Tag Archive for: myeloma

Advice for Setting Myeloma Treatment Goals and Collaborating on Care Decisions

Advice for Setting Myeloma Treatment Goals and Collaborating on Care Decisions from Patient Empowerment Network on Vimeo.

This animated video defines common myeloma treatment goals, provides an overview of available myeloma therapies, and includes advice for making care decisions and the importance of being your own advocate.

See More from Collaborate Myeloma

Related Resources:

Collaborate | Being an Empowered Myeloma Patient

Understanding Myeloma | How You Can Collaborate in Your Care

Understanding Myeloma | How You Can Collaborate in Your Care

The Benefits of Shared Decision-Making for Myeloma Care


Transcript:

Bianca: 

Hi! I’m Bianca, a nurse specializing in myeloma. In this video, we’ll discuss myeloma therapy and explain how to work with your healthcare team so that you can choose a care plan best suited for YOUR myeloma. 

I’d also like to introduce you to Suzanne. Suzanne is a patient advocate living with myeloma.  

Suzanne: 

Thanks, Bianca. I’m happy to share my own experience and to talk about how I worked with my healthcare team to decide on a care plan. 

When my doctor and I were first considering my options, we started by setting treatment goals. Bianca, can you define treatment goals?  

Bianca: 

Sure! Each patient is unique, so it may vary by person. You should collaborate WITH your healthcare team to determine YOUR treatment goals. Common goals of myeloma treatment may include: 

  • Reducing and managing your symptoms. 
  • Slowing the progression of the disease. 
  • Inducing remission. 
  • And, helping you live longer while maintaining quality of life.

Suzanne: 

Thanks for explaining this. When I discussed treatment goals with my doctor, I pointed out that I still wanted to be able to play pickleball with my friends, and to care for my young grandchildren. We reviewed options that could allow me to stay as active as possible and manage my myeloma at the same time. 

Bianca: 

That’s a great point, Suzanne. It’s also important to remember that your treatment goals can change throughout the course of your myeloma. Discuss them regularly, not just with your care team but also your care partner – that can be a family member or even a close friend. 

Suzanne: 

That’s right. I discussed my care with my husband AND my adult children – it’s important to keep your support team in the loop.  

And it’s also essential to understand the treatment options available to you. Bianca, what are the types of therapy available to treat myeloma? 

Bianca: 

There are a number of different classes of therapy, which include: 

  • Proteasome inhibitors; 
  • Immunomodulatory therapies or IMiDs; 
  • Monoclonal antibodies; 
  • Stem cell transplants; 
  • There are also the new and recently approved therapies such as bispecific antibodies and CAR (Chimeric Antigen Receptor) T-cell therapy; 
  • And, of course, clinical trials.   

Clinical trials can be a good option for patients at any stage of disease, often giving patients early access to a viable and cutting-edge therapy. When considering treatment, you should ask your doctor if there is a clinical trial that may be right for you.  

Suzanne: 

Good point! When I was deciding on a therapy, my doctor and I, along with my husband, discussed the risks and benefits of each approach as well as the potential outcome of each option. My doctor also walked through the clinical trials that were available to me. 

Bianca, given all of the options, how do you decide which therapy is appropriate for a particular patient? 

Bianca: 

That’s a great question. As mentioned in our previous video, results of in-depth testing, which determine if a patient has low-risk or high-risk myeloma, can affect your choices. Other factors that impact treatment decisions may include: 

  • Your age, overall health, and any pre-existing conditions. 
  • Potential side effects of the treatment. 
  • Previous therapies that may have been used to treat your myeloma. 
  • The financial impact of a treatment plan. 
  • And the patient’s lifestyle and preference.  

Suzanne: 

That’s right. And, you shouldn’t hesitate to weigh in on what YOUR preference is. Do your own research so you understand each treatment approach, then work WITH your healthcare team to determine what might be best for you.  

Bianca: 

That’s excellent advice, Suzanne. So, when making treatment decisions, you should: 

  • Work with your healthcare team to understand your treatment goals. 
  • Talk with your doctor about all available treatment options. 
  • And, inquire about any clinical trials that may be right for you. 
  • Then, discuss the pros and cons of each option. 
  • Bring a care partner with you to appointments and take time post-visit to discuss the proposed plan and treatment options. 
  • And, always remember that you have a voice in your care. Speak up and ask questions. You are your own best advocate. 

Suzanne: 

Great advice, Bianca. Don’t forget to visit powerfulpatients.org/myeloma to learn more. Thank you for joining us!  

Miguel’s Journey: Embracing CAR T-cell Therapy as a Latinx Myeloma Survivor



Miguel’s Journey: Embracing CAR T-cell Therapy as a Latinx Myeloma Survivor from Patient Empowerment Network on Vimeo.

Myeloma survivor Miguel wasn’t experiencing any symptoms when he received his shocking diagnosis. Watch as he shares his experience as a Latinx myeloma patient dealing with testing, multiple lines of treatment, and CAR T-cell therapy – and how to stay [ACT]IVATED in your care.

See More from [ACT]IVATED CAR T

Related Resources:

What Is the Impact of CAR T Therapy Access Barriers on Patients?

What Is the Impact of CAR T-Cell Therapy Access Barriers on Patients?

What Are CAR T Therapy Requirements for Care Partners?

What Are CAR T-Cell Therapy Requirements for Care Partners?

CAR T-Cell Therapy Follow-Up Monitoring | What Patients Can Expect

CAR T-Cell Therapy Follow-Up Monitoring | What Patients Can Expect

Transcript:

Being ACTIVATED in CAR T-cell therapy care is critical for patients and families. My name is Miguel and I want to share my story as a myeloma survivor and Latinx man. Even though CAR T-cell therapy has improved survival rates for myeloma patients, some disparities to treatment access still persist. 

I was 52 when I was diagnosed with smoldering myeloma, and my diagnosis came as a complete shock. I wasn’t experiencing any symptoms, and my doctor only discovered my condition after noticing that something looked abnormal in my blood work. After ordering further testing, my diagnosis was confirmed with a bone marrow biopsy. That was just the start of my long journey. My hematologist informed me that several rounds of chemotherapy would be best for my first line of treatment.

After I finished my rounds of chemo, my hematologist continued to monitor my tests closely for signs of recurrence. When my tests reached concerning levels, my doctor then recommended that I move forward with an autologous stem cell transplant – taken from my own stem cells.

Those were just my first two lines of therapy. My third line of therapy was an immunotherapy as part of combination therapy that worked for nearly two years. An allogeneic stem cell transplant – with stem cells taken from a donor – was recommended next. That second stem cell transplant kept my myeloma at bay for about two years. It was a nice break, and I was able to qualify for a CAR T-cell therapy when it came time to act on my fifth line of treatment. I had learned from my myeloma support group that patients need to have a lot of support to qualify for CAR T. 

Patients need to have a care partner to support them, and I was fortunate enough to have my sister stay with me to help me with my appointments and recovery. CAR T-cell therapy has made the future brighter for so many myeloma patients.

There have been a lot of recent advancements in CAR T-cell therapy for myeloma. I hope that sharing my story will make a difference for other myeloma patients who may have some mistrust of doctors. Remember, become empowered and stay [ACT]IVATED with these tips. 

[ACT]IVATION tips for CAR T patients: 

  • Ask your care team questions to learn about the status of your myeloma, treatment options, and what to expect during and after treatment.
  • Inquire if a clinical trial may be a potential treatment option for your myeloma.
  • Join a patient support group to offer and receive emotional support.
  • Stay updated about myeloma treatment options and research advancements. 

Being proactive is an essential step in your myeloma journey. Stay [ACT]IVATED by being informed, empowered, and engaged in your care.


Share Your Feedback

Create your own user feedback survey

[ACT]IVATED Myeloma Resource Guide for Veterans and First Responders

Download Resource Guide

ACTIVATED Vets and First Responders Resource Guide_KrinaPatel

Download Resource Guide

See More from [ACT]IVATED Multiple Myeloma


Share Your Feedback

Create your own user feedback survey

[ACT]IVATED CAR T-Cell Therapy Toolkit Checklist

See More From [ACT]IVATED CAR T

 

[ACT]IVATED CAR T-Cell Therapy North American Specialist Treatment Centers

See More From [ACT]IVATED CAR T

 

[ACT]IVATED CAR T-Cell Therapy Patient Plan

Thank you for taking this assessment. By answering the questions below, a custom patient plan featuring a collection of vetted resources will be emailed to you within 5 minutes. If you don’t see it, please be sure to check your spam. Stay [ACT]IVATED. 
 

 

See More From [ACT]IVATED CAR T

 

What Are Myeloma Risk Factors for Veterans and First Responders?

What Are Myeloma Risk Factors for Veterans and First Responders? from Patient Empowerment Network on Vimeo.

Veterans and first responders may come into contact with myeloma risk factors, but what are they? Expert Dr. Krina Patel from The University of Texas MD Anderson Cancer Center discusses environmental risk factors for 9/11 first responders and veterans, average age of onset for different veteran racial groups, and proactive patient advice.

[ACT]IVATION TIP

“…if you notice that anything is off, you’re not feeling well, or the lab, something is wrong with your labs, make sure to mention that to your physicians, they can first diagnose the correct thing, if something is going on. And two, if you actually have a diagnosis of cancer, I think talking to your teams that there are probably resources out there to help with a couple of things, I think one, even financial resources.”

