Tag Archive for: Rituxan

What Are Follicular Lymphoma Considerations for Watch and Wait?

What Are Follicular Lymphoma Considerations for Watch and Wait? from Patient Empowerment Network on Vimeo.

What do follicular lymphoma patients and providers need to be aware of during watch and wait? Expert Dr. Kami Maddocks from Ohio State University discusses what factors are monitored during watch and wait, common symptoms to be on the lookout for, and who patients can contact about concerns.

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:

One person is saying, “I’m in watch and wait currently. Is it possible that I’ll never need treatment, or how long do you wait, and what am I waiting for?”

Dr. Kami Maddocks:

That is a great question. There are patients in watch and wait who will never require treatment. Watching and waiting, we’re watching blood counts, watching the size of lymph nodes. So things that we’re watching for and you’re watching for are changes in lymph nodes size, so are they growing? Are they becoming more symptomatic? Is there a rapid change in them? Are we seeing a change in the blood counts? Are patients starting to have a drop in their blood counts which can happen if somebody’s spleen is getting bigger if they have lymphoma in their bone marrow and that’s progressing, watching for if the lymph nodes are causing a problem, you notice somebody have one in a location like the neck that’s starting to make swallowing difficult or changes in voice, that’s something you want to treat. And then there’s something called B symptoms that we watch for. So if the patient had night sweats…

…night sweats are like drenching night sweats, soak the bed, have to change clothes potentially sheets, fevers, so daily fevers that occur, or significant or rapid weight loss for no reason. All those are kinds of things that we want people to watch for. And we discussed a little bit too if patients start having extreme fatigue, not feeling well, not being able to eat, not having appetites if they have a new pain. And again everybody can have aches and pains. But if you’re having pain that’s not going away or some sort of symptom that’s not improving, those are all things we want to definitely have checked out.

Lisa Hatfield:

I imagine with some of your patients in that mode, there’s what I call the mental gymnastics of thinking, okay, I have this cancer, but I can’t do anything about it, and these symptoms are really vague that come up. So do you allow your patients just to contact you if they’re saying, “I think I have these symptoms, I’m nervous about this.” Can they come in and have a visit with you or contact you at any time?

Dr. Kami Maddocks:

Oh, yes. So we have a 24-hour triage line. I recommend that if patients have a question or concern, it’s better to ask us because if we don’t know about it, we can’t help is the first thing. Usually, we talk to the patient and say, “Okay, how long has this been going on” and see if it’s a red flag like you need to come in right now or is this something that maybe we might recommend getting a set of labs to look at certain labs to see if they’ve changed at all.

We might say, “Okay this seems like something we should actually see you for, but I want CT scans too so let’s order them, so I can have that information when you see me.” So, yeah, I think people should always call with any signs, symptoms, concerns, and then it can be addressed. Now, there are some things that we might say, “Okay, we think based on everything that new cough is probably more likely a respiratory infection. It’s okay to see your PCP.” But we also go through that as well. So yes, I think it’s always best to check in and not let something go.

Lisa Hatfield:

I’m guessing that’s challenging for some of those people in that mode, just thinking, well, I’m just waiting here, so that’s got to be a little bit more challenging.

Dr. Kami Maddocks:

I think you’re absolutely right. And sometimes there’s a benefit to…certainly like rituximab (Rituxan) therapy when there is a disease there, and it is a challenge to think that it’s not being treated.


Share Your Feedback:

Create your own user feedback survey

What Can Follicular Lymphoma Patients Expect With Remission?

What Can Follicular Lymphoma Patients Expect With Remission? from Patient Empowerment Network on Vimeo.

For follicular lymphoma patients, what can they expect to happen with remission? Expert Dr. Kami Maddocks from The Ohio State University explains how remission can vary among patients and shares an overview of potential treatments.

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 Are Follicular Lymphoma Considerations for Watch and Wait


Transcript:

Lisa Hatfield:

So one person says, “I’m currently in remission, what can I expect in my future? How long does remission last? And is treatment after remission the same as initial treatment?”

Dr. Kami Maddocks

So that is very dependent on what a patient receives. So there are different kind…of a lot of our treatments we look at median times. When patients have relapse, that can be a little bit different for single agent antibody therapy versus antibody in combination with chemoimmunotherapy for how long that treatment remission lasts. As far as we don’t typically reuse a treatment once we have used it before, although there is data in follicular lymphoma when patients receive single agent antibodies. So rituximab (Rituxan) alone, if they do well with that single agent immunotherapy for a long period, they may receive re-treatment with just that so long as they don’t have disease that requires more aggressive treatment.

Lisa Hatfield:

So is that more likely to happen then if a patient maybe wasn’t refractory to it, if they just stopped using it for some reason? Would that be more common for that to happen to go back on that same drug?

Dr. Kami Maddocks:

So with rituximab, we use it alone and in combination. So there are some patients that don’t necessarily have what we call a large tumor, and they don’t have a lot of lymph nodes, or they don’t have large lymph nodes, but they might be symptomatic from them, or the location might be problematic. And so once these lymph nodes get a certain size, they usually don’t have as good of a response to single agent antibody therapy. But there are patients who have small lymph nodes that aren’t as big but again are causing a problem that can get completely…you give a short course of the rituximab, and it can last for a very long time and then you would consider again using a short course of that rituximab.

The chemotherapies we have, we don’t reuse chemotherapy, for the most part. Some of that, for a while, there was bendamustine (Treanda) if patients got five, six, 10-year remissions out of it. Sometimes they would re-get that chemotherapy. But I think we’ve just seen so many newer therapies approved in the last five six years. Like the bispecifics, the EZH2 inhibitors, lenalidomide (Revlimid), CAR T, we had different PI3K inhibitors available for a while. And so I think it was just that you had the ability to offer a patient something that they never had before, and that is more appealing.


Share Your Feedback:

Create your own user feedback survey

Understanding Immune System Recovery Post Follicular Lymphoma Treatment

Understanding Immune System Recovery Post Follicular Lymphoma Treatment from Patient Empowerment Network on Vimeo.

