Tag Archive for: immune system

Expert Perspective | How Bispecific Antibody Therapy is Transforming Myeloma Care

 How has bispecific antibody therapy changed myeloma care? Tiffany Richards, a myeloma nurse practitioner, explains how bispecific antibody therapy works, who this therapy may be right for, and the important role of the care partner when caring for a loved one. 

Tiffany Richards, PhD, APRN-BC, AOCNP is a Nurse Practitioner in the department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center.

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

Care Partners | Tools for Self-Care and Managing Emotions

Care Partners | Tools for Self-Care and Managing Emotions

Transcript:

Katherine Banwell:

I’d like to start by learning a bit about you. Can you tell us about your role in the Myeloma Care Team? 

Tiffany Richards:

Yes. So, I’m a nurse practitioner and I’ve been here at MD Anderson for 20 years working with patients with plasma cell dyscrasia. And so, I work in collaboration with our nurse as well as our myeloma physician to not only evaluate patients, what their responses are to treatment but also to make sure that they’re tolerating treatment well, and then adjusting medication or providing supportive medications so that patients are better able to tolerate their therapies.  

Katherine Banwell:

Bispecific antibody therapy is a newer therapy. How has this option changed myeloma care?  

Tiffany Richards:

Between that and CAR T, it’s really offered our patients the opportunity to utilize the body’s own immune system to help fight the myeloma cells. I think the one nice thing that the bispecific antibodies have allowed is that you’ve had a group of patients that maybe weren’t candidates at that time for CAR T either due to other medical conditions or maybe because their disease isn’t at a place where we would be able to get them to CAR T.   

Either maybe their lymphocyte count was low, white blood cells, and so maybe the ability to collect those T cells would be impaired or the disease itself was rapidly progressing and so the patient would not be able to be off therapy in order to have those T cells collected.  

And so, the bispecific antibody allows us to utilize those T cells to go after the myeloma cells without having to go through the process of having to collect those T cells. And so, that has really changed for that group of patients. But also, we have a bispecific antibody therapy that doesn’t target the same receptor that the CAR T-cell therapies do. So, our CAR T-cell therapies target something called BCMA, which stands for B Cell Maturation Antigen.

That’s expressed on the surface of the myeloma cells, and there’s a bispecific that targets a different receptor called GPRC5D. It’s a lot of letters. But it’s a different target, and so even for patients who have had CAR T-cell therapy we can use that bispecific antibody now for those patients who have maybe progressed on CAR T.  And so, it’s allowed another treatment option for patients that they didn’t otherwise have.  

Katherine Banwell:

So, how many bispecific antibody therapies are available for people and how do they differ? 

Tiffany Richards:

So, we have three. So, we have two that target the BCMA; so, that would be teclistamab (Tecvayli) and elranatamab (Elrexfio). And then, we have a third one that targets the GPRC5D which is called talquetamab (Talvey). And so, we utilize the talquetamab if we wanna use a bispecific therapy that does not target the BCMA. And then, for patients who maybe wouldn’t be able to get to CAR T, we might use one of the BCMA therapies.  

And as far as differences between to the two BCMA, really, they’re pretty similar as far as response rates. They haven’t been compared head-to-head. And so, different centers might utilize one versus the other depending on what they have on formulary. So, I would just say, whatever one your center is utilizing that would be the one to go with. 

Katherine Banwell:

Why is a care partner required for patients who are undergoing bispecific antibody therapy?  

Tiffany Richards:

That’s a great question. So, it’s because of some of the side effects that we can see in patients who are undergoing bispecifics. So, similar to CAR T cell therapy, we can see what’s called cytokine release syndrome. We abbreviate that by CRS. And then, we also can see neurotoxicity. We don’t see it to the same degree that we see it with CAR T but patients can still experience it.  

So, cytokine release syndrome, you can get fevers. You can have a drop in the blood pressure, chills, increase in the heart rate. And so, because of that you have to be monitored closely because, if you would start to have cytokine release syndrome, we need to make sure that we’re properly intervening and we can utilize a different medication called tocilizumab (Actemra) to help quiet the immune system a little bit, quiet down those T cells. And so, you need to have somebody that’s with you at all times that knows you, and also, same with the neurotoxicity. Again, we don’t see it to this same degree that we see it with CAR T, but that doesn’t mean that it can’t happen.  

And so, you really need to have that care partner alongside of you. Plus, I think just with these immune therapies, it’s a lot of information that we’re giving patients.   

And so, it’s important to have that other person there to kind of hear what maybe you’re not able to catch. There’s a lot of information that’s being given to you and can be very overwhelming at times. And so, it’s important to have that second person there to kind of be another set of ears as you’re going through this journey. 

Advice for Being Prepared As a Bispecific Antibody Therapy Care Partner

 When a loved one is undergoing bispecific antibody therapy, how can a care partner prepare? Tiffany Richards, a myeloma nurse practitioner, provides key advice for care partners, including important questions that they should ask the healthcare team.

Tiffany Richards, PhD, APRN-BC, AOCNP is a Nurse Practitioner in the department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center.

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

Care Partners | Tools for Self-Care and Managing Emotions

Care Partners | Tools for Self-Care and Managing Emotions

Transcript:

Katherine Banwell:

When meeting with a patient and their care partner for the first time, what three key pieces of advice would you share with them? 

Tiffany Richards:

So, one is I would make sure that you understand the regimen; so, what days you’re going to be dosed, what day you’re going to go in the hospital, approximately what day you might be discharged, knowing that that discharge date can vary depending on how you’re doing with tolerating the therapy. I would also make sure that you understand what prophylactic medications you need to be on.  

So, oftentimes we’re putting you on an antiviral. We’re putting you on something to prevent a certain kind of pneumonia called PJP. And then, we also may put you on something to help you boost up your antibodies. So, oftentimes patients with myeloma, they’re low on their normal IgG antibodies just because we’re depleting all of these plasma cells in the bone marrow and so you’re not producing enough of these other antibodies. And so, we may have to put you on IVIG once a month.  

And so, making sure that you understand what it is that you all need to be taking, and then also making sure that you’re asking the questions as far as “How often do I have to come back?” 

“Do I have to get it all at an academic center or is it the opportunity to get it done locally?” I think that’s really important question because some local community providers, they’re willing to do the subsequent cycles of the bispecific. They may just not want to do that first cycle. And so, asking the question, “Can I get my subsequent cycles locally?” because to travel long distances, particularly depending on where you live geographically, it may be difficult to go back and forth to an academic center. 

Katherine Banwell:

Is there anything you’d like to add about caring for someone who is being treated with bispecific antibody therapy? 

Tiffany Richards:

Yeah. I think knowing as much as you can about how the drugs work is really important, and then also what you can do to help manage and mitigate some of the side effects is important. And then, making sure that you understand the schedule, and as well as the schedule for the supportive medications is also really important. When we transition patients from an academic center to community, we usually have a touchpoint with that community provider. We provide instructions.  

So, I think it would good to ask the academic center for a copy of the letter for the communication that’s being given, so that everybody is on that same page, can be really, really helpful. 

Advances in CAR T-Cell Therapy Side Effect Management

Are there new ways to manage the potential side effects of CAR T-cell therapy? Dr. Rahul Banerjee, a myeloma specialist and researcher, reviews common side effects of this treatment option and discusses new ways that providers are approaching the management of both short-term and long-term issues. 

Dr. Rahul Banerjee is a physician and researcher specializing in multiple myeloma and an assistant professor in the Clinical Research Division at the University of Washington Fred Hutchinson Cancer Center in Seattle, WA. Learn more about Dr. Banerjee.

Download Resource Guide

Related Resources:

CAR T-Cell Therapy for Myeloma | Challenges and Unmet Needs

CAR T-Cell Therapy for Myeloma | Challenges and Unmet Needs

Myeloma Research | Updates in CAR T-Cell Therapy

Myeloma Research | Updates in CAR T-Cell Therapy

How Long Is CAR T-Cell Therapy Effective in Myeloma? An Expert Explains

How Long Is CAR T-Cell Therapy Effective in Myeloma? An Expert Explains

Transcript:

Katherine Banwell:

Well, let’s talk about side effects of CAR T-cell therapy. What advances are being made in managing them?  

Dr. Rahul Banerjee:

Excellent question. So, I break the toxicities into short-term and long-term toxicity of CAR T therapy.  

And this is actually fairly similar for both, regardless of the underlying disease, whereas lymphoma, leukemia, or myeloma, very similar. Just so everyone, just to reorient the audience, short-term toxicities, people often talk about the big two. I’m going to say the big three, actually. The first one is cytokine release syndrome, or CRS, which is inflammation, typically causing fever, sometimes low blood pressure. 

The second is neurotoxicity, or ICANS. For reasons that aren’t entirely understood, sometimes all those chemicals from inflammation can cause people to feel a bit off mentally or cognitively. Sometimes people might not be able to talk, or might not talk correctly, or sometimes have issues along those lines. 

And the third, I would say, is low blood counts or infections. Both the lymphoma, leukemia CAR Ts and the myeloma CAR Ts very rapidly deplete the good plasma cells or the good lymphocytes, the good cells in the bone marrow, and so immediately you see patients at risk of infections. 

And for a complicated number of reasons, one of which being cytokine release syndrome, CRS inflammation within the bone marrow, where all these cancer cells are hiding, the stem cells in the bone marrow hide away, right? They kind of go into a bunker to stay away from all of this. And so patients often have low blood counts for significant amounts of time after CAR T therapy, something called hematotoxicity. 

Those are short-term toxicities. Long-term, very briefly, that risk of infection and low blood counts is still there, I would say, for up to a year after CAR T therapy, sometimes longer for some patients. There is a risk, obviously, of the original cancer coming back, in this case the myeloma, particularly myeloma. 

And three, there are rare delayed toxicities to be on the lookout for. So, one of them, for example, with cilta-cel or Carvykti, the numbers are hard to see what the rate is prospectively because it’s been going down with time.  

But rarely, I would truly in my heart say 1 percent of the time, if not less, patients can get Parkinsonism, where they’re not able to move as rapidly, they’re not able to, they’re kind of shuffling gait, having tremors, et cetera. Not the same as normal Parkinson’s disease, because the normal meds don’t work, just time is often the only thing that really makes a big difference. 

Technically, there’s a box warning on all CAR T therapies now about a risk of second cancers. That risk is not new to anyone who’s ever received any treatment for myeloma because from the very beginning, we tell people that these drugs have been linked to a potential risk of second cancers in the future.  

In terms of strides that are being made to improve that, I think we’re making a lot of improvement. So, I think the biggest thing that we’ve learned, and I remember when I was a trainee, for example, when I was in my medical training, the early days of CAR T therapy – actually out in Philadelphia, I trained at Penn – and there, we were scared about trying to tone down the inflammation.  

When these side effects happen in the short term, the goal is if the patient’s obviously having side effects, and the question is, “Can we not kill the T cells? Can we just dial that down?” Say, “Look, I’m happy the T cells are angry. I’m happy they’re killing the cancer, the myeloma in this case. But can you just dial it down a little bit with a medication called tocilizumab (Actemra), or corticosteroids like dex?” 

We used to be very nervous about doing that because we said, look, the patient’s put all this blood, soil, sweat, and tears right into this CAR T therapy, and we don’t want to do anything that can hinder the T cells from working.  

Now we know that that level of inflammation is not doing anyone any favors at all, and so we’re able to really start these medications to just dial down the immune system faster. As soon as someone has a fever, for example, at many centers, we do consider, within an hour or two, giving one of those medications. Don’t wait till they’re in the ICU, give it then.  So, I think just tweaking our algorithms has made probably the biggest difference, in my mind, to make CAR T safer.  

Other things that have helped, I think, are better understanding of why patients have these other toxicities and strategies to prevent it. And so, for example, the neurotoxicity risk, some of it is part of disease burden. We think that patients who have a lot of disease going into CAR T therapy may have more toxicities. So, giving better treatments as, quote-unquote, “bridging treatment” before CAR T therapy that we have better, newer treatments now, have sometimes helped to really debulk the disease before going to CAR T.  

That’s helped a lot with side effect management. In terms of long-term risk, the third thing that I really encourage all my patients and all my oncologist partners in the community to really push for is the infection risk and how do we prevent it? So, I think probably the biggest thing that we’ve recognized is intravenous immunoglobulin, which is IVIg, which is basically an antibody transfusion.  

When people donate blood, they also donate plasma, often, and the plasma contains antibodies against whatever they themselves have fought off circulating in the area – viruses, colds, et cetera. 

You can take all those antibodies, put them all together into a sterile bag, and give it to the patient who’s gotten CAR T therapy. Because the patient’s gotten CAR T therapy, assuming it’s working, which it normally does for several months, right, to knock out all cells, good and bad immune cells, that patient is not making any antibodies at all. They’re a sitting duck for infections. And so I would say IVIg, using that routinely now is not just the exception once they’re having infections, but even in the absence of infections, just giving it.  

Insurance companies are not happy with me when I suggest that because it’s expensive, but that’s the right thing to do for patients, and I think that has helped a lot, in my experience, for all of these immunotherapies, both CAR T and bispecific antibodies, to lower the risk of infections. 

Being Empowered | Why Care Partners Should Feel Comfortable Voicing Concerns

Dr. Craig Cole, a myeloma specialist, shares advice for care partners to feel empowered when engaging with the healthcare team, emphasizes the importance of communication, and provides suggested questions for the care partner to ask. 

Dr. Craig Cole is a multiple myeloma specialist at Karmanos Cancer Institute in Detroit, MI and in East Lansing, MI. Dr. Cole also serves as an associate professor at Wayne State University and at Michigan State University. Learn more about Dr. Craig Cole

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Myeloma Care Partners | Understanding Bispecific Antibody TherapyMyeloma Care Partners | Understanding Bispecific Antibody Therapy Bispecific Antibody Therapy | The Important Role of Care Partners

Bispecific Antibody Therapy | The Important Role of Care Partners 

Transcript:

Katherine Banwell:

How can care partners feel comfortable speaking up and voicing concerns about care? 

Dr. Craig Cole:

Yeah, I think the big thing is – that’s such a good question because I’m very passionate about patient empowerment, a lot of Ps in that statement. 

But it really is having good communication with your provider. And I think one important thing is slowing down your provider. Providers, doctors, nurse practitioners, and PA’s, these days there’s a lot happening in the clinic. There’s a lot going on. And to the provider this may be a very routine bispecific antibody initiation, but for you it’s the first time.  

Katherine Banwell:

Right.  

Dr. Craig Cole:

And so, making sure you slow them down, to slow them down. I think my patients know if I’m running late, it’s because I had to slow down and go through the process, and make sure it’s well understood and that you should feel comfortable. And these days to ask your doctor question, and question your doctor, to ask about these therapies, the side effects, and the efficacy of them.  

If your doctor doesn’t like it, or if your doctor gets angry, then it really is time to find a new doctor because the doctor is there to serve you and to help you and make – you have to make sure that everything, your questions are answered, and that you feel comfortable going home. If you feel uncomfortable going home, then turn around and ask them again. 

Katherine Banwell:

Yeah, yeah. And if not, the doctor – I suppose some of these questions could be answered by someone on the care team. 

Dr. Craig Cole:

Yeah, and a lot of places have bispecific teams. I mean, these are such common drugs these days that there are teams of people that are behind the scenes working. And some of the phone numbers are to the other people that are on the team so absolutely. 

Katherine Banwell:

Yeah. You mentioned empowerment. How do you empower care partners to engage in their loved one’s care?  

Dr. Craig Cole:

I think that the first thing that I do, this is what I personally do, is I write down everything. I write down the plan. I write down the mechanism of action. I’m a very visual person. And so, I write everything down for patients. And I think when they see me writing it down, and I hand the papers to them as I write things down that it shows them that it isn’t a one way street. It isn’t me talking to myself in medicalese about a patient. It really is a partnership. And I do this with the trainees that you never, ever walk out of a room without asking, “Are there any other questions?” And I think one very important question for care providers and patients to ask their providers is to ask, “Is there anything that I should have asked? 

