Tag Archive for: thrombosis

Are MPN Risks and Outcomes Impacted by Race or Ethnicity?

Are MPN Risks and Outcomes Impacted by Race or Ethnicity? from Patient Empowerment Network on Vimeo.

Are myeloproliferative neoplasm (MPN) risks and outcomes impacted by patient race or ethnicity? Expert Dr. Idoroenyi Amanam from City of Hope explains common MPN symptoms, potential risks and outcomes, and patient advice for optimal care.

[ACT]IVATION TIP:

“…if you have stroke, heart attack, blood clots, abnormal counts, please follow up with your doctor and ensure that you get referred to the appropriate specialist to help assist in managing your complications.”

Descargar Guía|Download Guide 

See More From [ACT]IVATED MPN

Related Resources:

Myeloproliferative Neoplasm Basics for Newly Diagnosed Patients

Myeloproliferative Neoplasm Basics for Newly Diagnosed Patients

What Does the Future of Myeloproliferative Neoplasm Care Look Like?

What Does the Future of Myeloproliferative Neoplasm Care Look Like?

MPN-Related Complications | Are BIPOC Patients at Higher Risk

MPN-Related Complications | Are BIPOC Patients at Higher Risk?

Transcript:

Lisa Hatfield:

Dr. Amanam, how does race impact clinical MPN outcomes or complications? And are certain populations more susceptible to getting MPNs?

Dr. Indoroenyi Amanam:

Yeah, it’s a great question. I think, because of the fact that symptoms are not as clearly related to MPNs as opposed to some other diseases. So the major presenting symptoms for MPNs are related to high blood pressure or some other vascular abnormality, including strokes or blood clots. It takes a little work to diagnose MPN patients, and half of them are asymptomatic. And if you actually look at diagnosis rates, they’re one to three cases per 100,000 per year. And so this is very rare, and I think that we don’t have enough data to really understand if some groups are at higher risk to develop MPNs than others.

We have looked at incidences of thrombotic events in patients who have MPNs, and we tried to see if there was a difference between racial groups, and we didn’t. We did see that if you’re younger, you do have a higher risk of thrombosis over time, but there was no difference for if you were white, Hispanic, or African American.

What we did find though, in a small single center study is that if you are non-white, there’s a higher risk of death over time. And I think we still need a lot of work to get a better understanding as why that’s the case. So far we don’t have a clear signal for that, but I do think, in the same…the story that we understand for healthcare in general, there are some differences in terms of access to care, the quality of care that’s delivered in, you can guess that possibly that’s one of the reasons why that that affects African Americans and Hispanics disproportionately in this setting as well.

So my activation tip for this question would be if you have stroke, heart attack, blood clots, abnormal counts, please follow up with your doctor and ensure that you get referred to the appropriate specialist to help assist in managing your complications. 


Share Your Feedback

Create your own user feedback survey

How Will Advanced Technologies Enhance Myeloproliferative Neoplasm Care?

How Will Advanced Technologies Enhance Myeloproliferative Neoplasm Care? from Patient Empowerment Network on Vimeo.

How will myeloproliferative neoplasm (MPN) care be enhanced by advanced technologies? Experts Dr. Krisstina Gowin from University of Arizona Cancer Center and Dr. Ana Maria Lopez from Sidney Kimmel Cancer Center share information about how technology can help manage symptom burden, risk factors, and the patient experience for enhanced MPN care.

Download Resource Guide

See More from MPN TelemEDucation

Related Resources:

How Has COVID-19 Impacted Myeloproliferative Neoplasm Care

How Has COVID-19 Impacted Myeloproliferative Neoplasm Care

Will Advancing Technologies Allow MPN Clinical Trials in Patient Homes

 
How Is Telemedicine Influencing Personalized Medicine for MPN Patients

Transcript:

Lisa Hatfield: 

So, Dr. Gowin, do you have anything to add on, how MPN care or just cancer care, in general, could change with different technologies? We didn’t touch a lot on things like artificial intelligence and that type of thing, and we can speak to that or any other type of technology that you’re familiar with.

Dr. Krisstina Gowin: 

Well, I think the artificial intelligence aspect is really going to change the paradigm again on how we’re designing, studies. And I think one of the biggest challenges that we have in myeloma and as well as myeloproliferative neoplasms, is to think about how do we optimally sequence our therapies to achieve best survival, right? And I think this is a wonderful problem to have. We have now not only one JAK inhibitor on the market, but several and more in the pipeline and several other therapeutic targets. And so now the question is which therapy and when do we employ it? So things like artificial intelligence will help us to answer that question with machine learning decision tree analysis, all of that is going to be answered through those kind of platforms. And so I think that is going to be a shift we will see in the next five years is many different machine-based learning algorithms to better understand those problems we cannot have tackled traditionally otherwise.

Sensors though is another one, right? And so a big thing in MPNs is not only addressing the blood counts and reducing risk of thrombosis, and to address symptom burden, but it’s really addressing lifestyle because it’s things like cardiovascular disease, stroke that really we’re worried about as some of the sequelae of having the disease and what we’re trying to prevent with therapeutics. And so even going back to the NCCN guidelines, it’s addressing cardiovascular risk factors as part of our core treatment goals. And so how do we really do that? And it’s really through lifestyle medicine and that’s where the sensors come in. And so now we have, these Fitbits and smartphones that connect to our Apple watches and we have Garmins and all these wonderful devices that are prompting us to move more, prompting us to be cognizant of our heart rate and stress response prompting us to meditate. And so I can envision those evolving over time and connecting to the EMR and being very seamlessly interwoven into our clinical trials. And we’re already doing that. In fact, we’re talking about doing one very soon in MPN patients. And so I think the sensors are gonna be another big way that we’re going to be integrating, into our clinical trials and symptom management tools.

Lisa Hatfield: 

That’s fascinating. Thank you for that. And Dr. Lopez, do you have anything to add about other technologies and how they may affect cancer care in the future?

Dr. Ana Maria Lopez: 

Sure. When Dr. Gowin mentioned the sensors, it just reminded me, we’re building this new building, patient care building and oncology will be there. And I did a tour recently, and we’re used to go to the doctor, you stop in, they get your blood pressure, they get your weight, et cetera. Here, you’ll walk in directly to your exam room and you check in at a kiosk, so you just kinda check in [chuckle] with a little robot kiosk, and then it’ll tell you where you’re going. You’ll go to Room 3, let’s say, and Room 3 will say, “Welcome, Lisa.” [chuckle] And so you know that you’re in the right place. And you’ll walk in, there’s your gown, you’ll sit in the exam chair, and the exam chair automatically is gonna take your vital signs. So it just seems, really these built-in aspects to the technology. And one of the things, again, what I just love about this work is that it’s a very interdisciplinary, multidisciplinary. And one of the projects that we were working on, which it ties into this, when I was in Arizona with the telemedicine program is we worked with the College of Architecture and with this concept of smart buildings.

Dr. Ana Maria Lopez: 

So it’s kind of like that. Why should you do these different sensors that detect, but that it could also detect. You might walk into the room and you might be really nervous as you might be really cold, and it would detect that and it would warm the room for you. Or you might be coming in and be having hot flashes and it would just cool the room for you. So the technology has so much potential to really improve the patient experience.

Lisa Hatfield: 

And that’s amazing to me. I think that would be incredible to walk into a building to have that experience, as long as it doesn’t take away the compassion and care I get from my providers. I am so fortunate to have extraordinary providers, so I don’t think it will ever take over that aspect of it, I think that is a fear people have, especially with artificial intelligence and that type of thing, I think it can only go so far. Can’t provide the humanness that’s required for patient care, so yeah.

Dr. Ana Maria Lopez: 

Yeah. These are tools. 


Share Your Feedback:

Create your own user feedback survey

How Are ET, PV and Myelofibrosis Monitored?

How Are ET, PV and Myelofibrosis Monitored? from Patient Empowerment Network on Vimeo.

MPN specialist and researcher Dr. Joseph Scandura reviews tools that are used to monitor patients with essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF), including routine blood work and symptom management

Dr. Joseph Scandura is an Associate Professor of Medicine and Scientific Director of the Silver MPN Center at Weill Cornell Medicine. Learn more about Dr. Scandura.

 

Related Programs:

 
Thriving With an MPN | Tips for Managing Worry and Anxiety

Thriving With an MPN | Tips for Managing Worry and Anxiety

What Are Common MPN Symptoms?

What Are Common MPN Symptoms?

How to Treat PV-Related Itching

How to Treat PV-Related Itching


Transcript:

Katherine Banwell:

I would imagine monitoring patients is different for each of the MPNs. So, how are patients typically monitored over time, and let’s start with essential thrombocythemia?  

Dr. Scandura:

Yeah. I think – again, it’s similar. You know, what’s near-term, what’s long-term? And so, in all of these diseases, thrombosis risk is a near-term risk. That’s something that I am monitoring in certain ways to help mitigate that risk. In ET and PV, I approach them similarly. Blood counts are certainly – these are diseases of the blood forming system. Certainly, monitoring blood counts I find helpful. But the reality of it is, in ET, there is not a clear linkage between blood counts and risks.  

And so, I like to keep the platelet count near normal if I can. But I also recognize that it may not be worth suppressing all of the blood counts to achieve that landmark, because it’s not clear that that’s really reducing the risk any more than just having somebody on a medication that helps control the blood counts. In polycythemia vera, different blood counts are very important. The red blood cells are kind of like part of the clotting risk. We know from clinical trials that keeping the red blood cell parameters within certain ranges reduces the risk of clotting. And so, what I monitor in polycythemia vera is the hematocrit. In women, I like to keep it below 42. In men, I like to keep it below 45.  

But I don’t just – I’m not a slave to the hematocrit. I am keeping an eye on the other blood counts and the other red blood cell parameters. So, for instance, what’s the size of the red blood cells? That tells me a little bit about what’s going on in the blood formation for that patient. And what’s the number of red blood cells? So, sometimes people can have very small red cells, because they’re a little iron-deficient and have a huge surplus of the number of red blood cells. And that tells me a little bit about how their blood forming system is responding to therapy.  

Iron deficiency in polycythemia vera is very prominent. I personally believe it’s a very major driver of symptoms in patients who are receiving phlebotomy as part of their care. And it’s something that I monitor and really counsel patients on. My goal is to make phlebotomy independent, but it can take a while.  

Everybody starts out iron-deficient, and then we take iron out of their body through blood with the phlebotomy. And that makes them more iron-deficient.  

Katherine:

Right. 

Dr. Scandura:

I monitor symptoms from patients, and sometimes that can tell me that their disease needs to be – their treatment needs to be tweaked a little bit, even something as simple as aspirin. People can sometimes have burning in the skin or itching that is sometimes responsive to changing the aspirin dose or how it’s given, once a day versus twice a day.  

And that simple thing can be a big change for a patient who’s kind of, literally, climbing out of their skin or wishing they could and to try and find something that is helping.   

I had a patient the other day. He had COVID. I said, “Oh, you should probably get this medication.” Do you have your primary care physician? Who’s taking care of you?” And he goes, “Well, to be honest with you, you’re my guy.” And so, it’s true. I see this patient a lot. And so, sometimes they forget. If I’m not paying attention to their blood pressure, the risks or treatment of diabetes, cholesterol, lipids, their screening programs for mammogram or colonoscopy, health maintenance issues, I do keep an eye on that in patients, because I do think it’s a part of the MPNs.  

I think that there are excess risks for patients for some of these factors. Certainly, if you think of it as three strikes, they get a strike for having an MPN. I don’t want them to have any other strikes. So, diabetes, hypertension, those are strikes that I can potentially, at least, treat or refer them to somebody to help comanage with me. And so, that’s kind of my general approach. 

Katherine:

What about patients who have myelofibrosis? Are they monitored more closely? 

Dr. Scandura:

Yeah, I think it depends a little bit on the patient. Patients with early myelofibrosis often don’t have any symptoms or near-term risks much different than those from ET or PV. As the disease can progress, then some of these patients have more profound problems with symptoms, which I may be trying to find a solution to make them feel better. And also, blood counts can become more of an issue.  

Transfusions in some patients who are very high white blood cell count, the spleen is often quite enlarged. Although, in my experience, most patients aren’t really bothered by the size of their spleen as the physicians are. But it is something where I think, on average, they’re monitored a little bit more closely to quite a bit more closely depending on the patient. 

Katherine:

What happens if someone suddenly has a change in blood counts? What do you do? 

Dr. Scandura:

Yeah. I mean, repeat it. That’s the first thing. Also, check what’s going on. It’s not uncommon in patients with MPNs that I’ll see them and the counts are a little bit out of whack, the white count is much higher than it’s been, and questioning them. “Oh, yeah. I had X, Y, or Z last week or the week before.” It used to be a upper respiratory tract infection, or they had a minor surgical procedure.  

And sometimes the responses to these things can be accentuated in patients with MPNs. And so, if that’s what of this story, I certainly would repeat it and let things calm down a little. And that’s often all it is. I’m much more of a monitor of the trends. So, one-time measure doesn’t generally excite me. It might make me want to have a follow-up a little more – in a shorter period of time. Of course, it depends on what the change is. But, for most of the changes that we observe, they’re relatively minor. And I will monitor them over time.  

If I see a trend where something is progressively increasing or decreasing over time, then I start thinking about what else is going on. And that’s always in the context of what’s going on with the patient. How are they feeling? What’s their physical exam like? What are the other laboratory values like?  

Katherine:

When is a bone marrow biopsy necessary? 

Dr. Scandura:

I would say a bone marrow biopsy is absolutely necessary at the time of diagnosis. I personally do not routinely monitor by bone marrow biopsy unless it’s part of a clinical trial.  

But I do perform a bone marrow or want to look at the bone marrow morphology if there is one of these changes or at least a trend that I want a little bit more information about. And so, if – or if it’s been a very long time since somebody has had a bone marrow. If it’s been five or ten years, then sometimes I may recommend we look just so we can collect a little bit more up-to-date information.  

But I don’t routinely do a bone marrow, but I will do it if there are laboratories that are kind of trending in the wrong direction, there’s symptoms, there’s physical findings that I’m just not sure about. And I think it would help me be more sure as to what’s going on and be able to discuss that with the patient. Sometimes, just to say, “Hey. Look, we were worried about this, but the bone marrow looks really good.”  

Is There MPN Research Underway to Help Understand Progression?

Is There MPN Research Underway to Help Understand Progression? from Patient Empowerment Network on Vimeo.

How and why do MPNs progress? MPN specialist Dr. Joseph Scandura shares an update on research being done to better understand–and possibly prevent–disease progression.

Dr. Joseph Scandura is an Associate Professor of Medicine and Scientific Director of the Silver MPN Center at Weill Cornell Medicine. Learn more about Dr. Scandura.

See More From Thrive MPNs

 

Related Programs:

 
What Are the Signs of MPN Progression?

What Are the Signs of MPN Progression?

What Are Indicators of MPN Progression?

What Are Indicators of MPN Progression?

Expert Perspective: Hopeful MPN Research and Development


Transcript:

Katherine Banwell:

Is there research being done on MPN progression to understand how it happens or even prevent or slow progression? 

Dr. Scandura:

Yeah. There’s a lot. I think there is a – from both the sort of basic laboratory using animal models to try to understand what are the kind of systems that are involved in how these diseases change. What genes are involved? How do they talk to each other? You know, these are not cells that live in a vacuum, right? They live in a special microenvironment. What are the signals that crosstalk between the MPN cells, the MPN stem cells, and their microenvironment?  

And so, there’s a lot of research on that and the basic side of things. In humans, there’s a lot that has been done over the years in terms of trying to understand what are some of the genetic features of progression. And I think we’re beginning to get a little bit of a better understand of what are the non-genetic things that are associated with progression.  

I was part of an effort from the MPN Research Foundation and still am.  

They have what they call the Progression Network, where they tried to put together a number of investigators from really across the world to share ideas about the nature of progression and how we might look at studying this and understanding ways to prevent progression.  

