Tag Archive for: residual disease

HCP Roundtable: Exploring CLL Mutations and Best Practices for Side Effect Management

HCP Roundtable: Exploring CLL Mutations and Best Practices for Side Effect Management from Patient Empowerment Network on Vimeo.

As the chronic lymphocytic leukemia (CLL) treatment landscape evolves, how can healthcare professionals deepen their understanding of mutation profiles, including the emergence of novel CLL mutations over time? What innovative approaches are transforming the management of CLL side effects? Additionally, how can barriers in CLL practice be removed to enhance physician-patient communication and promote shared decision-making? 

Dr. Jennifer Brown from the Dana-Farber Cancer Institute and Dr. Callie Coombs from the University of California, Irvine, share their expertise and best practices for CLL healthcare providers.

Download Resource Guide  | Descargar guía de recursos

See More from EPEP CLL

Related Resources:

Aicha Diallo

Peer Insights: Understanding Cultural Competence vs. Cultural Humility

Collaborate | Understanding Your Role in Your CLL Care

Collaborate | Understanding Your Role in Your CLL Care

Aicha Diallo

Peer Insights: Practicing Cultural Humility to Empower Your Patients

Transcript:

Dr. Nicole Rochester:

Welcome to this Empowering Providers to Empower Patients or EPEP Program. I’m Dr. Nicole Rochester, founder and CEO of Your GPS Doc. EPEP is a patient empowerment network program that serves as a secure space for healthcare providers to learn techniques for improving physician-patient communication and overcome practice barriers.

In this CLL roundtable, we are tackling exploring CLL mutations and best practices for side effect management. As the chronic lymphocytic leukemia treatment landscape evolves, how do CLL healthcare providers better understand mutation profiles, including the emergence of novel CLL mutations over time? What groundbreaking CLL therapeutic targets are emerging, tailored specifically to molecularly defined patient subgroups? And what innovative approaches are transforming CLL side effect management? These are just some of the things that we’re going to discuss today. We’re going to talk about the complexities of CLL mutations and the clonal evolution and resistance mechanisms in CLL.

We’ll discuss clinical trials and novel targets focused on molecularly defined patient subgroups. And lastly, we’ll talk about strategies for healthcare provider to healthcare provider communication regarding the management of side effects.It’s my privilege to be joined by Dr. Jennifer Brown, Director of the CLL Center of the Division of Hematologic Malignancies at Dana-Farber Cancer Institute, and the Worthington and Margaret Collette Professor of Medicine in the field of Hematologic Oncology at Harvard Medical School. Thank you so much for joining us, Dr. Brown.

Dr. Jennifer Brown:

My pleasure. Thank you for having me.

Dr. Nicole Rochester: 

It’s also my privilege to be joined by Dr. Callie Coombs, an Associate Clinical Professor at the University of California, Irvine. Dr. Coombs primary clinical focus is in the care of patients with chronic lymphocytic leukemia and small lymphocytic lymphoma. She has participated in multicenter studies examining the real world implications of novel therapeutic agents on the lives of patients, and has served as an investigator on a number of clinical trials. Thank you so much for joining us, Dr. Coombs.

Dr. Callie Coombs:

Thank you for having me as well.

Dr. Nicole Rochester:

So let’s jump in as we have a lot to discuss as it relates to understanding CLL mutations and best practices for side effect management in CLL. So we’re going to start with the complexities of CLL mutations. And the first question, I’ll start with you, Dr. Brown, how do CLL healthcare providers better understand mutation profiles including the emergence of novel CLL mutations over time?

Dr. Jennifer Brown:

Well, the first thing that’s important to recognize is that CLL is not defined by any particular mutation. The landscape is quite varied and we see a large number of different mutations at low percentages. Well, the second key point to remember is that there are different mutations at baseline and then there can be acquired mutations that include some of what we see at baseline, but also novel resistance mutations that we don’t ever see at base.

So at baseline, the most common mutations, which are somewhere in the 10 to 20 percent range of patients, although less than that if you have very early stage patients, affect the p53 gene, NOTCH1, SF3B1, and ATM. P53 is the most important because that one does influence our thinking about the patients and our choice of therapy in some cases. P53 can be altered in CLL in two different ways. Actually, the most common way is as a deletion, deletion of the short arm of chromosome 17 or 17P deletion. About 75 to 80 percent of patients that have that deletion will have a point mutation usually in the other p53 allele. So they have double knockout of p53.

A small percentage of people with the deletion will not have the mutation. And then a certain number of patients will have just the mutation without the deletion. And one of the things that I’ve been very interested in for a while that we’re still trying to understand better is the implications of these different combinations of the way p53 can be affected in people with CLL, and that it may, in fact, be more adverse to have both alleles knocked out than single, although we don’t have great data for that as yet because most of the data that we have has combined all of it together.

But it’s very important to test for the p53 mutation alone because even if patients have only that one, at present, we consider the treatment implications of it all similarly regardless of how the p53 gene is affected. And then NOTCH1 is a fairly common mutation that always worries us a lot, because it’s associated with Richter’s transformation, which is a very high-risk event, but we don’t know anything to do about that to try and prevent it or to alter our therapy based on it.