Is there anything that you think that people normally ask that we may have missed or not gone over?” Because again if it’s your first time using these medications, you may not have thought of everything and thought of all the questions to ask. So, asking your provider, but really having that two-way conversation, and I really do. I really try to make sure that before my – before we give any of these medications, that my patients are engaged, that they understand what we’re doing and why we’re doing it. And if they don’t, then we start all over again, and there is no fault in that at all.  

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

How can you best care for a loved one who is undergoing bispecific antibody therapy? Dr. Craig Cole, a myeloma specialist, provides key advice for care partners emphasizing the necessity of taking notes and for having a solid plan if issues arise, and he shares key questions to ask the doctor about bispecific antibody therapy.

Dr. Craig Cole is a multiple myeloma specialist at Karmanos Cancer Institute in Detroit, MI and in East Lansing, MI. Dr. Cole also serves as an associate professor at Wayne State University and at Michigan State University. Learn more about Dr. Craig Cole

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

Essential Monitoring Following Bispecific Antibody Therapy for Myeloma

Essential Monitoring Following Bispecific Antibody Therapy for Myeloma

Being Empowered | Why Care Partner Should Feel Comfortable Voicing Concerns

Being Empowered | Why Care Partner Should Feel Comfortable Voicing Concerns

Transcript:

Katherine Banwell:

Dr. Cole, what sort of questions should care partners be asking the care team when a loved one is undergoing bispecific antibody therapy? 

Dr. Craig Cole:

I think one of the big questions and – oh first I would say write everything down. Write everything down and have your care provider write things down or record them. Because I think it’s important to have that – have something written on hand. In our house we put everything on the – instructions on refrigerator with a magnet to make sure everyone sees it. But the one – one big question to ask is, “What are the – with this specific antibody that the patient’s receiving, what is the risk of the of the cytokine release syndrome?  

What’s the risk of the neurotoxicity that we talked about in the timeline?” Because those can be very different. “When should I worry? And how long should I be watching for these side effects?” The other thing is to have a solid plan of what to do if there are – if there’s any side effects. And so frequently that doctors or providers will write a prescription for steroids or Tylenol to take if any of those symptoms happen, but also to have a phone number to call a provider or to call the clinic if something were to change. Because again, these aren’t symptoms that you want to sit on where you say, “Oh, I have a fever, no big deal.” I mean it’s definitely good to call, and so, having a plan set. And I would make sure that you have that written down and then talk back, repeat back to the doctor or the provider that the plan is set.  

It’s not a forever plan. It’s just doing those first few doses of the bispecific. And also knowing sort of – I think a really good question is knowing the long-term efficacy of these. I mean these therapies are – work really, really well, but also knowing what are the chances of this working, of it not working? And I always like to have a plan B. “If this doesn’t work well, what are we going to do next?” And I think that’s a very fair question to providers. 

Katherine Banwell:

Dr. Cole, is there anything else you’d like to add about caring for someone who’s being treated with bispecifics? 

Dr. Craig Cole:

I think that the biggest thing is how incredibly exciting these medications are. I mean, there are – I went through and talked about a lot of the bispecifics for cancer, but there have been revolutionary biospecifics for macular edema, for hemophilia, the bleeding disorder. And these are revolutionary drugs in cancer. And really, it’s incredible that – how well these drugs fight cancer. And the fact that they use your own immune system, not someone else’s immune system, not some chemotherapy, but using your own immune system is incredible. And so, I always tell people to be really encouraged that the technology is this – if you’d have asked me this 10 years ago about a bispecific antibody I would say that’s impossible.  

And now we’re at the cusp of that. And the other thing is to be involved in clinical trials, that all these, a lot of – there are a lot of clinical trials and bispecifics because it is the big, exciting thing. And so, if you have the opportunity to participate in a bispecific clinical trial, I would definitely encourage that because it really is the cutting edge of medicine these days.  

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

 
Dr. Craig Cole reviews the side effects of bispecific antibody therapy, the symptom care partners should be monitoring for, and the importance and impact of early intervention if any issues arise.

Dr. Craig Cole is a multiple myeloma specialist at Karmanos Cancer Institute in Detroit, MI and in East Lansing, MI. Dr. Cole also serves as an associate professor at Wayne State University and at Michigan State University. Learn more about Dr. Craig Cole

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

Myeloma Care Partners | Understanding Bispecific Antibody Therapy

Myeloma Care Partners | Understanding Bispecific Antibody Therapy

Essential Monitoring Following Bispecific Antibody Therapy for Myeloma

Essential Monitoring Following Bispecific Antibody Therapy for Myeloma

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Transcript:

Katherine Banwell:

Do side effects vary from patient to patient? 

Dr. Craig Cole:

Yes, so they actually vary greatly from patient to patient and from drug to drug. There’s some bispecifics for some cancers that have low risks of cytokine release so low that they don’t even need to come to the hospital. And some of them have such a high risk of those cytokine release syndromes that people are in the hospital for a few days.  

The other thing is usually the more tumor someone has, the more disease and cancer they have, the higher those risks of cytokine release. And so, it does vary from patient to patient to and from medication to medication. 

Katherine Banwell:

What should care partners understand about caring for someone during therapy? 

Dr. Craig Cole:

One of the big things that care partners should look for or to be aware of are – is the timeline for a lot of those symptoms. The highest risk for the side effects, the things to look out for, the neurologic toxicity, the fevers, and shortness of breath, and things are in the first few days of each dose of receiving therapy.  

Some of those therapies actually because of the neurotoxicity, they don’t let anyone drive, any patients drive for the first few weeks after receiving a bispecific. So, knowing the timeline, that in those first few days, that you really have to check the temperature, have a plan, know who to call, watch for those symptoms. But as the weeks move on, like after the second dose, there’s much less toxicity, third dose, even less risk. Fourth dose and on is very rare to have any of those toxicities, and so then you can relax. And usually people are able to drive. So being aware of the timeline’s important. 

Katherine Banwell:

Yeah. Are there advances being made in the management of side effects for bispecifics? 

Dr. Craig Cole:

Oh yes, and so that’s the – that’s one of the really exciting things is the – is what I was just talking to one of our trainees about this, about the evolution of the bispecific antibodies have been to make them more effective, make them more sticky, make them engage those T cells more while decreasing the toxicities. 

And so the ones that we’re seeing that are in clinical trials now that hopefully will be approved soon have less of those side effects, less hospitalization, and actually have a longer frequency of being given. The other thing is that we’re really beginning to learn a lot about treating cytokine release syndrome, especially as severe cytokine release syndrome. So, there was a drug that was used to treat severe COVID called tocilizumab (Actemra).  

Katherine Banwell:

Yeah.  

Dr. Craig Cole:

And that was used when people came in with COVID symptoms which can be a lot like cytokine release. The would receive this medication to help control that. Now we’re using that to treat cytokine release syndrome.  

And there’s quite a bit of data, especially in multiple myeloma in using it prophylactically to prevent cytokine release syndrome. And there are studies that show that the usual rate in multiple myeloma, kind of the specialty that I have, the usual rate of cytokine release – some cytokine release is about 70 percent with using prophylactic tocilizumab, which is just an antibody against one of those cytokines, IL-6. It goes down to – up to about 25 percent, so 75 to 25.  

And really it has no adverse side effects and doesn’t do anything with the outcome or the effectiveness of the bispecific antibodies.  

Katherine Banwell:

Well, that’s an incredible difference, isn’t it? 

Dr. Craig Cole:

Yes, yes, that was really – the trick is trying to get insurance companies to approve it and to get hospital systems to approve it.  

But I am very confident that very soon as we get more data about using it prophylactically that they’ll be incorporating it into the guidelines. 

Essential Monitoring Following Bispecific Antibody Therapy for Myeloma

Why is a care partner essential for someone undergoing bispecific antibody therapy for myeloma? Dr. Craig Cole, a myeloma specialist, discusses the essential role of care partners following treatment, emphasizing the importance of monitoring for potential side effects. 

Dr. Craig Cole is a multiple myeloma specialist at Karmanos Cancer Institute in Detroit, MI and in East Lansing, MI. Dr. Cole also serves as an associate professor at Wayne State University and at Michigan State University. Learn more about Dr. Craig Cole

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

Being Empowered | Why Care Partner Should Feel Comfortable Voicing Concerns

Being Empowered | Why Care Partner Should Feel Comfortable Voicing Concerns

Bispecific Antibody Therapy | The Important Role of Care Partners

Bispecific Antibody Therapy | The Important Role of Care Partners 

Transcript:

Katherine Banwell:

What is the role of a care partner for someone undergoing bispecific antibody therapy? 

Dr. Craig Cole:

Yeah, the care partner is, I think, a critical component of someone receiving bispecific therapy. And their reason is really to do with the side effects and monitoring the side effects of the therapy. What’s the big side effect of the bispecific antibodies is again when those T cells engage the cancer cells and they find the cancer, they release chemicals to destroy the cancer immediately.  

And those chemicals are from the T cells, can cause people to feel very ill, or can cause them to feel very ill very quickly, or they can have fevers, and they can have difficulty breathing. And that’s called cytokine release syndrome. Cytokines are the chemicals that the T cells are using to kill the cancer cells.  

Release, meaning that T cells are releasing that, and syndrome mean that different things can happen to different people. And the highest risk for the cytokine release syndrome is usually within the first two to three treatments, usually in the first two or three days of the therapy. And a lot of times when people get the bispecific antibodies, sometimes it’s given in a brief hospitalization like an overnight hospitalization, but then they go home.

And then the trick is monitoring for that cytokine release syndrome, the fevers that can be associated with that, shortness of breath, low blood pressure. And in having a couple people observing, watching for those signs and symptoms are really important. Because if cytokine release syndrome isn’t addressed immediately, it can progress to worse outcomes, meaning that the blood pressure gets lower, the difficulty in breathing gets worse.  

If let completely go, people can end up in the intensive care unit which is very, very, very rare. But that’s why we address this as early as possible. The other side effect, and probably kind of the most subtle thing, are some of the neurologic things that can happen with the bispecific antibodies. So, it’s the neurologic toxicity, or some people call it ICANS. And that’s when some of those cytokines that we talked about that are from the T cells can cross the blood brain barrier and cause patients to be confused.  

They can have word finding difficulties. They can feel – almost have stroke-like symptoms. They’re temporary, but they definitely need to be addressed. And sometimes patients may not be aware that they can’t find the right word, or they want to speak, and the words don’t come out, or when they speak it’s the wrong words are coming out.  

And that’s a real, real big sign that you need to call your doctor immediately, or your provider immediately if you have those neurologic symptoms. So, watching for those side effects, so low blood pressure, the high fevers, and stroke like symptoms. It’s not a stroke, but it’s just those chemicals in the brain that can cause people to have some neurologic problems. And again, if you address those immediately, they are definitely reversible.  

Myeloma Care Partners | How Can You Support Your Loved One During CAR T-Cell Therapy?

How can care partners be informed and prepared when a loved one is undergoing CAR T-cell therapy? Myeloma expert Dr. Adriana Rossi explains the role of the care partner in each step of the CAR T process, how to understand and monitor for side effects, and shares key advice for self-compassion and self-care when serving as a care partner for a loved one. 

Dr. Adriana Rossi is Director of the CAR T and stem cell transplant program at the Center for Excellence for Multiple Myeloma at Mount Sinai Health System in New York City. Learn more about Dr. Rossi.

Download Resource Guide

See More from The Care Partner Toolkit: CAR T-Cell Therapy

Related Resources:

CAR T-Cell Therapy | Monitoring for Side Effects As a Care Partner

CAR T-Cell Therapy | Monitoring for Side Effects As a Care Partner

An Essential CAR T-Cell Therapy Team Member | The Care Partner

An Essential CAR T-Cell Therapy Team Member | The Care Partner

Where Can Myeloma Care Partners Find Out More About Financial Support?

Where Can Myeloma Care Partners Find Out More About Financial Support?

Transcript:

Jamie Forward:

Hello and welcome. I’m Jamie Forward. Today’s program is part of the Patient Empowerment Network’s Care Partner Toolkit Series focusing on the role of the care partner when a loved one is undergoing CAR T-cell therapy.   

Today, we’re joined by a myeloma specialist who works with patients and their care partners. Before we get into the discussion, please remember that this program is not a substitute for seeking medical advice. Please refer to your own healthcare team about what might be best for you. Let’s meet our guest today. 

Joining us is Dr. Adriana Rossi. Dr. Rossi, welcome. Can you please introduce yourself?   

Dr. Adriana Rossi:

Yes. Nice to be with you today. I am the director of the CAR T and Stem Cell Clinical Program at Mt. Sinai in New York.  

Jamie Forward:

Thank you so much. We’re glad to have you with us. Dr. Rossi, before we get into the role of care partners and the CAR T process, let’s talk about what CAR T-cell therapy is. Can you please give an overview of CAR T and how it works to treat myeloma?  

Dr. Adriana Rossi:

Absolutely. CAR T are genetically engineered cells. So, we generally use the patient’s own T cells, modify them to make them special killers just for that patient’s myeloma. And then, infuse them back into the patient over a process that I’m happy to go into in much more detail.  

Jamie Forward:

Sure. And, we’ll cover the process a bit later in the program. So, we can walk through that in just a bit. So, where does CAR T-cell fit into a myeloma treatment plan? 

Dr. Adriana Rossi:

Well, we originally had approvals in patients whose myeloma three or four times. But, in 2024, now the two commercially available CAR T products, one ide-cel (Abecma) and the other cilta-cel (Carvykti), are now both approved in earlier lines. So, we actually could potentially be eligible for CAR T after your first relapse. As long as you’ve had a number of therapies up front.  

Jamie Forward:

Okay. And so, when you say lines of treatment, that’s basically the number of therapies you’ve had so far? 

Dr. Adriana Rossi:

Right. The number of times the myeloma has come back. So, regardless of whether it’s one drug or three drugs together – we now often use four drugs together.  

But, we start off with a certain amount of myeloma and we treat it until it’s in remission as deep as we can. And then, we try to make that remission last as long as possible. Unfortunately, myeloma tends to eventually make its way back. That’s called the relapse. And then, you would start a new line of therapy. So, once the myeloma has come back after treatment, CAR T would be an option.  

Jamie Forward:

Okay. So, obviously a care partner is a part of this process, as is today’s focus of the program. So, can you walk us through the role of a care partner of a patient who’s receiving CAR T-cell therapy? 

Dr. Adriana Rossi:

Absolutely. And, many patients and their families will have experience with stem cells. I think the first thing to keep in mind is this is nothing like a stem cell transplant. Yes, there are cells that are collected. There’s chemotherapy and the cells are infused in a hospital setting. 

But, other than that, they are really very different experiences. And, given that’s what we would consider a long journey of CAR T through apheresis, which is the collection, then a bridging therapy while the cells are in manufacturing. Then, the hospital stay, and then the monitoring after. I think all of that is not a solo undertaking, and it really is essential to have one or more caregivers in that setting.

It’s really important to have a second set of ears at the consultation so that that amount of new information, all the big words, how things go together, meeting people is a little less overwhelming. The whole getting ready for the CAR T. There are a lot of different doctors’ appointments. We like to check that hearts and lungs are healthy. A dentist needs to check you out and make sure there’s no infection. So, just an overwhelming process. 

And, every step of the way, that’s going to be made easier if you have someone by your side.  

Jamie Forward:

Sure. It sounds like there’s a lot of coordination that takes place, as well. So, an extra set of hands is always useful there.  

Dr. Adriana Rossi:

Yeah. 

Jamie Forward:

Great. So, the care partner is a key member of the healthcare team as we established. So, who are the other members of the CAR T-cell therapy healthcare team?  

