grow. And because of that, there have been treatments that are developed against that particular genetic abnormality. So, it is referring to lung cancers that have that particular genetic abnormality. A prime example of this is lung cancers that have what we call an EGFR mutation. That means that there is this gene called EGFR that is abnormal, and that’s making the cancer grow.

Now, not everyone has a cancer gene that is driving that cancer. I would say about 30 percent or for 30 percent to 40 percent of patients would have an oncogene-driven cancer for which there may be treatments either as a standard treatment or in clinical trials. But the majority of patients do not have an oncogene, meaning that genetic abnormality where there is a targeted therapy option. So that’s the distinction we make. And I know this term or phrase is used a lot, but that’s what it means. And if you want to know if one has an oncogene-driven lung cancer, you would know based on the genetic test results.

Lisa Hatfield:

Okay. Great. Thank you. And just for clarification too, the genetic mutations are found in the cancer cells, not in their body, the cells? So that’s what the genetic testing is done just on the cancer cells. Is that correct?

Dr. Christina Baik:

Yes. Yes.

Lisa Hatfield:

Okay, great.

Dr. Christina Baik:

Thank you for clarifying that. That’s a very important distinction.

Lisa Hatfield:

Yeah. Thank you. So that leads right into the next question, and it’s kind of a lengthy question. And this person is asking, “Dr. Baik, you have done considerable research around EGFR exon 20 insertion mutations in non-small cell lung cancer, considering their association with poor survival outcomes, what are the survival implications of having EGFR exon 20 insertion mutations compared to other types of EGFR mutations?”

Dr. Christina Baik:

Now this gets a bit complicated, but not all EGFR lung cancers are the same. And there are patients who have what we call EGFR mutation that is a classical mutation. And I throw out that term, because that’s how it’s written on the Internet and a lot of papers. And then, so that’s one group, and the other group are patients who have this exon 20 insertion mutation. And the reason these are separated is because the treatments that work very well in the classical mutations do not work very well in this particular exon 20 mutation.

So when we look at all patients with EGFR mutation, it is true that the prognosis is poor in exon 20 patients just because there are no great targeted therapy options. That said, I am very hopeful that this is changing. There are a number of targeted therapies for exon 20 that are in trials, and I think these are going to be FDA-approved in the future, not too far off in the future, I believe. So I think the survival implications will start to hopefully equalize amongst all the EGFR-mutated lung cancer patients.

Lisa Hatfield:

Okay. Another question, Dr. Baik, from a patient, “For someone who is newly diagnosed with non-small cell lung cancer, what should be the top priorities in understanding and navigating treatment options, especially if I’m in a group that tends to have poorer outcomes?”

Dr. Christina Baik:

So the really, the priorities, when someone is first diagnosed, is really understanding what we’re dealing with and know what they’re dealing with. Now, if you don’t have all this information, I think it’s extremely important to ask. And if you get an answer that indicates that it’s not very clear, and they don’t know, then ask why.

And what are some of the follow-up tests that need to be done so that you can really know. So, I think really the knowledge is extremely important. One thing that I will expand on is that patients, when they’re first diagnosed with non-small cell lung cancer, they should get testing of the cancer, the genetic testing of the cancer to identify the subtype.

And often, it can’t be completed because there’s a lack of biopsy material. So that is often a common situation. So, if that’s a common situation, then ask, “Is there room for another test? Should I get another biopsy? Is there a blood biopsy that will be helpful in this setting?” So really be an advocate for yourself to complete the testing, because the treatment options are drastically different, whether there’s a genetic target in the cancer or not.

The other thing I would say in terms of poor outcomes is that, if you’re told that you’re in a group that has poor outcomes, I think the first question would be to ask why. “Why am I in that group? What makes…what about the cancer that makes it a poor outcome?” And I think this is a setting where you do want to especially get a different opinion potentially just so that the other doctors also agree that you’re in that group. And if so, are there other treatments outside of the standard treatments that you can take advantage of so that it could potentially work better?

Lisa Hatfield:  

Okay, thank you. So this patient mentioned that they are newly diagnosed. And I’m wondering if you can just in really general terms explain what treatment and monitoring might look like. For example, how often might this patient have to come in for treatment? What type of monitoring might be done? I know it has to be really general because every patient is so different. But can you just give a really kind of a high level overview of what that might look like for a newly diagnosed patient?

Dr. Christina Baik:

Yes. So I can speak for patients who have advanced non-small cell lung cancer, which is, unfortunately, still the majority of patients who are newly diagnosed. Most of them have advanced or metastatic lung cancer, meaning that the cancer has gone outside of the original area to other parts of the body. In those settings, if there is a genetic target on the cancer, often the treatment is a pill, which is great. And often, they are fairly tolerable, of course, there are exceptions, but for the most part it is a tolerable type of treatment. So for those patients who have a pill option and they’re tolerating it well, the pills are taken at home, right? They don’t have to come to a hospital for that.

Initially, at least in my personal practice, we do see patients fairly often just so that we can make sure the side effects are being managed and patients are doing well. So initially, it might be every other week type of visits for the first two or three sort of first visits, so the first month or two may be more frequent. But once we get to a point where the treatment is working and side effects are manageable, then I’m essentially seeing them every three months, and they can go. I have patients who travel internationally and just come check with me every three months or so.

So that’s one type of monitoring that we do and in this setting, we would get imaging often with CT scans, every three to four months or so, that number is not exact, there’s no science behind that three to four months. But we would…the point is that we will monitor the cancer using imaging, often with CT scans every three to four months in that setting. Now, and I do want to emphasize that that’s my own personal practice, every doctor does it a little bit differently in terms of intervals. Now, for patients who do not have a targeted therapy option, meaning there’s no pill option, then often patients are receiving some form of infusional treatment. And the infusional treatments vary in terms of the frequency. So some treatments are every three weeks, some are every four, some maybe every six. 

Lisa Hatfield:

Okay, thank you. That’s helpful. So another question, which is a big concern for many patients, I’m sure.  “What signs should I watch for that might indicate the cancer is coming back or progressing? And what steps can I take if I notice any concerning symptoms?”

Dr. Christina Baik:

Right. So the signs are often difficult to identify, mainly because the symptoms can be very similar to kind of normal symptoms, meaning so, for example, one thing we watch out for is back pain, for example, because we worry that the cancer may be going into the bone in the back. However, many of us have back pain chronically, so often it is difficult to tease out what’s what. That said, what I tell my own patients as a rule of thumb is if anything is persisting, so, for example, you have your usual back pain, you know that typically after some rest and some exercise it usually gets better after about a week. The severity may fluctuate a little bit, but most of muscle back pains will change or lighten up after some time if you strained it.

But if there’s a particular pain in a particular part of the body that is just not going away, that’s something to watch out for. Same thing with symptoms like cough, right? So I’ve had many patients who are diagnosed after COVID, where they would have this persistent cough after COVID, but they sort of blamed it on COVID and after a while, since it was not going away they sought medical care. So same thing there, if you had a viral infection, over time the cough should get better.

And if it’s not or if it’s getting worse, then we really have to…really need to investigate the reason for that. So anything that’s persistent, I would say that’s when you want to alert your team. What I tell my own patients is that just tell us, and we’ll figure it out for you or with you. Don’t try to kind of figure it out on your own. I do notice that I do have some patients who tell me that they feel bad about calling and “complaining,” right? And we tell them it’s not complaining, you’re just reporting to us, and we tell you it’s nothing to worry about just watch or something that you need to come in for. So really work closely with your oncology team.

Lisa Hatfield:

Okay, thank you. Well, that’s all the time that we have for today’s Start Here Lung Cancer program. Dr. Baik, you’ve been a phenomenal guest, thank you so much for sharing your experience and expertise with us.

Dr. Christina Baik:

Thank you so much. It’s been great.

Lisa Hatfield:

Yeah, we really appreciate your time. And thank you all for tuning in, it’s these conversations that make a world of difference for patients and their families, particularly when we can hear it from researchers on the ground floor. I’m Lisa Hatfield, and I’ll see you next time.


Share Your Feedback

Create your own user feedback survey

Lung Cancer Patient Expert Q&A: Dr. Christina Baik Resource Guide

Download Resource Guide

_START HERE Lung_Dr. Christina Baik Resource Guide

Download Resource Guide

See More from START HERE Lung Cancer

Lung Cancer Biomarker Disparities | How Precision Medicine and Research Can Help

Lung Cancer Biomarker Disparities | How Precision Medicine and Research Can Help from Patient Empowerment Network on Vimeo.

How can lung cancer research and precision medicine help with biomarker disparities? Experts Dr. Joshua Sabari from NYU Langone and Dr. Eugene Manley from SCHEQ Foundation discuss testing factors that need improvement, patient groups that show disparities, and how clinical trial participation can move research forward.