So at the moment it’s mostly something that we are aware of that we keep an eye on but not that changes therapy. And SF3B1, ATM, and this long list of other genes that can be mutated in just a few percent of CLL, and mostly what we know about them is some biology that’s been studied, and then the fact that the more of these mutations are mutated in a patient that is associated with a worse prognosis, just a total number.

But that’s not something also that really alters our therapy. And then when patients go through lines of therapy, they can sometimes acquire mutations in these genes. So a patient can acquire a mutation in p53 or in NOTCH after their second or third line of therapy. But the mutations that are hottest right now, or that people are most interested in are some of the mutations that occur as resistance to therapy. So in particular, that means BTK mutations.

Covalent BTK inhibitors have transformed the therapy of CLL, and they bind to the cysteine 481 residue of BTK. So that means, as you might imagine, that if you mutate that cysteine so that the inhibitor can’t bind, that will be associated with resistance. And that, in fact, is what has been found that the cysteine to serine mutation at 481 is the most common resistance mutation in patients on covalent BTK inhibitors.

And in the case of ibrutinib (Imbruvica), it makes the inhibitor into a much weaker and non-covalent inhibitor. In the case of acalabrutinib (Calquence) and zanubrutinib (Brukinsa), it probably abrogates all activity. And so that’s a mutation that we will sometimes look for in patients with clinical progression on those drugs. There’s also a mutation in BCL2 that can occur in patients in venetoclax (Venclexta).

So another example of an on target resistance mutation. The role of that one is a little bit less clear, and testing for it is not as widely available, but we’re still working on that. Resistance to venetoclax is probably more complicated than resistance to BTK inhibitors, although there’s also a subset of patients who will get BTK inhibitors who have novel mechanisms of resistance not related to BTK that we don’t really know anything about as yet.

And then finally, the non-covalent BTK inhibitors are becoming available, pirtobrutinib (Jaypirca) was approved for CLL in the United States in December for patients who’ve had covalent BTK inhibitors and venetoclax. And we’re starting to see different mutations in BTK at different sites, even though pirtobrutinib has activity against the 481 mutation. So there’s going to be a lot of activity in this area in the next few years probably.

Dr. Nicole Rochester:

Thank you so much, Dr. Brown, that was a very comprehensive overview of the mutations. Dr. Coombs, do you have anything that you want to add to what Dr. Brown said perhaps specifically around mutations associated with the progression of CLL?

Dr. Callie Coombs:

Sure. So, that’s a hard act to follow. She really took us through a whirlwind of everything mutation-related. I think what I would like to focus on in my answer is, well, what should we be testing for on a day-to-day basis in our CLL practices and what are some common misconceptions? So specific to TP53, I would say this is the most important test as far as all of the genetic tests that influences what we do day to day in the care of patients with CLL.

I test for this for my newly diagnosed patients who I think may be interested in enrolling in a clinical trial, first of all, so the standard of care in CLL is watch and wait, however, patients with higher risk disease may be eligible for trials looking at early intervention specifically the SWOG EVOLVE trial looking at early treatment. And so that’s one of the risk markers that can get a patient into the higher risk category of CLL where they could be eligible for a trial.

A common misconception I see is that 17p is the same thing as a TP53 mutation, it’s definitely not. So these are two different tests that have to be sent. 17p can be picked up on karyotype testing and on FISH testing where it looks for 17p deletion. However, mutations are a different test. And so I usually send a next gen sequencing assay that includes other genes.

However, you can test purely just for mutations in the TP53 gene, but again, that’s a sequencing test, so I’d like to convey that, somewhat a misunderstanding, but it’s such an important gene in CLL because when patients have TP53 aberrations, whether that’s 17p or a TP53 mutation or both, given that they can occur in isolation or together, these patients should never get chemotherapy, because they have extremely terrible responses to chemo, and that should not be part of the therapies offered to these patients.

The other interesting, I’d say controversy at least in 2024, is what is the role for mutation testing in the clinic in the setting of acquired resistance to inhibitors? So I think it’s very clearly important in the research setting where I think learning about the C481 mutation among others in the setting of covalent BTK inhibitors has shown us a lot about mechanism of resistance. But in the clinic, I don’t necessarily think that’s something that needs to be universally applied, given that it most of the time doesn’t affect what we would do clinically.

And so one example is a patient comes in progressing on ibrutinib, maybe about two-thirds of them may have a mutation in the C481S. However, if they’re clinically progressing, they need to switch therapy. And so I think an argument could be made in practice whether or not sending these mutation tests is beneficial, but research, clearly important, and I think it’s going to give us key insights into our therapeutic sequencing strategies going forward. So I’m certainly a proponent of doing the testing in a well-monitored setting, but I don’t think it’s ready for prime time to be applied completely broadly to our patients.