I think we do have some drugs now that show some promise in terms of being able to prevent progression. I think interferons have shown this in polycythemia vera in terms of a promise for improved long-term outcomes and delayed risk progression. I think that the gold standard randomized trials are maturing and are sort of bearing out some of the same findings that have been observed retrospectively, so sort of kind of looking back in time.  

But the difficulty is that it can take a long time for patients to progress. And you say, “Oh, that’s great.” And that is great. But, from a research – from a statistical side, it means things are really slow. If you have to wait 15 years to assess whether or not people progressed less in one treatment versus another, it’s really slow going. And so, we have to do a compromise of what’s – you know, what do animal studies say? What does retrospective analysis, when we might have people who started treatment 30 years ago, and now we’re just seeing how did it all work out? It’s not a perfect study, because biases can creep in, but it’s what we have now. And so, there’s a lot. And I think, increasingly, progression is being recognized as a goal of therapy, to prevent progression.   

Personally, it is one of my major goals, because I think we do a pretty good job at preventing clots with available treatments. But I don’t think we do a very good job at preventing progression, mostly, because we don’t exactly understand what’s driving that. And so, I think until we develop that deeper understanding and really invest the time and effort in terms of learning which approaches can help prevent progression, we’re going to continue to have these questions.  

When Should Stem Cell Transplants Be Considered for MPN Treatment?

When Should Stem Cell Transplants Be Considered for MPN Treatment?  from Patient Empowerment Network on Vimeo.

Dr. Jeanne Palmer, an MPN specialist, discusses when a stem cell transplant is an appropriate treatment option and provides an overview of how risk is assessed in MPN patients. 

Dr. Jeanne Palmer is a hematologist specializing in myeloproliferative neoplasms (MPNs) and bone marrow transplant at the Mayo Clinic in Arizona. Dr. Palmer also serves as Director of the Blood and Marrow Transplant Program and is Vice Chair and Section Chief for Hematology. Learn more about Dr. Palmer, here.

 

Related Programs:

 
Understanding Treatment Options for ET, PV, and Myelofibrosis

Understanding Treatment Options for ET, PV, and Myelofibrosis

What Are Treatment Options for Myelofibrosis?

What Are Treatment Options for Myelofibrosis?

What Are the Signs of MPN Progression?

What Are the Signs of MPN Progression?


Transcript:

Katherine Banwell:

When would you consider a stem cell transplant? 

Dr. Jeanne Palmer:

So, the stem cell transplant is based on disease risk. There is a number of ways we assess disease risk.  

The first two ones that were published a number of years back were the DIPSS score, which is Dynamic International Prognostic System Score, or the DIPSS Plus, which basically is the DIPSS and then you add to it a few other clinical features. This symptom score is based largely on things that we can see without even a bone marrow biopsy, so things like symptoms, age, number of white blood cells, whether somebody has anemia. And then the number of something called blasts, which is very immature white blood cells. The DIPSS Plus takes into account low platelets, need for transfusions, and chromosome abnormalities, which is the only test among that that needs to be from a bone marrow biopsy. 

Now, these were created prior to Jakafi being commercially available. So, we have to take a little bit of a grain of salt with those because of the fact that Jakafi probably has changed how long people can live with this disease. 

Now, more recently they’ve tried to account for these other molecular changes. So, when we take the genetic landscape of these diseases, we have the known driver mutations, so the JAK2 mutation which I have talked about, also calreticulin and MPL.  

These three mutations all affect that one pathway, the JAK/STAT pathway, so they all affect the pathway that drives the disease and they are known to be kind of mutually exclusive and definitely contribute to the formation of the disease. 

Some of these other mutations are called somatic mutations. They could be checked by things next generation sequencing or genetic analysis. There’s a number of different names that people use for this testing, but we look for mutations that are present and these mutations, number one, can sometimes tell us risk. So, there’s certain mutations that are high risk. Other times it can actually give us other opportunities for therapy, especially of the disease progresses. But these mutations are important to know for risk stratification. For example, if somebody has DIPSS score that is maybe not super high risk, but then they have one of these mutations, we know that that probably makes their disease a little bit more aggressive. 

And that’s when we think about transplant, is when we know that the disease probably has an average life – when somebody gets to the point in their disease where we estimate their life expectancy is around five years, recognizing that we’re not very good at this. That is the type of point when we start to think about transplant. But the timing of transplant is something that’s extremely difficult and a very personalized decision. It’s something that it’s really important to understand the disease risks, how we assess them and the caveats of these disease risk assessments as we move forward planning and timing of transplant and that’s something that is, again, a very, very important discussion to have at length with your physician. 

And I always recommend, there is quite a few of us out there who actually specialize in transplant for myelofibrosis and having discussions with somebody who really understands the biology of the myelofibrosis is important because it’s very different than a lot of the other diseases that are transplanted. 

Understanding Treatment Options for ET, PV, and Myelofibrosis

Understanding Treatment Options for ET, PV, and Myelofibrosis from Patient Empowerment Network on Vimeo.

MPN specialist Dr. Jeanne Palmer discusses the treatment options available for essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF). Dr. Palmer explains how a treatment choice is determined for each of the MPNs and how anemia is managed in patients with myelofibrosis. 

Dr. Jeanne Palmer is a hematologist specializing in myeloproliferative neoplasms (MPNs) and bone marrow transplant at the Mayo Clinic in Arizona. Dr. Palmer also serves as Director of the Blood and Marrow Transplant Program and is Vice Chair and Section Chief for Hematology. Learn more about Dr. Palmer, here.

Related Programs:

 
When Should Stem Cell Transplants Be Considered for MPN Treatment?

When Should Stem Cell Transplants Be Considered for MPN Treatment?

How Can Patients Navigate Care and Thrive With an MPN?

How Can Patients Navigate Care and Thrive With an MPN?

Expert Perspective: Hopeful MPN Research and Development


Transcript:

Katherine Banwell:

Much of the time the chosen treatment for MPNs manages the symptoms of the condition. I’d like to review the different types and classes of treatment for the three MPNs. So, let’s start with essential thrombocythemia again. When is it time to treat, and what are the options available? 

Dr. Jeanne Palmer:

Right. So, with essential thrombocythemia, that’s the disease that sometimes we don’t need to treat. 

So, we basically have a risk stratification system and this risk is based on age, history of a blood clot, the presence or absence of a JAK2 mutation. So, for example, if somebody is 28, does not have a JAK2 mutation, which is again one of those driver mutations, and never had a blood clot, they actually don’t necessarily need to do anything and just be monitored.  

Somebody who is less than 60 and has a JAK2 mutation or who is greater than 60 and does not have a JAK2 mutation, in that setting, a lot of times you can use aspirin. Now, it gets a little bit gray in terms of that over 60 without the JAK2 mutation with regards to whether at that point you really should start taking some medicine to lower the platelets. 

Now, if somebody has a JAK2 mutation, is greater than 60 or has had a blood clot, hands down they need to take medicine to lower the platelets, in addition to aspirin or whatever blood thinner they may need. So, for example, if you have a blood clot in a vein, a lot of times you need to take a blood thinner and that will be a lifelong thing. And again, we do these risk stratifications because we know there is a certain risk of clotting associated with the risk of essential thrombocythemia.  

So, for example, somebody who is less than 60 and does not have a JAK2 mutation, never had a clot, their risk of clotting is probably very close to that of the normal population. Whereas if you’re higher risk and have a JAK2 mutation and greater than 60 or have had a history of a clot, the risk of clot is probably about 4 percent per year. So, this is something that can vary quite widely, and even though that 4 percent per year on the short-term doesn’t sound like a lot, if you take it additive over years, that’s why we generally try to be aggressive about lowering the platelets.  

In lowering the platelets, the goal is to get less than 400 and doing that can be done through several different medications. The most commonly used medications is a drug called hydroxyurea, which has been around for a number of years, and a drug called anagrelide which is probably a little less commonly used, because it has some more GI side effects and headaches associated with it. 

In some cases, especially in younger patients with this disease, we can consider using interferon, which is an injection of a cytokine, which are one of the chemicals that regulates the immune system within the body. But this interferon can actually help lower the platelets and there is a question of whether it may affect the biology of the disease as well. 

Katherine Banwell:

Let’s turn to polycythemia vera or PV, what are the different options available for treating it? 

Dr. Jeanne Palmer:

So, for polycythemia vera, everyone needs to be on aspirin. 

And additionally, everyone needs to make sure to keep their blood count low, to manage their hematocrit, which is one of the measures of red blood cells. So, in men it’s generally recommended to keep below 45 and in women it’s recommended to keep below 42 percent. Now, the studied number was 45 percent and that was a study that was done, I don’t know, it was probably about 10 plus years ago, that actually showed that by keeping the blood hematocrit less than 45 percent you reduce the risk of having negative events like cardiovascular events and heart attacks. Because women tend to run with a lower blood count than men, it’s been extrapolated that 42 percent should be the number used for women. 

Now, this can be done by phlebotomy, which essentially is bloodletting.  

It’s kind of like donating blood except for that the blood unfortunately can’t be donated to anybody, it has to be discarded. But the phlebotomy is one way to do that, and the reason that works is because it makes somebody iron deficient. So, whereas if this is normal, if you’re iron deficient you become anemic. If your baseline hematocrit is here, making you iron deficient brings you back to normal. So, even though we always associate iron deficiency with anemia, iron deficiency in the setting of polycythemia vera is actually kind of a treatment of sorts. 

Now, once somebody gets above 60 and 60 seems to be sort of the magic age in these diseases, once somebody gets above 60, it is recommended that cytoreductive therapy is used, which means therapy or treatment that will bring down the red count. And again, for this one, hydroxyurea is an option as well as interferon. And there is recently an approval, actually FDA approval for a newer interferon called ropeginterferon or Besremi, which can help just bring down the red blood cells but it is the first interferon that’s actually been FDA approved for this indication.  

Katherine Banwell:

Are JAK inhibitors used as well? 

Dr. Jeanne Palmer:

They are. So, if somebody doesn’t respond well to hydroxyurea, the approval for ruxolitinib is actually for patients who have failed hydroxyurea. Although it’s something that we often consider especially in people who have a lot of symptoms. So, the itching, one of the things that can really help itching actually is Jakafi. If people have night sweats, they have weight loss, spleen related symptoms, those are the patients that will benefit from Jakafi. Additionally, if they are on hydroxyurea and can’t seem to get control of their blood count, Jakafi is a good option to help control the blood counts as well. 

Interferon is a very nice option because there’s great data that shows that you may actually be able to lower the percentage of JAK2 burden. 

So, we’d look at something called an allele burden, which is the percentage of cells that are involved – have the JAK2 mutation. Now, we don’t know whether lowering this percentage necessarily translates to long-term better survival, but I think there is enough data out there, and there is a good biologic underpinning for saying that this actually can help. But yes, Jakafi is another thing. 

And the really exciting thing is that there is a newer agent called rusfertide, which is a hepcidin mimetic, which is basically taking a protein in your body that helps metabolize iron and by making it externally and giving it to somebody that it can actually help bring down the hematocrit without having some of the other side effects we know with some of the other medications. That is currently in Phase III studies, so hopefully in the next couple of years we’ll see approval for that. 

Katherine Banwell:

Oh, that’s great news. And finally, how is myelofibrosis treated? 

Dr. Jeanne Palmer:

So, myelofibrosis is a little bit of a different animal. When you have something like essential thrombocythemia or PV, a lot of this is managing symptoms, preventing blood clots, but if you do appropriate treatment and management of these diseases you could probably live close to a normal life expectancy. 

So, I never typically pin a survival on it. With myelofibrosis, it’s a little bit different because there is a survival. Instead of saying you can live close to normal life expectancy, it backs up to saying how many years do I think you can live with this disease. Now, of course, we are horrible at predicting how many years anyone can live, so we have to take that all with a grain of salt. But we can at least sort of risk stratify people. 

And the first thing that’s really important is to figure out whether somebody is a transplant candidate or not and if, based on age, disease risk features, stuff like that, or whether we think they ever will be a transplant candidate. So, that kind of helps us sort of think about what your path moving forward is.  

Now, the current FDA-approved treatment for myelofibrosis, there are three JAK inhibitors approved, which is like Jakafi, which was the first approved one but there is also Inrebic or fedratinib and Vonjo or pacritinib and these have all been approved over the years. 

The role of JAK inhibitors and treatment of myelofibrosis is symptoms-based. So, for example, a lot of patients with myelofibrosis will have weight loss, night sweats, big spleens, really feeling fatigued and poorly and in this setting, the JAK inhibitor can be very helpful. And you don’t have to have a JAK2 mutation, a lot of times people say, well, I don’t have the JAK2 mutation so how can a JAK inhibitor help. So, the JAK inhibitor works on this pathway, which is called the JAK/STAT pathway, irrespective of mutation. 

So, if you are having symptoms and you have myelofibrosis, JAK mutation, excuse me, the JAK2 mutation does not predict who is going to have a response. And people who, regardless of which mutation you have, may actually benefit from it. 

So, the JAK inhibitors, though, are extremely effective at reducing symptom burden as well as reducing the spleen size. And we know that if a spleen is big and we can make it shrink that, that probably is a surrogate marker for living longer, and I think it’s because inflammation does a lot of wear and tear on the body. So if you can reduce the inflammation and the spleen shrinks, which generally go hand in hand, then you might help somebody live longer. It is not changing the biology of the disease, though, however, it doesn’t change the pathway and that this disease is kind of projecting ahead in terms of creating – it changes, as it goes along, may acquire new mutations or something like that which makes the disease become more serious. 

Right now, the approved therapies for it are JAK inhibitors and the Jakafi, ruxolitinib was the first one approved. Inrebic was approved several years back, or fedratinib. 

And then the most recent one that was approved is Vonjo or pacritinib and that’s a drug that is a JAK inhibitor that is actually very good for people with low platelets. The reason I bring that up is because if we think of what’s the biggest limiter of JAK inhibitors, JAK inhibitors bring down red blood cells, and they bring down platelets. So, when somebody has low platelets it’s very hard to use a JAK inhibitor, because we’re not really able to increase the dose well enough to get that inflammatory reduction because of the fact that the blood counts will drop too low. 

So, now drugs like Vonjo exist which, due to several other mechanisms associated with the drug are actually much more tolerated in somebody with low platelets. So, if you have low platelets, you can actually take the Vonjo, hopefully get the same degree of JAK inhibition to help the spleen shrink, help the symptoms get better without necessarily making the platelets substantially worse. A lot of times they do drop, it doesn’t help bring up the platelets, but it does help people tolerate more JAK inhibition, which ultimately will help with symptoms.  

Dr. Jeanne Palmer:

So, one thing I also wanted to add about myelofibrosis treatment is sometimes people present, they don’t have a lot of symptoms, they don’t have a lot of spleen related problems but they have anemia or low blood counts and these can be incredibly hard to treat. 

Even with symptoms and low red blood cell count or anemia or low platelets, it can be challenging to treat because many of these medications lower that. To treat the anemia there are several things that we can do. One of the first ones is using erythropoietin, and so there are many agents, they go by the names of like Procrit or darbepoetin alfa (Aranesp), that actually stimulate red blood cell growth by – like we give a recombinant hormone that helps red blood cells grow. This is normally something produced by the kidney. 

So, one thing that’s important before going on one of these injections is to make sure that the kidney is not already producing enough. So, for example, if the kidney said, oh geez, I really need more red cells and is making lots of this hormone, erythropoietin, giving more of it is not going to help the system. But in people who don’t have a really high level it can be very beneficial.  

The other thing that can help with anemia, specifically, is a drug called danazol.  

It’s been around for a very long time. There are multiple presumed mechanisms of action, but one of them is that it is kind of a testosterone derivative. So, this is a medicine that can often help increase red blood cells in probably about 40 percent of people, and it’s a pill that you take twice a day. 