Follicular lymphoma treatment may impact the immune system in different ways. Expert Dr. Kami Maddocks from The Ohio State University Comprehensive Cancer Center discusses how immune function may be impacted and how recovery is monitored.

See More from START HERE Follicular Lymphoma

Related Resources:

When Should Follicular Lymphoma Treatment Begin

When Should Follicular Lymphoma Treatment Begin?

What Should Follicular Lymphoma Patients Know About Beauty Products

What Should Follicular Lymphoma Patients Know About Recurrence


Transcript:

Lisa Hatfield:

So another person is asking, “How long does it take for the immune system to really start bouncing back after follicular lymphoma treatment? And what blood test results indicate a weakening immune system?”

Dr. Kami Maddocks:

Yeah, so this is a great question. It also can be a complicated question with many different answers. So one, it can depend on the treatment that a patient receives. Two, it can actually depend on their different parts to the immune system. So different parts of the immune system can recover at different time periods from treatment. So acutely, our neutrophils are something that often gets…they’re bacteria infection fighting cells. Those are the cells that during chemotherapy, when that count gets low and patients are counseled on if you have a fever during your treatment, you need to be evaluated and be seen because if you have an infection and a fever during chemo or some of these treatments, your blood counts are low, you might need to be in the hospital on IV antibiotics.

So those neutrophil parts of it are usually quicker to recover, so they drop with treatment and then recover pretty quickly with each cycle, including after an ended treatment cycle. Sometimes when patients have been treated with several different therapies, it can be harder for those cells to recover. They can stay lower for longer. Then there’s a component of the immune system, so we are ripping out the lymphocytes, because that’s what the cancers have.

And so things targeted. Chemotherapy in general kills the lymphocytes, but there also are targeted therapies like rituximab (Rituxan) bispecific antibodies CAR-T cells, those are particularly wiping…targeted towards proteins on the lymphocytes and wiping them out. Those can be for a more prolonged time. In general, we usually think of about a six-month period so patients can be at increased risk for viral infections in that six-month period may not respond as well to vaccines in that period.

But for some patients it takes longer and some patients recover quicker. It also can depend on where patients are at in their journey because every therapy that they’ve had can take a little bit longer to recover. The last part I’ll add is just sometimes when the lymphocytes are wiped out for a long time people’s proteins, their immunoglobulins that help fight infection get low. And so sometimes we actually will end up giving patients replacement of IVIG to help if they’re having lots of infections.


Share Your Feedback:

Create your own user feedback survey

PODCAST: Follicular Lymphoma Patient Expert Q&A: Dr. Kami Maddocks

Follicular lymphoma expert, Dr. Kami Maddocks discusses the latest in follicular lymphoma, meaningful highlights from the American Society of Hematology 2023 meeting and answers questions submitted by patients and care partners.

Dr. Maddocks is a hematologist/oncologist specializing in treating lymphatic diseases from The Ohio State University. Learn more about Dr. Maddocks.

See More from START HERE Follicular Lymphoma

Download Guide | Descargar Guía

Transcript:

Lisa Hatfield:

Welcome to the 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 healthcare team. Joining me today is Hematologist Oncologist, Dr. Kami Maddocks, Professor of Clinical Internal Medicine in the Division of Hematology at the Ohio State University Wexner Medical Center.

Dr. Maddocks specializes in treating patients with B-cell malignancies, including non-Hodgkin’s lymphoma, Hodgkin’s lymphoma, and chronic lymphocytic leukemia. Dr. Maddocks researches new therapies for these hematologic malignancies largely through evaluating new targeted therapies in clinical trials. Thank you so much for joining us, Dr. Maddocks.

Dr. Kami Maddocks:

It’s really a pleasure to be here with you, Lisa. Thank you so much for having me.

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. Joining us today are patients and care partners facing a follicular lymphoma diagnosis, some of which are newly diagnosed, in active treatment, watch and wait, and also living for years with their disease. No matter where you are in your journey, this program is designed to provide easy-to-understand, reliable, and digestible information to help you make informed decisions. I’m thrilled you’ve joined us.

Please remember before we start to download the program resource guide via the QR code. There is great information there that will be useful during this program and after. So let’s get started. Dr. Maddocks, there is a great deal going on in terms of emerging treatment options and clinical trial data in follicular lymphoma. Can you speak a bit to the exciting developments in follicular lymphoma treatment and the major highlights that are just coming out of the ASH 2023 meeting?

Dr. Kami Maddocks:

Yeah, I think it is really exciting where everything is at right now in the development of treatments for follicular lymphoma. I think one of the most exciting developments is in the immunotherapy treatments that we have. So just a year ago, we saw the approval of the first bispecific antibody in relapsed/refractory follicular lymphoma. So bispecific antibodies are immunotherapy. They target a protein on the lymphoma cell, the follicular lymphoma, but then they also target the T cell to activate it to engage the immune system to attack the lymphoma.

So these bispecific antibodies have been highly effective in relapsed/refractory follicular lymphoma. And what we saw coming out of ASH is some data looking at these in the frontline setting. So a lot of patients will get treated with immunotherapy with rituximab (Rituxan) antibody or chemoimmunotherapy with rituximab in combination with chemo therapies. And we saw some data looking at activity of these bispecific antibodies as the first-line treatment. In addition, currently right now, they’re approved as…the drug that’s approved is called mosunetuzumab-axgb (Lunsumio). That’s approved as a single agent in the relapsed/refractory setting.

And so there were some smaller trials looking at this in combination with other agents to see the outcomes that that produced. I think one of the exciting things is looking at it in combination with lenalidomide (Revlimid), which is an approved oral immunomodulatory therapy in relapsed/refractory follicular lymphoma. And then just lastly, I’ll say there were a few smaller studies looking at combinations of oral targeted therapies and immune therapies in the frontline setting as well.

Lisa Hatfield:

Great, thank you for that. And with so many of these developments, what do you think are the most important highlights for patients and families with current treatment with clinical trials? Anything that you can highlight upon for patients and families?

Dr. Kami Maddocks:

I think, really, when you look at follicular lymphoma or more recent approvals that were looking at bispecific antibodies, chimeric antigen receptor, CAR-T cells, have been approved in relapsed follicular lymphoma, EZH2 inhibitors, so targeted therapies lenalidomide. I mean, really, when you look at follicular lymphoma, we see that patients are living longer and longer. Follicular lymphoma for many patients is somewhat like a chronic disease.