Dr. Nicole Rochester:

Thank you, Dr. Coombs, and I appreciate you adding that additional practical tips and information specifically for our healthcare providers. And you kind of moved into the next topic, which was really around new diagnostic tools and technologies that are available to detect and monitor mutations. So I’m going to go back to you, Dr. Brown, to see if you have any additional information that you’d like to share about new diagnostic tools, technologies with regard to these mutations and any other tips perhaps for our healthcare provider audience.

Dr. Jennifer Brown:

Well,  really the only issue is what Dr. Coombs mentioned that it’s very important to get a next generation sequencing test to evaluate the p53 mutation, that it really is not well-evaluated by any other test, and is often missed because it’s thought that checking for the deletion is sufficient. So I would just reemphasize that point that she made very clearly. Other than that, we don’t really need any additional tools to monitor for mutations.

In the research setting we’re trying to do more and more sensitive assays to try and see when the earliest time that these mutations may emerge is and is there a way we could prevent that or, and just to better understand some of the biology, but it’s not really anything that’s needed in clinical practice. And we’re also not using the mutations to monitor residual disease. It turns out that the best way to do that is probably looking at the B-cell receptor itself, which is again, something that we’re studying in the research setting, but is not really something that needs to be done in clinical practices yet.

Dr. Nicole Rochester:

Wonderful. Thank you, Dr. Brown. We definitely want to leverage you all’s expertise in this area. And so my next question has to do with practices. And you’ve really kind of addressed this to some extent already. Are there any unforeseen or perhaps outdated practice-related barriers that may either hinder your work or that of your colleagues specifically related to better understanding CLL mutations?

Dr. Callie Coombs:

Yeah, I mean, I think in addition to what I mentioned about 17p and TP53, one type of mutation we haven’t talked about is assessing for the mutation status of IGHV.  So that’s actually something else that I’ve seen frequently missed as far as the routine testing of a CLL patient. But I do think it’s very important to send. Is it as important as when we were in the chemoimmunotherapy era where it would be hugely predictive for who had a long remission and who wouldn’t? Maybe not as important, but I do think if someone’s unmutated that still can really help inform certain aspects of their journey. One is the time that between diagnosis and when he or she’ll need their first treatment.

But two, also the expected length of remission should this patient embark upon a time-limited regimen such as venetoclax and obinutuzumab (Gazyva). But the separate question is, again, coming down to the practical aspect of how IGVH is tested. So another misunderstanding that I’ve seen is FISH tests look for the IGH locus. And so I’ve seen on recurrent occasions if that’s deleted, they say, “Oh, that’s a mutation.” Well that’s definitely not the same thing, and so it’s just to realize the IGHV test is a very specific test.

Some large facilities do it as an in-house test, I myself have been sending mine out to the Mayo Clinic, there’s other vendors where you can do it, but what they do is they specifically sequence IGHV and then compare the patient sequence to a consensus germline sequence to determine the percent of mutation, and it’s actually a good thing to be mutated with this gene, these are the patients that often have a longer time until they need their first treatment, if they need treatment at all, and then they generally have better responses to therapy. Though with BTK inhibitors, that difference is often becoming quite slim given that they work in both groups of patients.

Dr. Nicole Rochester:

Wonderful. Thank you so much, Dr. Coombs. So now we’re going to shift to talking about clinical trials and novel targets focused on molecularly defined patient subgroups. So, Dr. Brown, can you talk about any emerging CLL trials targeting specific molecular subgroups, and also how can CLL experts stay updated on these advancements in clinical trials?

Dr. Jennifer Brown:

So, as you heard from Dr. Coombs, there’s increasing interest in looking at high-risk patients in particular, and I think looking specifically at patients with p53 aberration in dedicated clinical trials, it’s become increasingly clear that the behavior of the disease when it’s higher risk based on p53 mutation, NOTCH mutation, IGHV status is quite different, particularly with time limited therapy compared to lower risk disease.

And so having dedicated trials that evaluate outcomes specifically in certain of these subgroups is increasingly important. We do have more trials than we used to focusing specifically on p53 aberration. My personal belief is that we would be well served to have trials separately in the IGHV groups that Dr. Coombs mentioned, although that has not gained as much traction.

And then what we are seeing is now that there are resistance mutations, it actually has turned out that some of the drugs that we use in that setting, venetoclax and pirtobrutinib, seem to have pretty similar activity in patients with and without the mutations. But as drugs are being studied in this context, there’s been an increasing tendency to study them in specific subgroups.

So patients who have the mutation and had clinical progression on a covalent inhibitor, patients who don’t have the mutation and had clinical progression, patients who may have come off their covalent inhibitor for adverse events who may not actually be resistant, what is their response to the next line of therapy? And so all of that is just helping us understand in a more nuanced way what the best benefit for patients will be as we look at these different subgroups of patients.

Dr. Nicole Rochester:

Thank you, Dr. Brown. Appreciate that. Dr. Coombs, do you have anything to add?