Another option, sometimes we use thalidomide or lenalidomide (Revlimid). These are medications that have been used quite frequently in the setting of multiple myeloma and even a little bit in myelodysplastic syndrome, so some other blood disorders.  

But in the setting of myelofibrosis, they can be helpful with anemia and sometimes are combined with prednisone or a corticosteroid. 

And then finally, in terms of drugs that are being tested and hopefully will be approved at some point in the future. There is a drug called momelotinib, which is another JAK inhibitor that actually has some mechanisms that may also help improve hemoglobin.  

So, this is something I’m really looking forward to and we anticipate may be approved by the end of the year. And finally, there is another drug called luspatercept. Luspatercept may work in the setting where your kidneys are already producing enough erythropoietin. So, the luspatercept is an injection that you receive once every three weeks.  

It is currently FDA-approved for the treatment of myelodysplastic syndrome but this is something that has been shown to have some efficacy in myelofibrosis as well. So, this could be another therapeutic option for patients with myelofibrosis. 

It is also important, especially for people who have polycythemia vera myelofibrosis to make sure that your iron has been checked and B-12 has been checked, because just because you have a bone marrow disorder doesn’t necessarily mean you don’t have a nutrition deficit that may be able to help improve your hemoglobin somewhat. But these are important things to talk to your doctor. I do not recommend just starting to take iron or B-12, however, if you’re anemic because in many cases you are not deficient and taking too much iron can actually be damaging.  

Katherine Banwell:

Yeah, that’s great advice.  

What Are Common MPN Symptoms?

What Are Common MPN Symptoms? from Patient Empowerment Network on Vimeo.

Dr. Jeanne Palmer, an MPN specialist, reviews the most common symptoms associated with essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF).

Dr. Jeanne Palmer is a hematologist specializing in myeloproliferative neoplasms (MPNs) and bone marrow transplant at the Mayo Clinic in Arizona. Dr. Palmer also serves as Director of the Blood and Marrow Transplant Program and is Vice Chair and Section Chief for Hematology. Learn more about Dr. Palmer, here.

 

Related Programs:

 
How to Treat PV-Related Itching

How to Treat PV-Related Itching

How Can Patients Navigate Care and Thrive With an MPN?

How Can Patients Navigate Care and Thrive With an MPN?

Thriving with an MPN What You Should Know About Care and Treatment

Thriving with an MPN | What You Should Know About Care and Treatment


Transcript:

Katherine Banwell:

Would you walk us through the common symptoms of each of the MPNs? Let’s start with essential thrombocythemia. 

Dr. Jeanne Palmer:

Right. So, there are a number of shared symptoms throughout all the diseases and when we start to figure out how to categorize them, they call into several different categories. The first one is inflammation-related symptoms. We know that the inherent pathway that’s dysregulated or that causes these diseases to happen can also result in significant inflammation in a person, that can result in things like fevers, night sweats, weight loss, and overall feeling really fatigued and poorly, which is something that it seems to be much more prevalent in patients with MPNs, all sorts of them, actually. 

The next set of symptoms is related to microvasculature, so all the little blood vessels. And sometimes we think, oh, maybe that’s because there’s too many red blood cells or platelets and the blood become viscous. It’s probably more related to the actual dysregulation of that JAK2 pathway, which is inherent to all the myeloproliferative diseases and as a result, the little blood vessels can clamp down and that can give people headaches, visual changes, numbness and tingling in the hands and feet, and even can cause sort of a painful rash called erythromelalgia in the body. 

So, these are things that can happen that are probably less appreciated side effects of the disease. And finally, there’s spleen-related symptoms. The spleen is in the left upper quadrant of the abdomen and it’s an organ that generally is about 12 centimeters in length, 10 to 12, but in patients with myeloproliferative diseases it can be enlarged. And as a result of an enlarged spleen people can have feeling like they get fuller early. So, if you’re eating a meal, all of the sudden you can only eat half of that meal versus the whole meal. 

Discomfort or pain in the left upper quadrant. Sometimes it’s much more noticeable when you like bend over to tie your shoes. And then sometimes people can actually, when the spleen gets really big, the blood flow can be impaired towards the end of it which can cause some of the spleen tissue to die, and that can be painful. So, these are things that if somebody does start to notice that they’re having fullness in the left upper quadrant, pain, stuff like that, that that may be related to spleen symptoms.  

Katherine Banwell:

What about PV or polycythemia vera, what are the symptoms? 

Dr. Jeanne Palmer:

So, all of these sorts of relate to all of the myeloproliferative diseases. So, one other one that I didn’t mention, and this is actually more in PV than others, is itching. Itching can be absolutely unbearable when somebody has PV. It’s particularly noticeable after taking a shower. So, a lot of times I’ve met patients who are like I haven’t been able to take a shower in years, because it causes such a high degree of itching. 

Katherine Banwell:

Why a shower? Is it different from having a bath?  

Dr. Jeanne Palmer:

Water on the body that can cause the problem. So, if people take hot showers, it’s even worse. Although I think that people sort of react to it differently. Usually what patients end up doing is more like sponge bath type of things, rather than actually being exposed to the water. 

 Taking colder showers or cooler showers can sometimes help mitigate that. But the itching, and even in the absence of a shower, people can have pretty severe itching, and that can also be one of the major side effects. 

Thriving With an MPN | Tips and Support for Navigating Care

Thriving With an MPN | Tips and Support for Navigating Care from Patient Empowerment Network on Vimeo.

Dr. Joseph Scandura, an MPN specialist, discusses the management and monitoring of essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF), and shares resources and support for managing day-to-day life with an MPN.

Dr. Joseph Scandura is Associate Professor of Medicine and Scientific Director of the Silver MPN Center at Weill Cornell Medicine. Learn more about Dr. Scandura, here.

 

Related Programs:

 
Thriving with an MPN What You Should Know About Care and Treatment

Thriving with an MPN: What You Should Know About Care and Treatment 

What Are the Signs of MPN Progression?

What Are the Signs of MPN Progression? 

How Treatment Goals Impact MPN Treatment Decisions

How Treatment Goals Impact MPN Treatment Decisions 


Transcript:

Katherine:

Hello, and welcome. I’m Katherine Banwell, your host for today’s webinar. This program is part of our Thrive series. And, today, we’re going to discuss navigating life with an MPN. 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. Joseph Scandura. Welcome, Dr. Scandura, would you please introduce yourself?

Dr. Scandura:

Hi. I’m Joe Scandura. I am Associate Professor of Medicine at Weill Cornell in New York City. I am a Physician Scientist. I actually run a lab studying MPNs and hematopoietic stem cells. And I am Scientific Director of the Silver MPN Center at Cornell.

Katherine:

Thank you so much for taking time out of your schedule to join us today. We start all of the webinars in our Thrive series with the same question. In your experience, what do you think it means to thrive with an MPN?

Dr. Scandura:

As a goal, I think it’s very simple, symptom-free and normal life expectancy. Thriving with an MPN is living your life as though you didn’t have an MPN.

Katherine:

And one part of thriving with an MPN is finding a treatment approach that manages your disease, the symptoms of your MPN, and that fits with your lifestyle. So, what are the factors that are considered when choosing treatment for patients with ET, PV, and MF?

Dr. Scandura:

Certainly, the goals of the therapy. So, is the therapy one that I would be looking to maybe delay progression or for long-term potential benefits, or is it something I need now to control short-term risks such as blood clots? The goals of the patient because some therapies may be more suitable to the goals of one patient than another. 

And the other – you know, there’s clinical features that may kinda push towards one approach versus another. Certainly, in a 20-year-old patient, I’m thinking about fertility. I’m thinking about a normal life expectancy. In a 90-year-old patient, I have a different set of concerns, multiple medications – what am I going to do that might be affecting their other comorbid conditions?

Katherine:

Right. Right.

Dr. Scandura:

I think about what are my near-term and long-term goals. So, obviously, age becomes a factor there. If I’m 95 years old, no matter what I do that person is not going to live 20 years. If that person’s 20 years old and they’re not living 30, 40, 50, 60 years, that’s a real shame. That’s a huge loss of life. So, that helps kinda point me in one direction or another. 

And, then, there’s different types of therapy. There are injectable agents. There are pills. There are drugs that have been used for a long time but don’t really have an FDA approval. There are drugs that are approved for certain indications. 

And, as physicians, we can sometimes stretch that based upon clinical judgment. So, I think a lot of that goes into the discussion I have with patients about therapy.

And that’s always – you know, I present to them what the options are, what I think the benefits might be, what the potential toxicities are, and then we discuss.

Katherine:

Right. I would imagine monitoring patients is different for each of the MPNs. So, how are patients typically monitored over time, and let’s start with essential thrombocythemia? 

Dr. Scandura:

Yeah. I think – again, it’s similar. You know, what’s near-term, what’s long-term? And so, in all of these diseases, thrombosis risk is a near-term risk. That’s something that I am monitoring in certain ways to help mitigate that risk. In ET and PV, I approach them similarly. Blood counts are certainly – these are diseases of the blood forming system. Certainly, monitoring blood counts I find helpful. But the reality of it is, in ET, there is not a clear linkage between blood counts and risks. 

And so, I like to keep the platelet count near normal if I can. But I also recognize that it may not be worth suppressing all of the blood counts to achieve that landmark, because it’s not clear that that’s really reducing the risk any more than just having somebody on a medication that helps control the blood counts. In polycythemia vera, different blood counts are very important. The red blood cells are kind of like part of the clotting risk. We know from clinical trials that keeping the red blood cell parameters within certain ranges reduces the risk of clotting. And so, what I monitor in polycythemia vera is the hematocrit. In women, I like to keep it below 42. In men, I like to keep it below 45. 

But I don’t just – I’m not a slave to the hematocrit. I am keeping an eye on the other blood counts and the other red blood cell parameters. So, for instance, what’s the size of the red blood cells? That tells me a little bit about what’s going on in the blood formation for that patient. And what’s the number of red blood cells? So, sometimes people can have very small red cells, because they’re a little iron-deficient and have a huge surplus of the number of red blood cells. And that tells me a little bit about how their blood forming system is responding to therapy. 

Iron deficiency in polycythemia vera is very prominent. I personally believe it’s a very major driver of symptoms in patients who are receiving phlebotomy as part of their care. And it’s something that I monitor and really counsel patients on. My goal is to make phlebotomy independent, but it can take a while. 

Everybody starts out iron-deficient, and then we take iron out of their body through blood with the phlebotomy. And that makes them more iron-deficient. 

Katherine:

Right.

Dr. Scandura:

I monitor symptoms from patients, and sometimes that can tell me that their disease needs to be – their treatment needs to be tweaked a little bit, even something as simple as aspirin. People can sometimes have burning in the skin or itching that is sometimes responsive to changing the aspirin dose or how it’s given, once a day versus twice a day. 

And that simple thing can be a big change for a patient who’s kind of, literally, climbing out of their skin or wishing they could and to try and find something that is helping.

I had a patient the other day. He had COVID. I said, “Oh, you should probably get this medication.” Do you have your primary care physician? Who’s taking care of you?” And he goes, “Well, to be honest with you, you’re my guy.” And so, it’s true. I see this patient a lot. And so, sometimes they forget. If I’m not paying attention to their blood pressure, the risks or treatment of diabetes, cholesterol, lipids, their screening programs for mammogram or colonoscopy, health maintenance issues, I do keep an eye on that in patients, because I do think it’s a part of the MPNs. 

I think that there are excess risks for patients for some of these factors. Certainly, if you think of it as three strikes, they get a strike for having an MPN. I don’t want them to have any other strikes. So, diabetes, hypertension, those are strikes that I can potentially, at least, treat or refer them to somebody to help comanage with me. And so, that’s kind of my general approach.

Katherine:

What about patients who have myelofibrosis? Are they monitored more closely?

Dr. Scandura:

Yeah, I think it depends a little bit on the patient. Patients with early myelofibrosis often don’t have any symptoms or near-term risks much different than those from ET or PV. As the disease can progress, then some of these patients have more profound problems with symptoms, which I may be trying to find a solution to make them feel better. And also, blood counts can become more of an issue. 

Transfusions in some patients who are very high white blood cell count, the spleen is often quite enlarged. Although, in my experience, most patients aren’t really bothered by the size of their spleen as the physicians are. But it is something where I think, on average, they’re monitored a little bit more closely to quite a bit more closely depending on the patient.

Katherine:

Yeah. You mentioned blood counts. And we know that lab results can fluctuate a bit. What happens if someone suddenly has a change in blood counts? What do you do?

Dr. Scandura:

Yeah. I mean, repeat it. That’s the first thing. Also, check what’s going on. It’s not uncommon in patients with MPNs that I’ll see them and the counts are a little bit out of whack, the white count is much higher than it’s been, and questioning them. “Oh, yeah. I had X, Y, or Z last week or the week before.” It used to be a upper respiratory tract infection, or they had a minor surgical procedure. 

And sometimes the responses to these things can be accentuated in patients with MPNs. And so, if that’s what of this story, I certainly would repeat it and let things calm down a little. And that’s often all it is. I’m much more of a monitor of the trends. So, one-time measure doesn’t generally excite me. It might make me want to have a follow-up a little more – in a shorter period of time. Of course, it depends on what the change is. But, for most of the changes that we observe, they’re relatively minor. And I will monitor them over time. 

If I see a trend where something is progressively increasing or decreasing over time, then I start thinking about what else is going on. And that’s always in the context of what’s going on with the patient. How are they feeling? What’s their physical exam like? What are the other laboratory values like? 

Katherine:

When is a bone marrow biopsy necessary?

Dr. Scandura:

I would say a bone marrow biopsy is absolutely necessary at the time of diagnosis. I personally do not routinely monitor by bone marrow biopsy unless it’s part of a clinical trial. 

But I do perform a bone marrow or want to look at the bone marrow morphology if there is one of these changes or at least a trend that I want a little bit more information about. And so, if – or if it’s been a very long time since somebody has had a bone marrow. If it’s been five or ten years, then sometimes I may recommend we look just so we can collect a little bit more up-to-date information. 

But I don’t routinely do a bone marrow, but I will do it if there are laboratories that are kind of trending in the wrong direction, there’s symptoms, there’s physical findings that I’m just not sure about. And I think it would help me be more sure as to what’s going on and be able to discuss that with the patient. Sometimes, just to say, “Hey. Look, we were worried about this, but the bone marrow looks really good.” 

Katherine:

Yeah. Can you talk about shared decision-making? Why is it so important for patients to work closely with their healthcare team on choosing a therapy?

Dr. Scandura:

Because these are therapies that last for a long time. And, hopefully, the patients and the relationship last for a long time. And so, I think that everybody has to be comfortable with the decision about a therapy. And my personal goal is to try to make sure that everybody understands the rationale for a therapy, the potential ups and downs with the therapy, which every drug has, every approach has, and what I’m kind of watching and monitoring. I’m a very – I think that communication relieves a lot of anxiety. I think that the unknown is far scarier than the known, even if it’s not perfect. And so, I think shared decision-making has a role in relieving some of the scariness of unknown. 

If we’re discussing to come to a decision, that means that my job is to give you the knowledge that I have so that you can tell me the knowledge about you and what you’re feeling and what you want back. And that back and forth is what helps me do a better job of taking care of the patient and helps the patient understand what’s going on and relieve some of the stress of the unknown. So, I think it’s a very synergistic approach. I don’t think I could practice medicine in another way.

Katherine:

Dr. Scandura, much of our MPN community is highly engaged in their care. What are some educational resources you would recommend for people who are seeking more information about their condition?

Dr. Scandura:

I think that there’s some basic information available from a variety of – for instance, the National Cancer Institute has some basic information. Leukemia & Lymphoma Society has some basic information. 

The MPN Research Foundation has some basic information. And then there are some information websites that are run by corporations, which are – I think they try to be even-handed in some of the discussion and has some good information there, too. I think the – none of these is a perfect source of information. I don’t think there is one source that you can go to answer every question that you could ask. 

