Tag Archive for: combination therapy

Should All MPN Patients Undergo Molecular Testing?

Should All MPN Patients Undergo Molecular Testing? from Patient Empowerment Network on Vimeo.

Dr. Gabriela Hobbs discusses the necessity of molecular testing for myeloproliferative neoplasm (MPN) patients, including the pros and cons of this in-depth testing for patients with polycythemia vera (PV) and essential thrombocythemia (ET).

Dr. Gabriela Hobbs is a hematology-oncology physician specializing in the care of patients with myeloproliferative neoplasms (MPN), chronic myeloid leukemia, and leukemia. Dr. Hobbs serves as clinical director of the adult leukemia service at Massachusetts General Hospital. Learn more about Dr. Hobbs.

See More From MPN Clinical Trials 201

Related Programs:

How Driver Mutation Research Is Advancing MPN Treatments

How Driver Mutation Research Is Advancing MPN Treatments

Advancing MPN Research: How Clinical Trials Work

Advancing MPN Research: How Clinical Trials Work

Advances in Myelofibrosis Research

Advances in Myelofibrosis Research


Transcript:

Katherine:

How useful is having a genetic panel done? Should all patients get molecular or genetic testing? 

Dr. Hobbs:

Great question. And I think that it is very important to have genetic testing.   

And genetic testing involves more than just testing the JAK2 mutation. So, we know that the JAK2 mutation is the most common mutation in patients with MPN. But that being said, there are other mutations that also occur such as the calreticulin mutation and the MPL mutation.   

And so, I think having genetic testing that at least tests for those three mutations is very important so that we can actually help a patient know that they have an MPN. In addition to those three main mutations, many clinicians now have access to what’s called extended next-generation sequencing, where there’s a panel that tests for many different genes at the same time and can test for a variety of other mutations.  

And this is particularly relevant for patients with myelofibrosis. As we know that having other mutations, like, for example, mutations in IDH or ASXL1 and others, can increase the risk of that disease in terms of its risk of transforming to leukemia or how long a patient may live with their myelofibrosis. 

And so, I do recommend having extended next-generation sequencing done at least at diagnosis.  

When I generally think about repeating that, if there’s something that looks like it’s changing within the patient’s disease, to be honest, also on the flipside of that argument, sometimes this next-generation sequencing will mostly contribute to adding anxiety and will not necessarily directly impact how a patient is treated. And this is particularly true in patients with PV and ET, where we’ll sometimes order these tests, and we get a bunch of mutations back, but we don’t know what to do with that information yet.  

And so, as a researcher – not a clinician – as a researcher, I think it’s very important to have that information so that we can then do studies and understand the patterns of mutations and how that affects outcome. But as a clinician, and you as a patient, you need to really be aware of how that’s going to impact the patient in front of you and how that may impact you as a patient. Do you want to know if you have these mutations if nothing can be done about it? So, I would say, take a moment to reflect upon what I said and also to ask your clinician, how is this information going to help me? Do I need to have this information?  

Maybe you want to have it done so that it’s in your record. But maybe you don’t necessarily want to know those results. And everybody’s very different. And I think it’s absolutely wonderful to talk to my patients about all the information. But there may be some patients that really are just, like, do the test but don’t tell me the results, because I know that I’m just going to be very anxious knowing that I have something that I can’t do anything about. So, just take a minute to talk about it with your doctors. I think that’s really important.  

Advances in Myelofibrosis Research

Advances in Myelofibrosis Research from Patient Empowerment Network on Vimeo.

What are the recent developments in the study and advancement of myelofibrosis treatment? MPN researcher Dr. Gabriela Hobbs discusses ongoing clinical trials for new JAK inhibitors, BET inhibitors, and anemia therapies, among others.

Dr. Gabriela Hobbs is a hematology-oncology physician specializing in the care of patients with myeloproliferative neoplasms (MPN), chronic myeloid leukemia, and leukemia. Dr. Hobbs serves as clinical director of the adult leukemia service at Massachusetts General Hospital. Learn more about Dr. Hobbs.

See More From MPN Clinical Trials 201

Related Programs:

Key Questions to Ask When Considering an MPN Clinical Trial

Key Questions to Ask When Considering an MPN Clinical Trial

How MPN Researchers Collaborate to Advance Patient Care

How MPN Researchers Collaborate to Advance Patient Care

What Are the Long-Term Effects of JAK Inhibitors

What Are the Long-Term Effects of JAK Inhibitors?


Transcript:

Katherine:

What about myelofibrosis, Dr. Hobbs? What advances are being made in the care of patients with this more advanced MPN? 

Dr. Hobbs:

Yeah. So, in myelofibrosis, I would say it is almost difficult to keep track of how many clinical trials are currently open. So, in 2011, we had ruxolitinib approved, or Jakafi. That was the first JAK inhibitor. Since then, we’ve had two more JAK inhibitors approved, fedratinib (Inrebic) and most recently pacritinib (Vonjo). And we’re currently awaiting the fourth JAK inhibitor to be approved, and that’s called momelotinib.   

And in addition to the JAK inhibitors, there are lots of other clinical trials underway right now that are either alone – a drug by itself or a drug in combination with ruxolitinib.  

So, there are several Phase III studies. And the reason why that’s important is that after Phase III we usually see a drug approval. So, we can expect, hopefully in the next couple of years, to see many more drugs available on the market to treat patients with myelofibrosis. Some of those include agents that block different pathways within a cell. And that includes a drug called parsaclisib. There’s a drug called pelabresib (CPI-0610), which is a BET inhibitor.  

There’s another drug called navitoclax (ABT-263), which is a cousin of venetoclax (Venclexta), which is a drug that we’ve been using a lot in leukemia. So, there’s lots of different drugs that are being used in combination with ruxolitinib. There’s also a drug called luspatercept (Reblozyl) that’s also been approved for myelodysplastic syndromes. And I suspect that that’ll be approved as well to help patients with anemia. So, really, there’s lots of drugs that are being studied right now. And I think the question that we’re all asking is, well, how are we going to use all of these different drugs? So, I look forward to seeing the results of those studies.  

Katherine:

Mm-hmm. Will some drugs work better for some patients and others not? 

Dr. Hobbs:

That is such a good question. And so, what I’m hoping to see is exactly that. I’m hoping to see that for patients, for example, with anemia, perhaps we’re going to be using luspatercept and momelotinib. Perhaps we’re going to see that patients with certain mutations may respond better to certain medications like the BET inhibitors or navitoclax or the PI3 kinase inhibitor, parsaclisib. But as of now, we don’t have enough information.  

We haven’t seen enough results of these studies to start to be able to know, you know, what is the patient that’s going to do better with two drugs versus one drug? And so, I think that over the next couple of years we’re going to start to have answers to those questions.  

Advances in Polycythemia Vera Research

Advances in Polycythemia Vera Research from Patient Empowerment Network on Vimeo.

What are the recent developments in the study and advancement of care for patients with polycythemia vera (PV)? Dr. Gabriela Hobbs reviews recently approved PV treatments as well as those currently in clinical trials.

Dr. Gabriela Hobbs is a hematology-oncology physician specializing in the care of patients with myeloproliferative neoplasms (MPN), chronic myeloid leukemia, and leukemia. Dr. Hobbs serves as clinical director of the adult leukemia service at Massachusetts General Hospital. Learn more about Dr. Hobbs.