See More from [ACT]IVATED Multiple Myeloma

Download Resource Guide

Related Resources

Key Advice for Myeloma Patients | Questions to Ask About a Care Plan

Understanding Myeloma | How You Can Collaborate in Your Care

Understanding Myeloma | How You Can Collaborate in Your Care 

Roadblocks for Black and Latinx Patients From CAR T Trial Access

Roadblocks for Black and Latinx Patients From CAR T Trial Access 

Transcript:

Lisa Hatfield:

Dr. Patel, a lot of patients wonder what might have caused my myeloma, and there are some environmental factors that are known to have the association with myeloma, like Agent Orange exposure, and now we’re seeing an increased instance of myeloma and other blood cancers in the 9/11 first responders. For patients who might have concerns about this, do you have any suggestions or thoughts on that?

Dr. Krina Patel:

Yeah, I’ve actually quite a few patients that come up to me that were in previous wars or veterans, and the first thing I talk about is the 9/11 first responders. So people ask me if this they’re born with myeloma, that’s the number one question, and I say, you know, likely not, most people do not have genes that they were given by their parents, that causes myeloma.

All of us have different susceptibility to cancer based on how our immune system repairs itself, how our plasma cells repair themselves, the micro-environment, but it’s also our exposures, and we know that there’s certain exposures like Agent Orange, as well as those first responders that went in here in terms of epidemiology, in terms of the number of patients that ended up with myeloma at a younger age, a much younger age. They’re in their 50s, for the most part, that tells us that this was not something that those folks are going to get. This really was based on that exposure, and that’s the hard part of saying that something causes something.

I think we know with 9/11, the numbers were so high that this was a…listen, this is something wrong, that whatever they were exposed to during that process led to their plasma cells becoming myeloma at a much younger age, and it seems that a lot of them had more aggressive disease than the indolent slow-growing myeloma.

We see that a lot of patients get. The other big question I get is, How can we say that something caused this, and then again, it comes back to how many people are exposed and then how many people actually got that disease, and that’s why it becomes so hard. But I know a lot of my patients think about Roundup or different petrochemicals and things like that that they’ve been exposed to, and I know that the government and folks are looking into it because a lot of my patients are getting letters from us and things like that just to say, “Listen, I was exposed to this, could this have caused my myeloma?”

And again, the majority of patients are not exposed to things at that level that really tell us that that’s what caused their myeloma, but I do think that if you are exposed to something like Agent Orange or major petrochemical spill or something that is worthwhile noting at least, even though I most likely won’t be able to tell you it definitely caused the myeloma. We do know that there are environmental exposures that are more likely to lead to cancer, you know, we have these hot spots in the U.S. where especially those petrochemical companies are, where there’s a much higher level of just cancer diagnosis, not just myeloma up to cancer in general, compared to other areas where we don’t have those industrial companies existing.

Lisa Hatfield:

Do you happen to have any tips for patients who maybe were a part of 9/11 event or even veterans or first responders of any type, any tips for them in general?

Dr. Krina Patel:

Yeah, I think that the activations have here is that if you notice that anything is off, you’re not feeling well, or the lab, something is wrong with your labs, make sure to mention that to your physicians, they can first diagnose the correct thing, if something is going on. And two, if you actually have a diagnosis of cancer, I think talking to your teams that there are probably resources out there to help with a couple of things, I think one, even financial resources.

When stuff like this happens, usually there are some financial resources that pop up, and two, the mental aspect of this. You got this while you’re doing something you’re supposed to be doing and helping others, and really finding patient groups which are out there as well, so that you get the resources for just the ability to talk to someone about what happened and being able to go through that process as well.

Lisa Hatfield:

Dr. Patel, are there any notable trends or patterns of the presentation and progression of myeloma and other blood cancers in veterans and first responders that differ from civilians?

Dr. Krina Patel:

Yeah, that’s a great question. I honestly don’t know if there’s been anything published that shows a difference, I tend to see my veterans are a little bit younger in general, average age for myeloma, 70 for Caucasian patients, it’s 65 for Hispanic patients and 66 for African American patients, right? So a lot of my veteran patients have been in their 50s, just a little bit younger than what I’ve seen with most other patients, and then in terms of patterns, not necessarily.

We think, oh, is it more aggressive? Is it not. I do have patients with aggressive disease, but I have patients that come in with MGUS or smoldering disease that eventually turns into myeloma isn’t necessarily high risk or aggressive, but again, I don’t know any data that’s out there that’s published, I think that would be worthwhile. But I will say my patients tend to be on the younger age.


Share Your Feedback

Create your own user feedback survey

PODCAST | Myeloma Patient Expert Q&A: Dr. Ola Landgren

 

START HERE bridges the gap between expert and patient voices, empowering myeloma patients to feel comfortable asking precise questions of their healthcare team.

In this webinar, Dr. Ola Landgren delves into the emerging and exciting therapies and clinical trials for myeloma, discusses the latest options for relapsed disease, and explores the current landscape of managing and monitoring multiple myeloma. Watch as Dr. Landgren answers patient-submitted questions and discusses another hot topic: the utilization of artificial intelligence in multiple myeloma.

Download Guide | Descargar Guía

See More from START HERE Myeloma

Transcript:

Lisa Hatfield:

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

Today I am honored and really excited to be joined by Dr. Ola Landgren. Dr. Landgren is chief in the Division of Myeloma and the Department of Medicine, and also serves as director of the Sylvester Myeloma Institute at the University of Miami’s Miller School of Medicine. Dr. Landgren, it’s such a pleasure having you today.

Dr. Ola Landgren:

Thank you very much for having me. It’s really a great pleasure to be here today.

Lisa Hatfield:

So in this program, first, we’ll get a high level update from Dr. Landgren on what the latest myeloma news means for you and your family. And then we will launch into some questions that we’ve received from you. Dr. Landgren. We’re at a pivotal moment in the history of multiple myeloma. We’re experiencing an unprecedented wave of progress marked by significant increase in new treatment options and ongoing research. We are very honored to have your expertise to guide us in understanding these advancements and providing clarity around all the evolving landscape of myeloma care.

So before we get started, to you at home, would you please remember to download the program resource guide via the QR code. This is where you’ll find useful information to follow before the program and after. So we are ready to START HERE. Dr. Landgren, can you speak to the emerging and exciting myeloma therapies and trials right now?

Dr. Ola Landgren:

I’ll do my best. There are so many things to talk about, and I don’t think we have 10 hours, so I will have to shorten it. But I would say that the past 12 to 18 months, we have had three new drugs approved in the field of myeloma. These are the bispecific antibodies. The first out of those three was the BCMA-CD3 targeted drug teclistamab-cqyv (Tecvayli). And in the middle of 2023, we had both talquetamab-tgvs (Talvey), and elranatamab-bcmm (Elrexfio) approved. Talquetamab has another target is GPRC5D with CD3. And elranatamab is similar to teclistamab with the BCMA-CD3 targeted bispecific antibody. These are amazing drugs. They have been found in patients that have been heavily pretreated to result in about 60 percent or more percent of patients responding.

So overall response rates ranging from 60 percent to 80 percent in various trials. We have now these drugs approved, they’re still only approved as single drug and there are new trials going, combinations of two of these or these drugs with other drugs such as daratumumab (Darzalex) or IMiDs, such as lenalidomide (Revlimid) or pomalidomide (Pomalyst). So a lot of drug development is ongoing as we speak. We also have the CAR T cells that are reasonably new drugs. We, you think about everything new every week there’s a new drug, but they are very new CAR T cells.

We have had them for about three or so years, three-and-a-half years. And, the two drugs that are approved in that setting is, ide-cel (idecabtagene vicleucel) [Abecma]. That was the first and then cilta-cel (ciltacabtagene autoleucel) [Carvykti], that was the second. They both go after BCMA similar to the two antibodies I mentioned, teclistamab and elranatamab because they are CAR T cells, that indicates that they are cells.

They come from the same person who’s going to receive them back as treatment. So you collect the cells from the blood and you manufacture them into to CAR cells. So chimeric antigen receptor T cells, and then you give them back. There are several new CAR T-cells in development. There are other targets in development, GPRC5D, for example. There are additional other targets and there are also dual targeted cell therapies in development.

There are also allogeneic CAR T cells in development and that means that you could have a product off the shelf. So someone could donate cells, they could be manufactured into CAR T cells, and then you could give them to technically any person, so it doesn’t have to be the same person collecting and then manufacturing, giving them back. So that would shorten the time window for production.

And there are a lot of other details also that are important in this context. The whole manufacturing process that’s currently four to six weeks is being improved. There are some technologies that can make the CAR T cells in 48 hours, but the turnaround time is maybe one to two weeks with all the control steps, but that’s still a huge improvement. And then you have the antibody drug conjugate if you want.

So then you have the belantamab mafodotin (Blenrep). That actually was the first BCMA targeted therapy we had in myeloma. And then the drug was approved on an accelerated approval study. But when the randomized study was completed, it turned out that it was not better than the control arm. The company took it off the market. And now what’s happening is that there are two new trials, and one of them was just reported in the beginning of February of 2024.