My MPN Center has a website with a bunch of QAs, and we just every now and then add a new one. And it’s just a really long list. So, these are questions our patients frequently ask us, and we sort of put answers there to help guide. But individual details are often more important than sort of generalizations. I find patient – go ahead.

Katherine:

Oh. I was just going to ask, what about the forums, patient forums that are available? Is that something you would recommend? 

Dr. Scandura:

What I kind of I find my patients do is they’ll go out and look for information, because patients with MPNs, thankfully, tend to live a long time. And they are often curious about their disease and want to do better and figure out how they can do better. And so, a lot of them will go to whatever sources are available. But, generally, they come back. So, we circle back; we regroup. And sometimes, it’s la-la land, a little bit crazy things, and sometimes it’s really interesting. 

I learn a lot, you know, what’s going on in terms of what are patients really reporting, because sometimes in a clinic visit people kind of don’t say everything, or they forget to say something or maybe just my experience. I don’t see every patient in the world, right? So, if it’s something that’s relatively rare, then I may not have seen it with a new drug or something like that. 

So, I can learn from that experience as well. So, I think it’s kind of like people go out. They can be like little honeybees and collect all the information from all the flowers out there. And then they come back, and we regroup in the nest. And we discuss and decide what makes sense, what’s relevant to them, and what might help with our decision-making.

Katherine:

Yeah. Managing the worry associated with a diagnosis or concerns even about progression can lead to a lot of anxiety and fear amongst patients. Why is it important for them to share what they’re feeling with their healthcare team?

Dr. Scandura:

I would say this. If our goals are to have people – I mean, this is what I say to patients – I want you to think about this disease when you’re here. And, then, when you’re not here, my goal is to have you not thinking about this disease because you’re feeling okay and you’re comfortable and confident in what’s going on. 

So, I want to make it a clinic visit disease. That’s not always possible. But, for many patients, it is. I don’t want somebody to become – to start thinking like a sick person when they’re not. I don’t want the diagnosis to be the disease, right? I want the person if they’re feeling well, to recognize that. Live your life; move on with things. But, at the same time, these kinds of diagnoses are scary. 

Katherine:

Yeah.

Dr. Scandura:

And so, it is normal with a new diagnosis or a change in the diagnosis to go through a period of time where you have to adjust. And so, that’s normal, and you have to work your way through it. Some people want to work that all out internally, and that’s good to a certain extent as long as they have good supports at home. But I often want to know how they’re doing, how they’re working through that so I can get a gauge of how it’s affecting their life and the duration where this adjustment is going on. 

So, somebody who’s still adjusting to a new diagnosis two years after the diagnosis, and they’re otherwise clinically well, that’s getting into the range where it’s not normal. You might need additional help. You might need counseling. And, in some patients, that might include some medications for a short period of time. The goal is to have the disease affecting you only in so far as it’s affecting you, not the idea of the disease.

Dr. Scandura:

So, that’s a – again, it’s a conversation. There are lots of resources. People, being individuals, deal with things in their own way, and I just try to help understand with them how it’s affecting their life. And, if it seems to be more than I would expect, I’ll tell them that. 

And then we can discuss that. It doesn’t mean we have to do something today, but I will tell them, “I think this is maybe a little bit more. Why are you so worried? I think you’re doing great.”

Katherine:

Yeah. Yeah. Can a social worker or somebody else on the healthcare team help with these emotional needs that patients have?

Dr. Scandura:

Absolutely. We have great social workers. I tap into them all the time. We also have a group of psychiatrists who are really interested in kind of psychiatry that’s related to oncology and the diagnoses and how it impacts care. I mean, this is New York City, so everybody has a therapist. But a lot of patients have preexisting connections to healthcare providers or support systems. I think, for some patients, groups are helpful.

Katherine:

We’d be remiss if we didn’t bring up financial concerns, treatment and regular appointments can really become quite expensive. Understanding that everyone’s situation is different, of course, where can patients turn if they need resources for financial support?

Dr. Scandura:

Yeah. It depends on what the issue is. So, one of the biggest areas that I found this can interfere with care is when we have copays that are really not reasonable and not affordable. And so, how do we fix that? How do we get access to an agent that might be beneficial for a patient but that – you know, and the insurance has approved it, but they’ve approved it with such a high copay that it’s just not an option anymore. 

And so, there are foundations. The PAN Foundation, we often will reach out to for copay assistance. And, actually, many companies have copay assistance programs for their individual drugs. And so, we have some of our nurses who are quite good at navigating these different agencies, and some of them are kind of drug-specific. 

And because we see a lot of patients with MPNs and the number of drugs is not that great, we’re pretty tapped into what are the options for copay assistance that might be helpful. And it often works. It doesn’t always work. I had a patient I saw pretty routinely, and I kind of like my certain group of labs that kind of make me feel like I have a good sense of what’s going on. But he was getting killed with the lab costs. And he mentioned this to me, and then I have to do what I tell my – I have three teenage daughters, right? And, when they were littler – smaller, younger, we spent a lot of time distinguishing needs from wants, right? 

So, this was one of those instances. What laboratory do I need to make sure that this patient is safe? What do I want because it makes me feel like I have a better idea of what’s going on? And maybe I can back off on those wants if I’m seeing the patient pretty frequently, which I happen to be at that time. And so, some of that is a conversation. 

And it depends on the specifics of the insurance and a little bit of back and forth and knowing how to kind of minimize that financial burden when that’s starting to compromise care.

Katherine:

Yeah. Let’s answer a few audience questions that we received in advance of the webinar. This one is from Sophie, “What complications can arise from an MPN during pregnancy?”

Dr. Scandura:

Well, look, pregnancy – here you have two things, one of them common and complicated and the other one uncommon and complicated. So, common is pregnancy, but every pregnancy is different. And there’s a lot of changes going on in the body, and there’s certain risks that can go along with that as well. So, clotting risks sometimes can be increased in pregnancy. And then you have an MPN, where you have a clotting risk on top of that. The pregnancy really changes what kinds of medications we can think about using. And so, there are certain medications that we use comfortably in patients that would be an absolutely forbidden medication in a pregnant woman. 

And so, it depends a little bit on what’s going on with the patient. But, if they have a history of clotting, then certainly, we would think about wanting to control the blood counts. It depends a little bit on what the disease is how we would do that. Interferons are commonly used in pregnancy, and they are safe in pregnancy and can improve the outcomes in some patients with pregnancy. 

But short of that, in patients, for instance, who are very thrombotic risk, sometimes we have to sort of balance the risk of having a clot and something that can interfere with the pregnancy and the risk of bleeding. So, it’s not uncommon that people are on blood thinners during pregnancy at some point, but it really depends on the individual patient. What we do here is we keep very close contact with the patients. 

And all of our patients are seen by the high-risk OB/GYN. So, it’s not the general obstetrics people who are monitoring the patient, so they’re much more closely monitored for complications of pregnancy. And we are seeing them more frequently during pregnancy to help, from the MPN side, to try to optimize and minimize the risks of clot. And that doesn’t end as soon as the baby’s out. If breastfeeding, their clotting risk is not normalized after pregnancy, as soon as the baby comes out. And so, you know, there’s an adjustment for several months afterwards where we’re still kind  of thinking about this person a little bit differently than we would if they were not or had not been recently pregnant. 

Katherine:

Yeah. We have another question. This one from Jennifer. She wonders, is there research being done on MPN progression to understand how it happens or even prevent or slow progression?

Dr. Scandura:

Yeah. There’s a lot. I think there is a – from both the sort of basic laboratory using animal models to try to understand what are the kind of systems that are involved in how these diseases change. What genes are involved? How do they talk to each other? You know, these are not cells that live in a vacuum, right? They live in a special microenvironment. What are the signals that crosstalk between the MPN cells, the MPN stem cells, and their microenvironment? 

And so, there’s a lot of research on that and the basic side of things. In humans, there’s a lot that has been done over the years in terms of trying to understand what are some of the genetic features of progression. And I think we’re beginning to get a little bit of a better understand of what are the non-genetic things that are associated with progression. 

I was part of an effort from the MPN Research Foundation and still am. They have what they call the Progression Network, where they tried to put together a number of investigators from really across the world to share ideas about the nature of progression and how we might look at studying this and understanding ways to prevent progression. 

I think we do have some drugs now that show some promise in terms of being able to prevent progression. I think interferons have shown this in polycythemia vera in terms of a promise for improved long-term outcomes and delayed risk progression. I think that the gold standard randomized trials are maturing and are sort of bearing out some of the same findings that have been observed retrospectively, so sort of kind of looking back in time. 

But the difficulty is that it can take a long time for patients to progress. And you say, “Oh, that’s great.” And that is great. But, from a research – from a statistical side, it means things are really slow. If you have to wait 15 years to assess whether or not people progressed less in one treatment versus another, it’s really slow going. And so, we have to do a compromise of what’s – you know, what do animal studies say? What does retrospective analysis, when we might have people who started treatment 30 years ago, and now we’re just seeing how did it all work out? It’s not a perfect study, because biases can creep in, but it’s what we have now. And so, there’s a lot. And I think, increasingly, progression is being recognized as a goal of therapy, to prevent progression. 

Personally, it is one of major goals, because I think we do a pretty good job at preventing clots with available treatments. But I don’t think we do a very good job at preventing progression, mostly, because we don’t exactly understand what’s driving that. And so, I think until we develop that deeper understanding and really invest the time and effort in terms of learning which approaches can help prevent progression, we’re going to continue to have these questions.

Katherine:

Yeah. Well, thank you for those answers, Dr. Scandura. And please continue to send in your questions to question@powerfulpatients.org, and we’ll work to get them answered on future programs. As we close out this conversation, Dr. Scandura, I would like to get your thoughts on where we stand with progress with MPN care. Are there advances in treatment research that you’re hopeful about?

Dr. Scandura:

Yeah. I think it’s a very exciting time, actually. I think that over the past 5 to 10 years the amount of new drugs that have been developed and tested in patients has grown exponentially. The number of companies that are targeting MPNs for their drug development has expanded dramatically. The number of clinical trials, good quality clinical trials has increased dramatically. And I think the success that’s coming out of that is we start seeing drugs now that are looking to be very, very effective. I don’t want to name individual drugs. 

But I know we have a number of clinical trials where we’re seeing things with these agents that we haven’t seen with our traditional therapies, meaning changes in the bone marrow that we haven’t seen before or a normalization of symptoms or blood counts in an area that has been challenging in the past. And so, we now have drugs and a number of drugs going for approval, a number of newly-approved drugs, even interferon, which is a drug that’s been around forever. Well, not forever. But, I mean, I guess forever, yeah, because it’s a natural product. 

So, as long as there have been humans, there have been interferons, even before humans. But now we have it. As a pharmaceutical, they’ve been around for decades. And we now have the first – even though we’ve been using it for decades, we have the first approved, FDA-approved interferon for polycythemia vera, which is I think a huge change. 

A company invested the money in getting FDA approval for an agent, and that means they have to – the bar’s higher, and they have to prove something that just using it off-label hasn’t. So, I think it’s a tremendously exciting time. I expect it’s going to continue. We’re going to continue to have improvements in care. There’s going to be combinations of drugs. I think that we’re going to see real advances over the next 5 to 10 years.

Katherine:

Well, Dr. Scandura, thank you so much for taking the time to join us today. 

Dr. Scandura:

It was a pleasure. It was nice meeting with you.

Katherine:

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

Thriving With MPNs: Your Role in Managing Your Treatment and Care

Thriving With MPNs: Your Role in Managing Your Treatment and Care from Patient Empowerment Network on Vimeo.

 How can patients thrive with a myeloproliferative neoplasm (MPN)? Dr. Jeanne Palmer discusses treatment approaches, strategies for managing disease symptoms and treatment side effects, and advice on how patients can be proactive in their care.

Dr. Jeanne Palmer is a hematologist specializing in myeloproliferative neoplasms (MPNs) and bone marrow transplant at the Mayo Clinic in Arizona. Dr. Palmer also serves as Director of the Blood and Marrow Transplant Program and is Vice Chair and Section Chief for Hematology. Learn more about Dr. Palmer, here.

 

Related Programs:

 
How Should You Participate in MPN Care and Treatment Decisions?

How Should You Participate in MPN Care and Treatment Decisions?

What Are the Signs of MPN Progression?

What are the Signs of MPN Progression

Expert Perspective: Hopeful MPN Research and Development


Transcript:

Katherine Banwell:

Hello and welcome. I’m Katherine Banwell, your host for today’s program. Today, we’re going to talk about how to live and thrive with an MPN. We’re going to discuss MPN treatment goals and how you can play an active role in your 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, joining us today is Dr. Jeanne Palmer. Dr. Palmer, welcome. Would you please introduce yourself? 

Dr. Jeanne Palmer:

Thank you so much. I am so happy to be here and to help participate in this. My name is Jeanne Palmer. I am a hematologist at Mayo Clinic in Arizona. I specialize in MPNs as well as bone marrow transplant, and I am thrilled to be here. 

Katherine Banwell:

Thank you for taking the time out of your busy schedule to join us today, Dr. Palmer. We start all of the webinars in our Thrive series with the same question and that is, what does it mean to you to thrive with an MPN? 

Dr. Jeanne Palmer:

I think living with an MPN can be very difficult. I think there is a number of things. First of all, there’s always the worry of what’s going to happen in the future. Many of these MPNs can start as fairly, for lack of a better term, as benign issues and can convert to something much more serious. So, I think living with that sort of timebomb in the back it can be extremely stressful. So, figuring out how to live with the fact that there is some degree of uncertainty. 

I think the other thing is making sure to understand your disease. These are very rare disorders and even if you go to a hematologist-oncologist specialist, a lot of times they don’t have all the information because they don’t see a lot of them every year. So, it’s really important to make sure that above and beyond that you understand what’s going on in your body so that when new things happen, new symptoms happen, you’re able to really address them as opposed to sort of living with something that may make you feel poorly that’s not being addressed.  

So, again, I think the biggest piece of this is seeing how do you live with uncertainty, and how do you make sure you understand your disease well enough that you know what’s going on in your own body. 

Katherine Banwell:

Yeah. That’s helpful to understand, especially as we move through today’s program, and we’re going to cover the three classic MPNs, polycythemia vera, essential thrombocythemia and myelofibrosis. One part of thriving with an MPN is managing the symptoms of the disease. Would you walk us through the common symptoms of each of the MPNs? Let’s start with essential thrombocythemia. 

Dr. Jeanne Palmer:

Right. So, there are a number of shared symptoms throughout all the diseases and when we start to figure out how to categorize them, they call into several different categories. The first one is inflammation-related symptoms. We know that the inherent pathway that’s dysregulated or that causes these diseases to happen can also result in significant inflammation in a person, that can result in things like fevers, night sweats, weight loss, and overall feeling really fatigued and poorly, which is something that it seems to be much more prevalent in patients with MPNs, all sorts of them, actually. 

The next set of symptoms is related to microvasculature, so all the little blood vessels. And sometimes we think, oh, maybe that’s because there’s too many red blood cells or platelets and the blood become viscous. It’s probably more related to the actual dysregulation of that JAK2 pathway, which is inherent to all the myeloproliferative diseases and as a result, the little blood vessels can clamp down and that can give people headaches, visual changes, numbness and tingling in the hands and feet, and even can cause sort of a painful rash called erythromelalgia in the body. 

So, these are things that can happen that are probably less appreciated side effects of the disease. And finally, there’s spleen-related symptoms. The spleen is in the left upper quadrant of the abdomen and it’s an organ that generally is about 12 centimeters in length, 10 to 12, but in patients with myeloproliferative diseases it can be enlarged. And as a result of an enlarged spleen people can have feeling like they get fuller early. So, if you’re eating a meal, all of the sudden you can only eat half of that meal versus the whole meal.  