It’s managed over time with periods where they get treatment and then don’t. And what you look at with all of our therapies is we really are looking at immunotherapy and targeted therapies, both in the relapsed setting, but also now in the frontline setting, as opposed to or in place of chemotherapy.

Lisa Hatfield:

Great. Thank you. So you probably have a number of patients who are in the watch-and-wait mode right now. When do you decide when you use these particular therapies? And if you use them earlier on, is there any chance of managing the follicular lymphoma longer or a longer remission?

Dr. Kami Maddocks:

That’s a great question. So from what we know from follicular lymphoma, it’s, as I mentioned, an indolent lymphoma, not curable, but very treatable. So many patients that are diagnosed with follicular lymphoma, the median overall survival is very long, and it’s more, again, like a chronic disease that we manage with treatment. So sometimes we recommend watch-and-wait because patients, there’s never been any study showing that early treatment with the therapies we had improved overall survival. So it’s a balance between deciding when patients have a need for treatment versus not exposing them to treatment that can cause toxicities, if we know that it ultimately doesn’t make them live longer.

But, of course, we want to both treat patients who need disease treatment either for symptoms or for things that are going on by the size of their lymph nodes. So when patients have low blood counts, when they have symptomatic lymph nodes, when they have lymph nodes that are potentially causing a problem to an organ, or we foresee that it could cause a problem to an organ, when they have certain burdens of disease or when they have enough and large lymph nodes that we think that there’s going to be a problem in the near future without treating is when we decide to initiate that treatment.

Lisa Hatfield:

So when you do have patients going through therapy, what are the typical side effects? And how do you help them manage those side effects of treatment?

Dr. Kami Maddocks:

Yeah, that’s a great question, and it’s very dependent, because we have so many different treatments now. It’s very dependent on the treatments that the patient’s getting. So things like single-agent antibody therapy with single-agent rituximab, most of the time, the biggest risk of that is the first time a patient can get it, they can have an infusion reaction. That’s managed as it’s happening. And then they tolerate that, in general, fairly well. That does wipe out the lymphocytes, as most of the treatments do, and puts patients at increased risk of infections, particularly viral infections for a period of time.

Chemotherapy, the most common chemo that we give for follicular lymphoma, I would say nausea, fatigue, and an increased risk of infection are kind of the bigger things. Bendamustine (Treanda) is a commonly used chemotherapy for follicular lymphoma, and that’s some of the big side effects from that.

Lenalidomide, the oral pill, so cytopenias infection, GI toxicity and rash are potentially the more common side effects of that. Less common, but we’re always concerned about blood clots, so most patients will take either an aspirin or a blood thinner, depending on their clot history when they’re on lenalidomide. The bispecific antibodies have a particular risk called cytokine release syndrome, so that immune systems activated, but it can almost get overactivated.

The most common symptom of that is fever, and so patients are counseled very closely on that. But activation of the immune system with that fever can also include changes in blood pressure or the need for some oxygen. Some of the CAR T-cell therapy has the same risk of the cytokine release, also has potential neuro side effects. And then longer term is just how long the patients’ immune systems take to recover. There can be risk for infections.

Lisa Hatfield:

Okay, thank you for that information, that overview. That’s great information for patients to have. So regarding clinical trials right now, are there any clinical trials that you are conducting or that you’re particularly excited about for patients that they might want to ask their providers about?

Dr. Kami Maddocks:

Yes, so we’re also looking at opening a trial for frontline follicular lymphoma that looks at the use of bispecific antibodies. So I think that’s very exciting, because in general, it’s a well-tolerated therapy. And I think if it gives us a chance to produce very good outcomes, but without the toxicity of chemotherapy in the frontline setting, that to me is super exciting for patients. We’re also looking at different bispecific antibodies. So they currently approved one target CD20. We have a CD19-targeted bispecific antibody that I also think is exciting to look at the potential for different targets because then once a patient has had one, you’re targeting something different, and the thought is that they might still be able to respond to a different one.

Lisa Hatfield: Yeah. So with bispecifics then, is that continuous therapy, or is that limited duration therapy?

Dr. Kami Maddocks:

It actually depends on the bispecific. So in follicular right now, the one approved is for a limited duration. When you look at a few of the others that have been approved and other lymphomas that are being studied in follicular lymphoma, there’s a little bit of a variation between continued treatment and limited-duration therapy. I think what’s exciting about a lot of the combination studies is they are more looking at a defined period of time with the combinations.

Lisa Hatfield: Which I’m sure a lot of patients love to hear that. Limited duration, there’s an end to this possibly, so yeah. 

Dr. Kami Maddocks:

Yeah. Nobody wants to be on treatment forever. 

Lisa Hatfield:

That is true. Yeah. Well, thank you so much for that important overview, Dr. Maddocks. It’s that time where we answer questions that we’ve received from you in the audience. Remember, as patients, we should always feel empowered to ask our healthcare providers any and all questions we might have about our treatment and prognosis. Please remember, this program is not a substitute for medical care. Always consult with your own medical team.

So we have a patient who is asking, “What is the recommended frequency and length of imaging PET-CT as a diagnostic tool?” And I wonder if it might be helpful, since we have a broad range of…our audience kind of runs the gamut of newly diagnosed to people who are in remission, people in watch and wait, maybe you can explain the frequency and length of imaging for those who are watching and waiting, and also for those who are maybe in remission, and those who are currently in active treatment.

Dr. Kami Maddocks:

Yeah, so I think this is a great question. I think the important thing about PET scans is to recognize where and when they have a role in follicular lymphoma, because they’re not used as monitoring tools in follicular lymphoma long term in either watch and wait, typically, or in patients who’ve had treatment. So follicular lymphoma, when patients have a diagnosis, we like to get a PET scan, because it helps us stage the follicular lymphoma a little bit better than just generalized CT scans. When patients are being monitored and they have a change in symptoms, a change on maybe routine CT scans and their physical exam in their labs, then you may want a PET scan. We always want to get one before a patient requires treatment, and then after a patient has treatment to help determine the response by PET scan.