Dr. Adriana Rossi:

Yeah. It’s really important to recognize just how big that team is. We always have the CAR T physician. That one’s easy. A physician is usually supported by nurse practitioners or physician assistants and nurses that are part of again, getting all of the appointments organized. In all of this, we tend to have CAR T coordinators. Both to make sure the paperwork and the insurance side of things are done. The clinical appointments. But, it’s also important to recognize, as we were talking about, coordination. Transportation. Sometimes, patients need to stay close to a center that’s far from home. 

So, social work and all of those folks become very important. And then, there are a number of different steps with different drugs. So, our pharmacists are very important. And then, beyond that, any of the other doctors that keep our patients optimized. So, if there’s a cardiologist, a pulmonologist, an endocrinologist. All of those physicians working together. 

Jamie Forward:

Sure. As you’re preparing for the CAR T process and you’re meeting with patients and their care partners, what sort of advice do you give them about the process as you’re setting the stage?  

Dr. Adriana Rossi:

Yeah. I think it’s very important to ask questions and never think there’s a bad question, or a stupid question, or whatever. There are no limits. I know this is a completely new language, and I think it’s important even if you’ve asked it before, keep asking until it’s clear.  

And, don’t ever think you’re bothering us or anything. I’ve heard that, and it just doesn’t compute on our end. We are here to teach and support. Secondly, to take time. I think it’s really important to not think, “Oh, I’ll do this, and then I’ll run off and do something else, and then I’ll come back.” Or, have other commitments. Really allow both the patient and the caregiver protected time to be together and to just go through everything that this journey requires. And, for the caregivers to look after themselves. I think it’s really important when you’re trying to take care of someone who has the label of patient, you need to take care of yourself, as well so that you can then be of use to the process.  

Jamie Forward:

That’s great advice. So, as I mentioned, now we’re going to sort of walk through the steps of the CAR T process and what happens in each step, and how the care partner can support the patient during this time. So first, is there a consultation once a patient has been approved for this therapy?  

Dr. Adriana Rossi:

Absolutely. There are several consultations. The first one, once the patient’s identified by a referring physician, they will come and meet with myself and again, the coordinators and several members of that team to make sure that it seems like a good fit. That this is the right time, and identify any steps that we can take to really set that patient up for success.  

Jamie Forward:

Okay. And, how can the care partner participate in this meeting? Are there key questions they should be asking? 

Dr. Adriana Rossi:

Absolutely. Again, this is the beginning of the journey, and they should absolutely be there. Mostly because a lot of the information, this may be the first time they’re hearing again, the words. The concepts. The timeline. So, do ask about when things are going to be happening. As the CAR T physician, I do this all day. So, it’s very clear in my mind, but until it’s clear for them, again, ask more questions. Ask for clarification. 

Be clear on what resources are available. If there’s something that there is a question like transportation, or sequential appointments, or children in the family. All kinds of things. Really be as curious and as vocal as you’re up for. 

Jamie Forward:

Right. Arranging for childcare and pet care is probably really important during this time. 

Dr. Adriana Rossi:

Exactly. 

Jamie Forward:

What about financial planning? Is that a good time to ask about insurance and who to coordinate with there? 

Dr. Adriana Rossi:

Absolutely. Again, you will meet with social work. But, if there are specific issues that we’ve already identified, specific resources, specific paperwork, we can get that started right away.  

Jamie Forward:

Okay. Great. So then, after that, once all of that has been squared away and you’re ready to go into the CAR T-cell therapy process, there’s the T-cell collection, correct? 

Dr. Adriana Rossi:

So again, to distinguish it from stem cells, I think it’s important to know it is a one-day collection for CAR T. 

There are no injections or other preparations ahead of time. There’s no minimum number of cells that we’re aiming to get. It really is a one-day commitment to collect the cells that we collect, because they’ll be then engineered and modified before they’re ready. And so, it’s not the ordeal that sometimes you have to go through for stem cells.  

Jamie Forward:

Okay. So, the care partner should just be there during that time to be a supportive loved one. 

Dr. Adriana Rossi:

Exactly. And, it can be a long day. You’re tethered to the machine for a few hours. And, when all goes well, it is an exceedingly boring experience. So, be entertaining and be nearby. Always helpful.   

Jamie Forward:

That’s great advice. So, once the cells have been collected, can you walk through the next steps? I believe there are bridging treatments involved. Are those administered inpatient or outpatient? 

Dr. Adriana Rossi:

Absolutely. Bridging therapy is the therapy the patient receives while the cells are out being manufactured. And really, the goal there is not to get rid of myeloma. It’s just to prevent it from growing. Because myeloma that is not cared for tends to grow quite quickly. There are options to do it inpatient. To do it outpatient. There are certain therapies that would require the patients come to our center. Others that are easily given with their local oncologist. So, we really try to find something that the myeloma will be sensitive to, and that will hopefully not be too toxic, so there’s not a big recovery or a big downtime as we are preparing for the hospital stay for CAR T. 

Jamie Forward:

Okay. And, how can care partners support the patient at home during this time? I would imagine it’s sort of an anxious time. 

Dr. Adriana Rossi:

Absolutely. Many times, the bridging is something that may be familiar. Like, we’re recycling drugs they’ve seen before.  

But, these could be brand new drugs. And, I think every time you’re experiencing a new cocktail, there is some learning of how will you react, and the anxiety that can come with that, as well. There are a few times when there are delays in the cells getting ready. So, it’s not a very exact day, and that waiting period, wondering will they really come on the day they’re expected absolutely could be an anxious time. I think keeping each other company and just actively working to be your healthiest self for whenever the CAR T is ready, and knowing that working with your physicians, we are all working behind the scenes to work to the greater success hopefully is helpful.  

Jamie Forward:

Okay. That’s great. And then, finally in the process, the cells are infused back into the patient. Since this is a critical time for patients, how can care partners best be prepared to help their loved one 

Dr. Adriana Rossi:

One of the most common side effects is something called CRS. 

Which patients experience as a fever. And, I think many times in blood cancers, we really worry about fevers, because those could be infections. I think it’s important to be prepared and expect the fever so that again, it’s not oh no, what is this? We were waiting for it. It tends to come at a very scheduled time dependent on the product. So, just reassuring. Remembering yes, there are toxicities, but they are expected. Plan for them.

The medical team will have an antidote. We’ll have steps that we take depending on what comes up. And, the reason for being in the hospital is exactly to allow the medical team to respond very quickly. Most of the time, very little happens, and that is wonderful. So, if anyone is feeling bored, that is great. Celebrate it with them. No news is good news during the couple weeks in the hospital.  

Jamie Forward:

Okay. And, how long is the patient monitored for side effects in the hospital following new infusion? 

Dr. Adriana Rossi:

So, depends on the product. Ide-cel tends to have very early reactions. And so, our policy is one week for ide-cel and two weeks for cilta-cel because there, most of the side effects are around seven days in. So, we wait for the inflammation to peak and resolve. And, once it’s safe, we aim to get patients home. But, once they leave the hospital, they should for at least a few weeks be very close to the CAR T center, and usually require two to three visits a week for that close monitoring. 

Jamie Forward:

What are the short-term side effects associated with CAR T-cell therapy?  

Dr. Adriana Rossi:

Absolutely. So, the T cells are part of the immune system. Their job is to grow and expand once they’re in the patient, and pick a fight with the myeloma, which will cause a certain level of inflammation. So, some inflammation is good. But sometimes, they overdo it, and it manifests itself as a fever. We call that cytokine release syndrome. Cytokines are the molecules T cells use to communicate with other members of the immune system. So, this is part of the process we are causing, but we want to keep it in check.  

And, in the early days, we were very hesitant to do anything that could harm these precious T cells. But, we’ve learned in time that all of the antidotes, including tocilizumab-bavi (Tofidence) and steroids, don’t harm the effectiveness of the CAR T. And so, we’re very quick to intervene early and intervene with as many tools as we need. And so, that’s really become mostly just the fever. If left untreated, it can lead to low blood pressure and maybe an oxygen requirement. 

Again, usually quite easily reversible. When the inflammation happens around the brain or the nerves, we call that neurotoxicity. Specifically ICANS, which is the confusion and neurological deficits that occur with CRS. Neurotoxicity also includes other things like a peripheral neuropathy, cranial nerve palsies like Bell’s palsy has been reported quite frequently. And then, very rarely, delayed neuromuscular toxicities, which again, by patient selection are becoming more and more rare.  

And, the last is low blood counts, which we’ve touched on as part of the reason patients need such close follow-up once they leave the hospital. They’re very much at risk for infections, because they’re not making antibodies. Their neutrophils, which is the infantry type white blood cells, are low. And, their T cells are going to be low from the process. 

Jamie Forward:

Okay. And so, for a care partner, what should they be looking for? And, when should they contact a member of the healthcare team? 

Dr. Adriana Rossi:

I would say contact us anytime there is a question. It’s not too specific. Certainly, any fever. Any sign or concern for infection. And, any neurologic deficit. If someone is not acting themselves, the caregiver’s usually in the best position to recognize that. 

Jamie Forward:

Okay. And, what are the long-term side effects?  

Dr. Adriana Rossi:

Yeah. We’re still learning. Beyond a year, really there shouldn’t be many. We continue to support the patient until recovery of those antibodies, and T cells, and neutrophils. So, there’s a lot of preventive things. Monitoring and time. And, there are these rare neurological toxicities that have been reported, but they’re much less than one in 1,000. And so, it’s hard to learn or to make any generalizations at this time. 

Jamie Forward:

Okay. And, as far as monitoring at home once someone gets back home, in the weeks that follow their time in the hospital, are there certain supplies they should have? It sounds like maybe blood pressure? Perhaps a scale? 

Dr. Adriana Rossi:

Yeah. So, blood pressure and temperature probably are the two more important ones. We actually do discharge patients with a log, and for those first few weeks, we really would like at least twice a day for these numbers to be monitored. And, it’s patient-specific. So, the less you need, the more we graduate out to fewer measurements and less monitoring.  

Jamie Forward:

When it comes to diet and nutrition, are there ways that care partners can help prepare or benefit to a highly nutritious diet? Is there anything related to diet and lifestyle that might be important to know? 

Dr. Adriana Rossi:

No. I think there’s no restriction. The important thing is when your appetite is low, your body needs calories. We’re asking your body to get a lot of work done, and it can’t do that without calories. So, don’t be too picky on only eating fruits and vegetables. If it’s ice cream three times a day, go for it. Make sure you’re meeting a caloric intake. Certainly, nutrition is better. The only dietary restrictions we have are really kind of similar to after a transplant where we’re trying to avoid germs. So, foods that can be cooked, peeled, or washed are really the focus. Things like berries and salads can easily have germs sneak in. So, we do try to avoid those. And again, it’s usually just for that first month or two. Recovery tends to be quick. 

Jamie Forward:

Okay. Great. So, have a lot of ice cream on hand. So, how do you know if the treatment’s working? 

Dr. Adriana Rossi:

Well, most patients will have an M spike or light chain change. So, we can follow that by blood tests. And, as with any other therapy, it’s usually a monthly check of those numbers. 

And then, we follow the paradigm we see in stem cell transplants at around day 100 doing a bone marrow biopsy and a PET scan. 

Again, up to 90 plus percent of patients, will have a complete remission on their blood tests within a month. But, we wait until day 100 to really let that protein have time. There’s a certain time to clear from the system. Check the cells in the bone marrow and really give you full credit for all your efforts.  

Jamie Forward:

Okay. Great. So, we’ve sort of touched on this before, but I think it bears reiterating. So, why is it so important that care partners let the care team know about any changes they see in their loved one? 

Dr. Adriana Rossi:

I think early intervention really leads to success. Most of the toxicities will respond very well to an early intervention. If left untreated, be it an infection, a neurologic finding, a cell count issue, the longer it happens, the bigger of a problem it is, and the harder it would be to turn around. 

So, something that could hopefully be a quick visit to the office could then become an admission to the hospital, and we’d really like to prevent that.  

Jamie Forward:

Okay. Great. So, let’s talk a bit about self-care for care partners. I think this can obviously be a really taxing time. Why is self-compassion essential during this time when you’re caring for someone else? 

Dr. Adriana Rossi:

Because many times, again, the focus is on the patient. But really, we need to recognize it’s stressful for all of us. And, the whole medical team is taking care of the patient. Very few people are paying attention to the caregiver. So, they really need to be able to ask for help. Hopefully, again, it’s not a one-man job. It’s rally the village around the person. We do ask for those first few weeks that it’s 24 hours a day the patient be with someone. But, it doesn’t have to be one person. So, have someone else come in, so you can go exercise, or go get a cup of coffee, or just spend time dedicated to self-care. So that then you can be as strong and as present as you can for the patient. 

Jamie Forward:

And, what are signs of burnout? How can care partners recognize that? 

Dr. Adriana Rossi:

Very hard to recognize, and usually it’s someone else who needs to point it out. But, emotional exhaustion I think is the most common, because it is such an emotionally taxing time. So, having a difficult time concentrating. Being irritable or pessimistic when sometimes the medical’s team’s like, “Everything’s going great.” And still, you’re like, “No. But, it’s not going to last.” Putting a negative twist is usually part of that. You just don’t have the reserves to look forward. And then, changes in sleeping, or eating, or regular habits can also be a flag. 

Jamie Forward:

And, what advice do you have for care partners to make time for self-care? When can they find those spaces for themselves? 

Dr. Adriana Rossi:

I think the biggest thing is to not think that it’s being selfish or that you’re taking away from the partner. 

Think of it as something you are doing for the patient. You are not useful if you’re burnt out and if you’re spent. So, self-care really is a giving activity of strengthening yourself so that you can then be of most use to the patient.  

Jamie Forward:

I think that makes good sense. So, there are obviously social workers at the centers, and obviously these larger CAR T-cell therapy centers have a number of resources. So, what is available to help care partners during this time?  

Dr. Adriana Rossi:

So, social work will meet with the patient and the caregiver to tailor resources, and plans, and support in any way that is specifically useful to them. 

Again, if there is specific paperwork that needs attention. If there are resources, for example, lodging, transportation. All of these things are really tailored to the needs of each individual.  

Jamie Forward:

Okay. And obviously, this isn’t for everybody, but support groups are always a good idea, even if online. 

Dr. Adriana Rossi:

Absolutely. And, we have a number of those. We’re lucky to have a group of social workers, and they each lead different groups. So, if one doesn’t seem to be a good fit, I also think keep looking. There are very specific ones like younger patients or patients of any particular group. But, there are also general patients. There are transplant-specific. And, more and more, there are CAR T-specific groups where patients share their experience. 

Jamie Forward:

Yeah. It’s always nice to know that you’re not alone in these situations.  

Jamie Forward:

So, are there in-home services that can be useful for CAR T-cell therapy care partners during this time?  

Dr. Adriana Rossi:

I’d have to say that’s probably very specific to geographic areas. I happen to work in New York where there are a lot of home services, and it’s very population-dense, and a lot of the services are driven to that. I imagine in parts of the country where there’s quite a bit of distance between the facilities, there are probably programs that are more structured to provide those services. So, that’s probably fairly program-specific. But generally, yes. I just don’t know what they are for each part of the country. 

Jamie Forward:

Before we move on to audience questions, I’d like to add that the Patient Empowerment Network has a wealth of resources available for care partners. You can find those at powerfulpatients.org or by scanning the QR code on your screen.  

Dr. Rossi, here’s a few questions we received in advance of the program from our members. We can start with William’s question. How can a care partner manage the emotional aspects when a loved one is going through CAR T? 

Dr. Adriana Rossi:

I think be patient. Recognize that it’s a really difficult time, even when everything goes according to plan and the medical is very pleased that there’s nothing untoward. It’s just a really stressful time for both of you. So, it’s where we go back to the self-compassion, as well. Take time for yourself and recognize your needs as a caregiver in addition. So, tapping in, again, other friends. A small circle rather than a one-person job. And, being really open with social work on what resources can be helpful. Asking for help, again, is a brave act.  It’s not a sign of weakness at all. 