Dr. Callie Coombs:

Yeah, so I echo all of Dr. Brown’s comments, and I think I’m the person that is bringing all the practical aspects of CLL care because it’s, she’s so thorough. I just always like to contribute a few little pearls. So, pirtobrutinib has been an exciting drug, to see it become available for our double refractory patients. So the current FDA indication is for patients failed by not only a covalent BTKi but also venetoclax. But it’s the first BTK inhibitor that we can effectively use in the setting of a prior BTK inhibitor.

And that’s because of this unique aspect where instead of forming a covalent bond at the C481 residue, it binds reversibly, and we can still see activity. But the practical aspect is that that’s not an effective strategy when you have a patient progressing on, say, ibrutinib, you can’t switch them to acalabrutinib (Calquence) or zanubrutinib (Brukinsa) because of their shared mechanism of resistance. They’re all covalent inhibitors. They all share the same mechanism of resistance.

And so that’s one thing I’d like to bring up. However, there’s a very different and very common clinical situation that I encounter really a lot in my clinic, which is intolerance. And so that’s where it would be a very effective strategy to switch a patient from one covalent drug to another. And so literally in the past couple weeks of clinic, I’ve had patients with chronic long-standing toxicities to ibrutinib that perhaps went underrecognized where I say, “Hey, you’ve had…noticed your blood pressure has gone up a lot. Let’s switch you over to acalabrutinib,” or other patients, “Oh, you’ve had issues with atrial fibrillation…let’s try switching you to zanubrutinib.” Because the rates are a lot lower and a lot of patients can have improvement or just complete resolution of the prior side effect.

And so I hope that that emphasizes this is something that we think about every day, and switching is appropriate in the setting of intolerance. It’s not appropriate when you’re staying in the covalent class to switch in the setting of progression. But pirtobrutinib being a non-covalent inhibitor is certainly very effective after a covalent. And I think once we see readout of some of the ongoing Phase III trials, we may be able to use it in that setting under an approved FDA label, though that is to be seen in the future.

Dr. Nicole Rochester:

Awesome. Thank you. Thank you to both of you. And that leads us very nicely into our next topic. And so we’ve been talking about improving CLL treatment efficacy, we’ve talked about mutations, we’ve talked about really providing better outcomes for our patients by using therapies that are very specifically designed for the molecular characteristics of their disease. But along with all those therapies, of course, come potential side effects. And so, Dr. Coombs, I’m going to start with you and then we’ll go to Dr. Brown. Are there any strategies that you can share with our healthcare provider audience around innovative approaches or protocols that have been implemented to mitigate and manage the CLL side effects from the treatment?

Dr. Callie Coombs:

Well, I think it comes down to your internal resources, but I would say taking care of CLL patients is clearly a team effort. And so it’s not just me, but also a team of additional practitioners that I work with. So I’d like to emphasize how important pharmacists are because I’ve definitely seen some side effects that come about because a patient is now on a medication that interacts with whatever their CLL therapy is, which drives up the levels of the drug and then brings out certain toxicities so they can help us identify these.

If, perhaps I missed it or didn’t ask the patient about a supplement, et cetera. Next is nurse practitioners and oncology nurses. And so number one is it’s a team-based approach, and I think it’s certainly very important to have protocols internally. But also to just realize what the common toxicities are and how can we mitigate these.

One of the most common reasons that I’ve seen for patients stopping a drug prematurely actually is venetoclax. It very commonly causes neutropenia. And I’ve seen the drug given up on very early without any growth factor support, and so I think if you become educated and experienced with using drugs, you can realize there’s very clear strategies in improving patients with neutropenia, by supporting them with growth factor and getting them through whatever their defined plan course of venetoclax may be.

And then BTK inhibitors have a whole smattering of side effects as well where perhaps working with cardio oncologists can help in addition to other strategies depending on exactly what side effect the patient may encounter. So in summary, definitely a team-based effort and growing experience with the common side effects helps I think all comers with strategies to help prevent or mitigate such side effects.

Dr. Nicole Rochester:

Thank you so much, Dr. Coombs. Dr. Brown, do you have some additional best practices you’d like to share with regard to the management of treatment side effects?

Dr. Jennifer Brown:

Well, I agree completely with Dr. Coombs. I would just add that I think it helps a lot when you warn the patients ahead of time about things that may happen but that often go away or that you can manage. So, for example, headaches often happen early on when you initiate acalabrutinib but they go away typically very quickly. And so if patients know that, then they’re much less worried, and then you can talk to them about the strategies, because caffeine or acetaminophen (Tylenol) will often help with that. If you warn them that they may have some joint aches or pains, that can also help, since those are often transient.

With venetoclax, warning them about some nausea or diarrhea, and then we often manage that by subsequently moving the drug to the evening after they’re done with their ramp up, or initiating an antiemetic, things like this. And then oftentimes many patients who have that in the beginning, it doesn’t persist throughout the whole time that they’re on the drug. Sometimes the diarrhea may, but many times it doesn’t. So getting the patients through that early phase with the close management. Which again, it helps, have your team help with that, the nurse practitioners, et cetera, and then hopefully things settle out and everyone’s happy.