See More From MPN Clinical Trials 201

Related Programs:

Resources for Accessing MPN Clinical Trials

Resources for Accessing MPN Clinical Trials

The Risks and Benefits of Participating in an MPN Clinical Trial

The Risks and Benefits of Participating in an MPN Clinical Trial

Should All MPN Patients Undergo Molecular Testing

Should All MPN Patients Undergo Molecular Testing?


Transcript:

Katherine:

There was recently an interferon approved for use in patients with PV. What other studies are showing promise for patients with PV?   

Dr. Hobbs:

Yeah. So, we as a community, there’s been a lot of excitement about this new interferon that was approved, the ropeginterferon (Besremi) study. And there are still some ongoing studies utilizing ropeginterferon to see if we can use it differently.  

Because currently the way that that drug is approved is that it has to be titrated up very slowly to get to the maximum dose. So, that’s something that is still ongoing. In addition, there’s a new drug that’s being studied called Rusfertide (PTG-300) from a company called Protagonist. And this drug has been very interesting. It acts through iron metabolism.  

And so far in preliminary results, it has shown that a lot of the participants that receive this medication no longer need phlebotomy. And I think what’s exciting about this is that phlebotomy is a very archaic way of treating patients.  

And I hope that we can stop utilizing it. So, it’s nice to have a compound that’s specifically asking that question. And the other thing to keep in mind is that this drug has been used in combination with other drugs, which is really reflective of how participants or patients show up to clinics.  

Some patients are not going to be on any medications. Some patients may be on hydroxyurea (Hydrea).  

Some patients may be on an interferon. Some patients may be on ruxolitinib (Jakafi). And these trials allow participants to be on a variety of different medications. So, that’s an exciting new compound. 

How Driver Mutation Research Is Advancing MPN Treatments

How Driver Mutation Research Is Advancing MPN Treatments from Patient Empowerment Network on Vimeo.

How do driver mutations affect MPN care? MPN researcher Dr. Gabriela Hobbs shares an update on what’s being learned about the JAK mutation and how researchers are working towards targeted therapy for MPNs.

Dr. Gabriela Hobbs is a hematology-oncology physician specializing in the care of patients with myeloproliferative neoplasms (MPN), chronic myeloid leukemia, and leukemia. Dr. Hobbs serves as clinical director of the adult leukemia service at Massachusetts General Hospital. Learn more about Dr. Hobbs.

See More From MPN Clinical Trials 201

Related Programs:

Advances in Essential Thrombocythemia Research

Advances in Essential Thrombocythemia Research

Advances in Polycythemia Vera Research

Advances in Polycythemia Vera Research

Advances in Myelofibrosis Research

Advances in Myelofibrosis Research


Transcript:

Katherine:

There have been huge developments in the last 10 to 15 years in the field of MPN. So, I’d like to dig a little deeper. We hear about the common driver mutations in MPNs like JAK2, CALR, and MPL. How are these being studied , and what is being discovered?  

Dr. Hobbs:

Yeah. So, it’s amazing how in the last 15 years really so much has been discovered. You know. The JAK2 mutation was first published out in 2005 and calreticulin in 2013. So, those are relatively recent discoveries. And I think a lot of efforts has been put into learning about what these mutations are doing and how they lead to disease. And so, we have the JAK inhibitors, which block the signaling through a pathway called JAK-STAT. And all of these mutations will activate that pathway within cells.  

And so, many of the approved drugs, for example, ruxolitinib (Jakafi), fedratinib (Inrebic), and pacritinib (Vonjo), work on blocking that pathway.  

But since then, we’ve also learned that there are other mutations and other pathways that are likely involved in the development of myeloproliferative neoplasms and also their progression. And so, what we’re seeing now is that many of the clinical trials that are being conducted don’t just target the JAK-STAT pathway or the pathway that’s influenced by these main mutations.  

But also block other pathways to try to really block all the variant expression of signaling in the myeloproliferative neoplasms. And so, we’re trying to attack it by many different angles.  

Katherine:

Yeah. Is there a possibility of specific targeted therapies at MPNs similar to those in AML such as FLT3 inhibitors? 

Dr. Hobbs:

Absolutely. So, similarly to AML, we know that we have mutations in similar types of genes called tyrosine kinases. So, these are enzymes that are turned on and always active. And so, I think there is definitely hope that we can develop some targeted agents. For example, ruxolitinib or the other JAK inhibitors are similar. They’re tyrosine kinase inhibitors where they block an enzyme, specifically the JAK2 enzyme.  

But I think that we can definitely do better and develop more specific inhibitors, for example, a molecule that just blocks the JAK2 mutation and not just every JAK2 molecule in every cell. Similarly to AML, there are mutations, for example, in enzymes called IDH.  

And we have IDH inhibitors for AML. And there are some studies that are using IDH inhibitors for MPN. So, I think we’re going to continue to see more targeted therapies specific to the mutations that occur in MPN. 

How MPN Researchers Collaborate to Advance Patient Care

How MPN Researchers Collaborate to Advance Patient Care from Patient Empowerment Network on Vimeo.

MPN specialist and researcher Dr. Gabriela Hobbs discusses how collaboration and data sharing among researchers around the world impact MPN treatment advances.

Dr. Gabriela Hobbs is a hematology-oncology physician specializing in the care of patients with myeloproliferative neoplasms (MPN), chronic myeloid leukemia, and leukemia. Dr. Hobbs serves as clinical director of the adult leukemia service at Massachusetts General Hospital. Learn more about Dr. Hobbs.

See More From MPN Clinical Trials 201

Related Programs:

Advancing MPN Research: How Clinical Trials Work

Advancing MPN Research: How Clinical Trials Work

The Risks and Benefits of Participating in an MPN Clinical Trial

The Risks and Benefits of Participating in an MPN Clinical Trial

How Driver Mutation Research Is Advancing MPN Treatments

How Driver Mutation Research Is Advancing MPN Treatments


Transcript:

Katherine:

I’d like to start by discussing your role as an MPN researcher. You’re on the front lines for advancements in the field. What led you to there, and why is it so important to you?  

Dr. Hobbs:

Many things in my life led me to becoming an MPN clinician. First, I wanted to be a clinical investigator since I was very little, and I read a Louis Pasteur book about – you know. And I was fascinated by the fact that you could be both a scientist and a clinician. And after that, I had phenomenal teachers and mentors. And I was really always drawn to patients with hematologic malignancies. I thought that that interaction was very intense and intimate.  

And I was honored to be a part of that interaction. And then from a research perspective and from a scientific perspective, I very clearly remember seeing when the first targeted therapy, Imatinib, was approved when I was an undergrad. And I just thought that was the most fascinating thing. And so, I’ve basically continued to feel that way as I’ve gone through my training, and I’m thrilled to be able to have actually become an MPN clinician so many years later.   

Katherine:

With the American Society of Hematology or ASH meeting taking place this month, it demonstrates how researchers work together around the world to advance care.  

Can you share with the audience how this collaboration works?  

Dr. Hobbs:

Yeah. So, the American Society of Hematology meeting – or the ASH meeting – is really one of my favorite events of the year.  