Jamie Forward:

Sure. And, I think it’s often that people will offer help, and you tend to decline because you think you can handle it early on. And, it is just so much easier to say yes. Say, “Yes. Bring over dinner.” Or, “Yes. I’d love you to come over for two hours while I go out and have a pedicure.” So, yeah. Always say yes when people ask you if they can help, because people want to help. 

Dr. Adriana Rossi:

Exactly. And then, it is that group activity, and it’s a shared experience. 

Jamie Forward:

Yeah. Okay. So, Marianne asks this question. She says how do the aftereffects of CAR T-cell therapy compare to those of stem cell transplant? 

Dr. Adriana Rossi:

Yes. Very different experiences. I think that’s one of my first and loudest messages. Stem cell transplants are really tough. Melphalan (Alkeran) is a very tough drug. The hair loss, the nausea, the weight loss we really do not see with CAR T.  

So, we mentioned you have to have your cells collected. You do get some chemo before getting the cells back. But, that’s as far as they are similar. The chemotherapy that you get before CAR T is called lymphodepletion. It only quiets down the T cells. It’s not a rebooting of all of the marrow the way we do with melphalan. And, the side effects are again, mostly driven by inflammation. So, fevers and neurologic deficits. Remembering that the fevers and CRS are expected in about 80 percent of patients. The neurologic side effects are in under 5 percent. So, much more rare. And, it’s usually with transplant, by day 100, if people were working before their transplant, they start to think of going back. With CAR T, I have patients who are 30 days out asking to go back to work, because they’re bored at home. You really just feel better much sooner.  

Jamie Forward:

Okay. That sounds like a pretty dramatic difference. And, here’s the last question we have from Debbie. She wants to know does the caregiver need to stay at the hospital room with the patient,  or are they only allowed during visiting hours?

Dr. Adriana Rossi:

I think that one is very specific to the center. At Mt. Sinai, we do have specific visiting hours. And, a few exceptions have been made for overnight depending on the specific circumstance. But, most of the time, that is a time the caregiver can go home, and sleep, and be ready at the time of discharge when we really do need them 24 hours.  

Jamie Forward:

Okay. That’s good to know. So, it’s center-specific. Great. So, before we end the program, I’d like to get your closing thoughts on the role of the care partner in the CAR T-cell therapy process. What message do you want to leave our care partner audience with? 

Dr. Adriana Rossi:

I think mostly to please reach out to us. 

We are there not only to take care of the patient, but the global patient experience. So, we are there to support the caregivers, as well. So, please ask questions. Many times, I’ll have had a conversation with a patient many times, and then the caregiver joins later and is hearing everything for the first time. So, please ask questions until everything is clear. And, remember to look after yourself. 

Jamie Forward:

That’s great advice. Thank you so much, Dr. Rossi. We appreciate you being here today. 

Dr. Adriana Rossi:

Thank you.  

Jamie Forward:

And, thank you to all of our collaborators. To access tools to help you become a proactive care partner, visit powerfulpatients.org. Thanks for joining us.  

Evolve | What You Should Know About Advances in CAR T-Cell Therapy for Myeloma

How is CAR T-cell therapy revolutionizing care for people with myeloma? Dr. Rahul Banerjee, a myeloma expert and researcher, shares an overview of how CAR T-cell therapy is evolving, important factors to consider when choosing to undergo this treatment, and how advances in research are impacting myeloma care. 

Dr. Rahul Banerjee is a physician and researcher specializing in multiple myeloma and an assistant professor in the Clinical Research Division at the University of Washington Fred Hutchinson Cancer Center in Seattle, WA. Learn more about Dr. Banerjee.

Download Resource Guide

Related Resources:

How Can Myeloma Patients Access CAR T-Cell Therapy Clinical Trials?

How Can Myeloma Patients Access CAR T-Cell Therapy Clinical Trials?

Will CAR T-Cell Therapy Be Approved for Earlier Lines of Myeloma Treatment

Will CAR T-Cell Therapy Be Approved for Earlier Lines of Myeloma Treatment?

How Is CAR T-Cell Therapy Research Advancing Myeloma Care?

How Is CAR T-Cell Therapy Research Advancing Myeloma Care? 

Transcript:

Katherine Banwell:

Hello and welcome. I’m your host, Katherine Banwell. Today’s program is part of the Patient Empowerment Network’s Evolve series, which was developed to provide you with the latest treatment information and research, helping you to feel empowered and confident during conversations about your care. In today’s program, we’re going to hear from an expert in the field discussing CAR T-cell therapy and the evolving treatment landscape. 

Before before we get into the discussion, please remember that this program is not a substitute for medical advice. Please refer to your healthcare team about what might be best for you. Joining us today is Dr. Rahul Banerjee. Dr. Banerjee, it’s so good to see you. Welcome. Would you please introduce yourself? 

Dr. Rahul Banerjee:

Of course. Thank you, Katherine, for taking the time to interview me. Thank you to everyone who’s listening. My name is Dr. Rahul Banerjee. I’m an Assistant Professor of Medicine at the Fred Hutchinson Cancer Center in Seattle, Washington.  

I specialize in multiple myeloma, which is a form of blood cancer that we’ll talk about, and other precursor conditions, for example, smoldering myeloma and AL amyloidosis. I have a heavy interest in CAR T therapy and bispecific antibodies and other types of immunotherapy for blood cancers, and I’m sure we’ll be speaking about that as well. It’s great to be here. 

Katherine Banwell:

And thank you so much for taking time out of your busy day. I’d like to start by talking about your role as a researcher. You’re on the front lines for advancements in the myeloma field. What led you here, and why is it important to you? 

Dr. Rahul Banerjee:

Absolutely. So, the short answer is that I love the art and the science of oncology, of cancer care. I feel like it really gives us a great chance to work with patients and really help them through the difficult chapter of their life. And the science, particularly in malignant hematology, is a word we use for blood cancers like myeloma, lymphoma, et cetera. There’s a lot of fascinating science about how do we train the immune system to overcome cancers of immune cells? 

In terms of why myeloma, you’ll probably hear this from many physicians who speak on this program. Part of it is the patients, and I love treating my patients with myeloma. They have the most interesting stories for me. Many of them are older and have been through a lot, and I worked with them for years for diagnosing myeloma, treating them through myeloma, monitoring for relapses, and so forth. 

And also the mentors whom I had. So, when I was a trainee in fellowship, I was not sure where I would go. I knew it was somewhere within blood cancers, but I had a mentor, Dr. Nina Shah, who is phenomenal, and she did a lot of work in multiple myeloma in particular, including around CAR T therapy, and I kind of molded myself, imprinted myself on her. And now several years later, here I am kind of working in this similar space, focusing again at the intersection of multiple myeloma and these immunotherapies, like CAR T therapy. 

Katherine Banwell:

We’ve been talking about CAR T-cell therapy for a number of years now. How has this treatment revolutionized care for patients? 

Dr. Rahul Banerjee:

Absolutely. So, the biggest benefit of CAR T therapy, I think, is how well it works. So, I’ll predominantly focus on multiple myeloma, but of course there are CAR T therapies that are approved for different kinds of lymphoma, and B-cell leukemia as well. But in brief, CAR T therapy, we train the immune system to fight back against the myeloma, against the cancer that’s there. And it’s phenomenal, because it’s the only living drug we have, really, in our toolbox. Everything else is dependent on the dose, right? As soon as the drug is out of your system, it stops working. CAR T-cells don’t work that way. They can live and expand and proliferate to get rid of the threat within. 

And at least in myeloma, for example, CAR T therapies regularly have response rates, meaning complete response rates, how quickly they knock all the myeloma parameters down to zero basically in the 70, 80, 90 percent range. Many patients achieve measurable residual disease, or MRD negativity, meaning by all the best tools available in 2024, no sign of the myeloma anywhere.  

That doesn’t mean cure, to be fair, but that’s wonderful. With other conventional therapies, to have one drug with one infusion have that big of an impact on the myeloma is phenomenal.  

The other benefit of CAR T therapy is that it is a one-time infusion. It doesn’t mean that patients can get one-time CAR T and never have to see a cancer doctor again because again, at least in myeloma, I don’t consider CAR T to be uniformly curative for patients. However, if you look at the studies, and there’ve been two randomized studies in myeloma of CAR T therapy versus standard therapies. One was a KarMMa-3 trial with a drug called ide-cel, a CAR T drug also known as Abecma. And the other one was CARTITUDE-4, which was a study of cilta-cel versus standard treatment. And cilta-cel is a kind of CAR T also known as Carvykti.  

Both studies saw improvements, dramatic improvements in the response rate, how many patients had the myeloma numbers come down, but also durations of response and progression-free survival. How long patients were alive and disease-free for.  

Literally just a month ago, we found out officially that the cilta-cel trial and the CARTITUDE-4 study, CAR T actually prolonged overall survival compared to standard therapies, which is what patients are looking for.  

But for me, what I love about CAR T the most is not just that you’re in remission for longer, or with cilta-cel you live longer, but that you live better. Both studies, and I’m very happy to see this incorporated, what we call patient-reported outcomes, which is an area of research of mine. Patient-reported outcomes are, as you can imagine, outcomes that are reported by the patient. Not the blood test for the myeloma, but how are patients feeling in terms of their quality of life, in terms of their fatigue, in terms of their pain.  

And in both studies, off the charts. CAR T does way better and way faster in terms of improving quality of life than standard treatments, to the point where the studies, for example, often for both of them, if I recall correctly, the first time point where they looked and compared the two arms was three months after CAR T. And already by then, night and day difference.  

The patients who got CAR T, the first month a little bit rocky, but after the first month or two, they’re doing better, because they’re not on myeloma treatments continuously. They’re still getting blood work checked and so forth, but they’re not on treatment. It’s a one-time infusion and monitoring thereafter. 

And I love that about it, and I think that explains a lot of the quality of life benefits that we see.  

For my other patients with myeloma, outside of CAR T, there is never a scenario where they’re observed. The vast majority of patients because myeloma is considered incurable, we keep them on some form of maintenance treatment, and those come with side effects. Those come with financial toxicity. That’s the word we use for high out-of-pocket costs. They come with what I would call time toxicity. That’s time spent on the phone coordinating shipments or coming in for this infusion or that infusion. 

CAR T has much less of that, and I think that’s phenomenal. So, I would say that it’s revolutionized the field, not just scientifically from the things that you’d expect a researcher to care the most about – for how long, you know, how deep are their remissions and how durable are their remissions, but also for the things that I care about. The art of oncology is how our patients are living, and that’s why I think CAR T is revolutionary. 

Katherine Banwell:

Dr. Banerjee, would you walk us through the currently approved CAR T-cell therapies for myeloma and share how these treatments work to combat myeloma? 

Dr. Rahul Banerjee:

Absolutely so. So, there’s two FDA-approved CAR T therapies right now for multiple myeloma. One is ide-cel, also known as Abecma. One is cilta-cel, also known as Carvykti. The mechanics of them are pretty similar. Both involve T-cells being collected from the patient and then turned into CAR T-cells, cancer fighting cells in a lab, then put back into the patient after a small dose, what we call lympho-depleting chemotherapy, that creates a void and makes room for the T cells. 

And then the T cells come in and are able to activate and proliferate and respond and kill off the myeloma. In terms of the two drugs, the differences between them, we might come back to that a bit in terms of just how they’re approved. Cilta-cel is currently approved as early as first relapse. So, second line treatment onwards in myeloma for patients who have disease that is refractory to lenalidomide (Revlimid) or a similar class of drugs.  

So, lenalidomide is Revlimid. So, if someone’s been on Revlimid and it stopped working, they could technically go to cilta-cel, which is Carvykti, as soon as second line. 

Ide-cel or Abecma is approved for third line treatment, meaning at least two prior relapses or two types of therapy that were stopped unexpectedly because of not working or toxicities or so forth. And those patients would have had to have received both an iMiD, like Revlimid, which is lenalidomide, a proteasome inhibitor, which is like Velcade or bortezomib, and a CD38 directed monoclonal antibody like daratumumab, which is also called Darzalex or Darzalex Faspro.   

Katherine Banwell:

CAR T therapies have been in use for several years now. As a clinician, what challenges are you facing with this treatment option? 

Dr. Rahul Banerjee:

Absolutely. So, I would say threefold, is the best way to describe it. So, I think the first and most practical, like the most important one is obviously access. It’s very easy for me to talk about CAR T therapy. I had the privilege of working at a big center, where I was just last week attending on our CAR T service. We’re big enough to have a CAR T service. 

Most patients are treated in the community, and they don’t have access to a doctor who’s personally familiar with CAR T. And so for them to get to CAR T requires a move to a big academic center, a big tertiary care center. Obviously, I think this talk is geared more towards the U.S. audience, but most of the world has no access to CAR T, period, except for research protocols, and that makes things really tough. So, I think that’s one unmet need in general, where those patients who cannot get to CAR T never see me, and that’s a big disparity in the field.  

Two, I would say that the autologous CAR T products we have, autologous is the word that we use for the T cells that are coming from the patient themselves. Both Abecma and Carvykti, again, it’s the patient’s own cells that are being taken out, turned into CAR  
T cells, and put back. That manufacturing process takes time. Typically, I tell patients to expect about one to two months, closer to two months often, between the day that the T cells are taken out and the day they’re put back in again, what we often call the quote unquote “vein-to-vein interval.” And that’s hard because for some patients that’s not practical. 

The myeloma is, you know, so aggressive, behaving aggressively, that they cannot wait. There’s no drug I can give them where I can wait two months for the T cells to be manufactured. We’re getting better at it. The drug company is working better at it. There are a lot of investigational products that are not FDA approved yet that are looking at rapid manufacturing, where can you grow these  
T cells and the CAR T cells in two days or one day, instead of several weeks? I think that’s really interesting. 

There are studies of allogeneic CAR T cells, where the T cells are pre-manufactured from a healthy donor and manipulated so they won’t cause any other problems, but will attack the myeloma. So, a lot of research happening there, but that’s the problem for patients where they cannot get to CAR T therapy.  

And then the third unmet need would be – I’m seeing more of this, unfortunately, right? CAR T therapy is not considered curative for myeloma. Have I met people who are doing great years out from myeloma? Yes, from CAR T therapy, and I love that. In the original LEGEND-2 study, the study that led to the CARTITUDE-1 study that led to the approval of cilta-cel, which is Carvykti. 

If I recall correctly, about 20 percent of patients on that first Chinese study years ago are alive and disease-free five years later. That’s not enough, right? I don’t want it to be 20 percent, I want it to be 100 percent. And so we have work to be done there in terms of what do we do when the myeloma, the CAR T cells stop working, or they’re out of the system and are no longer there to fight back. What do we do next? And I think that’s another unmet need still. Despite all the advances, what to do after CAR T, no one really knows. 

Katherine Banwell:

Yes. Well, how is success measured for CAR T-cell therapy? 

Dr. Rahul Banerjee:

It’s a great question. So, traditionally, the metric in multiple myeloma has been achieving a complete response. So, having the M-spike, the antibodies that are produced by the myeloma cells, come down to zero.  

So, either the M-spike, or for some patients, that may be the kappa or the lambda, which are fragments called light chains of antibodies, coming down to the normal range. The hard thing about those is that they take time. I’ve had patients who get CAR T therapy, and their bone marrow is completely clear, including MRD, measurable residual disease, as I’ll talk about in a second, but the M-spike takes months. 

I had one patient where it took a year and a half for the M-spike to finally come down to zero. And it’s frustrating because I would say that, look, in my heart, you’re in remission, but if I were a lawyer, I wouldn’t be able to say that, because technically the M-spike is not quite zero yet. Your body’s just recycling that antibody. It’s not all the way down to zero, and that makes things tough. So, I don’t think that’s a great barometer to use.  