[ACT]IVATION TIP

“…we really have to more universally test everyone equally to really have an impact on outcomes.”

Download Resource Guide  | Descargar guía de recursos

See More from [ACT]IVATED NSCLC Biomarkers

Related Resources:

Hope Unleashed: Advancing Therapies for Defiant Mutations in Lung Cancer

Hope Unleashed: Advancing Therapies for Defiant Mutations in Lung Cancer

Maximizing Biomarker Equity: Leveraging Partnerships to Close Biomarker Disparities in Lung Cancer

Maximizing Biomarker Equity: Leveraging Partnerships to Close Biomarker Disparities in Lung Cancer

How Can We Leverage Lung Cancer Biomarker Data to Address Health Disparities?

How Can We Leverage Lung Cancer Biomarker Data to Address Health Disparities

Transcript:

Lisa Hatifeld:

So, how can advancements in precision medicine be made more inclusive and equitable to ensure that biomarker-driven treatments benefit diverse populations equally? Second part is, what do you see as the most pressing research priorities in understanding and mitigating these biomarker disparities?

Dr. Joshua Sabari:

So I think first and foremost, testing is key. I mean, educating clinicians, healthcare providers, that every single patient, no matter what clinical characteristic that may be, age, sex, ethnicity, race needs to be tested broadly with the same mutational sort of profile or same biomarker profile. Having somebody in your office who never smoked, those patients generally will have broad panel and next generation sequencing. If you have an 85-year-old patient who is a former heavy smoker, the rate of mutational testing comes down.

So I think we need to remove that bias, that those clinical biases that we have, that we carry with us on a day-to-day basis. We need to test all patients with lung cancer regardless of any clinical characteristic. And what I tell my fellows, my residents, and what I talk to patients about is really all you need is lungs to develop lung cancer.

We need to remove that stigma and when we remove that stigma, we will be testing more broadly in our practices. There are also a lot of systemic biases, a lot of racism that exists, that prevents clinicians, I believe, from doing the best thing for their patients. And if you look at clinical trial enrollment in this country and that’s something that we do need to improve in order to develop better treatment options for our patients, particularly our patients of Latin American descent or Black Americans in the United States.

We need to enroll more patients of more diverse backgrounds onto our trials. Otherwise, we’re only limiting our treatments to specific or small percent of our patient population. So to be honest, I don’t know how well our EGFR inhibitors work in Black patients. I know it’s approved and we utilize it, but we don’t have nearly as much data prospectively treating novel therapies.

A lot of our trials have inclusion rates as low as 2 percent to 3 percent. And we know that our Black patient populations make up 13 percent to 15 percent of our practices. So I think more needs to be done to align our enrollment on trial, I think from institutional policies as well as governmental. So the FDA has really made a forceful statement here to pharmaceutical companies that if your data is not inclusive of a U.S. patient population, this will have ramifications for approvals in the future.

So a lot needs to be done in the sense of education both from the healthcare provider and…but also from the patient, and to really motivate patients to enroll in trials. And one positive that I’ve seen from the patient support groups, the advocacy groups, particularly EGFR Resisters Group, for example, we’ve seen a tremendous sort of push for patients to enroll on trials, again, to benefit themselves as an individual patient diagnosed with EGFR mutant lung cancer, but also to help those who come before or after them in their journey with lung cancer.

Lisa Hatfield:

And, Dr. Manley, do you have anything to add to that?

Dr. Eugene Manley:

I think he hit most of it, but I will say that you have to test everyone because there are people that have risk factors for lung cancer and those that don’t. And like, one of the leading risk factors is history of smoking, but there’s a significant population of specifically Asian females that don’t smoke. Even recently, that have been showing that Black women that don’t smoke also have increased rates of lung cancer. And these are, we don’t know why.

So we still need to be able to test all these patients across all the indications and maybe cross-reference with stress income, socioeconomic status and really try to determine maybe if there are certain specific drivers and what we didn’t talk about. We know that there are some epigenetic changes that may occur due to stress. We also know that there are some changes in tumor mutational burden, some stuff out of MSK. And I think there is some stuff that even shows differences in the immunomarker frequency and response in Black populations. So, we really have to more universally test everyone equally to really have an impact on outcomes.


Share Your Feedback

Create your own user feedback survey

Hope Unleashed: Advancing Therapies for Defiant Mutations in Lung Cancer

Hope Unleashed: Advancing Therapies for Defiant Mutations in Lung Cancer from Patient Empowerment Network on Vimeo.

What promising studies in lung cancer mutations are there that patients should know about? Expert Dr. Joshua Sabari from NYU Langone discusses common lung cancer mutations, incidence rates, promising and potential studies, and proactive patient advice.

[ACT]IVATION TIP

“…no matter what your lung cancer type is, no matter any clinical characteristic, you need next generation sequencing done, biomarker testing done, to identify these mutations. And even when we identify the mutation, I think as a group, as an academic community, we need to do more to study novel therapeutics and to better understand the biology of these mutations so that we can get better treatments to our patients.”

Download Resource Guide  | Descargar guía de recursos

See More from [ACT]IVATED NSCLC Biomarkers

Related Resources:

Maximizing Biomarker Equity: Leveraging Partnerships to Close Biomarker Disparities in Lung Cancer

Maximizing Biomarker Equity: Leveraging Partnerships to Close Biomarker Disparities in Lung Cancer

Lung Cancer Biomarker Disparities | How Precision Medicine and Research Can Help

Lung Cancer Biomarker Disparities | How Precision Medicine and Research Can Help

How Can We Leverage Lung Cancer Biomarker Data to Address Health Disparities?

How Can We Leverage Lung Cancer Biomarker Data to Address Health Disparities

Transcript:

Lisa Hatfield:

Dr. Sabari, can you speak to some of the more defiant mutations in lung cancer, and what promising studies are we looking at right now?

Dr. Joshua Sabari:

Yeah, so I think in lung cancer the most common mutations that we see are EGFR and KRAS. EGFR mutations are a quite broad range of alterations. The most common are in exon 19 deletion and exon 21. These are the location of the mutations. They make up about 80 percent to 85 percent. We have phenomenal treatments in the frontline setting, but most patients only remain on therapy for about two years before there is progression. So, we need to better understand resistance mechanisms, and we need to better understand the next generation of therapies that are available.

In contrast to EGFR, KRAS is equally as common. We see this in about 25 percent to 30 percent of the patient population. Unlike EGFR, KRAS is almost exclusively seen in people who’ve smoked in the past. And there are many different KRAS mutations or alleles. There’s KRAS G12C where we have two FDA-approved match targeted therapies in the second-line setting.

But we need better options, better opportunities for our patients in the frontline setting. And for KRAS, we’re not doing as well as we should, right? KRAS mutations, most people have about a 30 percent to 40 percent chance of responding to therapy. And the median time on treatment is in that six to seven month range. So this is a defiant mutation. It’s a mutation where we need to do better and we need to really develop the next generation of inhibitors for our patients.

So I guess the activation tip here is, again, no matter what your lung cancer type is, no matter any clinical characteristic, you need next generation sequencing done, biomarker testing done, to identify these mutations. And even when we identify the mutation, I think as a group, as an academic community, we need to do more to study novel therapeutics and to better understand the biology of these mutations so that we can get better treatments to our patients.


Share Your Feedback

Create your own user feedback survey

Equity in Action | Addressing Biomarker Disparities in Lung Cancer

Equity in Action: Addressing Biomarker Disparities in Lung Cancer from Patient Empowerment Network on Vimeo.

How can biomarker disparities be reduced in lung cancer patients? Experts Dr. Joshua Sabari from NYU Langone and Dr. Eugene Manley from SCHEQ Foundation discuss approaches that are being used for community engagement and further interventions that can be used to reduce disparities.

Download Resource Guide  | Descargar guía de recursos

See More from [ACT]IVATED NSCLC Biomarkers

Related Resources:

Understanding Non-Small Cell Lung Cancer: Types, Biomarkers, and Treatment Insights

Understanding Non-Small Cell Lung Cancer: Types, Biomarkers, and Treatment Insights

Navigating Lung Cancer Biomarker Testing | Challenges and Solutions for Timely Access

Navigating Lung Cancer Biomarker Testing | Challenges and Solutions for Timely Access

When Should Lung Cancer Patients Receive Biomarker Testing?

When Should Lung Cancer Patients Receive Biomarker Testing?

Transcript:

Lisa Hatfield:

So, Dr. Manley, are there any promising approaches or interventions aimed at reducing biomarker disparities that you’ve currently been exploring or are advocating for?