The other one was around the ASH meeting in 2023. These two trials show that if you combine it with other drugs, the most recent one was with bortezomib-dexamethasone (Velcade-Decadron), that was superior with the belantamab mafodotin with bortezomib-dexamethasone versus daratumumab with bortezomib-dexamethasone. So I think we will probably see this drug coming back to the myeloma field. It is currently available as compassionate use, so physicians can prescribe it, but these trials will most likely, I would think, lead to FDA approvals with these combinations.

And lastly, I would say that other exciting trials, there are so many trials going on, but another thing that I think is interesting and exciting is also the use of antigens. And you can use mRNA and things like that. So these are like the vaccines. You can either, take a patients’ myeloma cells and look what they have on the surface, you can make more traditional vaccines or you can use more sophisticated newer technologies just like how the COVID vaccines were developed. And you can inject these sequences and then they will translate into spike proteins where the immune system could go after myeloma cells.

We don’t yet have a product like that in the myeloma field, but there are a lot of biotech and groups that are working to see. Moderna, was actually initially a cancer vaccine company and then COVID came and they turned into a COVID company, and now they’re be back again in the cancer field. So that’s a little bit of a summary of a lot of the exciting news that’s out there.

Lisa Hatfield:

Thank you. And do you have any comments about the sequencing of some of these? So with both CAR T and some of the bispecifics approved, obviously if a patient comes in and they need something right away, they’ll take whatever is first available. But all things being equal, if a patient says, well, I can, I have both CAR T accessible and bispecifics accessible. There are some patients out there, I’ve spoken with some who are wondering, is there a benefit to sequencing one before the other, or are there any trials looking into that?

Dr. Ola Landgren:

There are studies that have allowed patients to go on treatment with one of these modalities. For example, the bispecific antibodies with the prior exposure to a CAR T-cell therapy. There are also trials with CAR T-cell therapy that has allowed patients who have been exposed to prior antibodies, either bispecifics or the conjugated antibody drug conjugates, Belantamab mafodotin. So if you look at those studies and see how the numbers compare, if you are not exposed or you are exposed, I think the data is not entirely clear-cut.

There is no definitive study. Some data suggests that maybe it’s not that different, but then there are some studies that suggest that if you go to the antibody first that maybe that would lower the efficacy of the CAR T cell. So some people have for that reason said the CAR T cell should be done first. To make it even more complicated, there are some studies that have then taken time into the equation. So that means that you could have the patient treated with the antibodies for BCMA and CD3, and the antibody is given successfully for a long time, for many years. And eventually, unfortunately, the antibody may stop working.

Now, if you switch back to back to a CAR T-cell therapy without any other therapy in between, some studies indicate that that’s less likely to be beneficial. But if you instead do another target, say you did GPRC5D/CD3, or you did a completely different therapy with small molecules or you did carfilzomib (Kyprolis), or you did venetoclax (Venclexta), or IMiDs, or different types of combinations that are out there, been around for a long time, and you get good mileage out of those combinations.

Now, if that stops working, if you now go to this other therapy, you go back to the CAR T cell, that will suggest that the results are not that different. So I think that there are aspects that we don’t fully understand. I personally believe, based on what I’ve seen, based on what I know from treating thousands of patients with myeloma for almost 30 years I’ve been a doctor, I think time is probably very, very important. So if you go back to back from one therapy to the other, that’s less likely to be beneficial. If you go from one therapy, and it stops working and go to the other drug with the same target.

But I would say it’s not that different from how we think about IMiDs or proteasome inhibitors. If you were to go single drug with a proteasome inhibitor and you switch to single drug with another proteasome inhibitor, or the same thing with an IMiD, that’s less likely to work versus if you went to something else in between. So we just need to generate more data and learn. Lastly, I want to say that in my experience, from all I see in my clinic at the current time, I think the choice that patients make is based on personal preference and to some degree also the situation of the patient. I saw a patient yesterday, 50 years old, who came from another country and has relocated to us here in Miami and asked, what are the options?

And we talked about CAR T cells, we talked about bispecifics. And considering all the different factors that CAR T cell would imply that we had to give some other combination therapy for two or three cycles while we harvest the CAR T cells and manufacture the CAR T cells and then plan for the admission and give it, and also that the patient was not really very happy about the side effects in the hospital with CAR T cell. That patient shows the bispecific, but I’ve also seen other patients in the same situation saying, I’d rather do these different steps for two or three months, I stay in the hospital, and then I enjoy being off therapy.

Actually, I saw another patient just a few days ago, a gentleman in his upper 70s who we had the same conversation, and he had picked the CAR T cells. And I saw him with his wife and he has been off treatment for two years doing excellent. So different patients make different decisions. And I think that is just how the field is evolving. So I think we should be open to individual patient’s priorities and what they want, and we should just offer everything. And of course, we can guide if a patient wants us to give direction, but I think presenting it and let patients be part of the decision-making, that’s the future of how medicine should be practiced.

Lisa Hatfield:

Thank you so much for that explanation. I’m going to segue into a comment that I always make to myeloma patients. As Dr. Landgren was explaining all of these treatment options, he is on top of all the latest and greatest news and therapies. I always recommend to myeloma patients newly diagnosed or otherwise to seek out at least one consult from a specialist. If you have difficulty accessing care, then a lot of places can do video conferencing, but even that one consult to see a myeloma specialist is so important in your care and treatment options. So I’ll just throw that out there, Dr. Landgren, as a myeloma specialist that you are, we appreciate your expertise in explaining that so well.

Dr. Ola Landgren:

I agree 100 percent with what you said, and I would like to add to that and say, going to a specialist center and it doesn’t have to be here, can really really help. It can be a lot of small things. There is data indicating that survival is longer for patients who have access to specialists. That has been published in the Journal of Clinical Oncology. The Mayo Clinic has published that, I think it was more than one year longer survival.

That by itself is, of course, very strong, but I also think that there are a lot of the small things like the different types of pre-medications, the drugs that are given around myeloma drugs. Could you decrease the dose of some of these drugs like the dexamethasone? Could you get rid of Benadryl if you give the antibodies? These may look as small things, but they can make a huge difference for quality of life.

We have a lot of people coming for second opinions, and we always say if you live closer to someone that you trust, you should go back and be treated there. You can always reach out to us. We are happy to be involved. You have us as a backup. We can be your quarterback if you ever need us. I think that is absolutely the best advice for every patient. Go and get feedback and if you’re not sure about the feedback you get, you could always have two different quarterbacks and you could ask them. I don’t think having 10 or 20 is going to help, but having one or two second opinions, I think is a good decision.

Lisa Hatfield:

That’s really helpful information, thank you, Dr. Landgren. So I think we’re going to shift a little bit to managing and monitoring multiple myeloma. Once you’ve had a patient go through the induction therapy, what kind of monitoring do you complete for your myeloma patients and in particular those who have reached a certain level response and are maybe on maintenance or continuous therapy, what type of tests do you do and how often regarding labs, imaging, bone marrow biopsies?

Dr. Ola Landgren:

There are a lot of different ways, obviously, of practicing medicine. So every center has developed models that they feel very comfortable doing. So I like details. I like to know things. I like to check things. I’m not excessive in ordering invasive tests, but I like to know. Also, I like to make sure the patient not only has good long-term clinical outcomes, but also good quality of life. And to me, I try to minimize the intrusiveness of what we do. So, for example, if I give a combination therapy where there is an injection or infusion, say week one, week two, week three, and then there is a week off. I recognize that if you do labs during that week off, you will have a better yield and understanding of how these three different injections or infusions actually have moved the disease forward and suppressed the disease.

But in my mind, I think that week off is a very important week off for the patient. So I would rather do testing the third day of the treatment at the treatment unit. So if it’s week one, week two, week three, I would draw the myeloma labs that same day. And that would give the patient six more days off from injection, infusion that third week and the whole fourth week off. So I would give the patient 13 days off.

Again, these are small things. These are things I’ve thought about a lot. I’ve practiced medicine for many years and I recognize that having time off like that, many patients travel, they go on vacation, they do different things. So I don’t want to just randomly put a blood test in the fourth week just because I want to check after week one, two, three, and then have the assessment.

I sort of underestimate the benefit of the therapy and then I start the next cycle, say back to back cycle two and cycle three and so forth. I would typically do blood tests once a month following these principles. I do baseline and I would do the last day of injection or infusion. For a newly diagnosed patient, you ask me, I would for baseline always do bone marrow biopsy and an aspirate. I would always do a PET-CT for every patient as my default. Sometimes we end up doing MRI. So that could be other things that are happening, but that is what we do for the majority of our patients.

After we have completed four to six cycles of treatment for patients that are candidates for consideration of transplant with chemotherapy with melphalan (Alkeran), we would usually do a biopsy after four to six cycles and we would use that to determine what’s the optimal mobilization protocol for stem cells. When we do that, we would run a MRD test.

We would run our in-house flow cytometry test that we developed when I used to work at Sloan Kettering and we have developed that here in Miami as well. We work closely with Sloan Kettering, and we have set up this assay in collaboration in the new 2.0 version. We will also send the aspirate for the clonoSEQ at Adaptive Biotech, which is the DNA-based sequencing for MRD. We would send the patient for collection of stem cells.