And it really highlights what you said. It is such a positive environment, and it’s so exciting to use that opportunity to talk to my collaborators from across the globe. And I really think that that’s where the scientific community shines because really all of us are actually trying to figure out how to work together and overcome sometimes a lot of obstacles – bureaucratic obstacles, regulatory obstacles – to make sure that we can share data, do it the right way. But really we always have one thing in mind.  

And that is to be able to advance the care that we give our patients. And so, that collaboration and really that collaborative environment is always very positive. And I always come back home very energized from that. And then just seeing all my colleagues presenting all the wonderful things that they are working on and getting updates on their research is just an exciting environment.   

 Katherine:

In your view, why is it essential to present and share data at these larger conferences like ASH? 

Dr. Hobbs:

So, for many different reasons. I mean, there are many different ways of presenting data that can be done through just publishing a paper. But the nice thing about conferences – and especially large conferences – is that you really get an opportunity to present work in progress. And some of these research projects may not end up turning into bigger projects or they may not become bigger trials. But all of them have at least an opportunity to learn something from them, whether or not they worked or they didn’t work.   

Oftentimes when things are published in journals, especially the high-impact journals, we are seeing trials that had positive results. But sometimes we don’t see those smaller trials that never went anywhere. And so, having a forum when we can discuss work that’s ongoing, discuss about projects that are maybe having issues, all those things actually really help us to change our research questions or develop new research questions based on what’s working and also really what’s not working. And so, having this large forum to present all of that data, I think, is really, really important to helping us design future clinical trials and projects. 

Tools for Choosing Myeloma Therapy

Tools for Choosing Myeloma Therapy from Patient Empowerment Network on Vimeo.

When faced with choosing a myeloma treatment, what should be considered? This animated video reviews factors that impact treatment decisions, provides a list of questions to ask your healthcare team about therapy and advice for engaging in your myeloma care.

See More From Innovative Myeloma Therapies

Related Programs:

What Is Myeloma CAR T-Cell Therapy?

Immunotherapy: Which Myeloma Patients Is It Right For?

Immunotherapy: Which Myeloma Patients Is It Right For? 


Transcript:

Whether a patient is newly diagnosed with myeloma or is facing a relapse, choosing a treatment approach can feel overwhelming.   

Shared decision-making is a process where patients and healthcare providers communicate and collaborate to make care decisions. This approach encourages patients to take a more active role in their care and treatment and can help them feel more confident when choosing a therapy. 

So, what can impact myeloma treatment decisions? 

  • There are patient-related factors, such as a patient’s age, fitness level, and pre-existing conditions. 
  • And, disease-related factors, including the aggressiveness of the patient’s disease and its location in the body. 
  • And, treatment-related factors, such as past treatments a patient has received or if they are refractory to medicines. 

How can you play a role in making treatment decisions?  

You can start by making a list of questions in advance of your appointment. This can help you to organize your thoughts before you meet with your healthcare team.  

And, when working with your doctor to choose a treatment, consider asking the following questions: 

  • What type of myeloma do I have?  
  • Are there test results that may impact my treatment choices? 
  • What are the risks and benefits of each treatment option? 
  • What approach do you recommend and why?  
  • How is the treatment administered, and what side effects might I expect? 
  • What are my options if this treatment stops working? 
  • Are there newer treatment options available to me, including immunotherapy?  
  • And, is there a clinical trial that might be right for me?  

It’s also a good idea to bring a friend or loved one to your appointment for support to take notes and help you recall information. Afterwards, discuss the appointment together – you can use this time to talk about your care plan and do your own research to learn more about your options.   

The patient portal is another useful tool in your care. You can use it to view lab and test results. And you can use the messaging feature to communicate with your healthcare team when you have more urgent questions to address before your next visit. 

Now that you know more about how to make myeloma treatment decisions, how can you take action? 

  • First, consider a second opinion or a consult with a specialist. 
  • Then, ensure you have had all relevant myeloma testing. 
  • Next, understand and participate in treatment decisions. This includes learning about your options, so you can weigh the pros and cons of each approach. And be sure to speak up and share your personal preferences and goals with your care team. 
  • Communicate regularly with your healthcare team – don’t wait to share information only when you have an appointment.  
  • And finally, bring a friend or loved one to appointments and always write down any questions or concerns in advance. 

Visit powerfulpatients.org/myeloma to learn more about myeloma and access tools for self-advocacy. 

What Are the Phases of Myeloma Treatment?

What Are the Phases of Myeloma Treatment?  from Patient Empowerment Network on Vimeo.

Myeloma specialist Dr. Mark Schroeder explains the phases of myeloma treatment, defining induction, consolidation, and maintenance therapy for patients with myeloma.

Dr. Mark Schroeder is a hematologist at Siteman Cancer Center of Washington University School of Medicine in St. Louis. Dr. Schroeder serves as Associate Professor in the Department of Medicine. Learn more about Dr. Schroeder.

See More from Engaging in Myeloma Treatment Decisions

Related Resources:

How Does Disease Staging Affect Myeloma Treatment Choices?

Expert Perspective Why Myeloma Patients Should Weigh in on Their Care Decisions

Understanding Myeloma Treatment Types

Transcript:

Katherine Banwell:

Let’s talk about the different phases of therapy for myeloma, and I’m going to ask you for some more definitions. What is induction therapy? 

Dr. Mark Schroeder:

Induction therapy is the first treatment that we’re starting for myeloma. It’s oftentimes a combination of a number of chemotherapies that our goal is to get control of the cancer quickly, so reduce the burden of the cancer in a patient’s body.  

Oftentimes, when patients present with myeloma, that’s when the burden of cancer is the highest. So, induction therapy is a combination often of three or four different drugs given over the course of about three to four months to treat the myeloma and get initial control. 

Katherine Banwell:

What about consolidation therapy? What is that? 

Dr. Mark Schroeder:

So, after you have had a response to induction therapy, your oncologist might talk about, “Well, let’s deepen that response.” That’s when we think about consolidation. So, it’s going to be poten – most of the time is a change of therapy from the three or four drugs that you were treated for in the myeloma. An example of consolidation would be going through a stem cell transplant or more chemotherapy after stem cell transplant. So, that’s a change in therapy, and it ends up deepening the response, killing more of the cancer. 

Katherine Banwell:

And what about maintenance therapy? 

Dr. Mark Schroeder:

So, after you have gone through induction, you have control of the myeloma, we’ve deepened that response with consolidation, we know that myeloma is a cancer that tends to come back. And we know from experience that continuing some of the drugs that we used in induction at low doses are effective to try and prevent it from progressing or coming back, and it extends that period of time – and that’s maintenance therapy. It’s using some of the drugs we used to initially treat myeloma at lower doses to continue to suppress low levels of the cancer. 

Finding an MPN Treatment Approach That Is Right for You

Finding an MPN Treatment Approach That Is Right for You  from Patient Empowerment Network on Vimeo.

Appropriate and effective treatment is an essential part of thriving with an MPN. Dr. Joseph Scandura reviews the goals of MPN treatment and factors that should be considered when choosing a therapy.

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:

 
Thriving With an MPN | Tips for Managing Worry and Anxiety

Thriving With an MPN | Tips for Managing Worry and Anxiety

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 Banwell:

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 kind of 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 kind of 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. 