I think MRD, which is again, a bone marrow test that we can spend more time talking about another day, is helpful. And at the one-month mark, achieving MRD negativity.  

So, if you look in the bone marrow, by all the best tests available in the United States in the year 2024, and you can’t even find one out of a million cells that are myeloma, that’s obviously great to see.  

That doesn’t mean that if it doesn’t happen, the patients are going to do poorly. There’s a lot more to CAR T than that. So, I think MRD negativity is probably the best short-term barometer. I think the best long-term barometer would be progression-free survival, which is, again, the medical word of saying how long is someone alive and in remission for. 

With cilta-cel, which is Carvykti, again, in the CARTITUDE-1 study, which led to its approval, its first approval for four prior lines of therapy. There, most patients, the median progression-free survival was 35 months. So, almost three years. That’s amazing, right? Three years of mileage of coming in for blood work, some supportive care, IVIG, which is a type of antibody transfusion, but not needing myeloma treatment. That’s great. That’s the bar that we’d be looking for from an efficacy perspective. I think the other thing, just to talk about it, I’m sure we’ll come back to it, would be safety, right? Because these cells, these therapies do come with side effects.  

And so I think the other bar by which, in the future, I may compare CAR T therapies as more come to market, would not just be this progression-free survival, which is how long are patients in remission for and disease-free and alive. Not just MRD negativity, which is how the bone marrow biopsy looks at day 28, one month after CAR T. But also the safety profile. Are we seeing any scary, concerning, delayed toxicities? And if we don’t, that would be another bar for success in my mind, especially as we move CAR T to earlier lines, where other treatments are available. 

We have to make sure that the benefit-risk ratio for CAR T remains favorable. 

Katherine Banwell:

Yes. Well, let’s talk about side effects of CAR T-cell therapy. What advances are being made in managing them? 

Dr. Rahul Banerjee:

Excellent question. So, I break the toxicities into short-term and long-term toxicity of CAR T therapy. And this is actually fairly similar for both, regardless of the underlying disease, whereas lymphoma, leukemia, or myeloma, very similar. Just so everyone, just to reorient the audience, short-term toxicities, people often talk about the big two. I’m going to say the big three, actually. The first one is cytokine release syndrome, or CRS, which is inflammation, typically causing fever, sometimes low blood pressure. 

The second is neurotoxicity, or ICANS. For reasons that aren’t entirely understood, sometimes all those chemicals from inflammation can cause people to feel a bit off mentally or cognitively. Sometimes people might not be able to talk, or might not talk correctly, or sometimes have issues along those lines. 

And the third, I would say, is low blood counts or infections. Both the lymphoma, leukemia CAR Ts and the myeloma CAR Ts very rapidly deplete the good plasma cells or the good lymphocytes, the good cells in the bone marrow, and so immediately you see patients at risk of infections. 

And for a complicated number of reasons, one of which being cytokine release syndrome, CRS inflammation within the bone marrow, where all these cancer cells are hiding, the stem cells in the bone marrow hide away, right? They kind of go into a bunker to stay away from all of this. And so patients often have low blood counts for significant amounts of time after CAR T therapy, something called hematotoxicity. 

Those are short-term toxicities. Long-term, very briefly, that risk of infection and low blood counts is still there, I would say, for up to a year after CAR T therapy, sometimes longer for some patients. There is a risk, obviously, of the original cancer coming back, in this case the myeloma, particularly myeloma.  

And three, there are rare delayed toxicities to be on the lookout for. So, one of them, for example, with cilta-cel or Carvykti, the numbers are hard to see what the rate is prospectively because it’s been going down with time.  

But rarely, I would truly in my heart say 1 percent of the time, if not less, patients can get Parkinsonism, where they’re not able to move as rapidly, they’re not able to, they’re kind of shuffling gait, having tremors, et cetera. Not the same as normal Parkinson’s disease, because the normal meds don’t work, just time is often the only thing that really makes a big difference. 

Technically, there’s a box warning on all CAR T therapies now about a risk of second cancers. That risk is not new to anyone who’s ever received any treatment for myeloma because from the very beginning, we tell people that these drugs have been linked to a potential risk of second cancers in the future.  

In terms of strides that are being made to improve that, I think we’re making a lot of improvement. So, I think the biggest thing that we’ve learned, and I remember when I was a trainee, for example, when I was in my medical training, the early days of CAR T therapy – actually out in Philadelphia, I trained at Penn – and there, we were scared about trying to tone down the inflammation.  

When these side effects happen in the short term, the goal is if the patient’s obviously having side effects, and the question is, “Can we not kill the T cells? Can we just dial that down?” Say, “Look, I’m happy the T cells are angry. I’m happy they’re killing the cancer, the myeloma in this case. But can you just dial it down a little bit with a medication called tocilizumab (Actemra), or corticosteroids like dex?” 

We used to be very nervous about doing that because we said, look, the patient’s put all this blood, soil, sweat, and tears right into this CAR T therapy, and we don’t want to do anything that can hinder the T cells from working.  

Now we know that that level of inflammation is not doing anyone any favors at all, and so we’re able to really start these medications to just dial down the immune system faster. As soon as someone has a fever, for example, at many centers, we do consider, within an hour or two, giving one of those medications. Don’t wait till they’re in the ICU, give it then.  So, I think just tweaking our algorithms has made probably the biggest difference, in my mind, to make CAR T safer.  

Other things that have helped, I think, are better understanding of why patients have these other toxicities and strategies to prevent it. And so, for example, the neurotoxicity risk, some of it is part of disease burden. We think that patients who have a lot of disease going into CAR T therapy may have more toxicities. So, giving better treatments as, quote-unquote, “bridging treatment” before CAR T therapy that we have better, newer treatments now, have sometimes helped to really debulk the disease before going to  
CAR T.  

That’s helped a lot with side effect management. In terms of long-term risk, the third thing that I really encourage all my patients and all my oncologist partners in the community to really push for is the infection risk and how do we prevent it? So, I think probably the biggest thing that we’ve recognized is intravenous immunoglobulin, which is IVIg, which is basically an antibody transfusion.  

When people donate blood, they also donate plasma, often, and the plasma contains antibodies against whatever they themselves have fought off circulating in the area – viruses, colds, et cetera. 

You can take all those antibodies, put them all together into a sterile bag, and give it to the patient who’s gotten CAR T therapy. Because the patient’s gotten CAR T therapy, assuming it’s working, which it normally does for several months, right, to knock out all cells, good and bad immune cells, that patient is not making any antibodies at all. They’re a sitting duck for infections. And so I would say IVIg, using that routinely now is not just the exception once they’re having infections, but even in the absence of infections, just giving it.  

Insurance companies are not happy with me when I suggest that because it’s expensive, but that’s the right thing to do for patients, and I think that has helped a lot, in my experience, for all of these immunotherapies, both CAR T and bispecific antibodies, to lower the risk of infections.  

Katherine Banwell:

Can you share any updates in CAR T-cell therapy research? 

Dr. Rahul Banerjee:

Sure. So, several. I would say the first, I alluded to very briefly earlier, was how do we make that vein-to-vein time shorter? So, the vein-to-vein is, again, from the moment that the T cells are collected to when they’re put back into the patient.  

And so a lot of research is how do we make that better? We have rapid manufacturing protocols that, again, where the T cells are taken out and manufactured within a couple of days and brought back into the patient. They’re still testing for safety and sterility and everything that needs to be done, so it’s not overnight, but still, one or two weeks’ worth of vein-to-vein time is way better than one to two months.  

The allogeneic CAR T cells that are coming from a healthy donor, those are fascinating, right? Because if the cells are pre-manufactured, there’s no risk of them not manufacturing or manufacturing in an odd manner, what we call auto-specification. They’re ready from a healthy donor, ready to go. 

And one of the studies that was presented last month at our International Myeloma Society meeting in Brazil, the time, the median time from when the patient went on the study to when they started that lympho-depleting pre-CAR T therapy was one day, and that means they got CAR T therapy within one week of going on the study. That’s phenomenal. And so I think that research is ongoing.  

There are some side effects about allogeneic, healthy donor CAR  T-cells in terms of making sure the T cells stick around and don’t cause other issues. Sorry, for another day. I think that’s one area of research.  

I think the other big area of research is how do we make CAR T-cells work for longer for more people? There are easy ways, and there are controversial ways to do that. So, I think the easy way is can we use MRD negativity or other tests to identify who is at very low risk of relapse and monitoring them appropriately.  

There are patients where, in the future, we may talk about doing some form of post-CAR T maintenance therapy. Again, the bar for that should be set pretty high, and it is set pretty high because as I mentioned earlier, many patients prefer CAR T therapy, not just because of the deeper remissions and longer remissions, but because it truly is time away from treatment. But they’re still coming in for the IVIg and the blood work and seeing a doctor, et cetera, but they’re not getting daily treatment with lenalidomide, which is Revlimid, or pomalidomide, or Pomalyst, or something like that. And that’s wonderful. 

However, there may be some patients where some form of strategy will use one of those medications or experimental medications. There’s a newer class of the lenalidomide, Revlimid, pomalidomide, Pomalyst called CELMoDs.  

They’re not approved yet, but drugs like iberdomide or mezigdomide that work better, and not just against the myeloma. They actually make T cells stronger, believe it or not. 

And so in the future, there may be scenarios where we recommend for certain patients that, hey, in your particular case, after CAR T therapy, to keep the myeloma away, I’d recommend using this form of maintenance, a pill or something like that, just low dose to, again, keep the myeloma at bay and keep the T cells, in this case, the CAR T cells strong. So, I think those are all areas of exciting research.  

And then the last thing I would say is, we have several novel CAR T therapies that are being studied that may work better and safer than the existing products. Some of them, which are in early phase studies, actually target two proteins at once. The idea, going back to one of the earlier topics I mentioned, is that if the CAR T cells see that protein BCMA, they immediately destroy the cell that’s holding that. But what if the cell learns to turn off BCMA? All of a sudden, it’s invisible to the CAR T cells, and you’re right back to starting square one again. 

And so the idea is that there are CAR T cells that are being developed that target two proteins at once, kind of what’s called origating, where if it sees this or this, it’s immediately able to attach and bind that cell.  

The idea being that it’s easy for a myeloma cell – not easy. It’s possible for a myeloma cell, just by dumb luck, bad luck for the patient, to mutate in a way that shuts off that one protein. For it to simultaneously be able to do that for two separate proteins at once, the odds are much lower.  

And so the idea with dual targeting is you may be able to knock out more cells more durably, or even knock out the myeloma precursor cells that aren’t quite myeloma cells, but are there, what we call stem cells under the hood that are still malignant? So, a lot of those areas, I think, are really fascinating. Obviously, we need a lot more research in those particular areas before they’re ready for prime time. 

Katherine Banwell:

Yes. What is GPRC5D? 

Dr. Rahul Banerjee:

Great. Yes, so it’s a mouthful, is the best way to describe it. It’s an interesting protein. It’s a protein that, I alluded to that, it’s a second target that’s being targeted by some CAR T cells, for example, either in addition to BCMA or by itself. Several companies are looking at that in trials.  

GPRC5D is interesting because we don’t know what it’s for. BCMA, we know exactly what it’s for and why good and bad plasma cells, again, the bad plasma cells being the myeloma, why they express it. No one knows for GPRC5D.  

In the field, we’ve put almost all of our myeloma eggs into the BCMA basket, historically speaking, and all the approved immunotherapies, CAR T and bispecifics in myeloma, with one exception, which is talquetamab or TALVEY, all target BCMA. So, we need to not put all our eggs in that basket. And so GPRC5D targeted therapies, again, there was one approved bispecific antibody, which was not the focus of today’s talk, called talquetamab or TALVEY.   

There are investigational CAR T products that target GPRC5D as well. At least from the limited experience that we have, there seems to be no correlation between them.  

In the future, do we know that someone needs to get BCMA therapy first and then GPRC5D therapy later? We don’t know that, actually, and that’s one of our areas of research and one of my areas of research as well. What if we do it the other way around, right? What if we give GPRC5D first and then do BCMA? We don’t know. 

So, I think one of the big questions in the field is sequencing, which is the word of like, what drug do we use first and when? We’re working on a paper, the International Myeloma Working Group is working on a big, extensive paper to try to put all the evidence together, but that’ll be a moving target.  

Katherine Banwell:

Yes. Patient participation is essential in advancing myeloma research. How do clinical trials impact care? 

Dr. Rahul Banerjee:

Absolutely. So, phenomenally and importantly, I think it’s a short answer. It’s worth noting that clinical trials come in all shapes and sizes. People often assume that clinical trial means, by default, a Phase I study, first time in human being, a quote-unquote “guinea pig.” That’s true for a minority of studies, and that’s very important for us to understand how best to make the drug work better. I would say the vast majority of trials that I put my patients on are not like that. They’re often bigger Phase II or Phase III studies. 

As an example, you know, both Abecma and Carvykti, those were already approved in later lines, but to get them approved in earlier lines, we had to run a study of using them earlier versus not using them earlier. So, that’s a good example where the drug is FDA-approved, it’s just the sequencing of it that’s new.  

There are trials of supportive care. We’ll be opening a study, I alluded to this, the side effects of GPRC5D targeted therapies with taquetamab. We’ll be opening a study that randomizes patients to one of four different supportive care strategies to figure out which one actually works to make the taste issues better. Because we don’t know until we try, right? If we don’t do a rigorous study, and we just go by, “Oh, I had one patient once where this worked, and one patient once where this worked,” that’s not a scientific way of answering questions, and we’re not really able to advance the field to help all patients.  

So, that’s where I think clinical trials come in handy. That, and I alluded to all these newer investigational CAR T therapies that might actually work better and be safer than the existing one. They’re all coming through investigational trials.  

So, trials is kind of how we get these drugs, one, these newer ones to market, but also how we learn to make them better. And we have trials, all sorts of trials, looking at all sorts of, again, not just new drugs, but also where to put the drugs, right? Where to sequence the CAR T therapies, or what supportive care strategies do we use? And the trials are kind of the linchpin of making the field work better. Again, not just for some patients who happen to do well, but for all patients. The only reason we’ve found out what works for all patients is by doing clinical trials.  

Katherine Banwell:

I’d like to get to a few questions that we received from our audience members prior to the program.  

Dr. Rahul Banerjee:

Yes, of course. 

Katherine Banwell:

This one is from Jennifer. Why do CAR T-cell transplants not last longer? Is it possible to do a second transplant if the first CAR T transplant stops working?  

Dr. Rahul Banerjee:

It’s a great question. So, it’s an excellent question. So, the only thing I would say semantically is, right, in my head, CAR T cells are not transplants per se, because I’m not changing the bone marrow. So, I would transplant a stem cell transplantation, separate from CAR T.   

Why do CAR T cells stop working? So, the short answer is we can speculate. We don’t know for sure for any individual patient. Three different buckets are involved. Three different things can happen. One, the T cells can stop working or disappear from circulation. So, that’s possible. CAR T cells don’t last forever. That’s actually okay, right? People often wonder like, “Man, I wish the CAR  
T cells could last forever.” I don’t know that I’d want that because as I alluded to, for as long as the CAR T cells are there, patients are immunocompromised, and so that can certainly interfere with the quality of life.  

So, it’s not necessarily true that the CAR T cells need to be there forever, but if they’re not there, or if they’re exhausted, which is actually a true scientific word for them not being able to activate and kill that cell when they recognized a protein, the BCMA, that’s one problem.  

The second problem is the myeloma cells can mutate. I alluded to this briefly earlier, where the cells can learn to shut off the protein, BCMA, or they can mutate the protein in just a way that the CAR T is no longer able to bind.  

And the third is something called the tumor microenvironment. And this is a little bit more complicated, kind of a grab bag of different things here. The idea is that myeloma cells have a lot of tricks, and they can use all of the cells around them to make it hard for the CAR T cells to get in, to get into the bone marrow and kill them all. And the T cells can be shut off before they even get there.  