As far as monitoring in patients who are on watch and wait or monitoring in a patient who’s received treatment, routine PET imaging is actually not recommended. It’s not recommended by the guidelines. And honestly, they’re not usually approved by insurance for routine monitoring, because they’re not recommended. So what is recommended is seeing your physician, usually at a three to six-month window, depending on where you are in your journey. And then usually, for routine monitoring, CT scans are done. If somebody’s in watch and wait, oftentimes they might initially be done at three months, then six months, then even yearly, and same with after treatment.

Lisa Hatfield:

All right, thank you. So another person is asking, “How long does it take for the immune system to really start bouncing back after follicular lymphoma treatment? And what blood test results indicate a weakening immune system?”

Dr. Kami Maddocks

Yeah, so this is a great question. [chuckle] It also can be a complicated question with many different answers. So one, it can depend on the treatment that a patient receives. Two, it can actually depend on their different parts to the immune system. So different parts of the immune system can recover at different time periods from treatment. So acutely, our neutrophils are something that often gets…they’re bacteria infection fighting cells. Those are the cells that during chemotherapy, when that count gets low and patients are counseled on if you have a fever during your treatment, you need to be evaluated and be seen because if you have an infection and a fever during chemo or some of these treatments, your blood counts are low, you might need to be in the hospital on IV antibiotics.

So those neutrophil part of it are usually quicker to recover, so they drop with treatment and then recover pretty quickly with each cycle, including after an ended treatment cycle. Sometimes when patients have been treated with several different therapies, it can be harder for those cells to recover. They can stay lower for longer. Then there’s a component of the immune system, so we are ripping out the lymphocytes, because that’s what the cancers have.

And so things targeted. Chemotherapy in general kills the lymphocytes but there also are targeted therapies like rituximab bispecific antibodies CAR T-cells those are particularly wiping…targeted towards proteins on the lymphocytes and wiping them out. Those can be for a more prolonged time. In general, we usually think of about a six-month period so patients can be at increased risk for viral infections in that six-month period may not respond as well to vaccines in that period.

But for some patients it takes longer and some patients recover quicker. It also can depend on where patients are at in their journey because every therapy that they’ve had can take a little bit longer to recover. The last part I’ll add is just sometimes when the lymphocytes are wiped out for a long time people’s proteins, their immunoglobulins that help fight infection get low. And so sometimes we actually will end up giving patients replacement of IVIG to help if they’re having lots of infections.

Lisa Hatfield:

All right. Thank you. Another good question, and this comes up with many blood cancers or a lot of cancers. Should patients be mindful of beauty products such as shampoos, soaps, and sunblocks when in remission for follicular lymphoma?

Dr. Kami Maddocks:

That’s another great question. I am not aware of any data connecting those specific things.I think patients definitely should be wearing sunblock, because we know that a lot of patients with blood cancers can get secondary malignancies. So being careful of being…we also know, I should say, even patients who are getting treated can have a more sensitivity to the sun. So being careful with sun precautions, either avoiding the sun or wearing sunblock, making sure you’re covered when you go outside. I’ll even say I’ve seen a few patients who during treatment have gotten bad windburns. So your skin definitely can be more sensitive when you’re receiving therapies.

Lisa Hatfield:

All right, thank you. Let’s see here. This patient is asking if you are in remission for a long period of time after follicular lymphoma treatment, can you technically be cured in some cases, or are you considered to still have the cancer?

Dr. Kami Maddocks:

So that is a great question. There’s a term that’s used in follicular lymphoma called a functional cure. So we have patients that essentially get treated, and they live long enough that they die from something else without their follicular lymphoma ever relapsing. So while we say from what we know if somebody lives long enough that this disease is likely going to relapse at some point, there are patients that will be treated, and the disease will never come back.

Lisa Hatfield:

Okay. That’s helpful, thank you. Can patients facing follicular lymphoma be considered immunocompromised if they’re in remission?

Dr. Kami Maddocks:

I think this kind of goes back to when we talked about the immune system recovery that this can be a little bit of a complicated question, because it depends a little bit on the treatment that they got, how far out from the treatment they are and how many treatments they’ve had in the past. So, in general, if I have a patient that has received therapy, their counts have recovered, they in general look like…their lab work looks like their immune system, then in general I would say that they have an immune system that’s likely similar to somebody who didn’t have the follicular lymphoma, and they’re going to be able to fight infections and respond to vaccines.

I think what we do know is sometimes when patients get rituximab maintenance or obinutuzumab (Gazyva) maintenance or some of the chemotherapies there are some patients that can have a longer time that they’re immunosuppressed. So I think this is always something that’s good to ask your doctor for. In your specific situation with the treatment you received, when do you expect to have a regularly functioning immune system?

Lisa Hatfield:

For follicular lymphoma, what are the predictors of transformation and relapse, and what symptoms should patients be looking out for and tell their doctor about?

Dr. Kami Maddocks:

Yeah, so I think this is a great question. As far as just in everybody predicting when they’re going to progress, when they’re going to relapse, we don’t actually have great ways to do that right now. One of the things that has been shown to potentially predict things is for patients who do receive treatment if they have an early relapse, that suggests that their disease is going to behave more aggressively. As far as looking for relapse, things that people want to look for, not all patients will have symptoms but certainly if patients feel any lumps or bumps if they start…I like to tell my patients if you…patients usually know if something’s wrong.

So everybody’s going to have aches and pains, everybody’s going to have the normal infections, but if you’re not feeling well significant fatigue night sweats fevers are always something that we look for but that’s not something that everybody has. New pains, not feeling well, just kind of the inability to feel like you can keep up with what you’re doing daily, those are always things that you should at least call in to see if you should be evaluated. It’s important to know that follicular lymphoma patients are followed. As I said, you are followed forever. We do also watch your blood counts to make sure that we’re not seeing changes in blood counts, changes in lactate dehydrogenase which is a non-specific marker but something that we follow in lymphoma.