Dr. Nicole Rochester:

Wonderful. I just want to emphasize two things. One that each of you said. One is this idea of a team-based approach, which is important in the treatment of all diseases, but of course very important in the treatment of the cancer. And also this idea of educating our patients so that they know ahead of time what to expect and really involving them as part of the team. So I really appreciate those, both of those points.

Well, it’s time to wrap up our roundtable. I have really enjoyed this conversation and I’d like to get closing thoughts from each of you. So I’ll start with you, Dr. Coombs. What is the most important takeaway message you’d like to leave with healthcare professionals who may be listening as they watch this program and understand better about CLL mutations, clinical trials, and managing side effects?

Dr. Callie Coombs:

So what is the most important thing, there’s so many, I would just say CLL is a chronic disease that affects our primarily elderly patients, and so it’s a marathon, not a sprint. However, with all of the advances that we’ve had in excellent drug therapies, despite these resistance mutations, patients can attain many, many, many years of high quality of life. But it’s incumbent upon us as their providers to help ensure that quality of life through effective management of side effects that may be encountered over the course of their time on therapy for the patients that do need therapy.


Share Your Feedback

Empowered AML Patient: Ask the AML Expert

Empowered AML Patient: Ask the AML Expert from Patient Empowerment Network on Vimeo.

For acute myeloid leukemia (AML) patients, how can they get the best care no matter location? Watch as expert Dr. Catherine Lai and AML patient Sasha Tanori discuss advancements in AML detection and treatments, the use of remote monitoring, questions to ask if you suspect you have AML, how AML can vary by age, and clinical trials access for optimal care.

See More from Best AML Care No Matter Where You Live

Related Resources:

What Treatments Are on the Horizon for Acute Myeloid Leukemia Patients?

What Treatments Are on the Horizon for Acute Myeloid Leukemia Patients?

What Role Does a Multidisciplinary Team Play in AML Care?

What Role Does a Multidisciplinary Team Play in AML Care? 

How an AML Survivor’s Resilience Saved Her Life 


Transcript:

Sasha Tanori:

I want to start off by saying, thank you so much for joining me, Dr. Lai, I greatly appreciate it.

Dr. Catherine Lai:

Thank you for having me.

Sasha Tanori:

Dr. Lai, early on before my diagnosis, AML, many of my doctors I saw dismissed my symptoms and attributed them to me being plus-sized. Can you share with us how detecting AML has evolved over the last several years?

Dr. Catherine Lai:

Yes, and I’m sorry to hear that, but what I would say about the diagnosis is that how we diagnose patients with AML, unfortunately, hasn’t changed significantly in the sense that we still have to rely on our standard techniques with the bone marrow biopsy. But what I would say is that the technology for how we risk-stratify patients and subsequently treat patients has improved because we have a better understanding of the molecular characteristics of AML now, and so it has helped us in terms of being able to identify more targeted treatments, where patients are more likely to respond and help us with both our short-term and our long-term plan.

Sasha Tanori:

Right, got it. My next question is, can you speak on how monitoring and treating AML has changed during the pandemic?

Dr. Catherine Lai:

Yeah, so unfortunately, as you experience it, you spent your induction in the hospital for several weeks, and when you’re able to be in the hospital with support, either from friends or from family, it makes the experience much, much easier and with COVID, especially at the height of the pandemic, we weren’t allowed our hospital. And I know several of my colleagues as well, the hospitals weren’t allowing any visitors and that put a lot of stress on the patient, on family members, on the staff, the nurses, the physicians, really the whole care team. Just because we were needing to spend extra time to make sure that everybody was updated, so either if we couldn’t do it on FaceTime, having to make sure other phone calls later, which is just…it is what it is. And we made the best of the situation. Currently, we are allowing to have a limited visitor policy, which is helpful. I think the other thing that has really changed is what we consider when we’re starting treatment, if patients obviously need induction chemotherapy and need to be in the hospital, we don’t change the recommendation based on that, but if there are patients who…

Dr. Catherine Lai:

There are options whether or not the patient is done inpatient versus outpatient, I think that that’s a huge consideration in terms of quality of life and how we manage those patients.

Sasha Tanori:

Can you speak to the advances and treatment options for high-risk AML patients?

Dr. Catherine Lai:

Yes, so fortunately, we have made a lot of progress in the AML space, that is one thing that is really exciting, I would say. Since 2017, there have been nine FDA approvals for AML, and prior to 2017, and we have been using the same chemotherapy for the last 40 years. Now, that’s not for lack of trying. There are many leukemia physicians who have been working at this for the duration of their careers, but AML just is very heterogeneous, and it’s very smart. It’s smarter than we are, and it’s constantly changing, and so that has made it challenging in terms of being able to treat it. So, there are newer treatment options, both modifications to traditional chemotherapy as well as other targeted therapies that have improved the landscape for AML and high-risk AML in particular. That’s awesome.

Sasha Tanori:

Dr. Lai, I think another factor that played a role in my diagnosis is somewhat being delayed is my age, I was 24 at the time, what are some questions others who suspect they have AML should ask to rule out the diagnosis?