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.  

How Clinical Trials Advance MPN Treatment and Research

How Clinical Trials Advance MPN Treatment and Research from Patient Empowerment Network on Vimeo.

MPN expert Dr. Angela Fleischman provides a deeper understanding of how clinical trials advance myeloproliferative neoplasm (MPN) research and treatment, explains safety protocols in place for trials, and addresses common misconceptions associated with clinical trial participation. Dr. Fleischman also shares an update on emerging MPN research.

Dr. Angela Fleischman is a physician scientist and assistant professor in the Department of Medicine at the University of California, Irvine. Learn more about Dr. Fleischman.

See More From MPN Clinical Trials 201

Related Programs:

Understanding Common MPN Clinical Trial Terms

Understanding Common MPN Clinical Trial Terms

MPN Clinical Trial Safety, What Are the Protocols?

MPN Clinical Trial Safety, What Are the Protocols?

How Can You Access an MPN Clinical Trial?

How Can You Access an MPN Clinical Trial?


Transcript:

Katherine:

Hello, and welcome. I’m Katherine Banwell, your host for today’s program. Today, we’re going to discuss how clinical trials advance research for myeloproliferative neoplasms, or MPNs, and we’ll talk about what MPN patients should know about participation. 

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. Angela Fleischman. Dr. Fleischman, welcome. Would you please introduce yourself? 

Dr. Fleischman:

Thank you very much for the invitation. Hi, everyone. My name is Angela Fleischman. I’m what’s called a physician scientist, meaning, I do research as well as see patients, and my focus for my entire career thus far has been on myeloproliferative neoplasms, specifically their role of inflammation in MPN. And I am at the University of California, Irvine in Southern California. So, nice to be here today. 

Katherine:

Well, thank you so much for joining us and taking the time. Before we get into the discussion about clinical trials, because you’re so heavily involved in research, let’s talk about the latest developments in the field. What MPN clinical trials are you excited about right now? 

Dr. Fleischman:

So, I would say, there’s a lot of new clinical trials in the field for myelofibrosis, which is the most severe form of myeloproliferative neoplasm. 

There tend to be more clinical trials because that’s a patient population in – I don’t want to say in more need, but they do have more need in terms of necessitating better treatments. 

Drugs that are quite far along in clinical trials – and in order for a drug to make it to market, one needs to go through multiple clinical trials to demonstrate the safety, as well as efficacy. Things like a BET inhibitor are very, very promising in moving forward in clinical trials. Other medications for other diseases, such as polycythemia vera, not anymore in clinical trials, but excitingly, newly FDA-approved, was ropeginterferon for polycythemia vera. 

So, that’s a real exciting development for Polycythemia Vera patients. 

And now, we have – outside of the context of clinical trials, because I want to talk about what’s actually available to patients now, we now have three JAK inhibitors available for myelofibrosis patients. And really, since 2011, we had only had one, and then, more recently, a second JAK inhibitor, but now, we have three. So, now we’re moving into an era where we can tailor a specific JAK inhibitor for a specific myelofibrosis patient, depending on what their particular needs are. So, I think that that’s very promising. And then, there are lots of clinical trials combining JAK inhibitors with new drugs. 

Katherine:

So, how does it work? How do clinical trials advance MPN research and treatment? 

Dr. Fleischman:

Well, there are multiple stages of clinical trials. One needs to have some rationale for testing a specific drug in patients. You just can’t say, I just want to take something off the shelf and see if it works for myeloproliferative neoplasms. 

There could be different ways that things sort of enter into clinical trials, either preclinical data from in vitro, meaning, in the lab, in the liquid media, with cells, that makes somebody think that it might work in humans, or that it works in a similar disease to myeloproliferative neoplasm. So, it’s a little bit of a stretch, but a very rational stretch, to then test it in a new population. 

First and foremost, safety needs to be evaluated, because as physicians, one of our primary objectives is to do no harm to patients. So, at very early stages of clinical trials, the primary objective is to see what the appropriate doses, what’s tolerated, what the side effect profile is. 

And then, moving on to efficacy. So, maybe it’s tolerated, but does it actually work at the next stage of clinical trials. Then, a much larger clinical trial would be to do a head-to-head comparison between, in most cases, standard of care versus drug X. 

And I think, for clinical trials, in particular, for myeloproliferative neoplasm, it’s very important to understand what the stated, primary end point is, in particular, for myelofibrosis patients, that myelofibrosis patients may have different problems. Some myelofibrosis patients, their primary issue may be anemia. And so, if they’re looking for a clinical trial to address their anemia, they would probably want to be looking for one whose primary end point is transfusion, freedom from transfusions, or improving the anemia, not necessarily – there was another trial that’s primarily looked at spleen reduction, but they didn’t have an enlarged spleen, that, necessarily, wouldn’t be appropriate for the patient. 

So, I think it is particularly important in myeloproliferative neoplasm to identify what the primary end point is, and whether what you’re going for is that primary end point. 

Katherine:

Mm-hmm. Any advances that are being done in MPN research require MPN patients to participate in clinical trials, right? 

Dr. Fleischman:

Of course. 

Katherine:

So, to start, let’s talk about where clinical trials fit into the treatment plan for ET, PV, and MF patients. When should a patient consider participating in a clinical trial? 

Dr. Fleischman:

Okay, well, I guess a patient could really consider participating in a clinical trial at any point if they had a very altruistic philosophy, that understanding that their participation may not necessarily help them at this moment in time, but may help others in the future, and we’ll gain knowledge about myeloproliferative neoplasms. 

That’s one approach. 

Another approach, which is probably a more usual approach, is when a patient has already tried standard therapies and they haven’t quite worked for them, or they’re in a class where, maybe, we don’t have really great standard therapies for somebody. 

For example, a myelofibrosis who may not be doing too well and may not necessarily be a candidate for a transplant, I think that’s a very reasonable population to go out and seek clinical trials, because there’s really not necessarily a great standard of care treatments for that patient population, or ET or PV patients who have tried standard of care and, maybe, can’t tolerate standard medications, or they’re just not working for them. 

But really, anytime somebody can do a clinical trial, if that’s what they feel is important to them.  

Katherine:

What are the benefits and risks of a trial participation? 

Dr. Fleischman:

So, the benefits are that you’re getting a drug that, potentially, is better than standard of care, that could be standard of care five to 10 years from now, but you’re getting it early.  

As investigators, ethically, we can’t start a clinical trial if we believe that the drug that we’re testing might have negative side effects on the patient, or maybe worse than standard of care. I mean, ethically, that’s not appropriate. So, ethically, we believe that what we’re testing may be better than what we’re currently giving patients, but we don’t know that. So, that’s the purpose of a clinical trial. 

So, a clinical trial, it’s a new drug. So, could have side effects that are on unanticipated, including death. I mean, that’s just the reality. That would be a very uncommon scenario, but it’s an unknown, so it’s an unknown. 

Other things that I think are very important to discuss are the financial implications of a clinical trial. On the pros, one could be getting a free drug that is outside of standard of care, and many of the tests that are done for the purposes of the research are covered. However, drugs, say, if it’s a combination drug, standard of care plus a new drug, the standard of care drug is usually billed to insurance. And so, the patient would need to pay for that, or if there are studies that would be considered standard of care, the patient would need to cover them. 