Discomfort or pain in the left upper quadrant. Sometimes it’s much more noticeable when you like bend over to tie your shoes. And then sometimes people can actually, when the spleen gets really big, the blood flow can be impaired towards the end of it which can cause some of the spleen tissue to die, and that can be painful. So, these are things that if somebody does start to notice that they’re having fullness in the left upper quadrant, pain, stuff like that, that that may be related to spleen symptoms. 

Katherine Banwell:

What about PV or polycythemia vera, what are the symptoms? 

Dr. Jeanne Palmer:

So, all of these sorts of relate to all of the myeloproliferative diseases. So, one other one that I didn’t mention, and this is actually more in PV than others, is itching. Itching can be absolutely unbearable when somebody has PV. It’s particularly noticeable after taking a shower. So, a lot of times I’ve met patients who are like I haven’t been able to take a shower in years, because it causes such a high degree of itching. 

Katherine Banwell:

Why a shower? Is it different from having a bath?  

Dr. Jeanne Palmer:

Water on the body that can cause the problem. So, if people take hot showers, it’s even worse. Although I think that people sort of react to it differently. Usually what patients end up doing is more like sponge bath type of things, rather than actually being exposed to the water. 

Taking colder showers or cooler showers can sometimes help mitigate that. But the itching, and even in the absence of a shower, people can have pretty severe itching, and that can also be one of the major side effects. 

Katherine Banwell:

Much of the time the chosen treatment for MPNs manages the symptoms of the condition. I’d like to review the different types and classes of treatment for the three MPNs. So, let’s start with essential thrombocythemia again. When is it time to treat, and what are the options available?  

Dr. Jeanne Palmer:

Right. So, with essential thrombocythemia, that’s the disease that sometimes we don’t need to treat. 

So, we basically have a risk stratification system and this risk is based on age, history of a blood clot, the presence or absence of a JAK2 mutation. So, for example, if somebody is 28, does not have a JAK2 mutation, which is again one of those driver mutations, and never had a blood clot, they actually don’t necessarily need to do anything and just be monitored.  

Somebody who is less than 60 and has a JAK2 mutation or who is greater than 60 and does not have a JAK2 mutation, in that setting, a lot of times you can use aspirin. Now, it gets a little bit gray in terms of that over 60 without the JAK2 mutation with regards to whether at that point you really should start taking some medicine to lower the platelets. 

Now, if somebody has a JAK2 mutation, is greater than 60 or has had a blood clot, hands down they need to take medicine to lower the platelets, in addition to aspirin or whatever blood thinner they may need. So, for example, if you have a blood clot in a vein, a lot of times you need to take a blood thinner and that will be a lifelong thing. And again, we do these risk stratifications because we know there is a certain risk of clotting associated with the risk of essential thrombocythemia. 

So, for example, somebody who is less than 60 and does not have a JAK2 mutation, never had a clot, their risk of clotting is probably very close to that of the normal population. Whereas if you’re higher risk and have a JAK2 mutation and greater than 60 or have had a history of a clot, the risk of clot is probably about 4 percent per year. So, this is something that can vary quite widely, and even though that 4 percent per year on the short-term doesn’t sound like a lot, if you take it additive over years, that’s why we generally try to be aggressive about lowering the platelets.  

In lowering the platelets, the goal is to get less than 400 and doing that can be done through several different medications. The most commonly used medications is a drug called hydroxyurea, which has been around for a number of years, and a drug called anagrelide which is probably a little less commonly used, because it has some more GI side effects and headaches associated with it. 

In some cases, especially in younger patients with this disease, we can consider using interferon, which is an injection of a cytokine, which are one of the chemicals that regulates the immune system within the body. But this interferon can actually help lower the platelets and there is a question of whether it may affect the biology of the disease as well. 

Katherine Banwell:

Let’s turn to polycythemia vera or PV, what are the different options available for treating it? 

Dr. Jeanne Palmer:

So, for polycythemia vera, everyone needs to be on aspirin. 

And additionally, everyone needs to make sure to keep their blood count low, to manage their hematocrit, which is one of the measures of red blood cells. So, in men it’s generally recommended to keep below 45 and in women it’s recommended to keep below 42 percent.  Now, the studied number was 45 percent and that was a study that was done, I don’t know, it was probably about 10 plus years ago, that actually showed that by keeping the blood hematocrit less than 45 percent you reduce the risk of having negative events like cardiovascular events and heart attacks. Because women tend to run with a lower blood count than men, it’s been extrapolated that 42 percent should be the number used for women. 

Now, this can be done by phlebotomy, which essentially is bloodletting. It’s kind of like donating blood except for that the blood unfortunately can’t be donated to anybody, it has to be discarded. But the phlebotomy is one way to do that, and the reason that works is because it makes somebody iron deficient. So, whereas if this is normal, if you’re iron deficient you become anemic. If your baseline hematocrit is here, making you iron deficient brings you back to normal. So, even though we always associate iron deficiency with anemia, iron deficiency in the setting of polycythemia vera is actually kind of a treatment of sorts. 

Now, once somebody gets above 60 and 60 seems to be sort of the magic age in these diseases, once somebody gets above 60, it is recommended that cytoreductive therapy is used, which means therapy or treatment that will bring down the red count. And again, for this one, hydroxyurea is an option as well as interferon. And there is recently an approval, actually FDA approval for a newer interferon called ropeginterferon or Besremi, which can help just bring down the red blood cells but it is the first interferon that’s actually been FDA approved for this indication. 

Katherine Banwell:

Are JAK inhibitors used as well? 

Dr. Jeanne Palmer:

They are. So, if somebody doesn’t respond well to hydroxyurea, the approval for ruxolitinib is actually for patients who have failed hydroxyurea. Although it’s something that we often consider especially in people who have a lot of symptoms. So, the itching, one of the things that can really help itching actually is Jakafi. If people have night sweats, they have weight loss, spleen related symptoms, those are the patients that will benefit from Jakafi. Additionally, if they are on hydroxyurea and can’t seem to get control of their blood count, Jakafi is a good option to help control the blood counts as well. 

Interferon is a very nice option because there’s great data that shows that you may actually be able to lower the percentage of JAK2 burden. 

So, we’d look at something called an allele burden, which is the percentage of cells that are involved – have the JAK2 mutation. Now, we don’t know whether lowering this percentage necessarily translates to long-term better survival, but I think there is enough data out there, and there is a good biologic underpinning for saying that this actually can help. But yes, Jakafi is another thing. 

And the really exciting thing is that there is a newer agent called rusfertide, which is a hepcidin mimetic, which is basically taking a protein in your body that helps metabolize iron and by making it externally and giving it to somebody that it can actually help bring down the hematocrit without having some of the other side effects we know with some of the other medications. That is currently in Phase III studies, so hopefully in the next couple of years we’ll see approval for that. 

Katherine Banwell:

Oh, that’s great news. And finally, how is myelofibrosis treated? 

Dr. Jeanne Palmer:

So, myelofibrosis is a little bit of a different animal. When you have something like essential thrombocythemia or PV, a lot of this is managing symptoms, preventing blood clots, but if you do appropriate treatment and management of these diseases you could probably live close to a normal life expectancy.  

So, I never typically pin a survival on it. With myelofibrosis, it’s a little bit different because there is a survival. Instead of saying you can live close to normal life expectancy, it backs up to saying how many years do I think you can live with this disease. Now, of course, we are horrible at predicting how many years anyone can live, so we have to take that all with a grain of salt. But we can at least sort of risk stratify people. 

And the first thing that’s really important is to figure out whether somebody is a transplant candidate or not and if, based on age, disease risk features, stuff like that, or whether we think they ever will be a transplant candidate. So, that kind of helps us sort of think about what your path moving forward is.  

Now, the current FDA-approved treatment for myelofibrosis, there are three JAK inhibitors approved, which is like Jakafi, which was the first approved one but there is also Inrebic or fedratinib and Vonjo or pacritinib and these have all been approved over the years. 

The role of JAK inhibitors and treatment of myelofibrosis is symptoms-based. So, for example, a lot of patients with myelofibrosis will have weight loss, night sweats, big spleens, really feeling fatigued and poorly and in this setting, the JAK inhibitor can be very helpful. And you don’t have to have a JAK2 mutation, a lot of times people say, well, I don’t have the JAK2 mutation so how can a JAK inhibitor help. So, the JAK inhibitor works on this pathway, which is called the JAK/STAT pathway, irrespective of mutation. 

So, if you are having symptoms and you have myelofibrosis, JAK mutation, excuse me, the JAK2 mutation does not predict who is going to have a response. And people who, regardless of which mutation you have, may actually benefit from it. 

So, the JAK inhibitors, though, are extremely effective at reducing symptom burden as well as reducing the spleen size. And we know that if a spleen is big and we can make it shrink that, that probably is a surrogate marker for living longer, and I think it’s because inflammation does a lot of wear and tear on the body. So if you can reduce the inflammation and the spleen shrinks, which generally go hand in hand, then you might help somebody live longer. It is not changing the biology of the disease, though, however, it doesn’t change the pathway and that this disease is kind of projecting ahead in terms of creating – it changes, as it goes along, may acquire new mutations or something like that which makes the disease become more serious. 

Right now, the approved therapies for it are JAK inhibitors and the Jakafi, ruxolitinib was the first one approved. Inrebic was approved several years back, or fedratinib. 

And then the most recent one that was approved is Vonjo or pacritinib and that’s a drug that is a JAK inhibitor that is actually very good for people with low platelets. The reason I bring that up is because if we think of what’s the biggest limiter of JAK inhibitors, JAK inhibitors bring down red blood cells, and they bring down platelets. So, when somebody has low platelets it’s very hard to use a JAK inhibitor, because we’re not really able to increase the dose well enough to get that inflammatory reduction because of the fact that the blood counts will drop too low. 

So, now drugs like Vonjo exist which, due to several other mechanisms associated with the drug are actually much more tolerated in somebody with low platelets. So, if you have low platelets, you can actually take the Vonjo, hopefully get the same degree of JAK inhibition to help the spleen shrink, help the symptoms get better without necessarily making the platelets substantially worse. A lot of times they do drop, it doesn’t help bring up the platelets, but it does help people tolerate more JAK inhibition, which ultimately will help with symptoms.  

Dr. Jeanne Palmer:

So, one thing I also wanted to add about myelofibrosis treatment is sometimes people present, they don’t have a lot of symptoms, they don’t have a lot of spleen related problems but they have anemia or low blood counts and these can be incredibly hard to treat. 

Even with symptoms and low red blood cell count or anemia or low platelets, it can be challenging to treat because many of these medications lower that. To treat the anemia there are several things that we can do. One of the first ones is using erythropoietin, and so there are many agents, they go by the names of like Procrit or darbepoetin alfa, that actually stimulate red blood cell growth by – like we give a recombinant hormone that helps red blood cells grow. This is normally something produced by the kidney. 

So, one thing that’s important before going on one of these injections is to make sure that the kidney is not already producing enough. So, for example, if the kidney said, oh geez, I really need more red cells and is making lots of this hormone, erythropoietin, giving more of it is not going to help the system. But in people who don’t have a really high level it can be very beneficial. 

The other thing that can help with anemia, specifically, is a drug called danazol (Danocrine).  

It’s been around for a very long time. There are multiple presumed mechanisms of action, but one of them is that it is kind of a testosterone derivative. So, this is a medicine that can often help increase red blood cells in probably about 40 percent of people, and it’s a pill that you take twice a day. 

Another option, sometimes we use thalidomide or lenalidomide (Revlimid). These are medications that have been used quite frequently in the setting of multiple myeloma and even a little bit in myelodysplastic syndrome, so some other blood disorders.  

But in the setting of myelofibrosis, they can be helpful with anemia and sometimes are combined with prednisone or a corticosteroid. And then finally, in terms of drugs that are being tested and hopefully will be approved at some point in the future. There is a drug called momelotinib, which is another JAK inhibitor that actually has some mechanisms that may also help improve hemoglobin. 

So, this is something I’m really looking forward to and we anticipate may be approved by the end of the year. And finally, there is another drug called luspatercept (Reblozyl). Luspatercept may work in the setting where your kidneys are already producing enough erythropoietin. So, the luspatercept is an injection that you receive once every three weeks.  

It is currently FDA-approved for the treatment of myelodysplastic syndrome but this is something that has been shown to have some efficacy in myelofibrosis as well. So, this could be another therapeutic option for patients with myelofibrosis. 

It is also important, especially for people who have polycythemia vera myelofibrosis to make sure that your iron has been checked and B-12 has been checked, because just because you have a bone marrow disorder doesn’t necessarily mean you don’t have a nutrition deficit that may be able to help improve your hemoglobin somewhat. But these are important things to talk to your doctor. I do not recommend just starting to take iron or B-12, however, if you’re anemic because in many cases you are not deficient and taking too much iron can actually be damaged. 

Katherine Banwell:

Yeah, that’s great advice.  

When would you consider a stem cell transplant? 

Dr. Jeanne Palmer:

So, the stem cell transplant is based on disease risk. There is a number of ways we assess disease risk. The first two ones that were published a number of years back were the DIPSS score, which is Dynamic International Prognostic System Score, or the DIPSS Plus, which basically is the DIPSS and then you add to it a few other clinical features. This symptom score is based largely on things that we can see without even a bone marrow biopsy, so things like symptoms, age, number of white blood cells, whether somebody has anemia. And then the number of something called blasts, which is very immature white blood cells. The DIPSS Plus takes into account low platelets, need for transfusions, and chromosome abnormalities, which is the only test among that that needs to be from a bone marrow biopsy. 

Now, these were created prior to Jakafi being commercially available. So, we have to take a little bit of a grain of salt with those because of the fact that Jakafi probably has changed how long people can live with this disease. 

Now, more recently they’ve tried to account for these other molecular changes. So, when we take the genetic landscape of these diseases, we have the known driver mutations, so the JAK2 mutation which I have talked about, also calreticulin and MPL.  

These three mutations all affect that one pathway, the JAK/STAT pathway, so they all affect the pathway that drives the disease and they are known to be kind of mutually exclusive and definitely contribute to the formation of the disease.  

Some of these other mutations are called somatic mutations. They could be checked by things next generation sequencing or genetic analysis. There’s a number of different names that people use for this testing, but we look for mutations that are present and these mutations, number one, can sometimes tell us risk. So, there’s certain mutations that are high risk. Other times it can actually give us other opportunities for therapy, especially of the disease progresses. But these mutations are important to know for risk stratification. For example, if somebody has DIPSS score that is maybe not super high risk, but then they have one of these mutations, we know that that probably makes their disease a little bit more aggressive. 

And that’s when we think about transplant, is when we know that the disease probably has an average life – when somebody gets to the point in their disease where we estimate their life expectancy is around five years, recognizing that we’re not very good at this. That is the type of point when we start to think about transplant. But the timing of transplant is something that’s extremely difficult and a very personalized decision. It’s something that it’s really important to understand the disease risks, how we assess them and the caveats of these disease risk assessments as we move forward planning and timing of transplant and that’s something that is, again, a very, very important discussion to have at length with your physician. 

And I always recommend, there is quite a few of us out there who actually specialize in transplant for myelofibrosis and having discussions with somebody who really understands the biology of the myelofibrosis and important because it’s very different than a lot of the other diseases that are transplanted.   

Katherine Banwell:

Yeah. Well, speaking of that, patients can sometimes feel like they’re bothering their healthcare team with their comments and questions. Why do you think it’s important for patients to speak up when it comes to symptoms and side effects?  

Dr. Jeanne Palmer:

Well, there is a lot of things. This is a disease, again, that we can direct our therapy many times towards symptoms, and so when we think about how do I direct my therapy, so how do I treat somebody, symptoms are an incredibly important part of it. And there is nothing worse than having a patient come and see me who I see every six months, because they’ve been pretty stable and they’re like, “Oh, for three months I’ve been feeling awful.” And you’re like, well, “Why didn’t you let me know, we could do something about this?” 