Dr. Catherine Lai:

So, Sasha, that’s a really good question. And what I would say is that, as you are aware, the median age of AML diagnosis is 68, so not to say that we don’t have young patients…I have plenty of young patients, but it doesn’t come to…it’s not a common thing to think about in younger patients right off the bat, the other thing that contributes to that is also AML compared to other cancers is an uncommon cancer. There are only 25,000 cases of newly diagnosed in the United States per year because it’s not as common in younger patients and because it’s not that common…doctors often want to rule out other simple things rather than just going straight to a cancer diagnosis though, unfortunately, that can lead to some delays, what I would say in young patients who are healthy is that they shouldn’t have low blood counts that can’t be explained for other reasons. So, I think having prompt attention in terms of if their blood counts are abnormal, to really understanding why they’re abnormal, and those are things that can be easily work up, and if all those things are rolled out, then you’re talking about doing a bone marrow biopsy I don’t like to do procedures for unnecessary reasons, but it’s one of those things that you can also…

I mean, I think if you have a physician who is the astute and is thinking about that, that you can…you can get to a diagnosis pretty quickly, I mean AML is a diagnosis in the name acute. It comes on acutely, so that means days to week, so I suspect you are probably feeling very well and over a very short prior of time felt very unwell, and you’re very in tune to your body, and that is very important because patients are smarter than we give them credit for, and so being persistent and knowing that something is wrong goes a long way. Again, I’m sorry that you had to deal with that, and I’m glad that they finally made the right diagnosis, but I think just awareness and education. While it is an uncommon disease, I think having a larger burden and strain that happen on younger patients because you haven’t been working for the majority of your life, and it takes a huge toll on what your potential is, both as a person, but economically and all sorts of things. So it’s a huge problem

Sasha Tanori:

Does prognosis of AML vary by age?

Dr. Catherine Lai:

So, yes and no. So let me answer that in two steps, so it does in the sense that older patients are more likely to have more comorbidities, so more medical problems, and so therefore have a higher likelihood of having complications, and also as patients get older, they acquire more mutations and more abnormality, so those molecular abnormalities, and so therefore, older patients then are become more challenging to treat as well. What I would say though, is that we typically risk-stratify based on molecular factors, so the different mutation than somebody has and the age and the comorbidities don’t necessarily play into that role of stratification, so for example, whether or not you’re receiving a transplant or not…age is a factor, if you’re kind of in that little risk category, the intermediate risk category, the other thing I would say is that for young patients, they are able to tolerate because many don’t have medical problems, so they are able to tolerate treatment better, so when I’m talking about numbers and likelihood of response and overall survival, those…all those mediums assume that somebody is in their mid-60s, and so I adjust the numbers because for younger patients that those numbers are likely higher…

Because they’re less likely to have complications.

Sasha Tanori:

Right. I had many medical professionals that participated early on in my care. Can you speak on the role of the multidisciplinary care team that plays in AML care?

Dr. Catherine Lai:

Yeah, this is…this is an excellent question. I would say that treating leukemia is a team sport, everybody has their role, and it’s not just one person, and this is part of why I love treating leukemia patients, is that we’re able to engage multiple players, everybody is good at their particular thing, and so one analogy is that…we’re kind of like a baseball team, is that you want everybody to be able to do their own…have their own position. What a standard for our center is that we have the leukemia physician, there’s a specific leukemia nurse, we engage our social worker very early on, and also our cancer nutritionists and physical therapist and occupational therapist so we all work together at different parts of the treatment journey to make sure the patient is getting everything that they need and the whole person is being taken care of.

Sasha Tanori:

Right. AML patients, just like anyone else, want to live and live a very long time. Are AML patients at risk for secondary cancers, and are there any studies that speak on this?

Dr. Catherine Lai:

Yeah, so I would say everything has its risk and benefits at the time of diagnosis, you need the chemotherapy in order to get into remission, and then if you need the transplant, whether or not you’re getting radiation and then further some chemotherapy before the transplant, so that’s not without risks, so especially in a young patient, for example, in your particular case, you’re at risk for secondary treatment-related MDS and other bone marrow-related disorders that could occur, most patients who are in their 60s who, if they live long enough would be at risk, but most of those patients will die of something else before you have that opportunity. As a young patient, the other thing to be aware of, especially with, given that you’ve had transplant, is that the increased risk of cardiovascular effects, as well as making sure in patients who have had your whole-body radiation, other effects in terms of their thyroid, lung function, and then screening earlier for other cancers. So in terms of looking at studies, we know that these risks are slightly increased and that monitoring starts a lot sooner, especially in young patients. So I think just being aware of what you need to do.

Dr. Catherine Lai:

We also have a survivorship clinic, which I think is really important to help understand, you know what your risks are, because once your leukemia is in remission, we don’t want you to develop other medical problems, so it’s important just for patients to be educated so that they know how to take care of their body at each stage of their…again, of their journey.