So, I think it, really, is important to discuss the financial implications. What money is it going to save you by participating, and may there be extra costs, or hidden costs, potentially, involved by participating? 

Katherine:

Yeah. Let’s talk about safety in clinical trials. Would you review the safety protocols that are in place before a clinical trial even begins? 

Dr. Fleischman:

So, before a clinical trial begins, there, usually, needs to be safety information in animals. Also, a lot of drugs have been tried in other diseases first. Either, they’re, have been studied in clinical trials and maybe not found to be very efficacious, but at least we have the value of the safety data in another population. 

So, we’re entering, again, into clinical trials with the understanding that it would not be harmful to humans with the data that we have available in animals, or in liquid culture. But again, we just don’t know that. And then, also, for many clinical trials, starting off at lower doses, and then, increasing the dose slowly in different cohorts of patients, to see what’s the maximally tolerated dose. 

As well as, when somebody is on a clinical trial, safety and side effects are very closely monitored, and even small side effects that likely have nothing to do with the drug, really do need to be investigated fully, just to make sure that they’re not related to the drug. 

Katherine:

Yeah. How do you know if the medicine is safe prior to starting a human trial? 

Dr. Fleischman:

That’s a great question. 

Based on what the molecule looks like, as well as, many times, they’ve been tested in animals to see – for example, for myeloproliferative neoplasm, it would be important to know, does it change a healthy rat’s blood count? Does it harm their liver? Those sorts of things, and safety information is usually available for a new drug. 

Katherine:

Are patients monitored more closely when they’re in a trial? 

Dr. Fleischman:

Yes, definitely. And for the purposes, mainly, of paying very close attention to even small side effects that, if somebody was not watched closely, may be missed because they’re so subtle. 

Katherine:

What if a patient decides to leave a trial? Does that negatively impact their care? 

Dr. Fleischman:

No, and I think that’s a very important point, that, ethically, as investigators, we cannot – and we do need to make it a point to communicate this fully with the patient, that when we’re asking the patient, or informing them about a potential clinical trial, we need to inform them that whether or not they participate will have nothing to do with the way that we treat them. We will treat them equally, regardless of whether or not they participate, as well as, anytime during the clinical trial, a patient has the absolute right, for whatever reason, they can decide to leave the clinical trial. That’s the most – I don’t say that’s the law, but those are the rules of clinical trials, as well as, a patient cannot be treated differently if they decide to leave a clinical trial.  

We have to be fair. I mean, this is – you have to be fair to all patients, and all patients deserve excellent treatment, regardless of whether they participate in the clinical trial. 

Katherine:

Dr. Fleischman, we’ve been talking about what happens when people participate in trials. But what if they don’t? Why is it crucial that patients participate in trials? 

Dr. Fleischman:

Because without participation in clinical trials, we are not going to further our understanding of myeloproliferative neoplasm. Many of the drugs that we use today in myeloproliferative neoplasms, as well as other diseases, the reason why we use them today is because people 10, 20 years ago participated in the clinical trial and demonstrated a benefit of these medications. So, people don’t participate, we’re not going to have new drugs for myeloproliferative neoplasms.  

Katherine:

All right. We know that much of the reason that people don’t participate is because of various stigma associated with clinical trials, and I’d like to talk about that with you. 

Let’s start with the word “experiment.” Why does this word not pertain to clinical trials? 

Dr. Fleischman:

So, I think the word “experiment” may have a negative connotation, and making the patient think, maybe they’ll say, a guinea pig. The only way that we can identify whether a drug is going to be beneficial is to test it out in humans with a particular disease. 

So, I mean, on one hand, it is an experiment, because we don’t know what’s going to happen, but we’re doing the experiment for the benefit of people who are suffering from the same disease. 

Katherine:

Yeah. Yeah. That’s a good explanation. What would you tell patients who are worried that they will receive a placebo? 

Dr. Fleischman:

So, that is part of a clinical trial, and it is also important to look how your clinical trial that you’re interested in is structured.  

So, some clinical trials do receive, or split into placebo, or active drug, and double-blinded means that the patient doesn’t know, nor the physician knows. So, no one knows, and that’s important because we don’t want to sway any subconscious things that, if you know you’re getting the drugs, then you’re going to say your symptoms are getting better, things like that. 

Again, ethically, in a clinical trial, we cannot not give somebody treatment that they – we can’t keep treatment from somebody. So, for example, if a person with polycythemia vera was a, per guidelines, should be on a cytoreductive agent, we cannot, ethically, treat them without a cytoreductive agent. So, it would be – they would have standard of care plus placebo, or drug X. 

So, maybe I’m not explaining this correctly, but if a placebo study is done, the placebo can’t take the place of something that we know is good for the patient. 

We can’t leave them hanging without any treatment, unless, for their specific situation, there’s not, necessarily, a known standard treatment, that it would be very reasonable to treat them with nothing.  

Katherine:

Another myth we often hear is that trials should only be considered if you have no other options. Why is that false? 

Dr. Fleischman:

I think there is a place for patients with no other options that – they may be more inclined to participate in, I want to say, higher risk studies, in which there’s less data to support a particular medication. But that’s why we look at these drugs in patients with no other options, because there’s no other reasonable thing to give them. 

But the patients with no other options may not be an accurate representation of the patient population, as a whole. So, it is important for people who may have other options, but maybe they want to think about, well, I do have a standard option, but maybe there’s something better out there for me, to participate in clinical trials. 

Katherine:

What if an MPN trial isn’t offered at the center where a patient receives care? What can they do?  

Dr. Fleischman:

Many times, specific clinical trials are only open at specific universities. And so, it’s very likely that your university, or the place where you receive care, may have a few clinical trials, or maybe one, or maybe zero for MPNs, but may not necessarily fit your exact circumstances. 

So, what I would recommend is, doing searching on your own, either through clinicaltrials.gov, or the MPN Research Foundation also has some nice resources, but doing some research on your own to identify some potential clinical trials that you’re interested in, and then go to your primary oncologist and say, “Hey, I printed these out. I think these might look really interesting to me.” 

And usually, on clinicaltrials.gov, they would have where they are, and you can actually, also, search for your state. So, maybe bring some that are close to you, and discuss with your primary oncologist the pros and cons of them. And then, ask your primary oncologist to make a referral to the location where they offer that specific trial. 

And a lot of times, you can – there’s a phone number you can call and be pre-screened. Say, “Hi, I’m a 55-year-old man with myelofibrosis,” and there are specific inclusion, exclusion, criteria that they can ask you. And if you don’t meet the inclusion criteria, then it’s not worth your time to go and have an actual visit, but if you do meet the inclusion criteria, then you could go and have an actual visit, and learn a little bit more.  

Katherine:

Oh, that’s great information. Thank you. Here’s a question we received from an audience member, prior to the program. Susan wants to know, “How can I get my community oncologist on board with trial participation? I’m interested in participating in a clinical trial that’s based in Chicago, and I’ll need her help in coordinating care with the team from a distance. Any advice for how to talk to my local doctor about that?”  