So, it’s one of those three things in general. Which one is it for an individual patient? Hard to say. And so hopefully in the future, we’ll have better diagnostic tests to be able to identify who is a patient where another BCMA targeted therapy would work well versus, “Oh no, these myeloma cells are no longer expressing BCMA, let’s move on to a different target like GPRC5D.” We’re not there yet. We’ll get there hopefully in a couple of years, is my goal.  

Then, you know, Jennifer, that’s a good question. Well, can we do a second CAR T? And that’s very practical. What I would say is if the first CAR T therapy did what we expected it to do, if it lasted for as many years as I would expect for Abecma, that’s typically 12 to 18 months. For Carvykti, that’s typically over 24 months.  

If it worked and then it stopped working, and then probably the T-cells are long gone, it’s reasonable to try CAR T therapy again. In general, I would recommend, I strongly recommend a different CAR T-cell therapy, even if it’s targeting the same class, like BCMA, it’s going from Abecma to Carvykti or vice versa. 

The risk with giving the same CAR T-cell product again is that even though the T cells are a patient’s own T cells, the protein is slightly foreign. Abecma was derived from decades of mouse research. Carvykti was made from decades of llama research, believe it or not. Again, there’s no mouse or llama involved with the actual products nowadays, but in making the sequence years ago that came to them, they are foreign. There are foreign sequences on them, and so everyone’s host immune system eventually recognizes these cells as foreign.  

And so if you were to give the exact same product again, immediately the body would reject it the second time around, because it recognizes them and has learned to recognize it as foreign. But changing to a different CAR T cell is very reasonable. 

And again, for these newer GPRC5D targeted CAR T cells that, again, don’t target BCMA the way that Abecma, ide-cel or cilta-cel, Carvykti do, but target a different protein entirely, for those patients generally there’s no restriction on whether they’d received a prior CAR T cells. Ideally, it should be at least several months away, at least like a year or so, but if that’s happened and they stopped working, very reasonable. In fact, some of the trials actually require that patients going on to the study of a GPRC5D product have had a prior BCMA therapy of some sort before, and so they’re kind of built into the architecture of things. So, I think it’s very reasonable.  

Obviously, there are some unknown unknowns, right? What do you do if the T cells are being manipulated twice, so to speak. Patients ask me about that. I will say just to put that fear to rest, in general, by the time that someone’s had myeloma come back after the first CAR T cells, I alluded to this, the CAR T cells are long gone. So, there’s nothing left. But again, every case is different, and future research will help us kind of figure out how best to do this. 

Katherine Banwell:

Yes. We have one more question from Rita. She wants to know if there is an age limit on CAR T-cell therapy. 

Dr. Rahul Banerjee:

It’s a brilliant question. So, in brief, no. It’s a short answer. My oldest patient who got the CAR T is in their 80s and handled it across all my years at UCSF and here at Fred Hutch. More important than chronological age is physiologic age in terms of how robust they are, how robust they feel in terms of going through this. What I recommend, I’ve heard of centers that do CAR T therapy for patients in their 90s, for example. So, I think it’s not so much age, it’s how fit they are. 

Fit is easy for me to say. What does that actually mean? Typically it’s a combination of, you know, like cognitively, how are people doing? Because obviously if someone has baseline dementia, which is pretty uncommon, for example, maybe CAR T is not the best use of their time. The biggest thing that I think gets some of my older patients in trouble is frailty.  

So, just not being as fit as they were before in terms of muscle strength, being able to move around quickly, and so forth, or cardiovascular issues. Because it’s worth noting that with CAR T therapies that we have now, probably 60, 70 percent of patients will get CRS, cytokine release syndrome. So, that’s fevers and low blood pressure, that’s what I said earlier.  

But if you think about it, a fever of 104 degrees Fahrenheit, your heart rate’s going to be like 130, 140 beats per minute, beating very fast for several hours, sometimes longer than that. If you’re having low blood pressure, it’s your heart that has to push blood to overcome that low blood pressure. Can someone’s heart handle that level of stress? As people get older, that obviously gets harder.  

So, if someone is already just 75 and were being considered for CAR T therapy, certainly I think it’s very reasonable to talk about it. I would ask someone where I may have them meet with a cardiologist beforehand, possibly a geriatrician as well beforehand, just to see how can we optimize their health to make CAR T as safe as possible for them? So, it’s definitely possible. 

Katherine Banwell:

Well, thank you for that, Dr. Banerjee. And those were all great questions. If you have more as part of the audience, please continue to send them to question@powerfulpatients.org, and we’ll work to get them answered on future programs.  

Dr. Rahul Banerjee:

Absolutely. 

Katherine Banwell:

Well, Dr. Banerjee, we learned that the field of myeloma care and CAR T-cell therapy is advancing quickly. As we close out the program, what are you excited about? Why are you hopeful? 

Dr. Rahul Banerjee:

Absolutely. So, I think I’m excited because, especially for CAR T therapy in particular, at our center, for example, we used to only be able to do like one or two patients per month. It was a very steady, low stream, because there were a lot of issues with manufacturing, and it was during the COVID pandemic, and there were supply chain issues, et cetera. 

And there were studies, and I remember them, very dark studies in 2021, 2022 even, that patients were more likely to die on the  
CAR T waitlist than they were to actually get CAR T therapy. That’s terrible. And we’ve come a long way since there, where our capacity, every year we’re doing more and more patients to CAR T therapy per month, which is wonderful. 

And so what I would say is that makes me hopeful, right? Because CAR T therapy, I’m not saying that everyone is required to get CAR T therapy. I have plenty of patients who hear about it and are like, oh, no thanks. I’d rather stay with what I’m getting right now with my local oncologist. And that’s okay, but I want it to be an option for as many patients as possible, and I think we’re making strides there. 

And so I think what I would say, the kind of closing words here is, we only can get you CAR T therapy if we know about your case and if you know about us. And so what I would say, I’m sure to people listening to this, you’re very motivated. And so you know the importance of having a myeloma specialist in your field. 

What I would say is my patients who do the best are the ones who are co-managed. They have a doc out in the community. I have patients from Idaho, Montana, Alaska, Eastern Washington, et cetera. They have an oncologist who knows them well, who knows their community well, easy to get to, can handle everything. They’re a jack of all trades and very good at it. And then they have me, who they see periodically for CAR T evaluation, just discussions about CAR T by telehealth, something along those lines. 

And I’m able to talk about CAR T therapy in detail and answer their questions. And so what I would say is, I’m hopeful that that trend will continue, and that if someone’s even interested in hearing about CAR T therapy, they don’t meet me for the first time when they actually need CAR T therapy right then and there. They meet me years beforehand to just talk about CAR T, so they know it’s an option for them. 

And that time – because we don’t have as long of a wait list as we do, and we used to do, if and when the time comes that we all talk, me, the patient, the caregiver, and the primary oncologist, and say, “Look, I think now is time,” we’re able to make it happen. 

Katherine Banwell:

Yes. That’s a promising outlook to leave our audience with, Dr. Banerjee. Thank you so much for joining us today. 

Dr. Rahul Banerjee:

Absolutely. The pleasure was all mine. Looking forward to seeing many of you on a future episode of this. 

Katherine Banwell:

And thank you to all of our collaborators. To learn more about CAR T-cell therapy and to access tools to help you become a proactive patient, visit powerfulpatients.org.  

I’m Katherine Banwell.

Thanks so much for being with us today.  

Myeloma Care Partners | Understanding Bispecific Antibody Therapy

What is bispecific antibody therapy? Dr. Craig Cole, a myeloma specialist, explains how bispecific antibody therapy works to kill myeloma cells, how the treatment is administered, and which patient type the therapy is most appropriate for.

Dr. Craig Cole is a multiple myeloma specialist at Karmanos Cancer Institute in Detroit, MI and in East Lansing, MI. Dr. Cole also serves as an associate professor at Wayne State University and at Michigan State University. Learn more about Dr. Craig Cole

See More from The Care Partner Toolkit: Bispecific Antibodies

Related Resources:

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

What Myeloma Care Partners Should Know About Bispecific Antibody Side Effects

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Proactive Steps for Supporting Your Loved One Through Bispecific Antibody Therapy

Bispecific Antibody Therapy | The Important Role of Care Partners

Bispecific Antibody Therapy | The Important Role of Care Partners 

Transcript:

Katherine Banwell:

Dr. Cole, let’s start with some basics. What is bispecific antibody therapy? And who is it right for? 

Dr. Craig Cole:

Yeah, in cancer medicine kind of to describe bispecific antibodies we need to really start with what T cell is.  

Because in cancer medicine the – really all of the bispecific antibodies engage T cells.   

So, T cells are a cell that’s in our bodies which help destroy cancer cells naturally. And so, the T cells, when we have any mutations in any of the cells in our body and it starts to become cancerous, the T cells come in and wipe it out before it even gets started. And so, part of the reason that people get cancer is that those cancer cells find a way to evade the T cells. And usually what they do is they hide. They’re able to masquerade as normal cells, and the T cells that should destroy them just slide right over them or check their ID and say, “Well, you’re okay,” and let them go.  

Then the cancer cells can grow. And so, what the bispecific antibodies do is that a regular antibody is shaped like a Y, and usually both ends are really sticky to stick to anything, usually bacteria, viruses. And that’s the antibody – is the way our immune system fights infection. And antibodies are sticky. They got two sticky ends. What they’re able to do in the laboratory is make one of the sticky ends to an antibody not produced by people but produced a laboratory. One sticky end is specific to the T cell. One sticky end is specific to the cancer cell. And when you give this drug, it brings the T cells that have been ignoring the cancer right up against the cancer cells. And so, all of a sudden, the T cells that destroy cancer that have been ignoring the cancer cells are suddenly made aware of the cancer cells.  

And as soon as they see those cancer cells, they begin to kill the cancer cells. And so, it brings the cancer hunting T cells together with the cancer cell so the T cells can destroy the cancer.  

Katherine Banwell:

Okay.  

Dr. Craig Cole:

And who is it right for? Most, if not all, of the bispecific antibodies that are approved now are for people that that have cancer that’s advanced, that has failed several therapies. And that’s the usual place where new drugs go is for the people who are most in need, the people who have exhausted a lot of other options. And so really it’s right for anyone who has advanced cancer, who needs new therapeutic options. 

Katherine Banwell:

How is this therapy administered and what is the frequency? 

Dr. Craig Cole:

Yeah, so usually for most by bispecific antibodies, they’re administered subcutaneously under the skin, and some are administered IV.  

Some are administered over long periods of time where people go home with infusion packs, and they get it over several days. And some of them are given once a week or every two weeks. And so, it really depends on what type of tumor is being – what the bispecific it is being used for and which tumor is directed towards. 

Does Diet Adjustment Impact Cancer Care?

 

How is cancer care impacted by diet adjustments? Oncology dietitian Dr. Cynthia Thomson from the University of Arizona discusses potential impacts of strategies for symptom management, immune response, nutrient deficiencies, inflammation, and diet patterns. 

Download Resource Guide

See More from RESTORE

Related Resources:

Age-Informed Nutrition and Intervention Strategies for Cancer Patients Across All Ages

Age-Informed Nutrition and Intervention Strategies for Cancer Patients Across All Ages

Nutritional Essentials: Supporting Behavior Change During Cancer Treatment

Nutritional Essentials: Supporting Behavior Change During Cancer Treatment

Transcript:

Lisa Hatfield:

How can adjusting your diet support you through your cancer journey? I’m excited to connect with a top expert to help us find the answers on this Patient Empowerment Network RESTORE program.  

Dr. Thomson, what are the key components of nutrition that patients should be thinking during their cancer treatment? 

Dr. Cynthia Thomson:

I think to start with, people need to think about what kind of symptoms they might be having during treatment and how can nutrition help them manage symptoms because I think symptom management is probably the most central nutritional issue in terms of making sure people can get through their cancer treatment in a timely manner that aligns with the prescription and optimizes their outcome after treatment. So I think managing those symptoms is one of the key components.

The second is to really do an assessment of what you’re eating and the quality of that diet. Are there nutrient deficiencies that need to be addressed and really honing in on specific nutrients or eating patterns that need to be evaluated and addressed. And then I think the third is really to move towards that long-term cancer survivorship eating plan. Where we’re really promoting a diet that’s more plant-based, a diet that is going to help reduce comorbidities, a diet that’s going to help you have the highest level of vigor and vitality after treatment. And so it’s kind of a continuum from symptom management, address those deficiencies and inadequacies in the diet, and then let’s go after optimization to reduce any risk of cancer recurrence as well as keep us healthy overall.

Lisa Hatfield:

Okay, thank you. And, Dr. Thomson, can you speak to the connection, if there is one, between the immune system and blood type and its impact on diet for cancer patients? And is this a consideration during care for you and your colleagues while supporting patients through their cancer journey? 

Dr. Cynthia Thomson:

Yeah, so I do think there are a number of different, what we call fad diets, like the blood type diet, that are intriguing. Could our blood type actually dictate what diet we should be on and what diet is optimal for our health? I would say that there’s just a total lack of evidence for the blood type specifically. And so will research evolve in the future? I don’t know. I think right now we need a lot more preliminary evidence that this matters, and this is an important factor before we would move forward.

In terms of the immune system, I think this is where diet is critical, as well as physical activity. There is a lot of evidence that when we get active and when we eat certain foods, we can modulate how our immune system responds. And in fact, our immune system is the checkpoint, to make sure cancer does not develop in our bodies or that when it does develop, we can counter it.

I think it’s really important to think about what we eat and our physical activity to enhance our immune system. And I’ll give you, One of the more common examples is adequacy of vitamin C and making sure that we have adequate vitamin C for our immune system to respond. We also know that people who are deficient in vitamin D can have lower immune response. It can happen with other nutrients as well, but on the other side, we have things like inflammation, which is a hallmark of cancer. And so If we can select dietary components that are anti-inflammatory that reduce the inflammatory response within our bodies, then that also can help to modulate our cancer risk. So I think this connection between lifestyle behaviors and the immune system is critical. It is the primary mechanism by which these health behaviors protect us and keep us healthy.

Lisa Hatfield:

Okay, thank you. I do have a follow-up question. As a cancer patient myself, and I know you’re a cancer survivor also, so we hear a lot about different diets, the blood type diet we just talked about, and then things like the keto diet, eat more protein, eat less protein, eat plant-based, importance of the health of the gut microbiome. Is there any evidence that any one type of diet works better than another when it comes to not just cancer prevention, but maintaining where we’re at with our cancer, whether it’s curing it or maintaining, stabilizing the cancer when it’s not curable? Do you have any comments on that? 

Dr. Cynthia Thomson:

Yeah, I mean, I think, first of all, there is a lack of randomized controlled trials, what we consider the gold standard, right? So, in terms of, can I say to you, we have 18 studies that say, the Mediterranean diet is the way to go. We don’t have that. What we do have is a lot of what we call epidemiological data.

So when we look at the diets of individuals who are either prior to their diagnosis of cancer, during their treatment, or after their treatment, we have increasing evidence that people who eat certain diet patterns, like a Mediterranean diet, like an anti-inflammatory diet, or score high on what we call the healthy eating index, where the quality of the diet is very plant-based, lower in fat, lower in processed foods, particularly processed meats. When those patterns are followed, we see better outcomes in terms of survival, but we also see that we can modulate certain inflammatory responses, oxidative stress, et cetera, that lead to cancer. So the mechanisms are there as well.

Lisa Hatfield:

Is there any evidence that there are cancer superfoods that might help stop the cancer or prevent cancer? I hear about blueberries being a superfood. Can you name any superfoods, or is there evidence to support that? 

Dr. Cynthia Thomson:

Well, I think we all have our own superfoods, right? Broccoli, because it’s a cruciferous vegetable, or garlic and onion, which are known to enhance immune response against viral infections, or, on and on and on omega-3 fatty acids. And it could go on and on. So I always hate to say that any one food is going to prevent cancer. That is so unlikely. If there is one, it must be hidden somewhere. I just can’t imagine and so it really and increasingly we know this.