Lisa Hatfield:

Okay. Thank you. And one follow-up to that question also. So are there follicular lymphoma specialists? If a patient is maybe in an area that doesn’t have a large academic center or a large cancer institution, do you recommend they see somebody who specializes in follicular lymphoma or can they see even for a consult or do you think that their local hematologist oncologist is very familiar with that themselves? Do you have recommendations?

Dr. Kami Maddocks:

Yeah, so that’s a great question. Local, I think follicular lymphoma is common enough that a lot of our general oncologists who see everything see follicular lymphoma. I think it never hurts of course to ask about clinical trials. So if that’s something that might be available. If it’s available, it might be worth going to a specialist for. If there’s concerns, I think it’s always a good idea to get a second opinion to make sure that a patient is comfortable.

I think if a patient seems to have a more aggressive behaving follicular or if they’ve had a lot of different treatments, that’s also if you’re seeing a general oncologist at a time, that it’s good to see if there are clinical trials or if a specialist has anything new or different.

Lisa Hatfield:

Okay. Thanks. So we have a person asking, “Does lymphoma recurrence always happen in an aggressive manner?”

Dr. Kami Maddocks:

That’s a great question. The answer is no on that, and in fact lymphoma recurrence doesn’t always need to be treated just because it does recur. So when you look at follicular lymphoma, there are patients who are in a watch-and-wait period. When they’re diagnosed, they’ll eventually progress to requiring treatment or most…well, there are patients who might not. Once they require treatment, they get a time period without…most of them will get a time period without disease.

There are patients who will…that you’ll find lymph nodes growing on CT scans maybe that you’re doing monitoring for, but the patient will otherwise feel well. They won’t have, necessarily, very big lymph nodes. Their blood counts will be okay and you may say, okay, just like you had watch-and-wait to start with, we’re going to watch and wait right now with this relapse, because you don’t have any indications that are saying we need to treat this.

And again that doesn’t necessarily make our patient live longer. So you want to balance their quality of life and toxicities of treatment. There are patients who will…when they relapse, they will have indications for treatments, and then there are patients who will potentially have more aggressive relapses and be very symptomatic or have larger lymph nodes.

Lisa Hatfield:

Okay. All right. So one person says, “I’m currently in remission, what can I expect in my future? How long does remission last? And is treatment after remission the same as initial treatment?”

Dr. Kami Maddocks:

So that is very dependent on what a patient receives. So there are different kind…of a lot of our treatments we look at median times. When patients have relapse, that can be a little bit different for single agent antibody therapy versus antibody in combination with chemoimmunotherapy for how long that treatment remission lasts.

As far as we don’t typically reuse a treatment once we have used it before, although there is data in follicular lymphoma when patients receive single agent antibody. So rituximab alone, if they do well with that single agent immunotherapy for a long period, they may receive re-treatment with just that so long as they don’t have disease that requires more aggressive treatment.

Lisa Hatfield:

Okay. So is that more likely to happen then if a patient maybe wasn’t refractory to it, if they just stopped using it for some reason? Would that be more common for that to happen to go back on that same drug?

Dr. Kami Maddocks:

So with rituximab, we use it alone and in combination. So there are some patients that don’t necessarily have what we call a large tumor, and they don’t have a lot of lymph nodes or they don’t have large lymph nodes but they might be symptomatic from them or the location might be problematic. And so once these lymph nodes get a certain size, they usually don’t have as good of a response to single agent antibody therapy. But there are patients who have small lymph nodes that aren’t as big but again are causing a problem that can get completely…you give a short course of the rituximab, and it can last for a very long time and then you would consider again using a short course of that rituximab.

The chemotherapies we have, we don’t reuse chemotherapy, for the most part. Some of that, for a while, there was bendamustine if patients got five, six, 10-year remissions out of it. Sometimes they would re-get that chemotherapy. But I think we’ve just seen so many newer therapies approved in the last five six years. Like the bispecifics, the EZH2 inhibitors, lenalidomide, CAR T, we had different PI3K inhibitors available for a while. And so I think it was just that you had the ability to offer a patient something that they never had before, and that is more appealing.

Lisa Hatfield:

Okay. Thank you. So you spoke a little bit about IVIG infusions before and this person is saying that, “I’m having to do IVIG infusions, which started years after my treatment due to my IgG numbers being low. Are nausea and headache side effects common?”

Dr. Kami Maddocks:

Yeah, so some patients can have nausea, headache, myalgia, body aches, some get fevers and some infusion reactions. For some patients, they can have that from the start, some patients can develop it. That’s always a good thing to talk to your doctor about. There are different products for IVIG, and sometimes patients are able to switch products. I will say IVIG can be very insurance-dependent, so it’s also sometimes what…the formulation that insurance…an individual’s insurance covers. But yes, these are side effects, they’re worth, if they’re getting worse or lasting a long time, making sure that it’s discussed with the physician prescribing it.

Lisa Hatfield:

Okay. All right. One person is saying, “I’m in watch and wait currently. Is it possible that I’ll never need treatment or how long do you wait and what am I waiting for?”

Dr. Kami Maddocks:

That is a great question. There are patients in watch and wait who will never require treatment. Watching and waiting, we’re watching blood counts, watching the size of lymph nodes. So things that we’re watching for and you’re watching for are changes in lymph nodes size, so are they growing? Are they becoming more symptomatic? Is there a rapid change in them? Are we seeing a change in the blood counts? Are patients starting to have a drop in their blood counts which can happen if somebody’s spleen is getting bigger if they have lymphoma in their bone marrow and that’s progressing, watching for if the lymph nodes are causing a problem, you notice somebody have one in a location like the neck that’s starting to make swallowing difficult or changes in voice, that’s something you want to treat. And then there’s something called B symptoms that we watch for. So if the patient had night sweats…

 …night sweats are like drenching night sweats, soak the bed, have to change clothes potentially sheets, fevers, so daily fevers that occur, or significant or rapid weight loss for no reason.  All those are kinds of things that we want people to watch for. And we discussed a little bit too if patients start having extreme fatigue, not feeling well, not being able to eat, not having appetites if they have a new pain. And again everybody can have aches and pains. But if you’re having pain that’s not going away or some sort of symptom that’s not improving, those are all things we want to definitely have checked out.