Dr. Fleischman:

So, that may be a tough one. So, many times, if somebody has to travel for a clinical trial, it does require some coordination. There are specific – and it’s clinical trial specific. There may be specific things that actually need to be done at the study site. For example, specific labs that would be drawn, and say, need to be frozen within two hours, or specific tests, for example, MRIs, if you need to look at the spleen size, you would need to do it on the same machine for everyone. 

So, there are specific things that have to be done at the location, or if it’s written to the protocol, you have to come to the location for a physical exam on this day and this day, and if it’s not within a two-to-three-day window, then there’s a deviation, and the data is not valid. 

So, what I would say is – sorry, this is a long answer here, but where certain things, if they’re written in the protocol that say a CBC could be drawn at any institution at week four, then that would be reasonable to have your primary oncologist do. But in the context of clinical trials, certain things are really set in stone as to the exact dates that needs to be done, and the exact location. And if they’re not done exactly, to a tee, then your data will not be – your data cannot be used for the analysis. 

Katherine:

Mm-hmm. But then, there’s also the issue of patients being willing and able to travel a distance to a teaching university where a clinical trial might be happening.  

Dr. Fleischman:

Correct, yes. And I think that, for some clinical trials, when the protocol is made, understanding that trying to minimize the trips to the actual site, and working the protocols, working some sort of wiggle room in the protocol, such that lots of stuff, or hopefully, lots of stuff, can be done remotely. But sometimes, it’s just not possible.  

Katherine:

Yeah. I’d like to turn our conversation to health disparities, Dr. Fleischman. Based on American history, some people believe that they won’t receive equitable or safe care if they participate in a trial. 

How can you reassure those people who are concerned they’ll be treated fairly? 

Dr. Fleischman:

Now, I think that this is a very important point, and something that there’s been a lot of emphasis, to try to improve diversity in clinical trials, because our American population is quite diverse. However, the participants that, in general, participate in clinical trials are, unfortunately, still have not a very diverse population in our clinical trials. 

I think what we need to first start doing is education, to reach out to underrepresented communities, to start to build the trust amongst these communities, to tell them about the value of clinical trials. And I think it’s going to take some time to build trust first, because it does take quite a bit of trust to participate in the clinical trial. 

But I don’t have a great answer for that, other than, we need to work hard to, first, build trust, and then, I think the diversity will come. 

Katherine:

Mm-hmm. How does holding on to some of these beliefs lead to limitations in care and create disparities? 

Dr. Fleischman:

So, and rightfully so, if a patient is scared, or has some reservations of participating in a clinical trial, they may – that’s offered to them, that they provide them with, potentially, something better than standard of care. They may be missing out on a potential opportunity. 

Also, potentially, if a patient, if they’re asked about a clinical trial and they have a negative connotation about them, they may lose trust with their physician, if they say, oh, my physician is asking me to participate in a clinical trial. 

This means that they’re thinking of me as an experiment, and maybe they’re not really thinking of me as patient. And so, they may not have that trust with their physician, and so, may not be as open, in terms of communication, with their physician.  

I think it all boils down to trust, and as physicians, we need to demonstrate that we are worthy of the patient’s trust, and we really are ingrained in us to treat every patient the same. I mean, that’s what our oath is. That’s what we’re supposed to do, and I think that the vast majority of patients, they have, ethically, are treating patients exactly the same, regardless of their circumstances. 

Katherine:

Yeah. Health equity means that no matter what a patient’s circumstances, whether it be race, income issues, lack of education, that they should have access to the best care. What is being done by the medical community to address this issue? 

Dr. Fleischman:

So, yes, this is a significant issue, and in particular, with myeloproliferative neoplasms, in whom there are lots of oral drugs – or with interferons, it’s injectable, but you get the prescription, and you give it to yourself – that there can be quite high copays, in some cases, exorbitant amounts, which, really, are not able to be paid for by the vast majority of people. 

So, many companies do have copay assistance programs. Also, foundations have copay assistance programs. So, I think that is, at least, one step in trying to make things more equitable, to get people who need a drug, their drug, at a very reasonable cost. Again, it does take some time, some legwork on the part of the patient, to seek out these programs, or to find an advocate for themselves to seek out these programs for them. 

Katherine:

Yeah. Would a healthcare team be part of that process, though? Would they be able to help the patient? 

Dr. Fleischman:

They will be able to help the patient in terms of saying, “Hey, there’s this program for this drug. Why don’t we fill out the form together?” Or, “Why don’t you call this,” you know. Many times, the patient needs to initiate the process. So, I think the healthcare team can sort of guide the patient in saying, this is what’s available, we can help. We can fill out our portion of the form, you fill out your portion of the form. But no, it does need to be – the patient needs to be an active participant in seeking out the support. 

Katherine:

Mm-hmm. Before we end the program, Dr. Fleischman, I’d like to close with some advice from you. What do you want to leave MPN patients with, relating to clinical trial participation? 

Dr. Fleischman:

I would say that MPN patients today are the key to our future treatments. 

Without participation in clinical trials today, there’s going to be no new drugs for myeloproliferative neoplasms. They’re just not going to appear. We need to test them in patients before them actually coming to market, and before really knowing whether they work or not. So, I would say that the MPN patients today are the key to the future of MPN treatments.  

Katherine:

Dr. Fleischman, thank you so much for joining us today. 

Dr. Fleischman:

My pleasure. As always, I really enjoy connecting with MPN patients, and I think this was a very important topic to discuss.  

Katherine:

Yeah. 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. 

Expert Perspective: Why Myeloma Patients Should Weigh in on Their Care Decisions

Expert Perspective: Why Myeloma Patients Should Weigh in on Their Care Decisions from Patient Empowerment Network on Vimeo.

Myeloma specialist Dr. Abdullah Khan shares key advice encouraging patients to participate in care and treatment decisions and discusses the importance of communicating symptoms and side effects to your healthcare team.

Dr. Abdullah Khan is a hematologist specializing in multiple myeloma and plasma cell disorders at the Ohio State University Comprehensive Cancer Center – The James. Dr. Khan is also an assistant professor in the Division of Hematology at The Ohio State University. Learn more about Dr. Khan.

See More from Engaging in Myeloma Treatment Decisions

Related Resources:

How Does Disease Staging Affect Myeloma Treatment Choices?

 
Expert Perspective Advances in Treating Relapsed and Refractory Myeloma

The Role of a Myeloma Specialist on Your Care Team

Transcript:

Katherine:

Let’s turn to decision-making, Dr. Khan. What is the role of the myeloma patient when making care and treatment decisions?  

Dr. Khan:

As a provider, my role is to inform the patient of the facts and the need for a decision. The purpose of the discussions is to determine the patient’s goals and preferences, because it’s essential the patient’s values of respected. The best outcomes occur when the medical facts align with the patient’s preferences. And this is a multi-disciplinary team approach.  

Katherine:

Why is it so important for patients to share any symptoms and side effects they’re having with their healthcare team?   

Dr. Khan:

I read something recently on an NIH website titled “What Do I Need To Tell the Doctor?” that I think answers this question well. And I’m quoting the article. “Talking about your health means sharing information about how you feel physically, emotionally, and mentally. Knowing how to describe your symptoms and bringing up other concerns will help you become a partner in your healthcare.”  