It’s a diet pattern, right? Like if you are, Lisa, putting blueberries on your cereal every morning, but that cereal is Cap’n Crunch or some other sugared cereal versus something that’s higher in fiber, like steel-cut oats or, you know. So I feel like we cannot look at foods in isolation because while one food may have a beneficial effect, we can easily counter that by making other choices that are not so healthy. And so we can eat this anti-inflammatory food and then counter it with a pro-inflammatory food. So it’s really the pattern of eating that we need to think about.

Lisa Hatfield:

Okay, thank you. You heard it here directly from the expert. Thanks for joining this RESTORE program. I’m your host, Lisa Hatfield. 

Overall Health and CAR T-Cell Therapy | Tips for Preparation and Recovery

 

How can you best prepare to undergo CAR T-cell therapy to aid in optimal recovery? This animated explainer video provides key advice for learning about CAR T-cell therapy, consulting with your care team members, and tips for recuperating after the process. 

See More From Thrive CAR T-Cell Therapy

Related Resources:

Planning for CAR T-Cell Therapy | Advice for Myeloma Patients

Planning for CAR T-Cell Therapy | Advice for Myeloma Patients

Current and Emerging CAR T-Cell Therapies for Myeloma

Current and Emerging CAR T-Cell Therapies for Myeloma

CAR T-Cell Therapy | Key Considerations for Myeloma Patients

CAR T-Cell Therapy | Key Considerations for Myeloma Patients

Transcript:

CAR T-cell therapy offers a groundbreaking approach for people living with myeloma, and taking steps to optimize your health can play a crucial role in your treatment journey. From preparing your body and mind before therapy to focusing on recovery afterward, there are actionable ways to support your overall well-being and, potentially, enhance outcomes. 

Here are some key steps to boosting your overall health when preparing to undergo CAR T-cell therapy: 

Start by learning about CAR T-cell therapy.

Take the time to understand how the treatment works and what to expect. Your care team can guide you through the process, from the collection of T cells to potential side effects and what to expect following therapy. Educational resources like those found on the Patient Empowerment Network website can also empower you with knowledge and confidence.  

Next, consider cost.

Confirm insurance coverage and make sure you understand the financial impact of CAR T-cell therapy. You can also meet with a financial counselor or a navigator at your medical center to see if there are any resources to assist with paying for therapy. 

Then, consult with your CAR T-cell therapy team.

When undergoing pre-treatment evaluation, be sure to get all of your questions answered and to understand what support will be available to you during the CAR T-cell therapy process.  

You should also build a support system.

Having a family member or friend who can accompany you to appointments and assist with your recovery is vital, and often required by the CAR T-cell therapy center. A care partner can be an advocate for you and help to ensure you feel supported throughout the process. 

It’s also important to plan ahead.

Coordinate with your employer for the time you’ll need to take off from work. And, if necessary, arrange for child or pet care so that you won’t have to worry about these logistics following treatment.  

And, last but not least, meet with other care team members:  

Consider a consultation with a nutritionist for advice on a diet that supports your body through the CAR T process, as well as safe handling tips for meals following treatment.  

A social worker can help you manage the emotional, logistical, and financial aspects of CAR T-cell therapy.   

And, meeting with a pharmacist may also be useful, as they can provide specific information about medications you will take before, during, and after treatment.  

After CAR T-cell therapy, maintaining your health is essential to boost recovery and to reduce potential side effects or complications. Here are some useful tips to aid in recovery: 

Focus on Nutrition.

Your body will need extra support as it heals. A balanced diet rich in vitamins and minerals can help boost your immune system.  

Stay Active.

Light exercise, such as walking or yoga, can help improve your strength and mental well-being. Consult with your doctor before starting any exercise routine.  

Monitor Your Mental Health.

Emotional health is just as important as physical health during recovery. Reach out for support if you’re feeling overwhelmed or anxious.  

Stay on Top of Follow-Up Appointments.

After therapy, your healthcare team will monitor your progress. Attend all follow-up appointments and keep track of your symptoms. If you notice anything unusual, contact your doctor immediately.  

CAR T-cell therapy is a powerful treatment and taking steps to prepare and care for yourself can make a significant difference in your recovery.

For more information and additional resources, visit powerfulpatients.org.

RESTORE Wellness Kitchen | Tomato Salad Sandwich

In this RESTORE video featuring a tomato salad sandwich recipe, explore the potential health benefits of ingredients such as tomatoes, olive oil, basil and how they may help support immune function, reduce inflammation, and contribute to overall well-being.

Download Resource Guide

See More from RESTORE

Related Resources:

How Art’s Healing Abilities Can Help Those Touched by Cancer

How Art’s Healing Abilities Can Help Those Touched by Cancer

What Is Whole Person Care Exactly?

Why Is It Important to Address Whole Person Care?

Why Is It Important to Address Whole Person Care?

Transcript:

Lisa Hatfield:

Welcome to our RESTORE kitchen! Today, we’re making a delicious and refreshing tomato salad sandwich.

Start by slicing vibrant orange and red tomatoes. Tomatoes are rich in vitamins A and C, which support immune function and overall health. Arrange the slices on a plate and drizzle with your favorite olive oil based Italian dressing (and maybe a little balsamic vinegar). The healthy fats from olive oil help absorb fat-soluble vitamins and antioxidants from the tomatoes.

Add freshly chopped basil for that perfect burst of flavor. Basil has antioxidants and anti-inflammatory compounds, which can help reduce oxidative stress and support the immune system.

Next, sprinkle Parmesan cheese over the tomatoes. Parmesan is high in calcium and protein, essential for maintaining bone health and muscle mass.

Lastly, take two slices of toasted whole wheat bread, which is a source of fiber and can help aid in digestion. Place the dressed tomatoes on one slice and top it with the other slice of bread, and there you have it – a simple yet nutrient-packed tomato salad sandwich. Enjoy!

What Are Common Follicular Lymphoma Treatment Side Effects?

What might follicular lymphoma patients experience for treatment side effects? Expert Dr. Brad Kahl from Washington University School of Medicine discusses common treatment side effects that patients might experience, some methods for dealing with side effects, and other precautions to help ensure optimal patient care. 

Download Resource Guide | Descargar Guía

See More from START HERE Follicular Lymphoma

Related Resources:

Defining Survivorship for Relapsed/Refractory Follicular Lymphoma

Defining Survivorship for Relapsed/Refractory Follicular Lymphoma

Addressing Vulnerabilities in Follicular Lymphoma

Addressing Vulnerabilities in Follicular Lymphoma

Why Does Follicular Lymphoma Relapse for Some Patients?

Why Does Follicular Lymphoma Relapse for Some Patients?


Transcript:

Lisa Hatfield:

What are the common side effects of the recommended treatments in newer therapies? And a really important question. Are there long-term side effects that I should be aware of or that a patient should be aware of?

Dr. Brad Kahl:

Yeah, the side effects are going to be different for all the different new agents that I mentioned. With the bispecific monoclonal antibodies, there’s a little bit of risk for something called cytokine release syndrome. When you’re first starting on the drug, sometimes the drugs are really potent at activating the patient’s immune system. And as that immune system is getting revved up, the immune system will release chemicals or cytokines, which can give you fevers and make you feel like you have the flu. It’s just your immune system responding.

And so that’s something that we have to watch for as we’re starting a bispecific, that’s usually a short-term problem. And it’s usually pretty easily managed with steroids or other drugs that can tamp down the immune system. And then once you’re past that risk for cytokine release syndrome, the bispecifics usually go pretty smooth, but the bispecifics do deplete your body of healthy B cells in addition to the follicular lymphoma cells.

So they do weaken the patient’s immune system some, and I’d say that’s the biggest risk that we have to worry about in patients getting a lot of these different treatments is just what it does to your immune system. And so we’re always telling patients to call us if you get a fever, infections in a patient on treatment can become a big deal. And that’s why we want those phone calls so we can figure out if you need to get seen, if you need to go to an emergency room, if we need to start on broad spectrum antibiotics immediately, if we need to bring in for fancy testing, because sometimes people can get kind of oddball or rare infections. 

So infections, infections, infections are the things we worry about the most with most of the treatments that we give to people with relapsed follicular lymphoma. That’s true of the CAR T-cell products, cytokine release syndrome. We also have to worry about some neurologic toxicity that can happen if that happens, that’s going to occur while the patient’s in the hospital with us getting those treatments. But again, these drugs will deplete the immune system for months and months, maybe even a year, maybe longer. So we have to just be super careful about infections in patients getting these different treatments that I mentioned today.


Share Your Feedback

Follicular Lymphoma Patient Expert Q&A: Dr. Brad Kahl

 

Dr. Brad Kahl from Washington University School of Medicine explores the transformative potential of emerging therapies for follicular lymphoma and their significance for patients and families. He also addresses the unique challenges of living with follicular lymphoma and its impact on patients’ lives today.

Download Resource Guide | Descargar Guía

See More from START HERE Follicular Lymphoma

Related Resources:

What’s the News on Follicular Lymphoma and Bispecific Antibodies

What Should Follicular Lymphoma Patients Know About Remission

What Can Follicular Lymphoma Patients Expect With Remission


Transcript:

Lisa Hatfield:

Welcome to this START HERE Patient Empowerment Network program. This program bridges the expert and patient voice, enabling patients and care partners to feel comfortable asking questions of their health care team. Joining me today is hematologist-oncologist Dr. Brad Kahl, Professor of Medicine in the Division of Oncology at the Washington University School of Medicine and Director of the lymphoma program at the Alvin J. Siteman Cancer Center in St. Louis, Missouri. Thank you so much for joining us, Dr. Kahl.

Dr. Brad Kahl:

It’s a pleasure. Thanks for having me, Lisa.

Lisa Hatfield:  

The world is complicated, but understanding your follicular lymphoma diagnosis and treatment options along your journey doesn’t have to be. The goal of Start Here is to create actionable pathways for getting the most out of your follicular lymphoma treatment and survivorship. No matter where you are on your journey, this program is designed to provide easy to understand, reliable, and digestible information to help you make informed decisions. And most of all, we’re asking questions from you. I’m thrilled you’ve joined us.

Please remember to download the program resource guide via the QR code. There is great information there that will be useful during this program and after. Let’s start here. Dr. Kahl, there is a great deal going on in the follicular lymphoma landscape, and I want to dig into that. But before we do, as is custom for this program, I’d like to start with a brief overview of this disease. What is follicular lymphoma? And can you break it down a little bit, the key differences between Hodgkin and non-Hodgkin lymphoma and how follicular lymphoma fits into that?

Dr. Brad Kahl:

Sure. The terminology can be kind of confusing to patients, so I’ll try to explain it. Hodgkin lymphoma is a specific kind of lymphoma. Non-Hodgkin’s lymphoma just means it’s not Hodgkin’s. So non-Hodgkin’s lymphoma is just a big, broad, descriptive term. It’s like saying automobile. But there are lots of different kinds of cars, obviously. So follicular lymphoma is a specific type of non-Hodgkin’s lymphoma. So it’d be like saying Chevy Malibu or something specific within that automobile term. So there’s like 100 different kinds of non-Hodgkin lymphoma. Follicular lymphoma is one of those. A

nd it’s kind of a unique answer biologically and clinically. Follicular lymphoma is characterized by this particular mutation inside the cells that sends a signal to the cells that says don’t die. So instead of being a disease of rapid cellular proliferation and growth, it’s more of a disease of slow cellular accumulation. If people can picture that, the cells are just accumulating slowly. So it’s kind of a slow-moving cancer. And probably when patients are diagnosed, they’ve probably had it for a long time already.

They just didn’t know it, because follicular lymphoma often doesn’t cause symptoms. And usually when we get a patient with newly diagnosed follicular lymphoma, the disease is very widespread. And that obviously makes people fearful. And so we spend a lot of time trying to reassure them that’s not a problem that’s typical for follicular lymphoma. Everybody wants to know their stage, of course. And I try to tell them, the stage doesn’t really matter that much in follicular lymphoma. In some cancers, the stage is a big deal. But those are cancers that you can kind of remove surgically.

But there’s really no role for surgery as a treatment in follicular lymphoma. The disease is typically very widespread in diagnosis, meaning it’s all over the body. And so when we do treat it, we pick treatments that will work everywhere. And our treatments tend to work just as well when the disease is at a more advanced stage. That’s why as the doctors, we don’t spend too much time worrying about the stage. It’s just not, it’s not as important in follicular lymphoma.

Lisa Hatfield:

Okay. Thank you. And just to clarify, when you mentioned that there is a mutation or often mutations in follicular lymphoma, is that in the cancer cells themselves, or is that in a mutation, like a BRCA mutation that a patient can be tested for? I presume it is.

Dr. Brad Kahl:

Right. That’s a great question. The mutation is specific to the cancer cells. So people are not born with this mutation. It’s not a mutation that you pass along in your family to children. It’s a mutation that is acquired in these cells at some point in the patient’s lifetime. Another confusing term is this whole idea of B-cell lymphoma or T-cell lymphoma.

And just to try to clarify that. So we have different kinds of lymphocytes in our body, and these lymphocytes, they have jobs to do as part of our immune system. And one kind of lymphocyte is a T cell, and that has specific roles in our immune system. And another kind is a B cell, and that has specific jobs to do in our immune system. Follicular lymphoma is derived from a B cell, a B-cell lymphocyte. So the…a B cell gets this mutation, and that turns it from a normal healthy B cell into a follicular lymphoma cell.

Lisa Hatfield:  

Okay. Thank you for explaining that and for that overview. That’s really helpful. I appreciate that. So, Dr. Kahl, you also mentioned treatments and how oftentimes it’s not a cancer where you can just remove the cancer. Can you talk about some of the exciting developments with treatments and new innovative therapies, and what are the most important highlights for patients and families?

Dr. Brad Kahl:

Yeah. There’s a lot to talk about here. So I’ll start with how we approach a newly diagnosed patient, and then we’ll go into how we approach patients who have relapsed disease. So the most often, or the most common way a follicular lymphoma patient comes to medical attention is they just either notice a lump from an enlarging lymph node, or some enlarged lymph nodes are just found incidentally because they’re having some testing for some other condition.

And so, like I said, very often patients don’t have symptoms. That’s very typical. Occasionally, the patients will have symptoms, and those symptoms might be pain from a large lymph node mass that’s pushing on something. Occasionally, they might have fevers or night sweats. They wake up in the middle of the night just drenching wet, or unexplained weight loss. Those would be symptoms that can occur in follicular lymphoma. But most patients who come to see us for the first time don’t have symptoms.

When we have a newly diagnosed patient and it takes a biopsy to make the diagnosis, we then need to do the staging evaluation. So that involves some sort of imaging. And nowadays that’s usually in the form of what’s called a PET scan, which gives us a good snapshot of the whole body. And it’ll show us enlarged lymph nodes. And then the PET portion of the scan will show us if the lymph nodes are metabolically more active.

So they show up as these bright spots on the PET scan. And that’s what allows us to stage the patient. It tells us where the disease is located and how much of the disease we see. And so I’m often telling patients, I don’t worry so much about the stage. I worry more about the disease burden. So the way I explain that to patient is, suppose I could take all the follicular lymphoma cells out of your body, and I made a pile. How big is the pile? And that’s actually, I think, more important than the stage in determining our initial strategy.

Because believe it or not, if we have a patient who comes to us with a new diagnosis of follicular lymphoma and they have no symptoms, and it turns out that their tumor burden is very low, we often will recommend an initial approach of no treatment, which is a strange thing for patients to hear. And we spend a lot of time trying to explain the rationale for that. So I’ll try to explain that to you now. Follicular lymphoma is hard to cure.