Dr. Eugene Manley:

I will take several angles on this. One thing is there has to be much more community engagement and involvement and really going to community groups, whether they’re faith-based, whether they’re barbershops, really going out where people are and letting them know about lung cancer, lung cancer disparities, biomarker testing, what you can do. The other way is also going to conferences where there are more diverse scholars that are attending. So a lot of these are STEMM meetings. They may not be specific in lung cancer, but if you can go out there and get the word out about lung cancer and the disparities, then they can go back to their families and talk about, you know, screening and testing and making sure that their family members are aware.

And then, you know, we just published a paper recently that shows the upstream part of biomarker testing is where are we starting at with our cell line? We just did a review of all the lung cancer cell lines. Of over 800 cell lines, majority were European-based. Only 31 cell lines in total were from Black African American populations. None were from Hispanic, none were from Native American, Pacific Islander, none from Alaska Native.

So just think about this. If that is our starting material for all of our biomarker testing and TCGA and databases, then everything we’re developing is on a population that already has great access and outcomes. But they don’t have the greatest disparities. So then you’re getting through doing all these trials, and then you have biomarkers, and you have immunotherapies coming out, and then you’re seeing adverse events in these diverse populations at the end because you don’t have the starting material.

Lisa Hatfield:

And, Dr. Sabari, after hearing Dr. Manley’s comments about that, how do you…or do you know of any approaches or interventions that are aimed at reducing these biomarker disparities? Because maybe they aren’t being acknowledged yet. Maybe they’re only being seen in certain populations.

Dr. Joshua Sabari:

Yeah, I think Dr. Manley hit it on the head. First off, we don’t even know the correct or true numbers for certain mutations in specific patient populations. And I just read an article about patients from Latin America, different rates of EGFR, ALK, and other mutations. You can imagine a study population from Africa, for example. And then obviously studying a population of Black Americans here in the United States as well.

We know that most of the cell lines, most of the data that we’ve had, particularly TCGA  (Tumor Cell Genome Atlas) is from a Caucasian or North European patient population. So I think we need to do better in that sense. I think equally as important, are clinical trial enrollment needs to diversify. Again, it’s mostly women. It’s mostly Caucasian women. We have very, very low rates of Hispanic patients enrolled on clinical trials, Blacks enrolled on clinical trials.

So I think we need to do better in that sense. One thing that we’ve really pushed for in academic medicine is to at least report who is being enrolled on trials so that we can understand is this data generalizable to my own clinical practice? And oftentimes if you look at the clinical characteristics of patients enrolled on the trial, it likely does not match what you see in your own practices.

So we need to do better in that sense. So I think the FDA, and especially pharmaceutical companies, are clearly looking to expand and broaden their inclusion criteria and also access to patients so that we can actually have a more diverse patient population that represents our country enrolled on these trials.


Share Your Feedback

Create your own user feedback survey

When Should Lung Cancer Patients Receive Biomarker Testing?

When Should Lung Cancer Patients Receive Biomarker Testing? from Patient Empowerment Network on Vimeo.

Biomarker testing is vital for non-small cell lung cancer (NSCLC) patients, but when should it happen? Expert Dr. Joshua Sabari from NYU Langone discusses cancer cell mutations and ideal timing for biomarker testing for the best patient care.

Download Resource Guide  | Descargar guía de recursos

See More from [ACT]IVATED NSCLC Biomarkers

Related Resources:

Understanding Non-Small Cell Lung Cancer: Types, Biomarkers, and Treatment Insights

Understanding Non-Small Cell Lung Cancer: Types, Biomarkers, and Treatment Insights

Navigating Lung Cancer Biomarker Testing | Challenges and Solutions for Timely Access

Navigating Lung Cancer Biomarker Testing | Challenges and Solutions for Timely Access

Equity in Action | Addressing Biomarker Disparities in Lung Cancer

Equity in Action | Addressing Biomarker Disparities in Lung Cancer

Transcript:

Lisa Hatfield:

And just from the patient perspective, does a patient need to be tested for these biomarkers throughout the course of their treatment, or is it done initially upon diagnosis or before second-line treatment?

Dr. Joshua Sabari:

Yeah, that’s a great question. You know, most mutations are clonal, meaning that they start in the original cancer cell and then the subsequent cells, daughter or son cells, also have that same mutation. So I would recommend doing next-generation sequencing up front in all patients. Now, some people have a specific mutation that we block with a targeted therapy. It could be pills. It could be an infusional targeted therapy. And that might change the sort of milieu or landscape of that mutational profile. So subsequently, after treatment, you may see acquired resistance or secondary mutations that will prevent the therapies from being effective. In those cases, I do recommend re-profiling.

So the most common example in lung cancer is the EGFR mutation, stands for epidermal growth factor receptor. We know that this mutation occurs in 20 to 25 percent of people diagnosed with non-small cell lung cancer. If you’re matched to a targeted therapy and don’t unfortunately have progression of disease, it may be very helpful to re-biopsy or re-sequence using both tissue and plasma to help us guide subsequent therapy. But if you do not have a targeted mutation and you’re treated with either chemotherapy and immunotherapy or immunotherapy alone, re-biopsy may not be as helpful in matching to further therapy.


Share Your Feedback

Create your own user feedback survey

How Can We Advance Equitable Access to Precision Medicine in Lung Cancer Care?

How Can We Advance Equitable Access to Precision Medicine in Lung Cancer Care? from Patient Empowerment Network on Vimeo.

With non-small cell lung cancer (NSCLC) precision medicine, what are disparities and strategies to equitable access? Expert Dr. Samuel Cykert from UNC School of Medicine discusses disparities, strategies to overcome disparities, and proactive patient advice toward optimal care.

[ACT]IVATION TIP

“…I know you do electronic health records, and as soon as this visit is done, you have data about my visit, so have you thought about creating a real-time registry to see how I’m progressing with my care and see how others are progressing with their care, whether to make sure that we don’t have missed appointments and to make sure that I’m not falling behind where I should be.”

Download Resource Guide | Descargar guía de recursos

See More from [ACT]IVATED Non-Small Cell Lung Cancer

Related Resources:

Enhancing Lung Cancer Care for Black and Latinx Patients | Tackling Challenges, Implementing Solutions

Enhancing Lung Cancer Care for Black and Latinx Patients | Tackling Challenges, Implementing Solutions

Improving Biomarker Testing Access for Rural Lung Cancer Patients

Improving Biomarker Testing Access for Rural Lung Cancer Patients

Empowering Lung Cancer Patients | Embracing Hope, Treatment, and Teamwork

Empowering Lung Cancer Patients | Embracing Hope, Treatment, and Teamwork

Transcript:

Lisa Hatfield:

Dr. Cykert, are there any disparities in access to biomarker testing for Black and Latinx patients with lung cancer compared to other racial or ethnic groups, and if so, what strategies or initiatives can be implemented to address these disparities and promote equitable access to precision medicine?

Dr. Samuel Cykert:

Yeah, biomarker testing followed up by precision medicine is really fairly new in the last half-dozen years, so there haven’t been a lot of studies done looking at how well we’re doing in different groups, but there’s a journal called The Journal of Clinical Oncology and precision medicine that published such a study in 2022.

And what that showed…and again, keep in mind that in a lot of…as they do in a lot of database studies, they are a couple of years behind, but what they showed in looking at the cases of over 20,000 patients, is that on first time testing, we talked about initial biopsies, when the initial biopsy is tested, there is actually about a 7 percent difference between Black and white patients with the white number being only 37 percent and the Black number being 30 percent, so that was low all the way around.

And then if you look at any biomolecular testing at any stage of the cancer, those numbers change to around 55 percent for white patients and 44 percent for Black patients, and I want to point out that for Asian patients and Latinx patients, the numbers were also low, but there weren’t enough patients in the database to achieve statistical significance, but it looks like things are going in the wrong direction there too, and when you think about it, in the state of the right now, those numbers ought to be close to 100 percent for everybody, at least in some of the basic markers like ALK and EGFR and PD-L1.

So there’s a lot of work to do. So there is a disparity. It has been documented, but we’re not getting perfect care to even anyone, and in the ACCURE (Accountability for Cancer Care through Undoing Racism and Equity) Study that I had described a little bit earlier, where we did an intervention, we created real-time transparency through up-to-date electronic health records and digital data of where patients were in their care, we were able to create a real-time registry to know what had been done for every patient, and in the case of precision medicine, this would be so easy, because you basically put every patient that’s had a lung cancer biopsy in the registry, then you have another column in the registry tested for X, tested for Y, tested for Z, and then you have a next column that says, treated for X, treated for Y, and treated for Z. We have the digital information now to do all this in real time, and we have to build the systems to do it.