Lisa Hatfield:

Okay. Thank you. I imagine with some of your patients in that mode, there’s what I call the mental gymnastics of thinking, okay, I have this cancer, but I can’t do anything about it, and these symptoms are really vague that come up. So do you allow your patients just to contact you if they’re saying, “I think I have these symptoms, I’m nervous about this.” Can they come in and have a visit with you or contact you at any time?

Dr. Kami Maddocks:

Oh, yes. So we have a 24-hour triage line. I recommend that if patients have a question or concern, it’s better to ask us because if we don’t know about it, we can’t help is the first thing. Usually, we talk to the patient and say, okay, how long has this been going on and see if it’s a red flag like you need to come in right now or is this something that maybe we might recommend getting a set of labs to look at certain labs to see if they’ve changed at all. We might say okay this seems like something we should actually see you for, but I want CT scans too so let’s order them so I can have that information when you see me.

So, yeah, I think people should always call with any signs, symptoms, concerns, and then it can be addressed. Now, there are some things that we might say, okay, we think based on everything that new cough is probably more likely a respiratory infection. It’s okay to see your PCP, but we also go through that as well. So yes, I think it’s always best to check in and not let something go.

Lisa Hatfield:

Okay. Thank you. I’m guessing that’s challenging for some of those people in that mode, just thinking, well, I’m just waiting here, so that’s got to be a little bit more challenging.

Dr. Kami Maddocks:

I think you’re absolutely right. And sometimes there’s a benefit to…certainly like rituximab therapy when there is a disease there, and it is a challenge to think that it’s not being treated.

Lisa Hatfield:

Okay. Thank you. This is I guess the last or second to the last question we have. Dr. Maddocks, can you speak to maintenance therapy and monitoring and follicular lymphoma, and what signs of infection should patients and care partners be aware of during treatment?

Dr. Kami Maddocks:

Yeah, so maintenance therapy, in follicular lymphoma is something, so maintenance antibody therapy after initial chemo…usually, chemoimmunotherapy is something that’s been studied that’s shown in some patients to provide a benefit as far as keeping disease away longer and a remission longer. But then it’s also been shown to be associated with a higher risk of infection, because you’re keeping those lymphocytes wiped out, particularly when it’s given as maintenance after certain chemotherapies in addition to the immunotherapy. So it can be a balance. I think maintenance isn’t something that every follicular lymphoma patient gets, but it’s something also that is used.

So that’s the first thing in discussion with your doctor, is this something that maintenance is recommended? Why or why not? Then watching during maintenance usually if people start to have more infections, which are oftentimes sinus respiratory infections, then we’re thinking about, okay, is this somebody that has low immunoglobulins? Do we need to check those? Are we worried about them? Needing to stop maintenance, potentially needing IVIG.

Lisa Hatfield:

Okay. Thank you. And then this is a Lisa question, “Do you know if there’s any data to suggest that the follicular lymphoma or any type of non-Hodgkin’s lymphoma has a familial or hereditary component?”

Dr. Kami Maddocks:

So there are very small number of…in non-Hodgkin’s lymphoma, there are some small familial components. Unlike, however, say breast cancer where there’s specific genes to test for, we don’t have that as a screening here in non-Hodgkin’s lymphoma.

Lisa Hatfield:

Dr. Maddocks, 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 very much, Dr. Maddocks.

Dr. Kami Maddocks:

Lisa, thank you so much for having me. It’s been a great conversation and hopefully it can help some people.

Lisa Hatfield:

I hope so too. I’m Lisa Hatfield. Thank you for joining this Patient Empowerment Network program.

What Follicular Lymphoma Treatment Side Effects Should Patients Expect?

What Follicular Lymphoma Treatment Side Effects Should Patients Expect? from Patient Empowerment Network on Vimeo.

Follicular lymphoma patients might experience different side effects, so what should patients expect? Expert Dr. Kami Maddocks from The Ohio State University Comprehensive Cancer Center discusses various treatments and common side effects that patients experience.

See More from START HERE Follicular Lymphoma

Related Resources:

What’s New for Follicular Lymphoma Treatment News and Developments

What’s New for Follicular Lymphoma Treatment News and Developments?

What Can Follicular Lymphoma Patients Expect for PET-CT Scans

What Are Predictors of Follicular Lymphoma Relapse or Transformation


Transcript:

Lisa Hatfield:

 So when you do have patients going through therapy, what are the typical side effects? And how do you help them manage those side effects of treatment?

Dr. Kami Maddocks:

Yeah, that’s a great question, and it’s very dependent, because we have so many different treatments now. It’s very dependent on the treatments that the patient’s getting. So things like single-agent antibody therapy with single-agent rituximab (Rituxan), most of the time, the biggest risk of that is the first time a patient can get it, they can have an infusion reaction. That’s managed as it’s happening. And then they tolerate that, in general, fairly well. That does wipe out the lymphocytes, as most of the treatments do, and puts patients at increased risk of infections, particularly viral infections for a period of time.

Chemotherapy, the most common chemo that we give for follicular lymphoma, I would say nausea, fatigue, and an increased risk of infection are kind of the bigger things. Bendamustine (Treanda) is a commonly used chemotherapy for follicular lymphoma, and that’s some of the big side effects from that.

Lenalidomide (Revlimid), the oral pill, so cytopenias infection, GI toxicity and rash are potentially the more common side effects of that. Less common, but we’re always concerned about blood clots, so most patients will take either an aspirin or a blood thinner, depending on their clot history when they’re on lenalidomide. The bispecific antibodies have a particular risk called cytokine release syndrome, so that immune systems activated, but it can almost get overactivated.

The most common symptom of that is fever, and so patients are counseled very closely on that. But activation of the immune system with that fever can also include changes in blood pressure or the need for some oxygen. Some of the CAR T-cell therapy has the same risk of the cytokine release, also has potential neuro side effects. And then longer term is just how long the patients’ immune systems take to recover. There can be risk for infections.


Share Your Feedback:

Create your own user feedback survey

What’s New for Follicular Lymphoma Treatment News and Developments?