When the patient is back, we will continue treating. So if you say we do it after four cycles, we would collect, if we do it after five or six, then we collect. After that, we would typically resume therapy and for the majority of our patients, we actually give around eight cycles of therapy, and we have seen that you can deepen the response. You don’t increase the toxicity, but you deepen the response for the vast, vast majority of our patients. When we have used our best therapies, we have done it that way…

We have even published on this, over 70 percent of our patients are MRD negative, and many of those patients, when they come to cycle eight, they ask, do I have to do the transplant? And that is a controversial topic. But I think there are two large randomized trials that have shown the same thing, that there is no survival benefit with transplant. But you can also say that there is, in those two trials, a progression free survival benefit, meaning that the disease would stay way longer with transplant.

But many patients say, if I reach MRD-negative, both those two trials show that if you’re MRD-negative without transplant, or you’re MRD-negative with the transplant, PFS was actually the same. And given that there is no survival, overall survival benefit, why would I subject myself to go to that? Why don’t I keep the cells in the freezer and go right to maintenance? And we will have a conversation with every patient, they would meet our transplant team, they would meet our myeloma expert team.

And the individual patient will make decisions. I think over time, more and more patients have chosen to keep the cells in the freezer. For patients that are MRD-positive, we would counsel towards transplant, but there are patients that don’t want to do that, and we are not forcing any patients to do that. We would give patient maintenance, and on some of our trials, we use the standard of care, which is lenalidomide maintenance.

And we are also developing new approaches where we have done daratumumab added once a month with lenalidomide. We have gone one year, and we have started to do two years of that. And after that, we would stop daratumumab and just do lenalidomide maintenance. Lastly, to answer your question fully here, we would do a PET-CT in the bone marrow after the eight cycles as a repeat, and we would offer a patient to check on maintenance on an annual basis, and this is in accord with the NCCN guidelines. So a lot of details here, but you asked me how we do testing.

Lisa Hatfield:

Yes. And one of the questions that comes up, too, regarding bone marrow biopsy, so you talked about patients kind of through the process of myeloma treatment, perhaps they’ve reached a point where they’re going to be for a while. Do you see a need for continued bone marrow biopsy, say, annually, or is there some benefit to using the newer tests that are being investigated, like mass spec testing and some of the newer ones, I think the EuroFlow? Do you think that that can be used to test for bone marrow biopsy? And how will that be used to monitor the myeloma if a patient is doing relatively well, or do you still like to do bone marrow biopsies on a regular basis? And I know every specialist is different in how they’ll answer that question.

Dr. Ola Landgren:

So what’s known in the literature is that there is no study that definitively has compared annual biopsies with these blood-based tests that you mentioned, showing that they can replace the bone marrow. Those tests or those studies have not yet been published and shown in a convincing way that we have done. This is how it is. It’s still an open question. We don’t know the answer for sure. So our take has been to offer patients to repeat it on an annual basis for maybe two or three and sometimes up to five years. I don’t think we would do biopsies every year for five, 10, 15 or more years. At some point, you have to ask yourself, what are we trying to chase here?

But I think the data we’re looking at that we have published on this and others have also show that if you are MRD-negative after completion of the eight cycles with or without the transplant, the patient that are MRD-negative one year later, they are more likely to be free from progression 10 years later, compared to the ones where you only check once and you don’t know what happened one year later. And that is frankly because there is a small group of patients where MRD-negative could bounce back into positive.

So to check after completion after eight cycles and to check after one more year on maintenance, I think gives us more confidence in thinking about if we eventually could step down and maybe even stop the maintenance at the long term. There is no study that definitively has proven that, but the data suggests that being negative after eight and do another year and even if you do two years out, those are very strong indicators that the disease will stay away long term.

So that’s our justification for offering it, but we would never force any patient. And I also want to say that we have thought about for a long time, how we can contribute to the field and how we can advance the field for blood-based tests. So we are here in Miami, developing a lot of these technologies, and I have made a promise that we will make all these available for all patients that come here to Miami as part of our standard workup. Because they are not clinically validated tests, they will have to be reported for now as research tests, but we will share the information with individual patients.

So we have three different platforms for now. And we are working on the fourth one. So one of them is the mass spec with MALDI, where we can screen the blood with lasers. And we can increase the sensitivity by maybe hundred times compared to existing immunofixation assays. The second is something called clonotypic peptides, which is a more sophisticated way to run mass spec, which is probably up to thousand times more sensitive than immunofixation. And the third technology we are doing or setting up right now is circulating cells that we sequence.

And this is the Menarini technology that is approved for certain other solid tumors. I think for GI malignancies, it’s FDA cleared, but we are doing it in myeloma. We are also looking for free circulating DNA. We’re working with New York Genome Center to set those types of assays up. So my thinking is, if we can offer every patient that come here to do it, and many of those patients will do an annual biopsy, we actually will have the database that can answer the question you asked me, if it can replace. There is no other way that this can ever be answered.

But having a large database, we actually can compare on a patient level, how the bone marrow biopsy with flow cytometry and sequencing, how that behaves in relation to the blood base. How does it perform? Is any of these better? Can they replace each other? So I think if we do this for one or two years, we will have the answer to the question. That’s why I want to do it.

Lisa Hatifeld:

So that kind of leads to the next question that is really an exciting area. I know it’s not necessarily new, but newer is artificial intelligence.  And I know I was reading an article about one of, that you and your colleagues have worked on a newer project and I don’t know if you pronounce it IRMMa or not, but using these large databases to help predict I think, it’s the response of treatment in some patients. So can you talk about that a little bit and tell us about that development and what developments are exciting with artificial intelligence in cancer, in particular myeloma?

Dr. Ola Landgren:

Yeah. So you mentioned the study we just published. We published a model that we call IRMMa and that stands for individual risk prediction for patients with multiple myeloma. So what we were thinking was at the current time, all the existing models are pretty much providing the average patient’s predicted outcome. So think about it is like it’s a probability measure. So you say, if I take this about therapy, what’s the predicted average outcome for patients that take this therapy, say, five years later? So on average, say 70 percent of patients are free from progression. That sounds pretty good. The problem is that you don’t know if you are in the group, 70 percent group that didn’t progress or if you’re in the 30 percent that did progress.

So where are you as an individual? So it’s almost like looking at the weather app on your phone. If it says it’s a 70 percent probability of sunshine and then you go outside and it’s raining, it’s because it didn’t say that it’s 100 percent probability of sunshine. So if you think about another situation would be, say, in a GYN clinic, if a woman were to come and ask the doctor, am I pregnant? Yes or no? You couldn’t say it’s 70 percent probability. You would say, yes, you’re pregnant or not pregnant.

So for myeloma, we have for a long time been living in these weather report systems where we say 70 percent or 30 percent. And we want to go in the other direction of the pregnancy test, where we actually can say for someone with this particular disease profile, with this treatment, this is where this is going to take us. We worked on this project for almost four years and we worked with a lot of other groups around the world that have a lot of data. And they have graciously agreed to collaborate with us and share their data sets. The beauty with this collaboration, there are many beauties of it, but one of them is that people don’t treat patients the same way.

And that actually has allowed us to say for patients that have a particular biological or genomic makeup, if you’re treated this way or that way or the other way or a fourth way and so forth, which of these different treatments would make patients have the longest progression and overall survival? So if you have a large database, you can actually ask those questions. So you can say that you profile individual patients in full detail and you put them in detailed buckets instead of grouping everybody together.

And now if you add a new case, if a new patient is being added and you say, which bucket would this individual fit? Well, this is the right biological bucket. You can then use this database to say out of all the different treatment options, which treatment option would last the longest, which would give the best overall survival? Other questions you could ask is also, for example, you have a patient with a certain biological workup or makeup. And you say, if I treat with these drugs, will the addition of, say, transplant, will that prolong progression for his survival?

And you can go into the database and the computer will then say, I have these many patients that have this genomic makeup and these many people that were treated with this treatment with transplant versus the same treatment without transplant. There was no difference in their progression or overall survival. So then the computer would say, it doesn’t add any clinical benefit, but there could be another makeup where the answer is opposite, but transplant actually would provide longer progression for his survival. I think the whole field of medicine is probably going to go more and more in this direction. So what we want to do is to expand the number of cases.

So we are asking other groups around the world, if they have data sets with thousands of patients, they could be added to this database and we could then have more and more detailed information on sub types of disease and more and more treatment. So it will be better as we train it with larger data sets. The model is built as an open interface so we can import new data. And that’s also important because the treatments will continue to change. So we, for example, say I have a patient that has this genetic makeup. I was thinking of using a bispecific antibody for the newly diagnosed setting.

How is that going to work? The computer will say, I don’t know, because we don’t have any patients like that in the database because that’s not the data, type of data that currently exists from larger studies. But let’s say in the future, if there were datasets like that, you could ask the computer and the computer will tell you what the database finds as the answer. But if you go for another combination, if that’s in the database, it would answer that too. That is where I think the field is going.

And lastly, I would say we are also using these types of technologies to evaluate the biopsies, the material. We work with the HealthTree Foundation on a large project where we are trying to use computational models to get out a lot of the biological data out of the biopsies and also to predict outcomes. So I think artificial intelligence is going to come in so many different areas in the myeloma field and probably in many, many other fields in medicine.