So, if there is something that doesn’t feel right, it’s very, very important to talk to your healthcare provider. I would much rather be bothered and handle something earlier on than miss something and really have a lot more catch-up to do afterwards. 

The other thing is symptoms may indicate a blood clotting event. We know that patients will have a higher risk of blood clotting. These are extremely important to identify early on because if they go unchecked, they can cause more damage. 

Katherine Banwell:

With many of the treatments available as pills now, patients have a role in self-administering their treatment regimen. What happens if a patient forgets to take a medication? Does it impact its effectiveness? 

Dr. Jeanne Palmer:

Generally no. I think the ones that would are certain blood thinners you really don’t want to miss and you don’t want to miss the doses on it. With drugs like Jakafi, if you miss one dose you probably won’t notice it, but if you miss multiple doses you can actually get very sick from that. So, some of these medications are really important to be consistent on. 

Now, I know this could be a challenge. I mean I don’t take very many medications and I sometimes have a hard time keeping track of what I take, so I know that this can be a difficult thing to do. So, one thing is if you really find you’re struggling with it, setting an alarm on your phone or your Apple Watch or whatever… 

Katherine Banwell:

…device. 

Dr. Jeanne Palmer:

Device you have can be a really helpful way of doing it. Also having a pill box. They make pretty amazing pill boxes these days that can account for taking drugs once a day, twice a day, three times a day. I’ve even seen them up to four times a day, although generally the most you’ll probably have to take a medicine for a myeloproliferative disease is twice a day. But those are different ways that can really help make sure you’re consistent about taking your medication. 

Katherine Banwell:

And if a patient misses a dose, do they need to call their healthcare team and let them know? 

Dr. Jeanne Palmer:

Not just for one missed dose. If like, for example, they’re run out and they say, “Oh, geez, I don’t have any and many of these drugs are specialty pharmacy,” so they need to be mailed, and you know that you’re going to be missing it for a while. Or let’s say you look at your pill bottle and go, “Oh shoot, I only have so many pills left,” it is helpful to call because a lot of times, for example, if somebody is on Jakafi and they know they’re going to run out of their pills four days before they’re going to get their next shipment in, then what I sometimes do is I lower the dose a little bit to make sure they maintain a dose throughout that time. 

But this is something you definitely want to do under the advice of a healthcare provider. You don’t want to just all of the sudden go, “Oh, well I’m going to run out so I’m just going to change my dose,” and kind of do that. 

Katherine Banwell:

Yeah, yeah. We received some audience questions prior to the program today. This one is from Jacqueline, “What can I do to minimize pruritus or itching due to PV? A typical histamine blocker like Claritin or Zyrtec has done nothing whatsoever.” 

Dr. Jeanne Palmer:

Yeah. Unfortunately, the itching of this is not as much mediated by an allergic type reaction or histamine. It’s a lot related to that microvasculature, those tiny little blood vessels. Things like avoiding hot showers, as we talked about, taking cooler showers or not even taking showers, just like cleaning yourself with a washcloth can be helpful. There are certain medications that we can use sometimes that help. 

Now, first of all, Jakafi is extremely effective for itching. Of course, it does have side effects. It’s not always approved for your disease, so for example, it’s not approved for essential thrombocythemia. But JAK inhibitors can be helpful in that setting. There are also medications like Gabapentin, which is a medicine that we use to treat peripheral neuropathy and that can actually be helpful because actually the itching, a lot of it is related to nerves not functioning right, so gabapentin can be helpful. 

And a really old-school medicine that I sometimes use, especially if the itching is most prevalent at night, is a drug called Doxepin and that’s been around for a very long time, but it can be extremely sedating and has to be used with caution, especially in patients who are older. 

Katherine Banwell:

Here is a question from Daniel. “How often should a person with PV have hematology appointments, and how often should you have blood tests?” 

Dr. Jeanne Palmer:

Well, that is something that you need to discuss with a provider, because everyone’s a little bit different. If I have somebody who I’m managing on a medication, they’ve been rock solid stable, it may be every few months that I check blood and maybe every six months that I see them.  

If I have somebody who have been particularly difficult to control and I’m sort of adjusting medications or they’re having symptoms, then I try to check blood more regularly, like on a monthly basis. But again, this is something that – I have checked blood as frequently as every two weeks, especially in somebody who has an extremely high red blood cell count that I’m trying to lower. I have checked blood as infrequently as every three months. Again, in somebody who is not undergoing treatment, say, for example, who has essential thrombocythemia, sometimes I check blood even less. So, it really is something that can vary from every two weeks to every six months. 

Katherine Banwell:

Okay. Katie had this question. “What are the signs of progression from PV to MF or AML, both clinically and in blood tests, and when do you need a new bone marrow biopsy to check for this happening?” 

Dr. Jeanne Palmer:

So, in terms of progression, there are several things that we see happen. 

I think most importantly is, let’s say you have PV, and you’ve always been on medication, and it’s been hard to control. And all of a sudden, you don’t need medication to control it anymore, or the same thing for essential thrombocythemia. You have been taking medication, and all of a sudden your platelets go down, and you don’t need to take drugs anymore. A lot of times people are like, “Oh, that means I’m fixed and I’m well,” not necessarily, you really need to make sure to talk to your healthcare provider and potentially get a bone marrow biopsy. 

Now, the other thing – sometimes the blood counts will actually drop too low, so you’ll have somebody who has PV, who has always been too high and then all of the sudden they come in, and their hemoglobin is very low, and they’re anemic, and that’s another situation where you do that. So, anytime the blood counts start to drop is concerning.  

Now, it’s a continuum, so the blood counts may drop as you’re at the point of transitioning but it doesn’t – it’s not like if your blood count is dropping you say, “Oh my God, I have myelofibrosis, I need a bone marrow transplant tomorrow.” That’s not necessarily the case. This is generally a transition type process. 

Also when the spleen starts to get enlarged. Now, the spleen can be enlarged even in the setting of just ET or just PV, so spleen enlargement does not necessarily mean you’re transforming, but it can be one of the things that we would see that would indicate that. 

Katherine Banwell:

Okay. 

Dr. Jeanne Palmer:

And then finally white blood cell count increasing can often be a sign of that. Now, in terms of progression to AML, that is generally something we’ll see in the blood. AML or acute myeloid leukemia, is indicated by the presence of blasts at greater than 20 percent. Now, many patients with myelofibrosis, in particular, but even PV and ET, may have blasts in their peripheral blood. Blasts are normal. If I did a marrow on every healthy person out there, they are going to have some blasts, because these are the first part of the development of white blood cells. So, they’re like baby white blood cells. But what the problem is, is when they start to grow too much. 

And so in the setting of myelofibrosis and even sometimes with these other diseases, the blasts will be in the peripheral blood primarily because the bone marrow is damaged and doesn’t hold them in very well. It becomes AML when it gets greater than 20 percent, so that blasts of greater than 20 percent in the peripheral blood or in the bone marrow but a lot of times we find it in the peripheral blood is where we indicate this has progressed to AML. 

Katherine Banwell:

Yeah. 

Dr. Jeanne Palmer:

Blasts of greater than 10 percent are also something that we really want to pay attention to, because that would suggest that the disease is starting to become more aggressive. Now, blasts vary, so for example, I’ve had patients go up to 11 and then drop back down to 3 or 4, and then they say around 3 or 4 or 5. So, you always want to make sure to double-check because one blast count at 11 percent, whereas it’s very important to address, may not necessarily reflect that you need to change in treatment at that time. Again, these blood tests, I always tell people, do not freak out over one blood test.  

Make sure you get at least a couple of them to really confirm what you are looking at. 

Katherine Banwell:

Thank you, Dr. Palmer. And to our viewers, please continue to send in your questions to question@powerfulpatients.org and we’ll work to get them answered on future webinars.  

Dr. Palmer, was we close out this conversation I wanted to get your thoughts on where we stand with progress in helping people live longer and truly thrive with MPN. What would you like to leave the audience with? 

Dr. Jeanne Palmer:

So, I think that the first thing is make sure you understand your disease. Don’t hesitate to ask for a second opinion. It’s always good to make sure you talk to someone who can really explain so you feel like when you go home you understand what’s going on in your body. Make sure you understand what symptoms to look for, what things to be aware of, because a lot of times people come in and they have no idea that, oh, these symptoms are actually related to their disease. 

The other thing to make sure is that you’re very honest with your provider on how you’re feeling. A lot of times people come in and they say, “Oh, how are you feeling?” “I feel fine,” but then they start to ask very specific questions and they’re like, “Oh yeah, I’m really tired, my fatigue is an 8 out of 10,” or something. 

So, make sure you’re really honest with your provider. When they ask you how they’re doing, this is not a social visit, this is a visit where they need to know your symptoms, so you don’t need to say I’m fine like you normally would if you were walking down the street. 

The next thing is to always make sure to know where there’s clinical trials because we are making enormous great leaps and bounds in this field. It’s a really exciting time for myeloproliferative diseases, and there’s a number of new drugs that are being tested and coming out. So, it’s always important, if the opportunity is available and you can do it, clinical trials are a great way to get treatment. 

Plus, you are giving back, because these are things that help us learn whether something works or not. So, you’re not as much a guinea pig, you never get a sugar pill. It’s one of those things you will always get the treatment you need and then they may add something to it or you may be in the situation where there is no treatment, so they try something. 

But clinical trials, I have to emphasize, are a great way to get therapy and really are how we know everything that we know about treatment for these diseases. 

Katherine Banwell:

Yeah. It sounds like there’s a lot of progress and hope in the field.  

Dr. Jeanne Palmer:

Oh, absolutely. 

Katherine Banwell:

Thank you so much, Dr. Palmer, for joining us today.  

Dr. Jeanne Palmer:

You are very welcome, my pleasure, and it’s always fun to do these things, so thank you for having me.   

Katherine Banwell:

And thank you to all of our partners. To learn more about MPNs and to access tools to help you become a proactive patient, visit powerfulpatients.org. I’m Katherine Banwell, thanks for joining us today. z

Expert Perspective: Hopeful MPN Research and Development

Expert Perspective: Hopeful MPN Research and Development from Patient Empowerment Network on Vimeo.

MPN expert and clinical researcher Dr. Abdulraheem Yacoub shares excitement about the future of MPN treatment and research, including an optimistic outlook for new approvals in the coming year. 

Dr. Abdulraheem Yacoub is a hematologist oncologist at the University of Kansas Cancer Center. Dr. Yacoub is an active researcher and is an Associate Professor of Hematologic Malignancies and Cellular Therapeutics. Learn more about Dr. Yacoub, here.
 
 

Related Programs:

 
How Treatment Goals Impact MPN Treatment Decisions

How Treatment Goals Impact MPN Treatment Decisions

Advice for Choosing MPN Therapy: What’s Right for You?

Advice for Choosing MPN Therapy: What’s Right for You?

How Should You Participate in MPN Care and Treatment Decisions?

How Should You Participate in MPN Care and Treatment Decisions?


Transcript:

Katherine:

I wanted to get your take on the future of MPN research and treatment. Are there new developments that you’re excited about and that make you hopeful?  

Dr. Yacoub:

Absolutely. So, again, I would like to take the last few minutes to advocate for the future. And the future can only come when doctors and patients and advocates work together to advance the science. We have few tools to treat patients and to help patients. We have a lot of unanswered questions. And the only way to answer them is by designing quality clinical trials, enrolling patients on trials, taking the risk, and trying to find new answers and new therapeutics. So, I always would like to advocate for patients to seek clinical trials whether with their doctor or whether they have to travel for it, and for doctors to consider that for their patients. That’s the only way to advance science.  

There are very important national and international studies going on right now. One of the – and first, I would like to emphasize is that we have had ruxolitinib (Jakafi) as the only therapy, or the first-line therapy for myelofibrosis for a decade now.  

Not everybody responds to it, not everybody responds to it for a long time. So, now we’re designing combination trials. So, there’s a few studies that we are trying to redefine, “Is ruxolitinib alone enough, or should we have a combination first-line therapy?” So, these are some of the more important questions being asked right now.  

And this is definitely one of the bigger moves in the field, is trying to redefine what is the first-line therapy for myelofibrosis. For polycythemia vera, we’re also exploring therapeutics that would reduce phlebotomy with things you can add to your medical care to reduce phlebotomy.  

So, that’s also going on. And it’s definitely a big leap forward for many of our patients. For ET, when we don’t have any actual drugs approved other than hydroxyurea (Hydrea) and anagrelide (Agrylin), we actually have trials with interferon going on.  

So, I would like to advocate for that. So, interferon succeeded and now approved for PV, but not yet for ET. We’re working on that. So, again, in every disease, we’re trying to design clinical trials to redefine what is the best treatment today.  

We’re also doing studies to understand the cancer. So, studies where patients donate their samples for research. These are very helpful, very important. And contributing to that always advances the science, and it’s low effort to the patients. So, if there’s a clinical trial that is offered to patients, I would strongly urge everybody to consider that favorably and contribute to science. 

That’s the only way we can help future patients and ourselves immediately, at moving the field forward.  

What Are the Signs of MPN Progression?

What Are the Signs of MPN Progression? from Patient Empowerment Network on Vimeo.

Dr. Abdulraheem Yacoub, an MPN specialist, explains how essential thrombocythemia (ET), polycythemia vera (PV), or myelofibrosis (MF) may progress from one disease to the next, including potential signs and symptoms of MPN progression. 

Dr. Abdulraheem Yacoub is a hematologist oncologist at the University of Kansas Cancer Center. Dr. Yacoub is an active researcher and is an Associate Professor of Hematologic Malignancies and Cellular Therapeutics. Learn more about Dr. Yacoub, here.
 

Related Programs:

 
What Are Treatment Options for Essential Thrombocythemia?

What Are Treatment Options for Essential Thrombocythemia?

What Are Treatment Options for Polycythemia Vera?

What Are Treatment Options for Polycythemia Vera?

What Are Treatment Options for Myelofibrosis?

What Are Treatment Options for Myelofibrosis?


Transcript:

Katherine:

We have a couple of questions from the audience. This one is from Sarah. She writes, “I’ve been living with essential thrombocythemia for three years, and have been relatively stable. Of course, I’m worried about progression to PV or MF. What is my chance of progression, and what are the signs of progression?” 

Dr. Yacoub:

That’s a very good question. And unfortunately, we’re very good at describing those numbers. Unfortunately, our tools at interfering are not as good. So, in general, patients with ET, statistically speaking, have a life expectancy that is not different from their age match peers. And Sarah’s story will be not too indifferent from her sisters and her mother, in terms of what’s going to happen to her long care and her health, provided she gets good medical care. The exception to that is that there is a transformation risk. For ET we caught around a 4 percent every 10 years in which ET will actually change into a different cancer, a higher risk cancer.   

Could be MF, could be MDS, could be acute leukemia. And that will be a much more serious diagnosis. So, it’s about 4 percent in 10 years. We do have a – or we extrapolate some of the data from other cancers. So, certain mutations are more favorable, certain mutations are more risky. And we try to forecast that but worried it’s really hard to predict that since it’s such a long journey with disease. The first symptoms or the findings, when patients start suspecting that their disease has changed, is that the pattern of symptoms that they have are different.   

They often become worse. So, they have more constitutional symptoms, more tiredness, more fevers, more night sweats, losing weight, not being able to eat a full meal, abdominal distension, the spleen gets bigger.  

So, these are some of the feelings that patients can experience that lead to this. Other objective things is when the blood tests change in a less favorable way. So, for patients with ET who always run at 800,000 platelet count, if they’re suddenly 200, and that’s in the normal range, but that’s actually not good news, because the cancer changed. And this change is not favorable. So, as the doctors run routine labs, if they see the sudden change in labs, that’s also abnormal.  

If the doctor can feel that the spleen gets bigger every time, that’s also concerning. If the patients suddenly have anemia or very high white cell count or immature white cells in the blood, that’s also a concern.  