What’s New for Follicular Lymphoma Treatment News and Developments? from Patient Empowerment Network on Vimeo.

Follicular lymphoma treatment options are expanding, so what’s the latest news? Expert Dr. Kami Maddocks from The Ohio State University Comprehensive Cancer Center shares treatment and research updates from the ASH 2023 conference.

See More from START HERE Follicular Lymphoma

Related Resources:

When Should Follicular Lymphoma Treatment Begin

When Should Follicular Lymphoma Treatment Begin?

What Can Follicular Lymphoma Patients Expect for PET-CT Scans

What Are Predictors of Follicular Lymphoma Relapse or Transformation


Transcript:

Lisa Hatfield:

Dr. Maddocks, there is a great deal going on in terms of emerging treatment options and clinical trial data in follicular lymphoma. Can you speak a bit to the exciting developments in follicular lymphoma treatment and the major highlights that are just coming out of the ASH 2023 meeting?

Dr. Kami Maddocks:

Yeah, I think it is really exciting where everything is at right now in the development of treatments for follicular lymphoma. I think one of the most exciting developments is in the immunotherapy treatments that we have. So just a little less than a year ago, this month, December of last year, we saw the approval of the first bispecific antibody in relapsed/refractory follicular lymphoma. So bispecific antibodies are immunotherapy. They target a protein on the lymphoma cell, the follicular lymphoma, but then they also target the T cell to activate it to engage the immune system to attack the lymphoma. So these bispecific antibodies have been highly effective in relapsed/refractory follicular lymphoma. 

And what we saw coming out of ASH is some data looking at these in the frontline setting. So a lot of patients will get treated with immunotherapy with rituximab (Rituxan) antibody or chemoimmunotherapy with rituximab in combination with chemo therapies. And we saw some data looking at activity of these bispecific antibodies as the first-line treatment. In addition, currently right now, they’re approved as…the drug that’s approved is called mosunetuzumab-axgb (Lunsumio). That’s approved as a single agent in the relapsed/refractory setting.

And so there were some smaller trials looking at this in combination with other agents to see the outcomes that that produced. I think one of the exciting things is looking at it in combination with lenalidomide (Revlimid), which is an approved oral immunomodulatory therapy in relapsed/refractory follicular lymphoma. And then just lastly, I’ll say there were a few smaller studies looking at combinations of oral targeted therapies and immune therapies in the frontline setting as well.

Lisa Hatfield:

And with so many of these developments, what do you think are the most important highlights for patients and families with current treatment with clinical trials? Anything that you can highlight upon for patients and families?

Dr. Kami Maddocks:

I think, really, when you look at follicular lymphoma or more recent approvals that were looking at bispecific antibodies, chimeric antigen receptor, CAR-T cells, have been approved in relapsed follicular lymphoma, EZH2 inhibitors, so targeted therapies lenalidomide. I mean, really, when you look at follicular lymphoma, we see that patients are living longer and longer. Follicular lymphoma for many patients is somewhat like a chronic disease.

It’s managed over time with periods where they get treatment and then don’t. And what you look at with all of our therapies is we really are looking at immunotherapy and targeted therapies, both in the relapsed setting, but also now in the frontline setting, as opposed to or in place of chemotherapy.


Share Your Feedback:

Create your own user feedback survey

Follicular Lymphoma Patient Expert Q&A: Dr. Kami Maddocks

Follicular Lymphoma Patient Expert Q&A: Dr. Kami Maddocks from Patient Empowerment Network on Vimeo.

Follicular lymphoma expert, Dr. Kami Maddocks discusses the latest in follicular lymphoma, meaningful highlights from the American Society of Hematology 2023 meeting and answers questions submitted by patients and care partners.

Dr. Maddocks is a hematologist/oncologist specializing in treating lymphatic diseases from The Ohio State University. Learn more about Dr. Maddocks.

See More from START HERE Follicular Lymphoma

Download Guide | Descargar Guía

Related Resources:

Relapsed/Refractory Follicular Lymphoma Treatments and Bispecific Antibodies

Relapsed/Refractory Follicular Lymphoma Treatments and Bispecific Antibodies

How Can Follicular Lymphoma Treatment Side Effects Be Reduced?

Understanding Follicular Lymphoma Disease Progression Symptoms and Monitoring


Transcript:

Lisa Hatfield:

Welcome to the 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 healthcare team. Joining me today is Hematologist Oncologist, Dr. Kami Maddocks, Professor of Clinical Internal Medicine in the Division of Hematology at the Ohio State University Wexner Medical Center.

Dr. Maddocks specializes in treating patients with B-cell malignancies, including non-Hodgkin’s lymphoma, Hodgkin’s lymphoma, and chronic lymphocytic leukemia. Dr. Maddocks researches new therapies for these hematologic malignancies largely through evaluating new targeted therapies in clinical trials. Thank you so much for joining us, Dr. Maddocks.

Dr. Kami Maddocks:

It’s really a pleasure to be here with you, Lisa. Thank you so much for having me.

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. Joining us today are patients and care partners facing a follicular lymphoma diagnosis, some of which are newly diagnosed, in active treatment, watch and wait, and also living for years with their disease. No matter where you are in your journey, this program is designed to provide easy-to-understand, reliable, and digestible information to help you make informed decisions. I’m thrilled you’ve joined us.

Please remember before we start to download the program resource guide via the QR code. There is great information there that will be useful during this program and after. So let’s get started. Dr. Maddocks, there is a great deal going on in terms of emerging treatment options and clinical trial data in follicular lymphoma. Can you speak a bit to the exciting developments in follicular lymphoma treatment and the major highlights that are just coming out of the ASH 2023 meeting?

Dr. Kami Maddocks:

Yeah, I think it is really exciting where everything is at right now in the development of treatments for follicular lymphoma. I think one of the most exciting developments is in the immunotherapy treatments that we have. So just a year ago, we saw the approval of the first bispecific antibody in relapsed/refractory follicular lymphoma. So bispecific antibodies are immunotherapy. They target a protein on the lymphoma cell, the follicular lymphoma, but then they also target the T cell to activate it to engage the immune system to attack the lymphoma.