Sasha Tanori:

Alrighty, after getting a bone marrow transplant three years later, I’m still dealing with graft-versus-host disease or GVHD, but there are other obstacles that I’m also facing. Does GVHD ever truly go away or is it something that I’m going to have to learn to live with?

Dr. Catherine Lai:

Yeah, I wish I had a magic answer for you. Our data is that it gives us guidance for each patient, but then also each patient as an individual and how they respond to different medications, and the nuances of that is…it can be different. So what I would say is that there are patients who you have chronic GVHD for years and it can eventually go away, and in some patients, they deal with it for a lifetime, you’re young enough, and I’m hopeful enough that at some point it will improve and get better. So I would be cautiously optimistic that things will improve.

Sasha Tanori:

I’m…I’m trying my best.

Dr. Catherine Lai:

It’s hard.

Sasha Tanori:

Yes, it’s very hard. Yeah, my care team suggested a clinical trial for a new drug focusing on improving my lung function, fortunately, my lungs improved on their own. Dr. Lai, not every AML patient is offered clinical trial as a care option, what advice you have for AML patients who are seeking clinical trial and what’s the best way to locate one?

Dr. Catherine Lai:

Yeah, so this is an area, a huge area of unmet need, I would say in general, across all oncology trials, and I think less than 10  percent of the patient population is on trials, there’s a lot of stigmas around clinical trials and are you getting… Are you getting a drug that we don’t know what’s gonna work, am I being…am I being tested? In oncology, I would say for the most part, we try to make trials where you’re being measured to the standard, so you’re getting the standard plus, or we’re trying not to…just in terms of doing what’s best for the patient, in general, I don’t offer trials to patients where I don’t think that there’s scientifically a rationale for those drugs, but to answer your question, the best place to look is on clinicaltrials.gov. That’s cumbersome. If you don’t know what you’re looking for, I can give you a lot of unnecessary information. There are a lot of other resources out there, The Leukemia & Lymphoma Society is a great resource. I know that they have online or people that you can talk to in terms of helping you direct specific clinical trials, I know depending on where you live in the country, there are other local New Chapters, oncology chapters that we have that can help patients find…

And have access to clinical trials, and then I think the biggest thing is just if a patient is with the community oncologist, having enough education to say, can I have a referral to an academic institution where they can ask those questions and get that information, and local community oncologists are fantastic, but they see everything, they see breast cancer, they see one cancer where the academic centers were specialized where all I see is leukemia and MDS kind of acute leukemias. So it’s just a different set of knowledge.

Sasha Tanori:

Okay, my next question is, I’ve had one telemedicine visit via my online portal, is the role of the telemedicine in AML care becoming more important?

Dr. Catherine Lai:

Yes, so what I would say…so this is my personal opinion, but in my opinion, that medicine compared to other industries tends to be a little bit farther behind, we’re not as quick to adapt the newest technology where COVID has helped, I think is at least in my practices, help utilize telehealth in the sense that there was a period of time where I was seeing fewer patients and then it really picked up because especially for patients who have a local oncologist but want a second opinion, the telehealth really offers that they don’t have to travel two hours to come see me to get that opinion. So what I would say is that it cannot replace the physical exam, it can’t replace a face-to-face discussion when you’re really talking about new diagnosis and therapy, because I really do think that that should be in person, but where… I have found that it’s been really helpful is if I’ve had an initial visit with the patient, and they either have a local oncologist, so I’m just checking in with them periodically, or if it’s to review results, say they’ve had a bone marrow biopsy and it’s…

They’re further along in their treatment, or if they’re just reviewing imaging results or something where I don’t necessarily need to see them have a physical exam and I’ve seen them recently, and so I do everything else that’s going on, but can I check in to review a specific part of information. I think that telehealth would have a role, and I hope it continues to have a role.

Sasha Tanori:

Yeah, yeah definitely, I agree. It’s really helpful in that sort of way, so you don’t have to actually leave the comfort of her home for something that’s not really super serious. You know?

Dr. Catherine Lai:

Exactly, yeah, I think what happens is patients do tend to…what I’ve noticed patients do is under-report, so it’s for… Not for infrequent visits, so for patients who are followed on a regular basis, it does allow there to be some ease of burden in terms of how we treat our patients.

Sasha Tanori:

Right. So a silent side effect that people facing cancer don’t always talk about is mental health. Are there any treatments or coping methods that you recommend for patients and care partners?

Dr. Catherine Lai:

Yeah, so I would say to get social work involved early on, I think there’s also…it’s silent, ’cause there’s a lot of stigma around it, is that is something that we should be talking about or not talking about or…I can handle it, that sort of thing, so I introduce our social worker very early to know that she is a resource for the patients, no matter how big or how small, just to try to get them used to that idea. What I would also say is just talking with as many people as possible as I’m sure you realize that the network and the community is small and everybody is willing to help each other out, so once you put yourself out there, you’ll realize that there are other resources out there, and you’re not alone in this journey, and what your cancer team offers you is different than what other patients who have gone through exactly what you’ve gone through can offer, and so I know that there are other resources out there in terms of societies that connect other patients who have the same diagnosis, so I would say it’s really just about education and talking and knowing that it’s okay to talk about your diagnosis and no matter what format that is, or if it’s a little bit now and a little bit later, and also just normalizing it, in the sense of the feelings you have are valid and normal, and if you don’t have those feelings is actually when I get worried about patients because you’re supposed to have certain reactions, you were a young patient and you were diagnosed with cancer.