So, that’s why it’s great or important to establish a baseline symptom burden.  A baseline spleen, a baseline bone marrow biopsy with mutation analysis, so that patients have a clear reference point to where they started, and if things change, they can always go back to that point and compare.  

What Are Treatment Options for Polycythemia Vera?

What Are Treatment Options for Polycythemia Vera? from Patient Empowerment Network on Vimeo.

MPN specialist Dr. Abdulraheem Yacoub provides an overview of available treatment options for patients living with polycythemia vera (PV). 

Dr. Abdulraheem Yacoub is a hematologist oncologist at the University of Kansas Cancer Center. Dr. Yacoub is an active researcher and is an Associate Professor of Hematologic Malignancies and Cellular Therapeutics. Learn more about Dr. Yacoub, here.
 
 

Related Programs:

 
How Treatment Goals Impact MPN Treatment Decisions

How Treatment Goals Impact MPN Treatment Decisions

Advice for Choosing MPN Therapy: What’s Right for You?

Advice for Choosing MPN Therapy: What’s Right for You?

How Should You Participate in MPN Care and Treatment Decisions?

How Should You Participate in MPN Care and Treatment Decisions?


Transcript:

Katherine:

You mentioned using interferons for ET. That’s something that you would also use for polycythemia vera. Yes?   

Dr. Yacoub:

Absolutely. So, the same principles will apply to polycythemia vera. We would like to treat the higher-risk patients more aggressively. Hydroxyurea (Hydrea) and interferon are also the first-line therapies in these patients.   

The good news in 2022 is that we actually finally have an FDA-approved interferon for our patients. Finally, after 50 years of using interferon, now, we have an FDA approval. So, the new interferon, ropeginterferon alfa-2b is a medication that was studied prospectively in Europe, and it has been approved and in clinical use in Europe under the brand name Besremi.  

And this year, it was approved in the U.S. for patients with polycythemia vera, which is a great achievement for the medical field and a great tool to help our patients. We have used other brands off-label in the past, but it’s glad now to get this confirmation from the FDA that this is a standard of care for all patients.  

And then beyond that, ruxolitinib or Jakafi, is also approved as a second-line option in patients who have had hydroxyurea as their first line.  

So, these are the medicines we use for polycythemia vera. We also use therapeutic phlebotomy. And the goal in high-risk polycythemia vera, or actually in all patient polycythemia vera, is to reduce their hematocrit.  

And we want it under 45 percent every day of the year. And we use the tools that we just discussed phlebotomy and medicines to achieve that, in addition to aspirin. So, that’s how PV is more unique than ET. Yes. 

What Are Treatment Options for Essential Thrombocythemia?

What Are Treatment Options for Essential Thrombocythemia? from Patient Empowerment Network on Vimeo.

MPN specialist Dr. Abdulraheem Yacoub reviews the various treatment approaches available to patients living with essential thrombocythmia (ET).

Dr. Abdulraheem Yacoub is a hematologist oncologist at the University of Kansas Cancer Center. Dr. Yacoub is an active researcher and is an Associate Professor of Hematologic Malignancies and Cellular Therapeutics. Learn more about Dr. Yacoub, here.
 
 

Related Programs:

 
How Treatment Goals Impact MPN Treatment Decisions

How Treatment Goals Impact MPN Treatment Decisions

Advice for Choosing MPN Therapy: What’s Right for You?

Advice for Choosing MPN Therapy: What’s Right for You?

How Should You Participate in MPN Care and Treatment Decisions?

How Should You Participate in MPN Care and Treatment Decisions?


Transcript:

Dr: Yacoub: 

So, let’s talk about essential thrombocythemia. This is among the other MPNs, the cancer with the lowest risk. Patients with essential thrombocythemia can have clots and can have bleedings. And they also often have symptoms because of their cancer.  

But they also enjoy a long life expectancy that is almost indifferent from patients who don’t have cancer provided they get good care. So, our emphasis is on focusing that their life quality is not touched by their cancer, and focusing on treating patients with symptoms, to ameliorate the symptoms and allowing them to have a decent and good quality of life. At the same time, we would like to reduce the risk of clotting and bleeding.  

And we have tools and medicines that are very effective at doing that in select patients who we define as high risk. And now there is a more clear definition of that. So, high-risk patients are patients who are over age 60 and have a JAK2 mutation, or patients who have already had a clot.  

That is not the majority of ET patients actually. The majority are not high risk. And those patients might not require therapy to reduce their platelet count. 

But for high-risk patients, we have tools to help them. So, hydroxyurea (Hydrea) is the most commonly used medicine in this setting.  

The goal of hydroxyurea is to reduce the platelet count. And we’d like to keep it under 400, sometimes under 600 under different circumstances. And that will reduce the risk of clotting and bleeding for our patients. The other option, which I also feel passionate about is interferon.  

Interferons are drugs that we’ve used for decades. They’re very effective. They’re safe in the right hands. And they do have advantages over hydroxyurea in terms of long-term safety. These are medications we can give to young patients, we can give to pregnant patients, we can give for long term without concerns of toxicity, and also they have a higher ceiling. Patients with interferon can achieve a disease control that we cannot achieve with hydroxyurea.  

And this will be beneficial long term treating those patients. So, these – yeah, and then aspirin therapy is always something we would like to include in this regimen.  

Katherine:

I was going to ask you about that. So, aspirin is still being used as a treatment?  

 Dr. Yacoub:

Absolutely. So, the standard of care is to use aspirin. Usually, one baby aspirin once a day, preferably in the morning is what we recommend. And that’s probably all the aspirin they need. We do not want them to take more than that either.  

How Treatment Goals Impact MPN Treatment Decisions

How Treatment Goals Impact MPN Treatment Decisions from Patient Empowerment Network on Vimeo.

Dr. Abdulraheem Yacoub, an MPN specialist, reviews treatment goals for patients with essential thrombocythemia (ET), polycthemia vera (PV), and myelofibrosis (MF). Dr. Yacoub goes on to explain factors that may impact treatment decisions, including the management of symptoms and side effects. 

Dr. Abdulraheem Yacoub is a hematologist oncologist at the University of Kansas Cancer Center. Dr. Yacoub is an active researcher and is an Associate Professor of Hematologic Malignancies and Cellular Therapeutics. Learn more about Dr. Yacoub, here.
 

Related Programs:

 
What Are Treatment Options for Essential Thrombocythemia?

What Are Treatment Options for Essential Thrombocythemia?

What Are Treatment Options for Polycythemia Vera?

What Are Treatment Options for Polycythemia Vera?

What Are Treatment Options for Myelofibrosis?

What Are Treatment Options for Myelofibrosis?


Transcript:

Katherine:

To give our patient audience some context before we get into the specifics of MPN treatment approaches, how would you define treatment goals?  

Dr. Yacoub:

Thank you, thank you. And I always like to highlight and emphasize that unlike many of the cancer syndromes that patients deal with, myeloproliferative neoplasms are unique. 

These are chronic cancers. There’s no finish line. And this is a disease you live with. It affects every day of your life, every activity of your future life. You plan your life events accordingly. Pregnancies and marriages and trips and all of that. So, this is a chronic cancer. And as we plan therapy, we always factor that in. We would like the cancer to have the least or almost no impact on your daily life.  

Whether it’s symptoms, whether it’s disability and dysfunction and inability to perform your daily functions, whether it’s actual physical symptoms that you’re having from the cancer, or whether it’s affecting complications that are hurting your health. So, we would like to focus on all of these, the medical aspect as well as the impact of the disease to everyday symptoms.  

This is a unique feature of these cancers. And it doesn’t really exist much in other diseases.  

So, as we approach our patients, we would like to get a good assessment of the disease burden to their lives. These can be symptoms. So, we actually have very good objective tools to measure symptoms, such as the MPN-SAF. It’s an objective tool to calculate the symptoms. So, we would like to get an objective baseline of symptoms. 

Because we do want to address the symptoms, regardless of the MPN subtype. We do want to master actually the symptoms because that is what patients feel every day, and we want to affect that early in the treatment. We also would like to get a good assessment of the disease complications. Have the patient suffered a clot or a hemorrhage or symptoms because of an enlarged spleen? Or were they unable to perform certain activities? Are they able to eat? Are they losing weight?  

So, we would like to see how is the cancer also causing them immediate morbidity, and we also would like to tackle the future. So, cancers tend to get worse with time. They tend to transform into a higher risk cancer. So, as we approach any of the MPN patients, we also talk about the future risk of the cancer turning into a more aggressive form of cancer.  

So, we would like if we can, for every patient to focus on these three pillars of their care: their immediate quality of life and symptoms, their immediate complications, and their future disease progression.  

And we would like to factor in that our treatments does not add more side effects to their lives. So, that’s the fourth pillar of how we take care of patients. So, these are the basic concepts that will apply today for all patients with all three diseases.  

Some patients will have more emphasis on one or the other. But this is something in our mind as doctors who treat MPN patients.  

Thriving with an MPN: What You Should Know About Care and Treatment

Thriving with an MPN: What You Should Know About Care and Treatment from Patient Empowerment Network on Vimeo.

 MPN specialist and researcher, Dr. Abdulraheem Yacoub, reviews factors that help guide care decisions for MPNs – essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF). Dr. Yaboub discusses the goals of treatment, shares tools for taking an active role in your care, and provides an update on promising new therapies for MPNs.

 
Dr. Abdulraheem Yacoub is a hematologist oncologist at the University of Kansas Cancer Center. Dr. Yacoub is an active researcher and is an Associate Professor of Hematologic Malignancies and Cellular Therapeutics. Learn more about Dr. Yacoub, here.
 
 

Related Programs:

 
The Latest in MPN Research: Updates from ASH 2021

The Latest in MPN Research from ASH 2021

Updates from ASH: How Biomarker Testing Has Changed MPN Care

Updates from ASH: How Biomarker Testing Has Changed MPN Care

Expert Advice for Finding an MPN Clinical Trial

Expert Advice for Finding an MPN Clinical Trial


Transcript:

Katherine:                  

Hello, and welcome. I’m Katherine Banwell, your host for today. Today’s program is about how to live and thrive with an MPN. We’re going to discuss MPN treatment goals, and how you can play an active role in your 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.

All right, let’s meet our guest. Joining me today is Dr. Abdulraheem Yacoub. Welcome, Dr. Yacoub. Would you please introduce yourself?

Dr. Yacoub:               

Hello, Katherine. And thank you very much for inviting me to participate in this very important and near and dear topic to my heart and to everything I do every day.

I’m a hematologist-oncologist at the University of Kansas. I practice hematology 100 percent of my time, and I dedicate it to patients with MPNs. I’m an active researcher through clinical trials at my own institution, as well as part of many national and international collaborations. We all strive to provide the best care and the updates for our patients. I’m also a Director of our hematology clinics in cancers at the University of Kansas, and I’m an Associate Professor of Medicine at the University of Kansas.

Katherine:                  

Well, thank you so much for taking time out of your very busy schedule to join us today. We appreciate it.

Dr. Yacoub:               

Absolutely, my pleasure.

Katherine:                  

To give our patient audience some context before we get into the specifics of MPN treatment approaches, how would you define treatment goals?

Dr. Yacoub:               

Thank you, thank you. And I always like to highlight and emphasize that unlike many of the cancer syndromes that patients deal with, myeloproliferative neoplasms are unique.

These are chronic cancers. There’s no finish line. And this is a disease you live with. It affects every day of your life, every activity of your future life. You plan your life events accordingly. Pregnancies and marriages and trips and all of that. So, this is a chronic cancer. And as we plan therapy, we always factor that in. We would like the cancer to have the least or almost no impact on your daily life.

Whether it’s symptoms, whether it’s disability and dysfunction and inability to perform your daily functions, whether it’s actual physical symptoms that you’re having from the cancer, or whether it’s affecting complications that are hurting your health. So, we would like to focus on all of these, the medical aspect as well as the impact of the disease to everyday symptoms.

This is a unique feature of these cancers. And it doesn’t really exist much in other diseases.

Katherine:                  

That’s helpful to understand as we move through today’s program. And we’re going to cover the three classic MPNs, polycythemia vera, essential thrombocythemia, and myelofibrosis.

So, for the person who has one of these conditions, can you help us understand the treatment approaches for each? Let’s start with essential thrombocythemia or ET.

Dr. Yacoub:               

Excellent. So, I’m going to start with some general concepts. So, as we approach our patients, we would like to get a good assessment of the disease burden to their lives. These can be symptoms. So, we actually have very good objective tools to measure symptoms, such as the MPN-SAF. It’s an objective tool to calculate the symptoms. So, we would like to get an objective baseline of symptoms.

Because we do want to address the symptoms, regardless of the MPN subtype. We do want to master actually the symptoms because that is what patients feel every day and we want to affect that early in the treatment. We also would like to get a good assessment of the disease complications. Have the patient suffered a clot or a hemorrhage or symptoms because of an enlarged spleen? Or were they unable to perform certain activities? Are they able to eat? Are they losing weight?

So, we would like to see how is the cancer also causing them immediate morbidity, and we also would like to tackle the future. So, cancers tend to get worse with time. They tend to transform into a higher risk cancer. So, as we approach any of the MPN patients, we also talk about the future risk of the cancer turning into a more aggressive form of cancer.

So, we would like if we can, for every patient to focus on these three pillars of their care: their immediate quality of life and symptoms, their immediate complications, and their future disease progression.

And we would like to factor in that our treatments does not add more side effects to their lives. So, that’s the fourth pillar of how we take care of patients. So, these are the basic concepts that will apply today for all patients with all three diseases.

Some patients will have more emphasis on one or the other. But this is something in our mind as doctors who treat MPN patients, we try to balance all these three pillars for every patient. So, let’s talk about essential thrombocythemia. This is among the other MPNs, the cancer with the lowest risk. Patients with essential thrombocythemia can have clots and can have bleedings. And they also often have symptoms because of their cancer.

But they also enjoy a long life expectancy that is almost indifferent from patients who don’t have cancer provided they get good care. So, our emphasis is on focusing that their life quality is not touched by their cancer, and focusing on treating patients with symptoms, to ameliorate the symptoms and allowing them to have a decent and good quality of life. At the same time, we would like to reduce the risk of clotting and bleeding.

And we have tools and medicines that are very effective at doing that in select patients who we define as high risk. And now there is a more clear definition of that. So, high-risk patients are patients who are over age 60 and have a JAK2 mutation, or patients who have already had a clot.

That is not the majority of ET patients actually. The majority are not high risk. And those patients might not require therapy to reduce their platelet count.

But for high-risk patients, we have tools to help them. So, hydroxyurea (Hydrea) is the most commonly used medicine in this setting.

The goal of hydroxyurea is to reduce the platelet count. And we’d like to keep it under 400, sometimes under 600 under different circumstances. And that will reduce the risk of clotting and bleeding for our patients. The other option, which I also feel passionate about is interferon.

Interferons are drugs that we’ve used for decades. They’re very effective. They’re safe in the right hands.

And they do have advantages over hydroxyurea in terms of long-term safety. These are medications we can give to young patients, we can give to pregnant patients, we can give for long term without concerns of toxicity, and also they have a higher ceiling. Patients with interferon can achieve a disease control that we cannot achieve with hydroxyurea.

And this will be beneficial long term treating those patients. So, these – Yeah, and then aspirin therapy is always something we would like to include in this regimen.

Katherine:                  

I was going to ask you about that. So, aspirin is still being used as a treatment?

Dr. Yacoub:               

Absolutely. So, the standard of care is to use aspirin. Usually, one baby aspirin once a day, preferably in the morning is what we recommend. And that’s probably all the aspirin they need. We do not want them to take more than that either.

Katherine:                  

And you mentioned using interferons for ET. That’s something that you would also use for polycythemia vera. Yes?

Dr. Yacoub:               

Absolutely. So, the same principles will apply to polycythemia vera. We would like to treat the higher-risk patients more aggressively. Hydroxyurea and interferon are also the first-line therapies in these patients.