Lisa Hatfield:

Could you share any examples of successful initiatives or programs aimed at improving the implementation of biomarker testing in lung cancer and what factors contribute to the success of these initiatives, and how can they be replicated or scaled in other healthcare settings?

Dr. Samuel Cykert:

I’ll have to plead my ignorance on this question because I haven’t talked to enough cancer centers on whether or not they’re creating real-time registries for whether all their patients with probable lung cancer are, [a] getting biopsied promptly, [b] getting biomarker testing, and then following those patients over time to see if they’re getting the treatments to match to that, so I know that at my own institution at the University of North Carolina Lineberger Cancer Center, we’re actively talking about building these systems, but we haven’t built them yet.

And so going back to the work that our UNC team has done in partnership with Greensboro Health Disparities Collaborative, we’ve done an intervention with real-time transparency in lung cancer treatment and breast cancer treatment, and real-time quality improvement and audit and feedback for accountability in those treatments and using navigation, particularly for high risk patients to make sure that they’re able to follow through with their diagnosis and treatment.

So with that combination in lung cancer, we got almost perfect care, 96 percent and 95 percent completing treatment, so there’s no reason that the same system cannot be applied to biomarker testing and biologic and immunotherapy, and we need to look at it and implement it and apply it as soon as possible, because when you think about all this, and I’m not just talking about cancer, but when you’re thinking about the whole picture, when you look at, for instance, Black, white disparities, whether it’s in cardiovascular care, whether it’s in diabetes, whether it’s in cancer care, if you look at the result of that in one year, if we brought up care to benchmark levels of the Black community on all those things, we would save 74,000 lives a year.

That’s incredibly impactful. And we need to quicken up the pace of doing this. I’ve been a disparities researcher and intervention researcher for over 20 years, and people really haven’t taken note of really doing interventions until the last five or six years. We need to pay attention, we need to move. It’s important. People’s lives depend on it. And care improved for everyone with these systems, it improved for white patients too. It’s not a zero-sum game.

Lisa Hatfield:

I’m wondering, as a patient, is there anything that I can do or that a patient can do to request or to ask if they use real-time data, that institution to help with the treatment or help with testing or whatever, is there a question the patient might be able to ask to ensure the real-time data is used? Because I imagine it’s not being used as often, so it could be, like you said, there probably isn’t a system in place.

Dr. Samuel Cykert:

Here’s my double activation tip. So at an institution, you don’t know if you have a problem until you look. So the first problem is, as I go back and look behind, am I making sure whether or not I’m seeing disparities, whether it’s a man, woman, Black, white, Latinx, do we have disparities in our treatment application and treatment outcomes in our institution? Because if we look at that, we can start brainstorming on how to possibly fix it, and then the second thing is, I know you do electronic health records, and as soon as this visit is done, you have data about my visit, so have you thought about creating a real-time registry to see how I’m progressing with my care and see how others are progressing with their care, whether to make sure that we don’t have missed appointments and to make sure that I’m not falling behind where I should be.

Lisa Hatfield:

Great, that’s perfect, thank you. Having the patients be…have that accountability too, to ask the question, if that exists, that real-time data, if there’s a way to use that. So thank you, I appreciate that myself personally, so thanks. 


Share Your Feedback

Create your own user feedback survey

Tailored Approaches to Lung Cancer | The Crucial Role of Biomarker Testing

Tailored Approaches to Lung Cancer | The Crucial Role of Biomarker Testing from Patient Empowerment Network on Vimeo.

How does biomarker testing factor into personalized non-small cell lung cancer (NSCLC) treatment? Expert Dr. Samuel Cykert from UNC School of Medicine explains different ways that biomarker testing is used in personalizing treatment approaches and proactive patient advice for biomarker testing.

[ACT]IVATION TIP

“…have access to personalized medicine, whether it’s a surgical biopsy or a radiologic biopsy by a radiologist, you always make the statement. I would like biomarker testing for my biopsy specimen, and I would like to consider the testing that goes along with research protocols too.”

Download Resource Guide | Descargar guía de recursos

See More from [ACT]IVATED Non-Small Cell Lung Cancer

Related Resources:

Catalyzing Lung Cancer Care | The Transformative Impact of Early Biomarker Testing

Catalyzing Lung Cancer Care | The Transformative Impact of Early Biomarker Testing

Closing the Gap | Ensuring Equitable Access to Lung Cancer Biomarker Testing

Closing the Gap | Ensuring Equitable Access to Lung Cancer Biomarker Testing

Unveiling Racial Disparities in Early-Stage Lung Cancer Treatment

Unveiling Racial Disparities in Early-Stage Lung Cancer Treatment

Transcript:

Lisa Hatfield:

How does biomarker testing contribute to the personalized treatment approach for patients with non-small cell lung cancer, particularly in identifying actionable mutations like EGFR, BRAF, and other mutations?

Dr. Samuel Cykert:

Yeah, great, great question. Because some of these biomarkers tell you that there’s a specific treatment that will really, really work for you, and some of the biomarkers tell you there’ll be specific treatments that don’t.

And so the importance of them have to do with, again, you talk about personalized treatment, personalized treatment is getting a treatment that works for you, getting a treatment that works for the genetic component of your tumor, and so it’s really, really important that you differentiate, because again, there are studies that show certain immunotherapy medicines like pembrolizumab (Keytruda) and nivolumab (Opdivo), that those medicines will work in certain situations, but in other situations, they really don’t, and there are other medicines, for instance,  tyrosine-kinase inhibitors that work, where in other situations, they don’t, and so it really is the definition of personalized medicine for lung cancer, knowing what’s going to work and what’s not going to work, and what your odds are in certain situations.

My activation tip is to have access to personalized medicine, whether it’s a surgical biopsy or a radiologic biopsy by a radiologist, you always make the statement. I would like biomarker testing for my biopsy specimen, and I would like to consider the testing that goes along with research protocols too.


Share Your Feedback

Create your own user feedback survey

Advances in Small Cell Lung Cancer Research | Hope for the Future

Advances in Small Cell Lung Cancer Research | Hope for the Future from Patient Empowerment Network on Vimeo.

What new treatments are being studied for small cell lung cancer (SCLC)? Dr. Triparna Sen, a leading researcher in the field, shares promising updates, including advances being made with LSD1 inhibitors, DDR (DNA Damage Response) inhibitors, and DLL-3 targeted therapies.

Dr. Triparna Sen is an associate professor in the department of oncological sciences and co-director of the Lung Cancer PDX Platform at the Icahn School of Medicine at Mount Sinai in New York. Learn more about Dr. Sen.

See More from Thrive Small Cell Lung Cancer

Related Resources:

Essential Small Cell Lung Cancer Testing

Essential Small Cell Lung Cancer Testing 

Expert Advice for Patients With Small Cell Lung Cancer

Expert Advice for Patients With Small Cell Lung Cancer

Understanding Small Cell Lung Cancer Treatment Options

Understanding Small Cell Lung Cancer Treatment Options

Transcript:

Katherine:

Dr. Sen, you are a leading researcher in the field. What is the latest research news that you can share with us about small cell lung cancer?

Dr. Sen:

There’s a lot of great research going on in my lab and labs all across the world. I think for the first time in a very long time, we are really trying to dissect the biology of small cell.   

It has been a research in making for many years. I think we have now really come to a point where we are really trying to understand the disease. I’ll go into a little more about the questions you are trying to answer. So, one of the main questions or one  of the main things that kind of is a hurdle to getting durable treatment options is that the frontline chemotherapy and immunotherapy doesn’t work as well as they should even for the approved regimens, which is the chemotherapy and the immunotherapy.  

The patients often do not have durable benefits. Even if patients have durable benefits, it’s only in a very minority of patient population which means in only about 10 to 15 percent of the total patient population actually do have any benefit from the frontline treatment. So, the main question that we are trying to answer is that why do these patients not respond to immunotherapy and chemotherapy in the frontline.  

What are the mechanisms of resistance to chemotherapy and immunotherapy? Primary resistance, what I mean by primary resistance is that patients who never respond. The disease comes back even while they’re getting the frontline chemo. So, the primary resistance, the mechanisms. Of course, when they have acquired resistance after the maintenance regimen when they come back, why are these patients having this acquired resistance to chemotherapy and immunotherapy? Because only when we understand resistance mechanisms will we be able to then come to the combination strategies.

That’s the next area of research is that once we understand the mechanism of chemotherapy and immunotherapy resistance is then coming up with effective combination therapy. So, what should we combine with immunotherapy in order to make immunotherapy better? I’ll give you an example from the research that we did. 

So, our lab focus is, as I said, on making immunotherapy better. What we understood is that there are certain epigenetic modifiers like LSD1.  