Lisa Hatfield:

Thank you so much, Dr. Landgren, for that broad overview of myeloma, especially relapsed and refractory myeloma. So it’s that time now where we answer questions we’ve received from you. Please remember that this is not a substitute for medical care. Always consult with your medical team. And we’re going to jump right into some questions that we’ve received from patients, Dr. Landgren, if you have a little bit of time to answer these questions for us.

Dr. Ola Landgren:

Of course.

Lisa Hatfield:

Okay. So broad questions. We try to make them broad so they apply to most people, but this patient is asking Dr. Landgren, what are the key biological processes driving disease, progression and evolution of multiple myeloma, and how can we target these processes to prevent disease relapse and improve long-term outcome?

Dr. Ola Landgren:

So that’s a very good question. So I think in a nutshell if you use genomics, which refers to the genetic changes that you can see in the plasma cells, there are certain features that the myeloma cells have. They have the copy number changes, that’s the gains and losses of chromosomes. You can find these if you do FISH and cytogenetics could be, for example, gain of chromosome 5 or gain of chromosome 7 or gain of chromosome 11. That would be part of the Hyperdiploidy disease, or you have loss of chromosome 13 or 13q deletions. We also refer to 17p deletion. These are copy number changes, they’re extra or loss of these chromosomes. But then you have also the structural variance where you have the translocations of chromosome 14, chromosome 14 harbors the IGH locus, which regulates the making of immunoglobulins.

Plasma cells make immunoglobulins. For reasons that are not entirely clear. The translocations in myeloma that include IgH, they are partnering up with oncogenes. There is a list of oncogenes, there’s MATH, there’s three MATHs, A, B, C. There’s FGFR3, MMSET, and there’s also Cyclin D1 that are on the list. So these are the different types of structural variants that you can see with FISH probes.

What people have understood less about are something called mutational signatures. And myeloma is made up by eight distinct mutational signatures that you can see in every single patient. And what that means is that you can, if you conduct whole genome sequencing and you look at all the base pairs, you can see there are certain number of combinations. C can be swapped for A and C can be swapped for G or C can be swapped for T, T can be A and T can be C and T can also be G.

Those are the combinations. So there are four different base pairs, but if you, because the DNA is double stranded, these are the only possibilities that mathematically that you can see. Now if you look for every base pair and you look on one base pair on the left and one on the right, we call that 5 and 3 prime, you look through triplicates, every of these base pairs can have these different swaps I mentioned. Mathematically, there are 96 different combinations that you can come up with. That’s it.

If you don’t go through the entire genome from left to right, you see that there are these recurrent eight signatures that are there in every patient. So although we don’t understand why they are and exactly how they function, the fact that you see them in every patient tells us that this has to have something to do with the biology of the disease. It must have a role in the control of the disease. We are starting to see that there is one signature that’s called APOBEC. That signature seems to be very important for resistance to treatments. And you can see that APOBEC can be more or less expressed.

And if APOBEC is very expressed, we see that there are lot of mutations in the cells. We have seen in patients with the chemotherapy that APOBEC can be very expressed. When we treat with four drug combinations, it can be very expressed. And what I’m saying, when I say it can be expressed, these are in the patients that relapse out of these therapies. We have also seen that in CAR T cells and bispecifics. So that makes me believe and our group believe that the cells use some form of what we call tumor intrinsic defense mechanism to protect themselves from whatever therapy we use.

It doesn’t matter if it’s immunotherapy, chemotherapy or small molecule therapy, there are some fundamental programs the cells can turn on. We need to understand that better and we are spending a lot of time trying to drill into this.

Lastly, I also want to say there was a fourth class of genomic events called complex events that you can see in myeloma, something called chromothripsis. That’s a very severe genomic lesion, is a ripple effect through the genome. There are a lot of havoc going on. And the first time we saw that, we thought this has to be something wrong with this sample. But when we look through more and more samples, we see that about a quarter of the patients actually have this chromothripsis.

So the bottom line is, it’s time to stop doing FISH, it’s time to do more advanced sequencing, ideally whole genome sequencing, but a step towards a whole genome could be to do whole exome sequencing. But there are companies saying that you can do whole genome sequencing for $1 in the future. So that’s really what needs to happen. We need to have better tools to better understand and then we can use this to better understand how to differentiate the therapy and have an individualized treatment. That’s what I talked about with the IRMA model.

Lisa Hatfield:

All right, well, thank you so much for that explanation. Dr. Landgren, can you speak to the advantages that bispecific antibodies offer over traditional therapies and how do you see their role in overcoming treatment resistance?

Dr. Ola Landgren:

Well, the bispecific antibodies is a novel way of engaging the immune system to go after the myeloma. So if you think about the other antibodies we have, we have three other antibodies. We have daratumumab, we have isatuximab (Sarclisa), we have elotuzumab (Empliciti), they are naked antibodies. They bind to the myeloma and on the backend of these antibodies, there is something called the FC receptor that attracts cells, NK cells, for example, also T cells, and they also attract, some of these antibodies also attract complement and they also by themselves send what’s called a death signal into the myeloma cell.

The bispecific antibodies are very different. They bind and they don’t send death signals, they don’t engage with the complement. What they do is that they have another arm sticking out that binds to the T cells. That’s a CD3 arm and there’s an open pocket. So when a T cell passes by, it grabs the T cell. And now you have a T cell linked to the antibody sitting next to the myeloma cell and the T cell will kill the myeloma. T cells can be very aggressive and kill the myeloma. You just hold them together, it’s like a matchmaker.

And if you think about how CAR T-cell therapy is designed, you take out the T cells, you manufacture them to have a special antenna receptor on their surface, and then you give them back again. And then they bind, this receptor binds to myeloma cells. So in the setting of a CAR T-cell therapy, the T-cell sits next to the myeloma cell, but that’s because the T cells were taken out of the body, manufactured to have this receptor that then finds the myeloma cell. But the bispecific antibody, that they don’t require the T cells to be taken out, to be modified this way.

You just use your existing T-cells in your body and these antibody just binds to the T cells and the myeloma cells in the body. So it’s sort of a little bit mimics what the CAR T cells do, but it does it in its own way within the cell, within the tissue in the body. You asked me for resistance mechanism and how they are better. Well, I think the best answer I can give you is to say that the overall response rate for the bispecific antibodies are very high. They are 60 to 80 percent single drug compared to the current trials. And if you look and see the trials that have led to approval for the other existing drugs, they were 20 or 30 percent.

So the overall response rate is much higher for the bispecifics than they were for the other existing drugs. We don’t really know exactly how to use them, I would say. What’s the optimal dosing schedule? We give them weekly, it may be every other week, and maybe monthly, eventually, I would think. And should they be combined with which drugs? That’s ongoing investigation. Other questions are, can they be stopped? Can you monitor patients off therapy for a long time? Will some patients never have the disease coming back? We hope so, but we don’t know. Or would it be patients could be off therapy for a long time, like with CAR T cell? Could that happen with the bispecifics? It’s possible.

And if you were to monitor with blood-based tests and you see that there is reappearing disease, would you then put patients back on the therapy? These are questions we…there are a lot of questions, we don’t have answers to all these, but that’s where I think the field is going. A lot of people, including us, are trying to investigate this.

Lisa Hatfield:

Okay, thank you. And we have a number of questions about MRD testing, so I’m going to try to combine those all together. Basically, what the questions are asking is how do you interpret MRD testing with regard to prognosis, treatment response, and maybe even like treatment, ongoing treatment? How do you use those results in your clinic or any comments you might have on the MRD test?

Dr. Ola Landgren:

So MRD tests have been around for quite some time. We have been pioneers pushing it. We have worked on it for over 15 years. We worked with the FDA to see if MRD could eventually become an endpoint for drug approval, that’s work in progress. The FDA will make those decisions. There are a lot of trials that use MRD as a secondary endpoint to see how it correlates with progression-free survival. And there actually are some trials that have been using it as a cool primary or primary endpoint in the absence of FDA’s decision to accept it. But that is probably going to change in the future. We will see.  What have we done in the clinic? Well, we have used it in the same way as we have done with PET-CTs and the regular blood work. So if you use SPEP IFE light chains and you see there is residual disease after you have delivered your planned treatment, people have used what’s called consolidation therapy.

So we have done the same with the MRD test. If there is someone who has a little bit of disease left, we have tried to see if we could make that patient MRD-negative. We have also used it as a tool to build more reassurance. I mentioned before for patients who get this new combination therapies, if they are not very keen on jumping right to chemotherapy with Melphalan and transplant, if they want to collect the cells and keep them in the freezer, using the MRD as a tool to guide for reassurance.

Looking at the randomized trial showing that MRD negativity with or without transplant seems to have the same progression-free survival and in the absence of overall survival, either way, that has been published. But we would always say to patients, there are no definitive studies that have shown that this is how it is. It’s still an area of investigation. So if a patient wants to sort of do everything by the traditional book, we would give every step in the therapy and not pay attention. But a lot of patients say, I would rather monitor, and if I have to do these more toxic therapies, I wouldn’t do it. But I will use MRD to build confidence in myself.