I think I really like that end, “partner in your healthcare.” The patient’s symptoms and suggest disease or disorder in the body. If there are concerns, this may prompt a clinic visit or the patient may be advised to go to the closest ER or urgent care depending on the urgency of the situation. But in other cases, the healthcare team may help provide reassurance that the symptom can be continued to be monitored more resolution, or it can be evaluated in more detail if it persists or worsens.  

Katherine:

What about side effects? Why is that important for patients to share any side effects they may be having?  

Dr. Khan:

Side effects may be a result of the disease itself. It might be a marker of the side effects from the treatment. Or I’m focused on the multiple myeloma, but there’s every other organ system in the body that also needs help. So, the myeloma might be doing okay. The treatment might be doing okay. But, for example, we might have a lung toxicity from their pre-existing COPD or a heart toxicity from their pre-existing coronary artery disease. So, it’s very important to share all symptoms So, we can see how to properly assess it.   

Katherine:

And better care for the patient.  

Dr. Khan:

Right.  

Expert Perspective: Advances in Treating Relapsed and Refractory Myeloma

Expert Perspective: Advances in Treating Relapsed and Refractory Myeloma from Patient Empowerment Network on Vimeo.

Dr. Abdullah Khan, of Ohio State University Comprehensive Cancer Center – The James, reviews currently available treatments as well as those in development for patients with relapsed or refractory myeloma. 

Dr. Abdullah Khan is a hematologist specializing in multiple myeloma and plasma cell disorders at the Ohio State University Comprehensive Cancer Center – The James. Dr. Khan is also an assistant professor in the Division of Hematology at The Ohio State University. Learn more about Dr. Khan.

See More from Engaging in Myeloma Treatment Decisions

 

Related Resources:

How Does Disease Staging Affect Myeloma Treatment Choices?

Expert Perspective Why Myeloma Patients Should Weigh in on Their Care Decisions

Relapsed and Refractory Myeloma Defined

Transcript:

Katherine:

Are there any recent advances in treatment for patients with relapsed or refractory disease?  

Dr. Khan:

Currently and in the past 20 years or so, we’ve seen about 20 approvals for new drugs for patients with multiple myeloma. The way the approval process works it typically looks at the effectiveness of a drug in the relapsed refractory setting first. And after establishing the safety and efficacy, the therapies are moved earlier in the disease course.   

The great example of this are the anti-CD38 monoclonal antibodies daratumumab and isatuximab. They were first approved in the relapsed refractory setting in combination with other antimyeloma treatments. And due to their impressive effectiveness and relative safety, they’re already being used in the frontline setting for patients with newly diagnosed multiple myeloma.   

In the newly diagnosed setting, a commonly cited study is the phase two GRIFFIN trial. And that added daratumumab to the BRd, or bendamustine (Bendeka, Treanda), lenalidomide (Revlimid), dexamethasone backbone.  

And Europe, they completed the phase three study of adding isatuximab, the other anti-CD38 monoclonal antibody to the BRd backbone. And what we’re finding what was very effective in the relapsed refractory setting was actually adding to the efficacy of newly diagnosed treatment regiments. As a side note, these trials – there are also trials looking at daratumumab and isatuximab in the smoldering myeloma phase, so moving it even earlier.  

I think one of the most attractive new targets in myeloma is targeting this antigen called B-cell maturing antigen, and a number of therapies are being developed or are already developed for it. The first approved was belantamab mafodotin, and this is an antibody drug conjugate. 

So, when the antibody binds to BCMA on the multiple myeloma cells, it releases its toxic payload into the myeloma cell. And so, it’s very effective towards myeloma, and no other good cells or fewer other good cells are affected by it. To provide some numbers, in patients with a median of seven prior lines of treatments, meaning their myeloma had relapsed that many times, the response rate was about 30 percent. And a fifth of those patients had VGPR, very good partial response, or better response.  

There are also bispecific antibodies that target this myeloma marker, and we anticipate getting one approved soon in the U.S. called teclistamab. Teclistamab is an antibody that binds both CD3 on T cells of the immune system and B-cell maturating BCMA on the myeloma cells. 

So, the way this antibody kills myeloma is by activating the T cells, the immune system, and directly killing the tumor. So, this was recently published in the New England Journal of Medicine. And in people who were treated with at least five prior lines of therapy, the response rate was about 63 percent, and the median progression-free survival, or the time until the myeloma progressed, was about 11 months.  

We were very active in a clinical trial looking at the effectiveness of another antibody, a bispecific antibody, called Regeneron 5458. In a similar patient population, the response rates were 75 percent in the higher-dose level group, and right now it’s actually a bit too early to tell how long the progression free survival is or the duration of response. 

There are also other bispecifics in development targeting other myeloma markers ssuch as talquetamab, that binds to a marker called GPRC5D, and cevostamab, which binds to a marker called FcRH5. The response rates as single agents in patients with relapsed refractory multiple myeloma are 66 percent and 45 percent respectively. These are all incredible numbers for a single drug in the relapsed refractory setting.  

How Does Disease Staging Affect Myeloma Treatment Choices?

How Does Disease Staging Affect Myeloma Treatment Choices? from Patient Empowerment Network on Vimeo.

What are the stages of myeloma, and how does this affect care? Dr. Abdullah Khan, a myeloma specialist, reviews how myeloma is staged, which genetic markers may affect risk, and the impact of staging on treatment decisions.

Dr. Abdullah Khan is a hematologist specializing in multiple myeloma and plasma cell disorders at the Ohio State University Comprehensive Cancer Center – The James. Dr. Khan is also an assistant professor in the Division of Hematology at The Ohio State University. Learn more about Dr. Khan.

See More from Engaging in Myeloma Treatment Decisions

 

Related Resources:

 
Expert Advice for Newly Diagnosed Myeloma Patients

Myeloma Induction and Consolidation Therapy Defined

Relapsed and Refractory Myeloma Defined

Transcript:

Katherine:

How does staging affect treatment option?  

Dr. Khan:

Staging is done by two methods. The older method is the international staging system, abbreviated as ISS. And then there’s the newer revised ISS, or RISS. 

The patients are assigned stages one to three. To determine the ISS you need lab values for the beta-2 microglobulin and albumin. For the revised ISS, you add on the lab value for LDH, lactate dehydrogenase, and you also add in the chromosome risk profile. So, there are certain genetic changes that predict a more aggressive myeloma. And the ones added to the revised ISS staging system are translocation 4;14, translocation 14;16, and deletion 17p.  

So, that’s the ISS stage and the revised ISS stage. There are also other factors patient providers look into when determining the risk profile for patients. So, that might include other genetic changes. 

One that is gaining a bit more traction right now is something called gain 1q, or amplification 1q, so more than one copy of part a chromosome. Some patients might have myeloma that doesn’t start, and the bone marrow might be found outside of the bones. And that’s called extramedullary disease, and sometimes that’s kind of high-risk. And some people have so much bone marrow plasma cells that it actually spills into their bloodstream. So, they might have high circulating plasma cells. Anyway, this will give information on staging.  

And in terms of how it affects treatment option, I’ll give maybe two examples. Let’s say in case one we have a 40-year-old patient high-risk multiple myeloma. The high risk portends a poorer prognosis, meaning the outcomes might not be as good as someone with a standard myeloma. So, in that case, I might try to do or use the most aggressive treatment option in order to maximize treatment responses because I know the overall outcome is poor. 