Repressing these, repressing LSD1, with a small molecule inhibitor actually augments or benefits the response to immunotherapy. So now, we are looking at LSD1 inhibitors in combination with immunotherapy. That’s one area that we are focusing on. The second are that we published extensively on is DNA damage response inhibitors which really works in combination with immunotherapy and makes immunotherapy response better.  

Now, we are investigating that in the lab the combination strategies of combining these DNA damage response inhibitors with immunotherapy. So, combination strategies. I think always coming up with novel targets. I will mention there are many novel targets that are right now in the clinical trials actually showing really, really encouraging data.  

I’m talking about DLL3 targeted BiTEs or ADCs we have seen that are showing preliminary data. We have seen a really good really good response in patients. So, finding these targets that are very specific for small cell and that can work in these unique population of patients.  

So, DLL3 targeted agents. There are agents that target B7-H3. So, we are looking at these novel targets and where they could fit in the current therapeutic regimen. Finally, since small cell lung cancer is not a surgical disease, we have to look for other options to find biomarkers. So, liquid biopsy. Liquid biopsy, what I mean by that is understanding the disease not just from tissue but also from blood.  

There’s a lot of research that’s happening in understanding the biology of small cell from blood draws from these patients.  

So, the field of using liquid biopsy or understanding the disease from blood draws is one of the areas that many labs, including ours, are focusing on, and how we can utilize these blood samples to then monitor the disease and also understand the resistance mechanisms to various drugs. I think these are the areas that we are investigating and seems, to me, very important areas that we need to address in order to really manage small cell lung cancer.   

Katherine:

What do these advances mean for small cell lung cancer patients? Are you hopeful?  

Dr. Sen:

Oh, yes. Of course. We’re always hopeful. That’s the goal, right. The goal is to have effective therapies that work and that works for a long time. That also benefits the patients in terms of quality of life which means without very severe adverse effects.   

So, very hopeful. Because I think what was limiting us for all those years for the last 40 to 50 years is that we really did not understand the complexity of small cell lung cancer. It is a very complex disease. It is very different from non-small cell lung cancer which has these mutations that you can target drugs against. So, there are this EGFR mutations and KRAS mutations in non-small cell.  

But small cell, it’s not that. It is not a disease where we have these GATA function mutations that we can devise therapies against. It’s a very different disease. The disease is aggressive. The disease progresses fast, and it also changes its physiology very fast. So, I think for the first time, we really are trying to understand the biology. What that helps is then to come with very informed decisions about therapy.  

So, yeah, I’m very hopeful. Because I think we have now targets that we are actually seeing benefits in patients. I think the more and more we understand resistance mechanisms, we’ll also be able to manage that better.   

Katherine:

That’s very promising news. 

Understanding Small Cell Lung Cancer Treatment Options

Understanding Small Cell Lung Cancer Treatment Options from Patient Empowerment Network on Vimeo.

How is small cell lung cancer (SCLC) treated? Dr. Triparna Sen discusses treatment options for patients with small cell lung cancer, both first-line and second-line therapies, and the important role of clinical trials in patient care. 

Dr. Triparna Sen is an associate professor in the department of oncological sciences and co-director of the Lung Cancer PDX Platform at the Icahn School of Medicine at Mount Sinai in New York. Learn more about Dr. Sen.

See More from Thrive Small Cell Lung Cancer

Related Resources:

Essential Small Cell Lung Cancer Testing

Essential Small Cell Lung Cancer Testing 

Expert Advice for Patients With Small Cell Lung Cancer

Expert Advice for Patients With Small Cell Lung Cancer

Advances in Small Cell Lung Cancer Research | Hope for the Future

Advances in Small Cell Lung Cancer Research | Hope for the Future

Transcript:

Katherine:

How do test results impact care? 

Dr. Sen:

So, you know, once the doctor has confirmed the small cell lung cancer and we have confirmed what stage it is at – what I mean by staging is that it could be either a limited stage disease which is an early stage small cell, or it could be an extensive stage of small cell. The treatment for those two are quite different. So, if it is an early stage or limited stage, patients are usually treated with chemoradiation. If it is an extensive stage or a metastatic small cell, then patients are usually given a standard of care which is chemotherapy in culmination with immunotherapy which is an antibody against PD-L1.  

Katherine:

You’re talking about treatment options that are currently available for small cell lung cancer. What about targeted therapies?  

Dr. Sen:

There aren’t very many therapeutic strategies that are targeted therapies as we speak like we hear from non-small cell lung cancer.  

So currently, like I mentioned, the frontline treatment for small cell lung cancer is with chemotherapy and immunotherapy and maintenance with immunotherapy alone.  

Once the patient relapses, which often is the case – all patients actually have resistance to the frontline chemo-io (chemoimmunotherapy) at some point in time. Once they have a relapse disease, the second line of therapy until now is with either topotecan or irinotecan which are two topoisomerase inhibitors or with lurbinectedin which is in the second line.  

So, when it comes to targeted therapies, so far we a have seen, you know, the conventional way that we think about EGFR inhibitors or KRAS inhibitors, it hasn’t been the case so far with small cell lung cancer. It’s very limited in the current approved setting. But there are many clinical trials that are investigating several targeted therapies that are either targeting – I can speak about that more as I talk about research strategy. But there are many targeted agents that are targeting surface targets like DLL3, B7-H3, or SEZ6. There are other targets that are targeting things like DNA damage repair, proteins, or epigenetic regulators like LSD1. But so far in the approved setting, it is quite limited.  

Katherine:

When we look at what therapies are available, what treatment options are available, what are some typical side effects? How are they managed?  

Dr. Sen:

Some of the major side effects that you see, especially with a frontline chemo-io (chemoimmunotherapy), are very common like you see with other cancer types. Also, it’s usually myelosuppression.  

I think it is prevented or is managed either by dose reduction or treatment delays or treated with transfusion. There has been research that CDK4/6 inhibitors, trilaciclib, when treated with in combination with chemotherapy can bring down the side effects that we see with chemotherapy.  

Some of the immunotherapy related adverse events includes pneumonitis, colitis. They are usually treated with early steroids, treatment withholding, and also it could be leading to permanent discontinuation of the treatment if the adverse events are really severe. Those are mainly what we see we the chemo-io (chemoimmunotherapy) regimen that is given up front.  

Katherine:

Okay. What questions should someone be asking about their proposed treatment plan?  

Dr. Sen:

Right. So, I think, of course, first is what stage. The treatment will depend upon the stage of small cell. Usually, on the frontline, everyone is given chemotherapy and immunotherapy. 

It’s a systemic therapy that’s being given. But I think the patient should be asking questions like are there clinical trials available for me. Because there are multiple clinical trials right now in the frontline and the second line setting.  

So, I think definitely the patient should ask about the clinical trials that the qualify for. In terms of contributing to research, I think if there are options for them to either sign up for blood collection protocol or for tissue collection protocol, I think the patient should definitely enroll for that.   

Because that really helps our research strategy. But in terms of treatment, I think they should ask about available clinical trials that they qualify for.  

Katherine:

Let’s turn to clinical trials then. Patient participation, of course, is essential to finding new and better treatments. What would you say to someone who’s hesitant to participate in a clinical trial?  

Dr. Sen:

Yes. I mean, that’s often the thing. We hear about these novel drugs. They’re in trial. For a disease that’s that aggressive, I think once there is a relapse, I think clinical trials could be a very good option for patients. These are novel drugs that have come out of very robust research that we do in the lab. They can often work quite a bit. So, I think, of course, talk to your physicians. Talk to them at length about whether you do qualify for it. But if there is a trial at the center that you’re getting treated at and if the doctor advises that, I think enrolling in a clinical trial could be a very good option for patients, especially in the aggressive setting where there are not many options available for patients.  

As I mentioned here, research is my true north. I mean, all my lab does is understanding the biology of small cell. It’s extremely essential that we actually try to get the knowledge of the patient tumor. So, if you have availability of contributing either in terms of tissue or blood to research, I think I would advise and encourage patients to definitely contribute to that. 

PODCAST: What Non-Small Cell Lung Cancer Treatment is Right for You?

 

What’s the best approach for YOUR lung cancer? Dr. Isabel Preeshagul discusses the importance of engaging in your lung cancer care decisions, shares advice for working with your team to determine a treatment approach, and reviews factors that affect therapy options. Dr. Preeshagul also provides an update on the latest research and clinical trials.

Dr. Isabel Preeshagul is a thoracic medical oncologist at Memorial Sloan Kettering Cancer Center. Learn more about Dr. Preeshagul.