Lisa Hatfield:

Well, thank you so much, Dr. Landgren, these have been great questions, and I actually have another half sheet of questions that we don’t have time for, because that’s all the time that we have. Dr. Landgren, thank you so much, it’s been a pleasure talking with you today. So thank you for joining our Patient Empowerment Network START HERE program. This has been an excellent discussion. Thanks to all of you, for your questions and tuning in. My name is Lisa Hatfield. I’ll see you next time.

Dr. Ola Landgren:

Thank you very much for having me. Thank you.

CAR T-Cell Therapy Follow-Up Monitoring | What Patients Can Expect

CAR T-Cell Therapy Follow-Up Monitoring | What Patients Can Expect from Patient Empowerment Network on Vimeo.

What can CAR T-cell therapy patients expect for follow-up monitoring? Expert Dr. Krina Patel from The University of Texas MD Anderson Cancer Center discusses how long follow-up monitoring is typically carried out, issues that are monitored for, and proactive advice for patients to help ensure optimal care.

[ACT[IVATION TIP

“…for long-term side effects really is infections, number one, because even after I just saw a patient last week whose IgG level’s still less than 100 even a year after CAR T. We’ve just knocked out the good and the bad, and so it was just a higher risk of infection, so we try to prevent by giving IVIG regularly, and so again, any time you get an infection, just talk to your doctors, don’t say, ‘This is just a cold,’ just make sure that someone’s following.”

Download Guide | Descargar Guía

See More from [ACT]IVATED CAR T

Related Resources:

What Is the Impact of CAR T Therapy Access Barriers on Patients?

What Is the Impact of CAR T-Cell Therapy Access Barriers on Patients?

What Are CAR T Therapy Requirements for Care Partners?

What Are CAR T-Cell Therapy Requirements for Care Partners?

What CAR T Research Is Ongoing to Improve Treatment Response?

What CAR T Research Is Ongoing to Improve Treatment Response?

Transcript:

Lisa Hatfield:

Dr. Patel, if a patient is or has been part of a clinical trial involving CAR T, how long will that patient be followed under the clinical trial protocol for long-term effects, and this is especially important for people who see community oncologists and are wondering about any latent effects that they might experience, how long were those clinical trials follow those patients?

Dr. Krina Patel:

That’s a great question. So most trials will follow for at least two years just for toxicity, efficacy, now, most trials will follow until you’re relapsing, so that’s the point, is that we want to make sure this is working, that you don’t have any long-term toxicity, and when you relapse, we call that the progression-free survival, which is what most of the trials are looking at, and once you relapse, usually they’ll say, Okay, you’re coming off a trial because now you need other therapy and that could take years.

And however, for all CAR T products, because these are genetically modified, the FDA requires that you go into a long-term protocol where we’re monitoring for potential leukemias or lymphomas that T cells can cause, theoretically. So that is for 15 years, total. So everyone then is supposed to go on to that, now we can’t force you to go on to those, but it is something important because it’s come up recently that maybe some of these T-cell products are leading to leukemia or lymphoma, because we’re modifying those T cells could they themselves turn into a cell that causes cancer.

The theoretical risk has always been there, I will tell you that in reality, yes, there have been probably a handful of patients out of all the lymphoma and myeloma and leukemia patients who’ve been treated with CAR T where maybe it came from the T cell itself, the actual CAR T. The majority of other cases that have been reported, it’s been a low risk, it’s less than what we usually see in the general population of patients with blood cancers that get other blood cancers.

But when we see it, most of the time,  it’s not in the T cell where the CAR was in, but again, a handful have been, and that is really why as a group, we have to be really careful and make sure that some of the different. The way we make CAR T is very different amongst the products, and to make sure that one product versus another isn’t more likely to cause T-cell leukemias or lymphomas. So that’s the main reason why that 15-year protocol exists.

Lisa Hatfield:

And do you have any tips for patients who maybe have undergone CAR T therapy, are several years out and working with our community oncologist, what should they be watching for in terms of any late in side effects or long-term side effects?

Dr. Krina Patel:

So I think the activation tip here for long-term side effects really is infections, number one, because even after I just saw a patient last week whose IgG level’s still less than 100 even a year after CAR T. We’ve just knocked out the good and the bad, and so it was just a higher risk of infection, so we try to prevent by giving IVIG regularly, and so again, any time you get an infection, just talk to your doctors, don’t say, “This is just a cold,” just make sure that someone’s following.

And the other big thing is your blood count, so if your blood counts start doing something crazy, your white count’s getting high or too low, you’re not on any therapy, your hemoglobin is getting really low, your platelets are getting low, that’s where we want to make sure there’s not a secondary cancer, a secondary blood cancer involved. Again, T-cell leukemia myeloma was really rare, but we have seen 10 percent patients with MDS or AML in the relapse refractory population, so that is something else we would still want to watch out for and make sure we don’t miss that.


Share Your Feedback

Create your own user feedback survey

What Is the Impact of CAR T-Cell Therapy Access Barriers on Patients?

What Is the Impact of CAR T-Cell Therapy Access Barriers on Patients? from Patient Empowerment Network on Vimeo.

How do CAR T-cell therapy access barriers impact patients? Expert Dr. Krina Patel from The University of Texas MD Anderson Cancer Center discusses factors that create CAR T-cell therapy access barriers and the impacts of FACT accreditation and the REMS program.

Download Guide | Descargar Guía

See More from [ACT]IVATED CAR T

Related Resources:

What Are CAR T Therapy Requirements for Care Partners?

What Are CAR T-Cell Therapy Requirements for Care Partners?

CAR T-Cell Therapy Patient Eligibility | What Patients Should Know

CAR T-Cell Therapy Patient Eligibility | What Patients Should Know

What CAR T Research Is Ongoing to Improve Treatment Response?

What CAR T Research Is Ongoing to Improve Treatment Response?

Transcript:

Lisa Hatfield:

Dr. Patel, how do you perceive the impact of patient access barriers, such as geographical constraints and caregiver requirements on the widespread adoption of CAR T therapies for myeloma patients?

Dr. Krina Patel:

Yeah, so this is a huge, huge problem. I think I’m such a big CAR T fan because again, it works so well, it gives people this amazing quality of life, so once they can get it, it’s phenomenal, and people want it again, if they can get it again down the road. But you have to get there, and so the biggest barriers, I will say is insurance coverage, making sure your insurance, now most people who have insurance it will cover, but some people with Medicare, if they don’t have a secondary, that 20 percent that you have to cover is insane. I don’t think most people would not be able to do that. So that’s number one.

Number two is location. And again, if you can’t come somewhere, there’s about 200 centers in the U.S. that are FACT-accredited, and you have to be…it’s an acronym basically for anyone that does cell therapy, and you have to be accredited to be able to give CAR T, so unlike other therapies like bispecifics that are not under that jurisdiction, for CAR T, you have to have that designation, so there are quite a few centers around the country that have that.

But again, access to myeloma CAR Ts is still limited to a certain degree, and so finding a place that’s nearby and having a caregiver, these are all really, really important, but we’re hoping that in the future with better CAR Ts, we won’t have to worry about staying in the vicinity for 30 days. We’re actually trying to push already to say that a lot of our patients, the majority do really well, that after two weeks, they should be able to go back home, and we can work with their local oncologist, their local doctors to make sure everything’s okay.

I think the original CAR Ts were in lymphoma and there were some pretty significant side effects we saw, so even that can’t drive for eight weeks and all these other things that we all have to do, all our patients have to do now came from that, even though most of our patients don’t get neurotoxicity, right, in myeloma, we don’t see those things. So again, we’re trying to find novel ways to push back and say to the FDA, do we really need these patients to stay here this long when our data looks so much different and better than it does for other diseases?

So I think that’s part of it as well. Through some of our newer trials, we’re trying to see if we can decrease those recommendations that once they get approved, it won’t be a part of the REMS program and all these things that. If once it becomes part of the REMS program, we have to do it and it’s hard to reverse that. But I know a lot of the companies are, and myeloma groups are trying to get together to decrease some of that burden, because it might be a little bit too much for the majority of our patients where they don’t need it medically and I know it would help them get access to the drugs, but coming back to the novel CAR Ts, there are ways to make the CAR T in less than 48 hours now that people are testing, and if we can do that, that can really decrease your time of even having to come get your cells collected, then go back home for bridging therapy, then come back for the actual CAR T treatment.

And if we can give it to you faster and we can decrease that toxicity, then hopefully again, it could be two weeks and then we get you back home. So I think that’s the ultimate goal. I just don’t know when that will happen.


Share Your Feedback

Create your own user feedback survey

How Can Variable Patient Groups Be Addressed in CAR T?

How Can Variable Patient Groups Be Addressed in CAR T? from Patient Empowerment Network on Vimeo.

Can CAR T-cell therapy address variable patient groups? Expert Dr. Krina Patel from The University of Texas MD Anderson Cancer Center discusses variances in different myeloma patient groups, the KarMMa-3 study, and proactive advice for patients.

[ACT]IVATION TIP

“…if you are in a area, let’s say rural America where you don’t have access or you are in a minority population, African American, Hispanic, etcetera, or older, frail patients who are older that are considered vulnerable as well, absolutely make sure to talk to your doctors about these novel therapies because you still can get them safely and they will work. They can work. You just have to go to a center where they know how to adjust those types of therapies to make sure you get the best options out there as well.”