That’s not a trivial thing. And we’re just…we’re all here to help you and help the patients go through everything…

Sasha Tanori:

So for my last question is the future bright in AML treatment and can you speak about any exciting studies that you are working on, that AML patients and their families should stay tuned for?

Dr. Catherine Lai:

Yes, so I am excited. I am excited to say that I think in my lifetime, I will be a part of AML change and we have already seen it. I have mentors who are in their 60s, who have used the same therapies, they use them for the entirety of their career. And so as I mentioned, we only have your 9 FDA approvals. I think there are more coming… I think what I would like to mention is I think the use of immunotherapy, bone marrow transplant is the original immunotherapy, but as you know, there are many risks and benefits and complications, and so how we manipulate the immune system or how we use drugs to help manipulate the immune system, I think it’s a work in progress. It has been more successful in other cancers, not as successful in AML yet, but I think we will get there. The other thing would be, is how… We look at minimal residual disease. So, as you know, but for everybody else, we consider a complete remission is less than anything less than 5 percent blast or 5 percent leukemia cells but we know that anything greater than zero is bad, and you have more than zero, the disease will come back at some point.

So looking to how we monitor, going back to those molecular technologies and how we’re monitoring for residual disease so that we can detect disease faster, so I think really the concept of detection and prevention will come into a huge role because also if we can detect the disease relapse sooner, we’re treating less disease and then there’s less side effects and less toxicity, and then I think the last thing would be health outcomes of a lot of what we’ve been talking about just in terms of the whole picture and how we can better treat these patients I also think there’s a huge role for looking at each individual person and their age and their medical problems, and they’re a physiologic age as opposed to their chronological age and how we can best treat the patient so they can have the best outcome.

Sasha Tanori:

All right, well, thank you so much, Dr. Lai, for taking the time to speak with me and for all you’ve done for the AML community and our patient’s families, everyone.

Dr. Catherine Lai:
Thank you, thank you so much for having me. I’ve really appreciated you putting yourself out there… Thank you.

What Treatments Are on the Horizon for Acute Myeloid Leukemia Patients?

What Treatments Are on the Horizon for Acute Myeloid Leukemia Patients? from Patient Empowerment Network on Vimeo.

What can acute myeloid leukemia (AML) patients expect in the future of AML studies? Watch as expert Dr. Catherine Lai shares insight about the state of FDA approvals. what is in pipeline for AML treatment, and disease monitoring technologies for improved patient care.

See More from Best AML Care No Matter Where You Live

Related Resources:

Acute Myeloid Leukemia, Recommended Coping Methods and Mental Health

Acute Myeloid Leukemia, Recommended Coping Methods and Mental Health

What Questions Should I Ask If I Suspect Acute Myeloid Leukemia?

What Questions Should I Ask If I Suspect Acute Myeloid Leukemia? 

Are Acute Myeloid Leukemia Patients at Risk for Secondary Cancers?

Are Acute Myeloid Leukemia Patients at Risk for Secondary Cancers? 


Transcript:

Sasha Tanori:

Is the future bright in AML treatment, and can you speak about any exciting studies that you are working on, that AML patients and their families should stay tuned for?

Dr. Catherine Lai:

Yes, so I am excited. I am excited to say that I think in my lifetime, I will be a part of AML change and we have already seen it. I have mentors who are in their 60s, who have used the same therapies, they use them for the entirety of their career. And so, as I mentioned, we only have your nine FDA approvals. I think there are more coming…I think what I would like to mention is I think the use of immunotherapy, bone marrow transplant is the original immunotherapy, but as you know, there are many risks and benefits and complications. And so how we manipulate the immune system or how we use drugs to help manipulate the immune system, I think it’s a work in progress. It has been more successful in other cancers, not as successful in AML yet, but I think we will get there. The other thing would be, is how…we look at minimal residual disease. So, as you know, but for everybody else, we consider a complete remission is less than anything less than 5 percent blast or 5 percent leukemia cells but we know that anything greater than zero is bad, and you have more than zero, the disease will come back at some point.

So looking to how we monitor, going back to those molecular technologies and how we’re monitoring for residual disease so that we can detect disease faster, so I think really the concept of detection and prevention will come into a huge role because also if we can detect the disease relapse sooner, we’re treating less disease and then there’s less side effects and less toxicity, and then I think the last thing would be health outcomes of a lot of what we’ve been talking about just in terms of the whole picture and how we can better treat these patients I also think there’s a huge role for looking at each individual person and their age and their medical problems, and they’re a physiologic age as opposed to their chronological age and how we can best treat the patient so they can have the best outcome.