Download Program Resource Guide

See More From INSIST! Lung Cancer


Transcript:

Katherine Banwell:

Hello and welcome. I’m Katherine Banwell, your host for today’s program. Today, we’ll discuss the latest advances in non-small cell lung cancer care as part of our Insist series, which encourages patients to play an active role and insist on better care. Before we get into the discussion, please remember that this program is not a substitute for seeking medical advice. Please refer to your healthcare team about what might be best for you. Well, let’s meet our guest today. Joining me is Dr. Isabel Preeshagul. Dr. Preeshagul, it’s so good to have you with us. Thank you. Would you introduce yourself? 

Dr. Isabel Preeshagul:

Yes. Thank you so much for having me and for the very kind introduction. My name’s Isabel Preeshagul. I am a Thoracic Medical Oncologist at Memorial Sloan Kettering Cancer Center, and it is a huge honor to be here with you today. 

Katherine Banwell:

Well, we’re so glad to have you with us. I’d like to start with a question pertaining to our series title, Insist. Why is it essential for patients to collaborate with their providers on care treatment decisions? 

Dr. Isabel Preeshagul:

So, collaborating is so important, right? I always tell my patients this is not a dictatorship, right? This is a collaborative effort where I’m here to guide you, but you are the captain of the ship. 

You are the one that needs to make all of the decisions, and I’m here to make sure that the ship goes in a smooth direction, so making sure we have open lines of communication that the patients and their caregivers feel comfortable talking to me and my team and also vice versa and that we trust each other. It’s so important because we are going for a marathon, right? We’re not going for a sprint. This is a long-term relationship, whether we’re treating for cure or we’re treating you with palliative intent and it’s treatable but not curable. We’re going to be following with each other for a long time.  

Katherine Banwell:

A lung cancer healthcare team, of course, consists of a number of different providers. Would you tell us about the various members on a team? 

Dr. Isabel Preeshagul:

Sure. So, there is – there are the people that do the scheduling, that make sure that the CAT scan is scheduled, that the MRI is done, your chemo gets scheduled, all of that. The schedulers are super important and an integral part of our team.  

And then we also have our office coordinators  that answers the phone calls and passes along the messages and assists with scheduling and sort of sets expectations and is the face of the practice. Then you have an office practice nurse or an oncology practice nurse who is the doctor’s right hand, making sure that the patients get proper chemotherapy teaches, making sure that they understand about possible side effects, risks versus benefits, making sure medications are up to date, assessing symptoms.  

They are sort of the front line when it comes to any patient call they’re triaging, and they’re escalating or deescalating. That would be the office practice nurse. And then you have an advanced care practitioner, an APP. You either have a nurse practitioner or a PA that’s working with you that’s sometimes seeing patients independently, sometimes putting chemotherapy orders, you know, really serving as almost as another doctor. 

If for some reason there is something that the doctor’s not available to do, the doctor needs in a pinch, or my patients that are almost at long-term follow-up that are doing great that are just kind of coasting, I will share with my NP and make sure that they know her just as well as they know me. And sometimes there’s a fellow or there’s a resident or there’s a med student that’s part of the team as well because see one, do one, teach one. It’s really important to teach those that are coming after you and serve as mentors and really include them in part of the team and part of the decision-making. And then you have the doctor that just kind of oversees everything.  

Katherine Banwell:

Of course. How would you define treatment goals for people with lung cancer? 

Dr. Isabel Preeshagul:

So, the goal of treatment, I think, is really contingent upon someone’s stage, but it’s also contingent upon what’s important to the patient, right? So, we have patients that are stage I all the way to stage IIIC that we treat with intention to cure.

And patients that have stage IV disease, it’s treatable but not curable. So, I am very transparent with that as long as I have the information to have that discussion. With that being said, there are some patients with stage IIIdisease or stage I disease that don’t really want treatment and want to focus on quality of life. And that’s okay too. And in which case, you know, at some point, their cancer will likely progress. How quickly or when that will happen, we don’t know. Could they pass from something else? It’s possible. But you really need to talk about what’s important to the patient, because it’s not always cut and dry.  

Katherine Banwell:

As you mentioned, Dr. Preeshagul, there are several different support members on a team. What would you say to patients or even care partners who can sometimes feel like they’re bothering their healthcare team with their questions and comments? 

Dr. Isabel Preeshagul:

So, we do get that concern a lot. And I always say, “I’m here for you 24/7. And, if it’s not me, it’s someone that’s just as qualified to answer your questions no matter what.” 

“And I would rather get a phone call at 3:00 a.m. than get a phone call at 9:00 a.m., and you need to go to the hospital right now or God forbid something happened. I get a phone call from someone in the ICU that you went overnight and terrible things happened. So, I want the phone calls to come through to keep you out of the hospital and keep you from going south. So, call me.” And I never try to – I don’t try to outline contingency plans or criteria of what would warrant a call, because then you end up getting in trouble.  

I always just tell my patient, “Think about how you’re feeling now in front of me. If you’re feeling any different than how you feel at this very moment, call me.”  

Katherine Banwell:

Good advice. I’d like to turn to the clinical side of non-small cell lung cancer. What tests help you identify the type and stage of lung cancer?  

Dr. Isabel Preeshagul:

Obviously, you need a CAT scan. You need a CAT scan of the chest, abdomen, pelvis, and you need an MRI brain and a PET scan.  

Those are really the gold standards for determining clinical staging. In regards to pathologic staging, it’s really important to have tissue samplings. So, you biopsy a site of disease that’s concerning to you. If it looks like there’s only disease in the chest, you want to biopsy the site where there’s the tumor, and then you talk with your thoracic surgery or pulmonary team to determine the best way to sample the mediastinum for full staging.  

Katherine Banwell:

Why is an accurate diagnosis so important?  

Dr. Isabel Preeshagul:

So, an accurate diagnosis is so important because lung cancer is by no means black and white anymore. There are so many histologic subtypes that we are learning about. There are so many different molecular drivers that we are learning about. So, making sure you have the right diagnosis, full and next-generation sequencing testing, all of the imaging that you need could really make or break your treatment plan.  

Katherine Banwell:

Dr. Preeshagul, let’s talk about biomarker testing. How is biomarker testing for lung cancer different from hereditary genetic testing?

Dr. Isabel Preeshagul:

So, we do do hereditary genetic testing for lung cancer patients as well. So, I think let’s backtrack a little bit. When you’re doing on a patient, there’s germline mutations and there’s somatic mutations. And germline mutations are mutations that you might get from Mom and Dad that they got from their parents and so on and so forth that you could give to your children or your brother and sister or whatever. So, that’s germline testing that could be passed along.  

That would be like BRCA or any other APC gene, but we are learning more and more that there are mutations in lung cancer that do have a hereditary aspect to them. So, we are learning now that while we do somatic testing, which is to find a mutation that just spontaneously happened in your tumor all on its own, it’s really important to pair that with germline testing to make sure that there isn’t some kind of heritable mutation that’s also driving this lung cancer.  

Katherine Banwell:

You mentioned hereditary genetic testing. Should family members of people with lung cancer undergo genetic testing then just to be reassured? 

Dr. Isabel Preeshagul:

So, if there is a germline mutation, then they should – the family members should be referred to a geneticist to have that discussion.   

Katherine Banwell:

What are common lung cancer biomarkers? 

Dr. Isabel Preeshagul:

So, we have nine biomarkers within approval right now, but there are so many. There’s more than I could even talk about today. But some of the more common ones are EGFR, ALK, ROS1, MET exon 14. You have KRAS, KRAS-G12C, which is a newer one. We have NTRK. We have RET. The list goes on, HER2. I could talk for – there’s not enough time on this Zoom video to talk about all of the mutations. But there are nine mutations with approvals as of now to date, this very moment. That could change tomorrow.  

Katherine Banwell:

Of course, it could. How do biomarkers in lung cancer affect treatment options for lung cancer patients? 

Dr. Isabel Preeshagul:

So, it used to only be in stage IV, but now we are learning that biomarker testing is really important from the get-go because we have induction or neoadjuvant protocols that are looking at giving targeted therapy before patients go to surgery. 

We know that there’s FDA approval for patients to get targeted therapy after surgery, and there’s a survival advantage there. So, make sure that you have next-generation sequencing testing regardless of your stage.  

Katherine Banwell:

Okay. That’s good advice. So, we’ve heard how testing and a patient’s individual disease can lead to more targeted options. And you just mentioned targeted therapies. How do they work? 

Dr. Isabel Preeshagul:

So, there’s many different targeted therapies that we have. Some of given as an infusion. For HER2, for example, we have TDXD, and we have T-DM1. TDXD is the only drug that’s FDA-approved in this setting. There are clinical trials looking at T-DM1. For EGFR Exon 20, we have another infusional drug called amivantamab (Rybrevant). For EGFR Exon 19 and Exon 21, we have a pill called osimertinib (Tagrisso). For KRAS, there’s a pill. For most of the driver alterations, it’s a pill, but some of them it does require infusional therapy. 