Download Guide | Descargar Guía

See More from [ACT]IVATED CAR T

Related Resources:

What Patient Types Are Good Candidates for CAR-T Therapy?

What Patient Types Are Good Candidates for CAR T-Cell Therapy?

Are CAR-T Clinical Trials Studying Use As a Frontline Therapy?

Are There Myeloma Trials Investigating CAR T for Frontline Therapy?

A Look at Promising Strategies to Improve CAR T-Cell Therapy Access

A Look at Promising Strategies to Improve CAR T-Cell Therapy Access

Transcript:

Lisa Hatfield:

Dr. Patel, how might the heterogeneity of patient populations impact the standardization and reproducibility of CAR T therapy outcomes across different clinical settings, and what initiatives are in place to address this variability?

Dr. Krina Patel:

Yeah, I think that’s a great question because again, this is a personalized therapy. So it depends on what your myeloma is like, the genomics, the genetics of your myeloma, how aggressive is it, plus your T cells, right? And so everybody’s genetic ancestry, etcetera, is very different. So the idea of a personalized medicine, more than just even across groups of people, it’s at the individual level. And I think when you talk about different races or ethnicities, we have seen some differences in our real-world data, in very relapsed/refractory patients, where people can get great response rates still.

So, for instance, Caucasian patients versus African American patients, our response rates are still high in the 80s and 90 percent, but the toxicity is a little bit higher in our African American patients. It’s still not high grade. It’s not anything that makes me say, I’m not going to give this, but the baseline inflammatory markers are a little bit higher. And so once we get the CAR T, our patients tend to get a little bit more CRS.

They end up in the hospital a little bit longer. Now, again, this is a multivariate analysis and we couldn’t find any other difference, but when we look at KarMMa-3, which is one of our big studies that led to ide-cel (idecabtagene vicleucel) [Abecma] being approved early, we actually had an outcomes of African American patients only that we looked at and that we presented just this past TCT, and response rates were actually a little bit better.

Again, you can’t compare them because the numbers aren’t there to power that to compare, but numerically the numbers were better in terms of response rate, in terms of progression-free survival, it was actually more months that it beat the standard of care and we didn’t see more toxicity.

And so I think we do need to look at these things and make sure there’s not one group of patients has a lower efficacy for some reason, and why is that and how can we improve that? And so far, we don’t really see that. And the other is the toxicity piece, to make sure that these therapies that do cause some strange toxicities that we’re watching and seeing who might be more vulnerable to those toxicities, who do we need to maybe even prevent, do prevention strategies for, but so far we haven’t seen it.

And then I think coming back to the individual, right?So again, all of us have these different T cells that have different mutations in them, and some folks, for some reason, even with less myeloma, their T cells just expand really fast and other folks, they don’t. And so in the future to get best outcomes, we need to see how we can turn the volume lower for those folks who have really sensitive T cells.

And for those who don’t, how do we, what else can we add in combination to actually increase those T cells so that they’re actually doing a better job at killing the myeloma, right? And including the microenvironment too. So I think there’s a lot of translational work as well as the epidemiology side of things to say, okay, how do we first diagnose the problem, find the problems, and then how do we figure out how to intervene to then improve outcomes for all our patients? I think the activation tip here is that if you are in a area, let’s say rural America where you don’t have access or you are in a minority population, African American, Hispanic, etcetera, or older, frail patients who are older that are considered vulnerable as well, absolutely make sure to talk to your doctors about these novel therapies because you still can get them safely and they will work. They can work. You just have to go to a center where they know how to adjust those types of therapies to make sure you get the best options out there as well.


Share Your Feedback

Create your own user feedback survey

What Patient Types Are Good Candidates for CAR T-Cell Therapy?

What Patient Types Are Good Candidates for CAR T-Cell Therapy? from Patient Empowerment Network on Vimeo.

For CAR T-cell therapy, what patient types are good candidates? Expert Dr. Krina Patel from The University of Texas MD Anderson Cancer Center discusses patient situations that qualify them for CAR T-cell therapy and shares proactive patient advice.

[ACT]IVATION TIP

“I think a lot of people have this misunderstanding that CAR T isn’t for everybody, but I will say it’s actually more likely that you’re going to be eligible for CAR T over auto transplant. So I think it’s just bringing it up, talking to them, and seeing a specialist to discuss which ones are the right one for you and when to go.”

Download Guide | Descargar Guía

See More from [ACT]IVATED CAR T

Related Resources:

How Can Variable Patient Groups Be Addressed in CAR T?

How Can Variable Patient Groups Be Addressed in CAR T?

Are CAR-T Clinical Trials Studying Use As a Frontline Therapy?

Are There Myeloma Trials Investigating CAR T for Frontline Therapy?

A Look at Promising Strategies to Improve CAR T-Cell Therapy Access

A Look at Promising Strategies to Improve CAR T-Cell Therapy Access

Transcript:

Lisa Hatfield:

Dr. Patel, given that CAR T is approved for earlier lines of therapy, can you describe the type of disease characteristics of patients that are likely to be considered first for CAR T?

Dr. Krina Patel:

Yeah, I think it’s a really exciting time, we got approval for two different CAR Ts that were approved in fifth line. So patients had to relapse four times before they could get to CAR T therapy. And now one of them, cilta-cel ciltacabtagene autoleucel [Carvykti], is approved in second line, so people have to relapse once before they can get it.

And the other CAR T, ide-cel (idecabtagene vicleucel) [Abecma], is approved in third line, so you have to relapse twice. There are reasons why that is the way it is, but both of these CAR Ts are pretty fantastic, and we’re really excited that more patients will now have access. As you can imagine, relapsing once, it’s already hard enough to then say, okay, I need a different therapy, but to go through four times before you can do something like CAR T is really, really important.

So I think the biggest characteristic, and I would say for my patients, it’s really their ability to keep their myeloma controlled for at least six to eight weeks, potentially, right? Because this is a personalized therapy, unlike most other myeloma therapies where we have to take the T cells out, we have to then send them to a lab to make the CARs, then it takes about four to six weeks to get them back. During that time, I just have to know that your myeloma can stay controlled or even improve with whatever bridging therapy we decide to do during that time.

And we know that when patients have myeloma that is on its way down, that it’s actually improving, by the time they get to the CAR T therapy, the infusion part, they tend to do better in terms of efficacy, but also have less toxicity. So there’s a few different toxicities that we can talk about with CAR T that are very distinct compared to most other therapies, that again, if you have less disease burden, the rates of that toxicity and the high-grade toxicity goes away, right? It’s much, much lower than if you have a lot of myeloma coming in.

So, again, for my patients who have disease that I know I have other therapies to keep it knocked down or to knock it down during that bridging, that really is the main difference between, can I take this patient to CAR T or not? But I think there’s some nuances too, again, that idea that one CAR T is approved in second line, another one is approved in third line, I do think they’ve never been tested head to head, so we don’t have data in a clinical trial, but in the real world, we’ve used both of these products, a lot of us have, and I think most of us will say that one of the products is probably stronger, it probably works better cilta-cel, and that is the one that’s approved in second line, which is great.

So for my fit patients who don’t have a lot of comorbidities, who do really have high-risk disease that I need to be as aggressive as possible and do something very different, hands down, it makes sense that cilta-cel is the right thing to do right at second line, but the toxicity is also a little bit higher with that, meaning that patients are more likely to potentially get some of these strange neurotoxicities that we see, that we didn’t really see before with other therapies, some of our myeloma patients get neuropathy and we think about that as neurotoxicity, but this is different.

This is more patients after 30 days of having had their T cells can all of a sudden get a facial palsy where they’re having drooping of their face and it can affect their eating and their speaking. Now those things are not fatal, we can treat it with steroids, things like that, but they can affect your quality of life. And if it doesn’t resolve, that can affect down the road, all the other therapies we want to give you, right? But the more dangerous one is something called delayed Parkinsonianism or delayed motor neurotoxicities.

And again, we know the best prevention of that is decreasing the myeloma burden before going to CAR T, but if we can’t do that or some patients can still potentially get this Parkinsonianism, we really want to make sure there’s a risk-benefit discussion, right? That we say, okay, this is why we should go in second line.

Again, the risk is less than 1 percent now based on how we’ve done things for prevention. But on the other hand, with ide-cel, most of our patients, even on dialysis, our patients that are getting CAR T and doing well, patients with heart failure, I’ve had a 90-year-old go through ide-cel without any issues and have great responses. So I think both of these offer, one of the first times they offer time without any therapy for myeloma. And so I would say this is something most of my patients should ask their physicians about, but really then it’s nuanced in terms of when we should do it and which product.

Lisa Hatfield:

Okay, thank you. And do you have an activation tip for that question, Dr. Patel?

Dr. Krina Patel:  

Yeah, so I think the activation tip here is bring it up, bring up CAR T to your doctors, right? I think a lot of people have this misunderstanding that CAR T isn’t for everybody, but I will say it’s actually more likely that you’re going to be eligible for CAR T over auto transplant. So I think it’s just bringing it up, talking to them, and seeing a specialist to discuss which ones are the right one for you and when to go.


Share Your Feedback

Create your own user feedback survey

Dr. Krina Patel: Why Is It Important for You to Empower Patients?