The good news in 2022 is that we actually finally have an FDA-approved interferon for our patients. Finally, after 50 years of using interferon, now, we have an FDA approval. So, the new interferon, ropeginterferon alfa-2b is a medication that was studied prospectively in Europe, and it has been approved and in clinical use in Europe under the brand name Besremi.

And this year, it was approved in the US for patients with polycythemia vera, which is a great achievement for the medical field and a great tool to help our patients. We have used other brands off-label in the past, but it’s glad now to get this confirmation from the FDA that this is a standard of care for all patients.

And then beyond that, ruxolitinib or Jakafi, is also approved as a second-line option in patients who have had hydroxyurea as their first line.

So, these are the medicines we use for polycythemia vera. We also use therapeutic phlebotomy. And the goal in high-risk polycythemia vera, or actually in all patient polycythemia vera, is to reduce their hematocrit.

And we want it under 45 percent every day of the year. And we use the tools that we just discussed phlebotomy and medicines to achieve that, in addition to aspirin. So, that’s how PV is more unique than ET. Yes.

Katherine:                  

And since myelofibrosis is a progressive condition, I imagine it’s more difficult to manage. So, what else is available for patients with myelofibrosis?

Dr. Yacoub:               

Correct So, myelofibrosis is the higher end of this spectrum of cancers.

It is a cancer that is associated with much higher symptom burden and impact on daily life. It is also associated with low blood counts, and some patients will require transfusions. It’s a major morbidity to our patients. And in addition, it’s a cancer that is associated with shortened life. So, patients with myelofibrosis will not live as long as their health would have allowed them. And some of them will live actually a much shorter life than they want or deserve.

So, myelofibrosis treatment requires a lot more considerations. So, for patients who are in good health, who have a cancer that is more aggressive, that would be imminently impacting their longevity, we start a discussion about a curative role of allogeneic stem cell transplantation very early in their course.

Because bone marrow transplantation can be curative, and those patients can live a long life after a successful transplant. So, this is a treatment modality that should be brought up very early for patients with higher risk myelofibrosis. There are approved JAK inhibitors, ruxolitinib and fedratinib (Inrebic). And we know that Ruxolitinib which has been approved for over 10 years can improve symptoms, can improve the spleen volume, can actually prolong lives for patients on it, and also makes the transplant more successful.

So, we should be offering that to the appropriate patients also early in their diagnosis, in a strategy where, in addition to that, we get them to a transplant. Fedratinib is approved in that setting. And we are very optimistic that by the end of this calendar year, we will have two other JAK inhibitors approved.

So, we look forward to those two drugs. Momelotinib and pacritinib for patients with special disease features.

[Editor’s Note: As of February 28, 2022, pacritinib (Vonjo) has been approved for the treatment of myelofibrosis patients with severe thrombocytopenia.]

And hopefully, by the end of this year, we will have a list of JAK inhibitors that we can choose from, which is great news for our patients.

Katherine:                  

Oh, we’re still fighting.

Dr. Yacoub:               

Yes, absolutely.

Katherine:                  

As with most conditions, all patients are different, of course, and what might work for one person might not be appropriate for another. So, how do you choose which treatment is right for a patient?

Dr, Yacoub:               

Excellent. So, and that’s actually the heart of what we define as the art of practicing medicine and being patient-centric and focused. And patients already have their own wishes and their needs. And everything should start with having a discussion with patients on what is their priority, and what are they trying to achieve.

And we do have to explain to them the tools we have, the interventions that can help them. But we also need to make sure they’re compatible with what they actually want and their goals in life. And sometimes what we doctors want is not exactly what the patients want. So, we always have to remind ourselves that patients are the drivers of their care. And they have the absolute right to be informed and to make informed decisions based on the options we advise them about.

So, that is always a centerpiece of healthcare. And then patients – Basically, we defined four pillars of care. We want to control their symptoms, we want to prevent complications, we want to modify the disease so it doesn’t transform, and we want our therapies not to have toxicities, not to have side effects that are worse than the disease. So, we bring that up to the table. And we also look at the patient. What is their symptoms? What did the disease cause them to be complications?

What is the risk that their cancer is actually going to progress quickly to hurt their lives? And how serious is the therapy we’re recommending? And we need to make sure that there is a good match between what we’re offering and what the disease is manifesting. So, for example, for patients who have a lot of symptoms, but they have low-risk cancer that they can live with for a long time, we focus on symptoms. We focus on treatments that improve their symptoms.

While with patients who have more serious diseases that are eminently life-threatening, we focus on an expedited path to a more aggressive therapy and a bone marrow transplant. And then we also try to match those therapies with the other patient’s wishes and needs and so forth. So, all these factors are important. We have more tools to try to prognosticate. So, prognosticate is the medical word that we use as forecasting.

We like to forecast the disease or the cancer. We try to predict the patient’s future. Fortunately, we actually have good tools to prognosticate now. We have models or calculators that factor in patients’ features, their symptoms, their age, their blood counts, their bone marrow findings, and their DNA mutations. And it gives us a score a risk score that can correlate with their life expectancy or their outcomes.

And we use those tools to guide us. So, there’s actually a tool we use to help patients reach that decision. It’s an objective tool to decide how serious is this disease and how seriously we should tackle it. It’s very applicable for patients with myelofibrosis, more sort of the other lower-risk cancers.

Katherine:                  

What about comorbidities? How do they fit into the treatment plan?

Dr. Yacoub:               

Very important.

So, again, it also goes back to finding the balance between how serious is the disease, how serious is the treatment, and how will the patient’s general health tolerate and factor in the choices patients make accordingly. So, myeloproliferative neoplasms do happen in a broad range of ages. And we have children, minors with MPNs, and we have elderly patients with MPNs. And it’s a continuous spectrum. And each individual patient will have their own health concerns and their own health comorbidities and their own wishes. And we always have to make sure that we match our therapies, the disease seriousness, and the patient’s wishes, which is also stemming from their own other health battles, too.

We cannot turn a blind eye to the other health issues going on. That plays a major factor as we choose to discuss bone marrow transplantation with patients. Because that’s when the medical comorbidities are often the first barrier to go through.

Katherine:                  

Are there specific biomarkers that may affect prognosis or treatment?

Dr. Yacoub:               

Yes. So, and we’re glad that actually myeloproliferative neoplasms are actually the model in medical oncology on how predictors can tell us a lot more about the patient’s future about the prognosis. So, early on in MPNs, we’ve developed models, like the International Prognostic Scoring Systems in many different iterations. And more recently, the Molecular Based International Prognostic Scores.

They factor in patient’s age, they factor in blood numbers, they factor in DNA abnormalities, they also factor in DNA mutations, including the common driver mutations JAK2 and CALR and MPL, as well as more novel mutations that we call higher risk mutations.

So, based on these models, we use these tools to predict how the cancer will behave, and how to approach it. This advancement has been an application for our MPN patients for a while, way ahead of all other fields of oncology. So, we’re proud that we can give our patients this tool before all other doctors were able to. Yeah.

Katherine:                  

That’s excellent. Dr. Yacoub, what is the role of the patient in their care? When does shared decision-making come into play?

Dr. Yacoub:               

Absolutely.

Patients are the drivers and the centerpiece of their health care. And patient self-advocacy is the most important tool. So, many of our patients are young and they will live with their cancers a lot longer than many cancer doctors will practice oncology. And they will have many doctors. Statistically, each MPN patient will have multiple doctors throughout their career. And they will hear different derivatives. And the science will change. And they will be given different counseling over the time. And their disease will change.

And they will have different needs as they go further. So, patients being involved in their wellbeing and their cancer care is important from the first day. And I always tell patients, “You need to start building your village from day one.” It is not just the patient, it’s your caregivers, it’s who else can help you.

Who else can advise you? You might want to also invest in a friend or a spouse or a child, to come to you and listen to some of those discussions so that they can advise you later on, “Why are you making different decisions?” So, we encourage patients to be very involved early on, to build their own village, and to seek care. We routinely ask for second opinions. We want patients to always hear the story and hear the same story from another doctor so that they hear the range of how we word the truth and how we word the facts.

And this way, they can have a better perspective. So, this is now a standard. Almost all patients should have two doctors, at least, the treating doctor and one doctor who’s an MPN specialist, who would give them another twist or another perspective to their health.

So, and that is always important. And then there are very good references and online resources for patients to tackle in, such as this seminar and other good places where patients can seek more information. They also can go to a clinical trial to find out what are the ongoing clinical trials and advancements.

There are structured patient symposiums nationally and regionally. So, and we strongly recommend that patients seek more opinions and more help and more resources and be very engaged with this disease, especially that it is a chronic cancer, and it’s not going to –

Katherine:                  

It’s not going away.

Dr. Yacoub:               

It’s just a new lifestyle. And they need to be as engaged with it as they can.

Katherine:                  

Absolutely. We have a couple of questions from the audience. This one is from Sarah. She writes, “I’ve been living with essential thrombocythemia for three years, and have been relatively stable. Of course, I’m worried about progression to PV or MF. What is my chance of progression, and what are the signs of progression?”

Dr. Yacoub:               

That’s a very good question. And unfortunately, we’re very good at describing those numbers. Unfortunately, our tools at interfering are not as good. So, in general, patients with ET, statistically speaking, have a life expectancy that is not different from their age match peers. And Sarah’s story will be not too indifferent from her sisters and her mother, in terms of what’s going to happen to her long care and her health, provided she gets good medical care. The exception to that is that there is a transformation risk. For ET we caught around a 4 percent every 10 years in which ET will actually change into a different cancer, a higher risk cancer.

Could be MF, could be MDS, could be acute leukemia. And that will be a much more serious diagnosis. So, it’s about 4 percent in 10 years. We do have a – or we extrapolate some of the data from other cancers. So, certain mutations are more favorable, certain mutations are more risky. And we try to forecast that but worried it’s really hard to predict that since it’s such a long journey with disease. The first symptoms or the findings, when patients start suspecting that their disease has changed, is that the pattern of symptoms that they have are different.

They often become worse. So, they have more constitutional symptoms, more tiredness, more fevers, more night sweats, losing weight, not being able to eat a full meal, abdominal distension, the spleen gets bigger.

So, these are some of the feelings that patients can experience that lead to this. Other objective things is when the blood tests change in a less favorable way. So, for patients with ET who always run at 800,000 platelet count, if they’re suddenly 200, and that’s in the normal range, but that’s actually not good news, because the cancer changed. And this change is not favorable. So, as the doctors run routine labs, if they see the sudden change in labs, that’s also abnormal.

If the doctor can feel that the spleen gets bigger every time, that’s also concerning. If the patients suddenly have anemia or very high white cell count or immature white cells in the blood, that’s also a concern.

So, that’s why it’s great or important to establish a baseline symptom burden.                                   

A baseline spleen, a baseline bone marrow biopsy with mutation analysis, so that patients have a clear reference point to where they started, and if things change, they can always go back to that point and compare.

Katherine:                  

Right. We have another question. This one from Victor. He says, “I was diagnosed with PV in 2018. And I’ve been treated with hydroxyurea. Recently, I’ve been very fatigued. I want to exercise, but I don’t have the energy to do much. Do you have any advice for boosting my energy?”

Dr. Yacoub:               

That is a very good question and very common question. So, the causes for fatigue in adults, in general, so many. And adding PV to that adds a few other reasons why one would be more fatigued. So, assuming that Victor follows with his doctor, and his primary care doctor has systematically went through all the possible causes for fatigue, and those were addressed.

Now that PV specific causes, A). Hydroxyurea can cause fatigue. So, maybe it’s the hydroxyurea dose. And that’s a side effect. And maybe that’s not the best medicine for him. B). Polycythemia vera can cause fatigue. Maybe we’re not controlling it enough. Maybe we need to dial up the dose of the medicine or dial down the dose of the medicine accordingly. And then there’s also the iron deficiency which we induce with PV and phlebotomy.

And whether we actually have taken Victor to become very low on iron, and that can cause fatigue. So, we have to evaluate the treatments, the disease, and the side effects of the interventions we’ve done. And those are the polycythemia vera specific factors that can add to the fatigue.

Katherine:                  

Here’s another question from the audience. This is from Sandy. She writes, are MPNs hereditary? Should my children or siblings be aware of their risk?

Dr. Yacoub:               

All right. Well, the answer to that question changed many times over the last 10 years. So, the answer changed from absolutely not, to very possibly maybe over the years. So, although we don’t think of cancers as inherited, it’s not passed from one parent to their children. But MPNs tend to run in families. And for 11 percent of patients with MPN, and that number has also increased over the years, have actually a first-degree family member with MPN. That is a big coincidence, it’s almost too high to be a coincidence. So, we are realizing that there is genetic makeup or clustering that can cause MPNs to happen more often in certain families.

So, how does this apply to patients? So, if a patient has MPN, that does not mean that their children or siblings will get MPN, it just means they’re more likely than the other people to have MPN, just because they all share the same genetic makeup. And they should be made aware. And they should maintain good health care and maintain the relationship with a primary and have routine labs and all that. But not necessarily that they will get cancer. This still is a very rare disease, and 11 percent of a rare disease still is a small number.

Katherine:                  

Thank you for answering those patient questions. I appreciate it.

Dr. Yacoub:               

My Pleasure.

Katherine:                  

And to our patients, please continue to send in your questions to question@powerfulpatients.org and we’ll work to get them on future programs.

So, Dr. Yacoub, as we close out our program and our conversation, I wanted to get your take on the future of MPN research and treatment. Are there new developments that you’re excited about and that make you hopeful?

Dr. Yacoub:               

Absolutely. So, again, I would like to take the last few minutes to advocate for the future. And the future can only come when doctors and patients and advocates work together to advance the science. We have few tools to treat patients and to help patients. We have a lot of unanswered questions. And the only way to answer them is by designing quality clinical trials, enrolling patients on trials, taking the risk, and trying to find new answers and new therapeutics. So, I always would like to advocate for patients to seek clinical trials whether with their doctor or whether they have to travel for it, and for doctors to consider that for their patients. That’s the only way to advance science.

There are very important national and international studies going on right now. One of the – And first, I would like to emphasize is that we have had ruxolitinib as the only therapy, or the first-line therapy for myelofibrosis for a decade now. Not everybody responds to it, not everybody responds to it for a long time. So, now we’re designing combination trials. So, there’s a few studies that are trying to redefine, “Is ruxolitinib alone enough, or should we have a combination first-line therapy?” So, these are some of the more important questions being asked right now.

And this is definitely one of the bigger moves in the field, is trying to redefine what is the first-line therapy for myelofibrosis. For polycythemia vera, we’re also exploring therapeutics that would reduce phlebotomy with things you can add to your medical care to reduce phlebotomy.

So, that’s also going on. And it’s definitely a big leap forward for many of our patients. For ET, when we don’t have any actual drugs approved other than hydroxyurea and anagrelide (Agrylin), we actually have trials with interferon going on.

So, I would like to advocate for that. So, interferon succeeded and now approved for PV, but not yet for ET. We’re working on that. So, again, in every disease, we’re trying to design clinical trials to redefine what is the best treatment today.

We’re also doing studies to understand the cancer. So, studies where patients donate their samples for research. These are very helpful, very important. And contributing to that always advances the science, and it’s low effort to the patients. So, if there’s a clinical trial that is offered to patients, I would strongly urge everybody to consider that favorably and contribute to science.

That’s the only way we can help future patients and ourselves immediately, at moving the field forward.

Katherine:                  

Seems like there’s a lot of progress in the field.

Dr. Yacoub:               

A lot of progress. I look forward to future events. I’m going to have a lot more tools to discuss. Hopefully, by this time next year, we’re going to have four JAK inhibitors, injectables for PV, interferon for ET, and a lot more things to go over.

Katherine:                  

That’s wonderful. Dr. Yacoub, thank you so much for taking the time to join us today.

Dr. Yacoub:               

You’re welcome. And it’s my pleasure. I feel passionate about this. And I’m happy to help.

Katherine:                  

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