But these are therapies that are targeted at the cells that harbor that mutation.  

Katherine Banwell:

Let’s turn to immunotherapy. What is it, and how does it work? 

Dr. Isabel Preeshagul:

So, immunotherapy is basically teaching your body to recognize cancer as foreign. So, when you have – I always kind of use this hand model. So, basically, a normal cell has, let’s say, three prongs. And then sometimes what happens is cancer will grow a marker called PD-L1 that makes it hide from the immune system. So, the body thinks that this is a normal cell. So, what immunotherapy does is it comes up and it sort of puts a cap on that PD-L1 so that the cell looks foreign again and the body can attack that cell and get rid of it. So, it’s almost like ramping up your immune system to recognize that marker and get rid of that cell. 

Katherine Banwell:

What is the regimen for immunotherapy, and how often is treatment administered? 

Dr. Isabel Preeshagul:

So, immunotherapy is approved in the neoadjuvant setting, which means before chemotherapy. It’s approved after chemotherapy, and it’s approved in the stage IV setting. There are many different regimens and many different dosings and many different drugs. But it’s typically given in your veins, either once every three weeks or once every four weeks for a certain amount of time. If it’s given in a curative setting and it’s given indefinitely or until there’s disease progression or intolerance in the stage IV setting.  

Katherine Banwell:

Okay. Let’s touch upon the side effects of these types of treatment. You’ve mentioned that there are so many, but what are some of the major side effects, and how are they managed? 

Dr. Isabel Preeshagul:

Side effects of immunotherapy can include pneumonitis, which is inflammation of the lungs, any kind of endocrinopathy like issues with your thyroid, issues with your pancreas like diabetes.  

It can cause colitis, which is diarrhea, inflammation of the colon, hepatitis, inflammation of the liver. It can cause cerebritis, inflammation of the brain. It can cause arthritis or arthralgias, inflammation of the bones. And it can also cause rash and fatigue. 

Typically, if it’s the thyroid, it’s managed with thyroid replacement hormone or a drug that would calm down the thyroid if it’s overactive. Pneumonitis is steroids. Hepatitis is sometimes treated with steroids. Colitis, steroids typically. Steroids usually come somewhere in there, usually not with the endocrinopathies, but the other itis’s, it’s typically – we start with steroids and go up from there. And the goal is to really recognize these toxicities before they become a problem and just at the glimmer of them just starting.  

Katherine Banwell:

So, would you consider these treatments to be personalized medicine then? 

Dr. Isabel Preeshagul:

So, it’s personalized in the sense that if someone has a high PD-L1 expression, there may be some data to demonstrate that they may benefit from immunotherapy or have a response. If someone can’t tolerate chemotherapy or is not interested in chemotherapy or has other reasons that may preclude them from getting it, it might be reasonable. So, in that sense, it is considered personalized.  

Katherine Banwell:

How would you define personalized medicine? 

Dr. Isabel Preeshagul:

To me, personalized medicine takes into account the biologic makeup of a patient’s disease like if they have a mutation and what their PD-L1 status is, what the histologic makeup of it. What’s their stage? And then, on the other hand, what’s important to that patient? If they’re a tailor, you want to make sure you’re not giving them a medication that’s going to cause neuropathy, so they can’t use their hands.  

If they enjoy playing the harp or the piano, same thing. If their goal is to continue to run marathons, you may want to avoid something that’s going to cause inflammation of the lungs and risk them for pneumonitis. Tailoring to make sure that the treatment is part of their life but does not become their life. 

Katherine Banwell:

If the test results don’t reveal one of the biomarkers you’ve been talking about, what other treatments are available?  

Dr. Isabel Preeshagul:

So, if I don’t have an FDA approval, then sometimes we look to see if there is a clinical trial in our early phase drug development program, and we talk about a clinical trial. If there’s no clinical trial and I don’t have an FDA approval, then we have to talk about what options are considered standard of care and how to make that work into the patient’s lifestyle.  

Katherine Banwell:

What about surgery? When is it used?  

Dr. Isabel Preeshagul:

Surgery is typically used in the curative setting with early-stage disease. We’re really trying to give patients some kind of chemotherapy or some kind of treatment before they go to surgery. It’s shown to improve outcomes. It just gives us a en vivo view of how the tumor will respond to the treatment. So, we typically use surgery in the curative setting. And, at times, it’s appropriate to use surgery for a metastasectomy when you have one little site that’s growing. Sometimes after a tumor board discussion, it might be reasonable to resect that area.  

Katherine Banwell:

Is radiation still used? 

Dr. Isabel Preeshagul:

Same thing. It can be used in the curative setting, typically for patients with stage IIIB or stage IIIC disease and combined with chemotherapy patients that are not considered surgical candidates, or it’s used in the palliative setting when patients have painful metastases. 

Katherine Banwell:

Would you define the B and C? You’ve mentioned that a couple of times.  

Dr. Isabel Preeshagul:

Yeah. 

Katherine Banwell:

We’re used to hearing Stage 1, 2, 3, 4. But what’s a stage IIIB and a stage IIIC? 

Dr. Isabel Preeshagul:

Yeah. Sure. Sure. So, it does get a little bit into the weeds here about the size of the tumor and the amount of lymph nodes and location of the lymph nodes. But basically, stage IIIA is considered resectable. That means – that could be the size of the tumor with no lymph nodes, or it could be a smaller tumor with a lymph node on the same side as the disease. Stage IIIB would be a lymph node right underneath the windpipe at the station 7. And stage IIIB also includes lymph nodes that have crossed over to the contralateral side. And stage IIIC would be lymph nodes that are maybe up at the contralateral supraclavicular space. 

Katherine Banwell:

Okay. Do treatment options change if the lung cancer returns? 

Dr. Isabel Preeshagul:

Yes, they do change depending on if this is the same tumor type that’s come back. It’s typically a different treatment algorithm, yeah.   

Katherine Banwell:

Okay. And should biomarker testing be done again if a relapse occurs? 

Dr. Isabel Preeshagul:

100 percent. Because it guides everything about a patient’s treatment. It’s super important.  

Katherine Banwell:

Okay. What are you excited about right now in lung cancer research? 

Dr. Isabel Preeshagul:

I am excited and overwhelmed by the fact that we have so many approvals and so much exciting data that was just presented at ASCO and World Lung and ESMO that it’s next to impossible to keep up. And I’m happy that we have that problem, and I’m happy that the patients have – there’s a spotlight on lung cancer when we were in the shadows. And now, I think we have the spotlight. 

And all of these approvals, you know, with it being Lung Cancer Awareness Month as well, I think is just so important. Just to make sure that we get the knowledge of these new approvals out there though, that is another struggle. 

Katherine Banwell:

Well, are there any current clinical trials that look promising to you? 

Dr. Isabel Preeshagul:

Yeah, I think there are many clinical trials. In the induction setting, there was some data that was just presented on ALINA looking at adjuvant alectinib (Alecensa). We just had a – we have approval for adjuvant osimertinib (Tagrisso) and the ADAURA trial.  

But we are learning more and more that as these targeted therapies have approval in stage IV, we’re trialing them in stage III, and then we’re going to trial them in earlier stages and earlier settings. So, this has been the pattern of how drugs get approved. So, yes, there’s lots of exciting data coming through. 

Katherine Banwell:

That’s excellent. Can you talk about antibody drug conjugates and where they fit into lung cancer care? 

Dr. Isabel Preeshagul:

Yeah. That’s a great question. I don’t think anyone knows the answer as to where they fit in just yet. 

We have probably over 300 antibody drug conjugates that are in development right now. And one of the more common ones that we use is trastuzumab deruxtecan (Enhertu), or TDXD, which is used in patients that harbor HER2 alterations in the stage IV lung cancer setting. It is basically almost like a Trojan horse. So, you have this antibody.  

It’s typically IgG1, immunoglobulin. And then you have a linker, and then at the end of that linker is the warhead or the chemotherapy agent. So, the antibody comes in towards the cancer cell looking very innocent. It binds to the cancer cell. And, once it binds, then everyone inside the Trojan horse or this warhead rush into the cell and get to do its damage. So, it’s a totally different mechanism. We’re trying to outsmart some of the bypass mechanisms that cancer cells develop. And this may be the new wave, but stay tuned, more to come.  

Katherine Banwell:

Right. So, it’s promising.  How can patients find out more about current clinical trials? 