So it’s this weird cancer in that it’s slow-moving. It often doesn’t make people sick, and we have good treatments for it, but curing it, like making it go away once and for all, proves to be kind of difficult. And studies in the past have shown if you have a patient who has no symptoms and is low tumor burden, that their prognosis is just as good if you leave them alone at the beginning. And many patients will not need any treatment at all for two years, three years, five years. I even have follicular lymphoma patients who I’ve been observing for more than 10 years that have never needed any treatment.

About two out of every 10 patients that are newly diagnosed can go 10 years without needing any treatment. So that’s why we’ll start that strategy for some patients. And that’s psychologically can be difficult for patients. You’re telling me I’ve got a new cancer diagnosis. You’re saying you have good treatments for it. And yet you’re saying you don’t want to use any of those treatments. And so it takes a lot of talking and explaining to try to get people comfortable with that.

Some people never get comfortable with that, I admit it. But some people get very comfortable with it. But it is a very appropriate initial strategy for a low tumor burden asymptomatic person just to observe and get a handle on the pace of the disease. If the disease starts to grow, or if the patient starts to get symptoms, we can start our treatment at that time. And the treatment is going to work just as well as it would have had if we started it last year, or two years ago.

So we feel like we’re putting the patient in no harm, no risk of harm by starting on this strategy of a watch and wait. On the other hand, some patients have high tumor burden, they have a lot of disease, or they have symptoms. And for those patients we need to start them on treatment because the treatment can put them in remission and get them feeling better. Right now, the most common frontline treatment in follicular lymphoma will be a combination of some chemotherapy and some immunotherapy.

The most commonly used regimen in the United States right now is a two drug regimen, a chemotherapy drug called bendamustine (Treanda), and an immunotherapy drug called rituximab (Rituxan). And you give that treatment every 28 days for six months. And it’ll put 90 percent of people into remission. And on average, those remissions last five plus years. And it’s a very, very tolerable treatment.  It’s not too bad as far as chemotherapy goes. There’s no, most people don’t lose their hair. They don’t get peripheral neuropathy, that sometimes chemotherapy drugs give.

It’s not too bad for nausea and things like that. I’m not saying it’s easy or it’s fun. It’s none of that. But as far as chemo goes, it’s not too bad. And it’s effective, it is very effective. And I’ve given that treatment and I have people who are still in their first remission 10 years later, so you can get, for some people can get these really long remissions. But the reality is most patients, their disease does come back, they do relapse at some point. And then we have to start talking about what to do for second line treatment or third-line treatments.

And that’s where things have really taken off in follicular lymphoma in the last few years, there are a number of brand new treatment options in play for relapsed follicular lymphoma that are very exciting, and proves that we’re moving away from chemotherapy. We have drugs that are oral, that are, we call them targeted agents, they hit like a molecular pathway inside the cell a lot, and they kill the cells a lot differently than chemotherapy does. And we have a number of new drugs that work through the immune system, and try to attack the lymphoma that way.

So when we have patients who relapse, probably the most commonly used second-line treatment right now is a combination of a drug called lenalidomide (Revlimid), which is a pill that’s used in a few different cancers. It works very well for certain cancers, and it works well in follicular lymphoma. And that’s given with the immunotherapy drug called rituximab. And that was proven in a study to be very effective. About 80 percent of people will respond to the regimen, and that remission on average lasts in the two to three-year range.

So that’s probably the most commonly used second line regimen right now in the U.S. for follicular lymphoma. And then there are a number of treatments that are now available in third-line and beyond that are new within the past, say three, four years. And these newer treatments that I’m about to describe are now being tested as second line treatments and even as first-line treatments.

So it’s possible that some of these treatments I’m about to describe will become in the future, our go to regimens for first line treatment or second line treatment. And we hope they do move up, because that means they’re, it means they’re even better than what we’ve been using. So probably the treatments that we’re most excited about right now in follicular lymphoma are the drugs called bispecific monoclonal antibodies. There are two that are now FDA-approved. One’s called mosunetuzumab-axgb (Lunsumio), and that was approved about a year-and-a-half ago.

And the other one’s called epcoritamab-bysp (Epkinly), and that was approved just a month ago. And basically these drugs are infused or injected under the skin, infused intravenously injected under the skin and their proteins that will literally stick to the lymphoma cells. And when it does that, it kind of coats the cancer cells. And then after these bispecific antibodies coat the tumor cells, they literally will trick the patient’s T cells or healthy T cells to come in and attack the cancer.

So it’s a way of trying to trick the patient’s own immune system to come in and start fighting the cancer. And these two drugs are very promising in the relapse setting. They work about 80 percent of the time to get some kind of response. About 60 percent of the time patients will go into complete remission, which means we can’t find any evidence for the lymphoma on scans. And they’re both so new that I don’t think we have a full understanding of how durable these remissions are going to be right now.

It looks that like about, if you do get a complete remission, that about half of those patients are holding that complete remission at two and three years. But we’re, we don’t know about four years and five years yet because the drugs are too new. And we expect that if, as these drugs move up and are tested in the second-line setting and in the first-line setting, they’ll work even better because the cancer cells tend to be easier to kill in earlier lines of therapy. Other agents that have moved into the relapse follicular lymphoma space would include CAR T-cell therapy.

This is a fairly sophisticated complicated approach where you actually will run the patient’s blood through apheresis machine and you will extract the patient’s T cells and those T cells get genetically modified in a lab and then expanded and then are shipped back to the center and then re-infused back into the patient. So now again, we’re tricking the patient’s T cells into fighting their B-cell lymphoma.

And there are three CAR T products that are now FDA approved for use in follicular lymphoma, and they have very high response rates. With seemingly good durability we’re now getting three and four-year follow-up for these CAR T products with about half of people still in remission. The CAR T products probably have a little more toxicity and a little more risk than the bispecifics. So I think most of us are thinking we would try the bispecifics before CAR T, but there might be certain patients where a CAR T strategy is more appropriate to use before a bispecific.

So we’re very excited to have these tools in our toolbox. It’s always good to have more options. And then I should just mention the small molecule inhibitors. So here’s an example. Just this past year there was approval for a small molecule called zanubrutinib (Brukinsa). It targets an enzyme called BTK or Bruton’s tyrosine kinase. This is a pill really well tolerated. It’s given in a combination with an immunotherapy drug called obinutuzumab (Gazyva). This zanubrutinib-obinutuzumab combination got FDA-approved just this year for recurrent follicular lymphoma.

The results look very good for that. It’s very well-tolerated. There’s another oral agent called tazemetostat (Tazverik), which was approved a couple of years ago. It targets a mutated protein in follicular lymphoma. This is, again, is a pill super well-tolerated, very few side effects. So, there’s just a few examples for you of all the different treatment options we have for follicular lymphoma that has recurred after initial treatment.

And believe it or not, the decision-making can be difficult when you have so many choices and so many good choices, that’s a good problem to have. And I find myself a lot of times spending a lot of time with the patient and their family as we talk through these different options, and we try to think what’s best for them at this point in time, talking through the pros and the cons, how active it is, what side effects do we need to be concerned about. And it’s a lot for patients to digest when you have so many choices. But like I mentioned that’s actually a good problem to have.

Lisa Hatfield:

I think you’re right. There’s a lot of hope in those options. I do have two follow-up questions. One of them is when you talk about lenalidomide or brand name Revlimid, CAR T bispecific antibodies, this new small molecule, are these all quality of life is so important for cancer patients. Are these all limited duration treatments for recurrent disease when there’s a recurrence of the disease or are they long-term treatments for the disease?

Dr. Brad Kahl:

Yeah, really good question. And the answer is different for every agent. So I’ll try to just kind of run through the list. For the CAR T products, the three different CAR T products, it’s like a one-time treatment and then you’re done because the cells that get infused will persist in the patient’s body for months and months and months. So they’re infused and then the cells will hang around a long time acting on the cancer. So for the CAR T it’s a one-time treatment. For the bispecifics, the mosunetuzumab-axgb product is a time-limited treatment that is done in less than a year. The epcoritamab-bysp is designed to be given indefinitely.

So those are, there are some pros and cons of those two agents, the two small molecules that I mentioned, the zanubrutinib is meant to be given indefinitely and the tazemetostat is meant to be given indefinitely. And then the first one I mentioned was the lenalidomide. That is in follicular lymphoma that it was developed to be given for 12 months in this setting. So the duration of therapy is unique for each of the different agents that I mentioned.

Lisa Hatfield:

Okay. Thank you for that overview of all those emerging therapies. That’s great to know for patients, Dr. Kahl. All right. It’s that time where we answer questions we’ve received from you. Remember, as patients, we should always feel empowered to ask our healthcare providers any and all questions we might have about our treatment, our disease, and our prognosis. Please remember, however, this program is not a substitute for medical care. Always consult with your own medical team.

So, Dr. Kahl, we have several patients who have submitted some questions. The first question is regarding emerging technologies. And I think that you probably have answered that very well actually in a discussion here. So the second question this patient had is how might future innovations build on the latest treatments to offer even better outcomes for patients? You, I think maybe have touched on that, but maybe speak to that a little bit more as far as longer remissions. Yeah.

Dr. Brad Kahl:

Right, right. So I think right now the main emphasis in research is to take some of these really promising drugs that were developed for relapsed follicular lymphoma and do two things with them, test them in combinations in the relapse setting to see if you can make them even more active. So an example of that would be take the drug lenalidomide, which is really active in the relapse setting and pair it with the drug mosunetuzumab-axgb, which is very active in the relapse setting, and pair them together and see if you can get better results than either drug alone.

So there are studies trying to answer questions like that at this time. And then the other area of major interest is to take these promising new treatments approved in the relapse setting and test them upfront. So there are studies being literally designed right now as we speak that will test bispecific monoclonal antibodies in the frontline setting.

So patients can envision being offered a chance to have a chemo-free strategy where they’re just getting a bispecific monoclonal antibody as their initial treatment. And there are studies that will test these drugs as single agents, and there are studies that will test these drugs in combinations with other agents in the frontline setting, like lenalidomide, for example. So we have no results from any of these trials yet, but these trials are just starting to enroll patients and this could fundamentally change the way we’re managing follicular lymphoma in the future if any of these new strategies turn out to be more promising than what we have done historically.

Lisa Hatfield:

Thank you. Okay. Another question, Dr. Kahl. How do outcomes differ for patients with relapsed/refractory disease compared to those who respond well to initial treatment?

Dr. Brad Kahl:

So that’s a really good question. And when we have a patient going through frontline treatment, we’re all really crossing our fingers that that first remission is incredibly durable. Because when the disease relapses, the remissions do tend to get shorter and shorter and shorter, which is frustrating for everybody.And so we love it when we get a nice long first remission. And in the older days when all we had to offer was chemotherapy and some different immunochemotherapy regimens, the remissions in second line and third line might be two years or one year.  It can get frustrating as you go through treatment after treatment after treatment. It’s hard on patients. The side effects start to accumulate. And that’s one of the reasons we’re so excited about all these new agents that we have for relapsed disease with the bispecifics and the CAR T products and the small molecule inhibitors like tazemetostat and zanubrutinib. Because it appears as though these remissions for relapsed disease might be getting longer than what we have seen historically. So there’s no question that dealing with relapsed follicular lymphoma is more difficult than dealing with frontline follicular lymphoma. But we’re optimistic that these newer treatments we have are improving outcomes for patients with relapsed disease.

Lisa Hatfield:

Okay. Thank you. And another question, which patients are considered the most vulnerable when it comes to follicular lymphoma and why, and what measures can be taken to better support these populations in terms of treatment and care? And I’m not sure if they’re talking about different age groups or ethnic groups or geographic groups like rural versus more urban areas, but if you can speak maybe to general terms to answer that question, that would be great.

Dr. Brad Kahl:

Yeah, right. Well, the first thing that comes to mind are older patients. Older patients are always more challenging to take through cancer therapies. The older patients are more fragile. They don’t tolerate the treatments quite as well. They don’t have the physiologic reserve. They’re more susceptible to complications and infections. So I always think when we have older patients that need treatment in follicular lymphoma, the doctor has to be extra, extra careful, sort of the Goldilocks principle. You don’t want the treatment too hot and you don’t want it too cold, too hot, it might work great, but you might get unacceptable side effects too cold, maybe no side effects, but not enough activity against the disease. So we’re always trying to get that patient the best remission we can get them, but doing the least amount of harm along the way.

So I think that takes a little bit of art, a little bit of experience to figure out how to get your older more fragile patients through follicular lymphoma therapy. And then I think the whole idea of patients who live in rural areas, that can often be challenging too, because they may be hours and hours away from medical care. So if they do have a complication of treatment, an infection, for example, it can be challenging to get them the care they need in a quick amount of time. So when I have patients who I know live way out in the country, far away from our center, I just, we always give them a card, it’s got our phone number and I’m like, you feel like something’s going wrong, call us. I don’t care if it’s 2 in the morning, you call us.

It’s not your job to figure out what’s going wrong. That’s our job. It’s just your job to describe to us what you’re experiencing and then we’ll figure out over the phone whether we want you to drive the three hours to come see us or whether we think you just need to go to the closest place, which might be 30 minutes away. So at least you’re in the hands of some medical professionals. And then they can call us with an update on what they’re noticing, what the tests are saying. So taking care of patients who live far away from the medical center poses some additional challenges.

Lisa Hatfield:

Okay. Thank you. And that’s a great takeaway for patients. If you have a question, call your provider. They can help take the stress away from making that decision yourself. 

Well, here’s a loaded question for you, Dr. Kahl. Why does relapse happen in the first place, and what are the changes in the body that signal when and if treatment is likely going to fail?

Dr. Brad Kahl:

Boy, we wish we understood why relapse happens in the first place. Last I mentioned, most of these treatments can get people into remission, which means that they can kill the vast majority of the cancer cells, maybe 99.9 percent of them, but for some patients, there’s just a few stubborn cells that remain behind. Maybe those cells are just sitting there, not growing at all, which follicular lymphoma cells can do.

And when the cells are not trying to divide, not trying to grow, they’re kind of protected from killing. They’re just sitting there doing nothing. And so we think it’s this property that how the cells kind of protect themselves. And so these rare cells that are just kind of sitting there, quiescently not growing, not dividing, these might be the cells then that just hang around for years and then contribute to that relapse five years down the road.

But I admit we don’t fully understand why one patient will relapse two years after a treatment and the next patient is still in remission 10 years later. These are things that we don’t fully understand. Every patient’s lymphoma is a little different, I’m afraid. So two people with follicular lymphoma, they don’t really have the same cancer, cancer, they are sort of like snowflakes. No two are alike. And so they can have different mutations inside the cells that’ll make the cancer behave a little differently from one patient to another. It might make it respond to treatment a little differently from one patient to another. And so what is true for one follicular lymphoma patient may not be true for another.

So if a patient’s symptoms are not being relieved, that might be a clue that the treatment isn’t working as well as we want it to. And then in some cases the only way to figure out if a treatment is working is by scanning. So we’ll have a before picture from a PET scan or a CT scan, and then we’ll take them through a few cycles of treatment, and then we’ll get another scan to prove that the treatment is working like we want it to work. And if it’s not working like we want it to work, then we’ll say, okay, this one isn’t working for you. Let’s go to the what we think is the next best option for you.

Lisa Hatfield:

Okay. Thank you. And just listening to you and hearing about all these nuances with follicular lymphoma, I would probably recommend as a patient myself with a different kind of cancer, seeking out at least a consult from somebody who specializes mostly in follicular lymphoma, at least a hematologist who can tease through some of these nuances to help you as a patient find the best treatments and therapies and quality of life. So just a little tidbit there. So, Dr. Kahl, thank you so much for being part of this Patient Empowerment Network START HERE program.It’s these conversations that help patients truly empower themselves along their treatment journey. And on behalf of patients like myself and those watching, thank you for joining us, Dr. Kahl.

Dr. Brad Kahl:

Thank you for having me.

Lisa Hatfield:  

I’m Lisa Hatfield, thank you for joining this Patient Empowerment Network program.


Share Your Feedback