Dr. Krina Patel: Why Is It Important for You to Empower Patients? from Patient Empowerment Network on Vimeo.

Why is it important to empower patients in their care? Expert Dr. Krina Patel from The University of Texas MD Anderson Cancer Center discusses her approaches and how she engages with her patients through treatment, care, and survivorship.

See More from Empowering Providers to Empower Patients (EPEP)

Related Resources:

Dr. Joshua Sabari: Why Is It Important for You to Empower Patients?

Dr. Samuel Cykert: Why Is It Important for You to Empower Patients?

Dr. Samuel Cykert: Why Is It Important for You to Empower Patients?

Dr. Eugene Manley: Why Is It Important for You to Empower Patients?

Dr. Eugene Manley: Why Is It Important for You to Empower Patients? 

Transcript:

Dr. Krina Patel:

So I think in myeloma, where our patients for the most part are not cured, they’re incurable and for the most part are on therapy lifelong. I think it’s really important that they have a community to go to, including their caregivers. There’s a lot of caregiver burnout that happens, patients, when they’re doing well or well, but when they relapse, it can be pretty dramatic and kind of take away everything again. And every time a patient’s relapsing, sometimes it feels hopeless.

And I think with all the therapies we have out there, this embarrassment of riches as we myeloma doctors like to say, we have to be able to get them through to have access to these drugs at the right time, make sure we decrease toxicity. But it’s a lot of information.

And I think for our patients, no matter how much time we spend with them, it’s just, it’s overwhelming. And I think it is for a lot of my colleagues who don’t just do myeloma all the time. I mean, it’s overwhelming for me half the time when I’m trying to see my patients and figuring out which is the next therapy. And so I really, at the first visit, talk to my patients about patient advocacy groups that are out there. And I even give them websites to go to.

At MD Anderson we’re trying to make videos for our patients so that while they’re waiting in the waiting rooms, they’ll have access to those, specifically, for CAR-T therapy and bispecifics. I think those are such great novel therapies, but they’re also high maintenance as I like to call them that there’s a lot of supportive care that’s needed for infection prophylaxis to make sure they don’t get secondary cancers, right?

All these complications that can happen, neurotoxicity, etcetera. And thankfully, for the most part, our patients do really well and they can get through it. But for those patients who end up with that, it’s really important they have this information, so they know when to contact us. And I think for my colleagues as well, we’re trying really hard to make sure we have better communication, for my patients that are in the community coming in for CAR T or for bispecific therapy, then going back to their doctors, their community doctors for the rest of their care.

So we have letters, that we come up with that we give to the patient as well as send to their doctor. We have phone numbers they can call that even if they’re back home, and they need to get ahold of someone that, they have a lifeline to say, I don’t know what to do. This is happening. And I think, it’s really important again for the patients and their caregivers to really understand, this is a lifelong journey, right?

This is not something that you’re just going to get a few cycles of treatment and then you go to survivorship clinic. And then hopefully we never have to treat again. And that this myeloma as of right now is still a continuous therapy and it could be, long periods of time between therapies. Or you might go on maintenance, for a long period of time before you need your next line of therapy, but this is a lifelong therapy that we’re going to have to do with, with everybody involved.

And I think, again, I can’t see every patient out there and most myeloma specialists can’t, but we’re happy to be a part of the team. And so really, when we can have access to things that the community might not, or be able to help in terms of, what combination is the best for this patient, and what dose reductions should we do for this specific patient?

Those are the things we would love to help our community doctors with to make sure outcomes for all our patients, those who are near us, but those who are also physically not close to us that we can still be able to help to make sure that they have the best efficacy, but also the best quality of life with this disease.

Understanding Myeloma | How You Can Collaborate in Your Care

Understanding Myeloma | How You Can Collaborate in Your Care from Patient Empowerment Network on Vimeo.

This animated video reviews the types of myeloma, essential testing following a myeloma diagnosis, and advice for working with your healthcare team for the best overall care.

See More from Collaborate Myeloma

Related Resources:

Collaborate | Being an Empowered Myeloma Patient

Myeloma Support and Resources | Why It’s Essential to Voice Your Concerns

The Benefits of Shared Decision-Making for Myeloma Care


Transcript:

Bianca: 

Welcome back! I’m Bianca, and I’m a nurse. I specialize in caring for people with myeloma. And this is Suzanne, a myeloma patient advocate.  

In this video, we are going to learn more about diagnosis and testing, and how test results may impact myeloma care. 

Suzanne: 

That’s right, Bianca. Most patients want to–and should– understand their diagnosis. Let’s start with the basics. Can you explain the different types of myeloma? 

Bianca: 

You bet. Let’s start with MGUS, which stands for monoclonal gammopathy of undetermined significance. MGUS typically has no signs or symptoms and is characterized by an abnormal protein in the blood or urine. This type of myeloma doesn’t require treatment but should be monitored regularly. 

Smoldering myeloma is a very slow-growing type of myeloma. It also does not present with symptoms. Patients with smoldering myeloma have a higher chance of needing treatment, so blood and urine studies are ordered regularly. 

And then there’s multiple myeloma, which is a buildup of plasma cells in the bone marrow that crowds out healthy cells, and causes symptoms and other problems in the body. Multiple myeloma requires treatment, and there are a number of approaches available. 

Suzanne: 

Thanks for explaining the difference. When I was initially diagnosed, I underwent a series of tests that included a blood test, bone marrow biopsy, urine test, and imaging.  

Bianca: 

Those are the standard tests when diagnosing myeloma. Your healthcare team should also order a more in-depth FISH test, which comes from the bone marrow biopsy sample, and FISH  testing can provide a better understanding of your disease. This is important because the results may impact your treatment options. 

Suzanne: 

Right—and it’s important ensure that you have had all necessary testing including this more in-depth test. You should also review the lab results with your healthcare team. You can ask questions like:  

  • Am I high-risk or low-risk?  
  • What do the results mean? 
  • How will the results impact my options for therapy? 
  • And, how often should testing be repeated? 

Bianca: 

That’s good advice, Suzanne. All of the information gathered during your diagnosis, or following a relapse, should be considered as well as your overall health when deciding on a care plan.   

And, as you’ve modeled, working with your healthcare team to make therapy choices is essential. This is a process called shared decision-making, which basically means that patients and their providers collaborate on healthcare decisions. Participating in this process encourages patients to engage in their care, helping them to feel more confident about the approaches they choose. 

Suzanne: 

That’s right! Working WITH my healthcare team makes me feel included and brings peace of mind when considering my options.  

Bianca: 

That’s the way it should be—you should always be at the center of your care. So, when considering a plan with your healthcare team, here are a few key steps: 

  • Start by understanding your diagnosis. 
  • Develop a good relationship with your healthcare team so that you can participate in your care. 
  • Ensure you have had all essential testing, including in-depth testing. 
  • Discuss the tests results with your doctor and ask questions about what they mean. 
  • And, as always, do research on your own and confirm what you’ve learned with your healthcare team. 

Suzanne: 

And don’t forget to visit powerfulpatients.org/myeloma to view more videos with Bianca and me. Thank you for joining us!  

Peer Insights: Fostering Clinic-Wide Engagement for Myeloma Clinical Trials

Peer Insights: Fostering Clinic-Wide Engagement for Myeloma Clinical Trials from Patient Empowerment Network on Vimeo.

Dr. Craig Cole from Karmanos Cancer Institute speaks to the success of clinical trials relying on not only provider endorsement, but also on the collective enthusiasm and involvement of all healthcare staff, which cultivates a patient-centric culture promoting myeloma trial participation and engagement.

See More from EPEP Myeloma

Related Resources:

Peer Insights: Maximizing Myeloma Patient Care

Peer Insights: Maximizing Myeloma Patient Care 

HCP Strategies for Navigating the Pre-trial Eligibility and Informed Consent Process

HCP Strategies for Navigating the Pre-trial Eligibility and Informed Consent Process 

How Can Myeloma HCPs Overcome Unforeseen Practice Related Barriers?

How Can Myeloma HCPs Overcome Unforeseen Practice Related Barriers? 

Transcript:

Dr. Craig Cole:

Some of the other barriers to clinical trials, the nurses and the other staff in the cancer center aren’t aware of the clinical trials, that when a patient goes through the clinic, they talk to more than just the provider. They talk to the treatment nurses, they talk to the intake people, they talk to the MAs, they talk to the scheduling people. And there was a study that was done a few years ago in looking at patients who were given consent forms and declined clinical trials. And they found that a lot of patients declined clinical trials, were because they said that, well, their doctor didn’t want them on the trial.

And when they looked further into that, they saw that, well, the doctor offered them a clinical trial, but when they discussed the clinical trial with a nurse practitioner, when they discussed that trial with a treatment nurse or the MA or any of the other staff, when they didn’t know about the clinical trial, that was considered well, if you don’t know about the clinical trial, it must not be good for me. And then they withdrew from the trial.

It really shouldn’t be left in the provider compartment. That excitement should be clinic-wide. And when you have that all-in approach where everybody’s involved, everyone’s excited about clinical trials, it produces a culture of clinical trials that everybody wants to be part of, and the patients then can jump on that bus and feel comfortable participating in the trial.