Dr. Isabel Preeshagul:

So, you can always ask your healthcare practitioner if there are any clinical trials at the institution that you’re at, but clinicaltrials.gov has all the clinical trials that are available nationally and internationally.  

You just type in your disease type. You can type in a couple keywords, EGFR maybe or ROS1 or stage IV, something along those lines, and then it should populate a list of clinical trials and what institutions have them open, if they’re still accruing or if they’re not, and a contact on that trial.  

Katherine Banwell:

If a patient is interested in a clinical trial, what kinds of questions should they be asking their healthcare about the trial? 

Dr. Isabel Preeshagul:

So, the first question to ask is, “Do we have any clinical trials that are appropriate for me?” If the answer is yes, “Are they appropriate for me now, or are they appropriate for me if what I’m on right now is not working?” 

So, trying to figure out where that will be, and if they are appropriate for you now, how can I get evaluated, and how can we get things underway? 

Katherine Banwell:

Yeah. What would you say to patients who are interested in participating in a clinical trial, but they’re nervous about it?

Dr. Isabel Preeshagul:

I think one thing that I love about being on a clinical trial is that there are more eyes are on you, because we are looking to get something approved, and we are just watching every single little granular detail. In a way, it’s almost like you’re being more micromanaged than if you were on standard of care because of just how many stops and checks there are, how many eyes are looking at your labs after the doctor and the nurse and the nurse practitioner, and the fellow take a look at everything. It’s 10 other people. So, it’s almost like it’s extra safe because of all of that. It’s exciting because you are hopefully getting tomorrow’s treatment today, right? 

You’re trailblazing the way for other people after you. So, I think it’s exciting, but, of course, it’s nerve-wracking. It’s something new. You don’t know if it’s going to work. But I have to believe that the way that clinical trials are designed now and the clinical trials that we choose to open here, we really hope are going to be pushing the space forward. 

Katherine Banwell:

Yeah. I’d like to get to a few questions that we received from audience members prior to the program. How do you help a family member that is an overwhelmed caregiver but refuses help? Any tips on how to provide support to this person?  

Dr. Isabel Preeshagul:

I mean, I think we see caregiver burnout thousands of times a day, unfortunately, and the first thing is knowing how to recognize it. And the second most important thing is taking the time away from the visit with the patient to address the burnt-out caregiver, because there is not enough time in any visit to ever – there’s never enough time in my mind to spend with a patient.  

I’m always pulled in a thousand different directions. And I think we all feel that. But taking the appropriate time to sit down and to say, “Hey. Listen. I recognize that you’re burnt out. I can see it. Who is in your corner helping you?” And just directing focus away from the patient just for a moment and to really focus on that caregiver and to rely on the social work team and the case manager and the support groups that your institution may have and to make sure that they know about those resources. 

Katherine Banwell:

Yeah. Here’s another question we received. “Can you share more information regarding treatments available for stage IV lung cancer and their side effects?” 

Dr. Isabel Preeshagul:

It depends on if this is non-small cell or small cell. It depends on if you have a driver alteration or not. So, I think that is a little bit challenging to talk about in just one session. But basically, you’re probably looking at some kind of targeted therapy if you have a mutation versus standard of care if you don’t have a targeted mutation versus a clinical trial. And I think those are kind of like the big baskets.  

Katherine Banwell:

When is a second opinion necessary? Dr. Isabel Preeshagul: A second opinion is necessary anytime you want a second opinion.  

Dr. Isabel Preeshagul:

There is no right or wrong time, any time. You’re just not jiving with your oncologist after the first day you met them, second opinion. You’re at the end of the line and you really want toknow more, second opinion. You’ve met two other doctors. You’re not jiving, third opinion. It’s always appropriate anytime you want. 

Katherine Banwell:

So, the patient shouldn’t feel obligated to stay with that one provider? 

Dr. Isabel Preeshagul:

Never. Never, never, never, never, never. No. Please don’t feel that way. There are no hard feelings. And, if there are, that’s not the right oncologist for you. It needs to feel like a perfect friendship. And, if it’s not that, it’s not the right thing.    

Katherine Banwell:

Before we close, Dr. Preeshagul, I’d like to get your final thoughts. What would you say to the audience about the future of lung cancer care and treatment? 

Dr. Isabel Preeshagul:

I do think that the future is bright because, as I mentioned, there is now this light that is shining in the lung cancer space. And things are getting approved. and discoveries are getting made faster than we can even keep up, which is exciting and overwhelming and daunting. But I am happy that, finally, this space is taking off, so I feel optimistic.  

Katherine Banwell:

Okay. All right. Well, I wanna thank you so much for taking the time to join us today, Dr. Preeshagul.  

Dr. Isabel Preeshagul:

Thank you so much for having me. These were wonderful questions, and I look forward to many more discussions with you. Thank you.  

Katherine Banwell:

And thank you to all of our partners. To learn more about lung cancer and to access tools to help you become a proactive patient, visit powerfulpatients.org. I’m Katherine Banwell. Thanks for being with us today.   

What Essential Testing Reveals About Your Non-Small Cell Lung Cancer

What Essential Testing Reveals About Your Non-Small Cell Lung Cancer from Patient Empowerment Network on Vimeo.

What do lung cancer test results reveal to your healthcare team about your disease? Dr. Isabel Preeshagul provides an overview of essential testing for lung cancer and explains the difference between germline and somatic mutations.

Dr. Isabel Preeshagul is a thoracic medical oncologist at Memorial Sloan Kettering Cancer Center. Learn more about Dr. Preeshagul.

See More From INSIST! Lung Cancer

Related Resources:

Insist on Better Lung Cancer Care | Tips for Essential Communication

Non-Small Cell Lung Cancer Treatment Options | Personalizing Therapy

Non-Small Cell Lung Cancer Treatment Options | Personalizing Therapy

Non-Small Cell Lung Cancer Treatment | Clinical Trials and Research Updates

Non-Small Cell Lung Cancer Treatment | Clinical Trials and Research Updates


Transcript:

Katherine Banwell:

I’d like to turn to the clinical side of non-small cell lung cancer. What tests help you identify the type and stage of lung cancer? 

Dr. Isabel Preeshagul:

Obviously, you need a CAT scan. You need a CAT scan of the chest, abdomen, pelvis, and you need an MRI brain and a PET scan.  

Those are really the gold standards for determining clinical staging. In regards to pathologic staging, it’s really important to have tissue samplings. So, you biopsy a site of disease that’s concerning to you. If it looks like there’s only disease in the chest, you want to biopsy the site where there’s the tumor, and then you talk with your thoracic surgery or pulmonary team to determine the best way to sample the mediastinum for full staging.  

Katherine Banwell:

Why is an accurate diagnosis so important? 

Dr. Isabel Preeshagul:

So, an accurate diagnosis is so important, because lung cancer is by no means black and white anymore. There are so many histologic subtypes that we are learning about. There are so many different molecular drivers that we are learning about. So, making sure you have the right diagnosis, full and next-generation sequencing testing, all of the imaging that you need could really make or break your treatment plan.  

Katherine Banwell:

Dr. Preeshagul, let’s talk about biomarker testing. How is biomarker testing for lung cancer different from hereditary genetic testing? 

Dr. Isabel Preeshagul:

So, we do do hereditary genetic testing for lung cancer patients as well. So, I think let’s backtrack a little bit. When you’re doing on a patient, there are germline mutations and there are somatic mutations. And germline mutations are mutations that you might get from Mom and Dad that they got from their parents and so on and so forth that you could give to your children or your brother and sister or whatever. So, that’s germline testing that could be passed along.  

That would be like BRCA or any other APC gene, but we are learning more and more that there are mutations in lung cancer that do have a hereditary aspect to them. So, we are learning now that while we do somatic testing, which is to find a mutation that just spontaneously happened in your tumor all on its own, it’s really important to pair that with germline testing to make sure that there isn’t some kind of heritable mutation that’s also driving this lung cancer.  

Katherine Banwell:

You mentioned hereditary genetic testing. Should family members of people with lung cancer undergo genetic testing then just to be reassured? 

Dr. Isabel Preeshagul:

So, if there is a germline mutation, then they should – the family members should be referred to a geneticist to have that discussion.  

Katherine Banwell:

What are common lung cancer biomarkers? 

Dr. Isabel Preeshagul:

So, we have nine biomarkers within approval right now, but there are so many. There’s more than I could even talk about today. But some of the more common ones are EGFR, ALK, ROS1, MET exon 14. You have KRAS, KRAS-G12C, which is a newer one. We have NTRK. We have RET. The list goes on, HER2. I could talk for – there’s not enough time on this Zoom video to talk about all of the mutations. But there are nine mutations with approvals as of now to date, this very moment. That could change tomorrow.