I do all this while acknowledging maybe the chances of having side effects might be higher, but that might be an acceptable tradeoff.  

In case two, I’ll flip to an 80-year-old with standard risk cytogenetics.  

So, I predict their myeloma to behave standard. In this case, I might try to use a regimen with a more acceptable safety profile, because the predicted response to treatment is anyways very good. So, I don’t want to hurt them in the process of getting their myeloma in remission.  

I’ll also say this. My practice pattern at The Ohio State University might be a little different than someone on the East Coast or West Coast, and that’s okay. We all have our experiences with the different treatment regimens, but we all have the same goal of being as aggressive as we can while being mindful of side effects. 

How Is CAR T-Cell Therapy Changing Myeloma Care?

How Is CAR T-Cell Therapy Changing Myeloma Care? from Patient Empowerment Network on Vimeo.

Dr. Abdullah Khan discusses how CAR T-cell therapy works to treat myeloma, the currently approved CAR T-cell therapies, and the outcomes related to progression free survival (PFS) for patients with heavily pre-treated myeloma.

Dr. Abdullah Khan is a hematologist specializing in multiple myeloma and plasma cell disorders at the Ohio State University Comprehensive Cancer Center – The James. Dr. Khan is also an assistant professor in the Division of Hematology at The Ohio State University. Learn more about Dr. Khan.

See More from Innovative Myeloma Therapies

Related Resources:

Immunotherapy: Which Myeloma Patients Is It Right For?

Immunotherapy: Which Patients Is It Right For?

How Is CAR T-Cell Therapy Changing the Myeloma Treatment Landscape?

How Is CAR T-Cell Therapy Changing the Myeloma Treatment Landscape?

What Are the Risks of CAR T-Cell Therapy?

Transcript:

Katherine:

Let’s talk about CAR T-cell therapy. How is CAR T-cell therapy changing the field?  

Dr. Khan:

Myeloma was a little late to the CAR-T game, but we’re very happy it’s here. The two products approved in myeloma are idecabtagene vicleucel, ide-cel for short, and ciltacabtagene autoleucel, or cilta-cel for short. 

So, the way CAR Ts work, they are customized T cells for each individual patient. You collect the T cell from the patient with myeloma. You reengineer them in the laboratory to produce proteins on their surface called chimeric antigen receptor. That’s CAR portion of the CAR T therapy. And these CARs recognize and bind specific proteins on the surface of multiple myeloma.  

So, these genetically modified T cells are then expanded or multiplied to make millions of cells. They’re sent back to the hospital where they were collected, where the patient is. And they’re infused back into the patient. The hope is that these modified cells, these CAR T cells, will continue to multiply in the patient. And with guidance from that engineered receptor, they will recognize and kill multiple myeloma very effectively. 

So, I can provide some numbers to the outcomes of the two approved CAR T cells – CAR T products in multiple myeloma. The first approved was ide-cel in patients with a median of six prior lines of therapy, a single dose of CAR T was able to produce an objective response rate – that’s how many people responded to the treatment – of 73 percent, and the median, the middle person, progressed after 8.8 months of getting this treatment. The other product, cilta-cel, was also studied in patients with a median of six prior lines of therapy, and the objective response rate was an astounding 98 percent.  

Katherine:

Wow.  

Dr. Khan:

And the median progression-free survival is actually not yet reached. So, these are remarkable results with heavily pre-treated myeloma. And the myeloma community’s very excited to actually bring these treatments to earlier lines of therapy such as a newly diagnosed patient with multiple myeloma. 

Advances in Myeloma Molecular Testing

Advances in Myeloma Molecular Testing from Patient Empowerment Network on Vimeo.

What is molecular testing, and how does it impact myeloma care? Dr. Abdullah Khan from the Ohio State University Comprehensive Cancer Center – The James discusses the specific markers found in cytogenetic analysis that determine a patient’s risk and may impact treatment choices.

Dr. Abdullah Khan is a hematologist specializing in multiple myeloma and plasma cell disorders at the Ohio State University Comprehensive Cancer Center – The James. Dr. Khan is also an assistant professor in the Division of Hematology at The Ohio State University. Learn more about Dr. Khan.

See More From INSIST! Myeloma

Related Programs:

What Tests Are Essential Before Choosing a Myeloma Treatment Approach

What Tests Are Essential Before Choosing a Myeloma Treatment Approach?

Understanding MRD and What It Means for Myeloma Patients

Understanding MRD and What It Means for Myeloma Patients

How Do Test Results Impact Myeloma Treatment Options?

How Do Test Results Impact Myeloma Treatment Options?


Transcript:

Katherine:

Have there been advances in molecular testing for myeloma patients?  

Dr. Khan:

Molecular testing is a broad term and can indicate checking genes, proteins, and other molecules. Even let’s say speaking outside of the world of myeloma, molecular testing can be used to determine in individuals if they have a high chance of developing cancers or other diseases.  

It can be done to confirm so cancer diagnoses using the tissue biopsy specimens. It can also be used to help plan treatment, find out how well the treatment is working, provide prognosis information, and other information. In the world of myeloma, there are – in the world of myeloma, there are researchers looking at all of these molecular changes that can happen with disease.  

Katherine:

So, how do the results of these tests affect treatment?  

Dr. Khan:

There’s a particular cytogenetic change called translocation 11;14 that’s found in maybe a quarter of all patients with newly diagnosed myeloma, and it predicts a high likelihood of responding to a new drug called venetoclax.  

In the clinical trial of venetoclax, when it was given to all patients with multiple myeloma, there was actually higher mortality in patients when given venetoclax in combination with bortezomib and dexamethasone. And this is despite a higher response rate by adding the venetoclax.  

The thought process was maybe those patients were not doing well because of higher chances of serious infections. But when they took the data and they looked at that subgroup of patients with the translocation 11;14, there was no such concern in that subgroup. So, in this case of having translocation 11;14, it actually giving you a new treatment option based on the findings of the molecular testing. 

We participate in a national clinical trial called MyDRUG, and that’s looking at other molecular changes to see if a more targeted treatment when added to the backbone of myeloma therapies translates to better outcomes.  

Another recent development in molecular testing is diagnostic testing for minimal residual disease, and that’s from the bone marrow in patients with multiple myeloma.  

The most commonly used test in clinical trials is the clonoSEQ test; it’s an FDA-cleared diagnostic test. The way it works it looks for specific DNA sequences on the receptors of the cancer cells. So, each cancer cell has like a genetic barcode.  

Using the liquid part of the marrow, we can look for those cells that harbor that genetic barcode. 

And the test is so sensitive, we can find one in a million cells in a patient’s bone marrow aspirate. So, it’s a very sensitive test, but it is not yet approved for making treatment decisions. One way we can use it though is for prognostic information. So, a patient attaining minimal residual disease-negative status or MRD-negativity, probably will do better than someone who has MRD-positive disease.  

And there’s an emerging concept called sustained MRD negativity. So, let’s take an example of someone getting MRD testing at one year and two years after their stem cell transplant. The patient who is MRD-negative at both the one-year and two-year marks will likely do better than the one who is MRD-negative at one year but turns positive at the two-year mark. 

So, these are some of the new developments in molecular testing in multiple myeloma.