So these bispecific antibodies have been highly effective in relapsed/refractory follicular lymphoma. And what we saw coming out of ASH is some data looking at these in the frontline setting. So a lot of patients will get treated with immunotherapy with rituximab (Rituxan) antibody or chemoimmunotherapy with rituximab in combination with chemo therapies. And we saw some data looking at activity of these bispecific antibodies as the first-line treatment. In addition, currently right now, they’re approved as…the drug that’s approved is called mosunetuzumab-axgb (Lunsumio). That’s approved as a single agent in the relapsed/refractory setting.

And so there were some smaller trials looking at this in combination with other agents to see the outcomes that that produced. I think one of the exciting things is looking at it in combination with lenalidomide (Revlimid), which is an approved oral immunomodulatory therapy in relapsed/refractory follicular lymphoma. And then just lastly, I’ll say there were a few smaller studies looking at combinations of oral targeted therapies and immune therapies in the frontline setting as well.

Lisa Hatfield:

Great, thank you for that. And with so many of these developments, what do you think are the most important highlights for patients and families with current treatment with clinical trials? Anything that you can highlight upon for patients and families?

Dr. Kami Maddocks:

I think, really, when you look at follicular lymphoma or more recent approvals that were looking at bispecific antibodies, chimeric antigen receptor, CAR-T cells, have been approved in relapsed follicular lymphoma, EZH2 inhibitors, so targeted therapies lenalidomide. I mean, really, when you look at follicular lymphoma, we see that patients are living longer and longer. Follicular lymphoma for many patients is somewhat like a chronic disease.

It’s managed over time with periods where they get treatment and then don’t. And what you look at with all of our therapies is we really are looking at immunotherapy and targeted therapies, both in the relapsed setting, but also now in the frontline setting, as opposed to or in place of chemotherapy.

Lisa Hatfield:

Great. Thank you. So you probably have a number of patients who are in the watch-and-wait mode right now. When do you decide when you use these particular therapies? And if you use them earlier on, is there any chance of managing the follicular lymphoma longer or a longer remission?

Dr. Kami Maddocks:

That’s a great question. So from what we know from follicular lymphoma, it’s, as I mentioned, an indolent lymphoma, not curable, but very treatable. So many patients that are diagnosed with follicular lymphoma, the median overall survival is very long, and it’s more, again, like a chronic disease that we manage with treatment. So sometimes we recommend watch-and-wait because patients, there’s never been any study showing that early treatment with the therapies we had improved overall survival. So it’s a balance between deciding when patients have a need for treatment versus not exposing them to treatment that can cause toxicities, if we know that it ultimately doesn’t make them live longer.

But, of course, we want to both treat patients who need disease treatment either for symptoms or for things that are going on by the size of their lymph nodes. So when patients have low blood counts, when they have symptomatic lymph nodes, when they have lymph nodes that are potentially causing a problem to an organ, or we foresee that it could cause a problem to an organ, when they have certain burdens of disease or when they have enough and large lymph nodes that we think that there’s going to be a problem in the near future without treating is when we decide to initiate that treatment.

Lisa Hatfield:

So when you do have patients going through therapy, what are the typical side effects? And how do you help them manage those side effects of treatment?

Dr. Kami Maddocks:

Yeah, that’s a great question, and it’s very dependent, because we have so many different treatments now. It’s very dependent on the treatments that the patient’s getting. So things like single-agent antibody therapy with single-agent rituximab, most of the time, the biggest risk of that is the first time a patient can get it, they can have an infusion reaction. That’s managed as it’s happening. And then they tolerate that, in general, fairly well. That does wipe out the lymphocytes, as most of the treatments do, and puts patients at increased risk of infections, particularly viral infections for a period of time.

Chemotherapy, the most common chemo that we give for follicular lymphoma, I would say nausea, fatigue, and an increased risk of infection are kind of the bigger things. Bendamustine (Treanda) is a commonly used chemotherapy for follicular lymphoma, and that’s some of the big side effects from that.

Lenalidomide, the oral pill, so cytopenias infection, GI toxicity and rash are potentially the more common side effects of that. Less common, but we’re always concerned about blood clots, so most patients will take either an aspirin or a blood thinner, depending on their clot history when they’re on lenalidomide. The bispecific antibodies have a particular risk called cytokine release syndrome, so that immune systems activated, but it can almost get overactivated.

The most common symptom of that is fever, and so patients are counseled very closely on that. But activation of the immune system with that fever can also include changes in blood pressure or the need for some oxygen. Some of the CAR T-cell therapy has the same risk of the cytokine release, also has potential neuro side effects. And then longer term is just how long the patients’ immune systems take to recover. There can be risk for infections.

Lisa Hatfield:

Okay, thank you for that information, that overview. That’s great information for patients to have. So regarding clinical trials right now, are there any clinical trials that you are conducting or that you’re particularly excited about for patients that they might want to ask their providers about?

Dr. Kami Maddocks:

Yes, so we’re also looking at opening a trial for frontline follicular lymphoma that looks at the use of bispecific antibodies. So I think that’s very exciting, because in general, it’s a well-tolerated therapy. And I think if it gives us a chance to produce very good outcomes, but without the toxicity of chemotherapy in the frontline setting, that to me is super exciting for patients. We’re also looking at different bispecific antibodies. So they currently approved one target CD20. We have a CD19-targeted bispecific antibody that I also think is exciting to look at the potential for different targets because then once a patient has had one, you’re targeting something different, and the thought is that they might still be able to respond to a different one.

Lisa Hatfield: Yeah. So with bispecifics then, is that continuous therapy, or is that limited duration therapy?

Dr. Kami Maddocks:

It actually depends on the bispecific. So in follicular right now, the one approved is for a limited duration. When you look at a few of the others that have been approved and other lymphomas that are being studied in follicular lymphoma, there’s a little bit of a variation between continued treatment and limited-duration therapy. I think what’s exciting about a lot of the combination studies is they are more looking at a defined period of time with the combinations.

Lisa Hatfield: Which I’m sure a lot of patients love to hear that. Limited duration, there’s an end to this possibly, so yeah. 

Dr. Kami Maddocks:

Yeah. Nobody wants to be on treatment forever. 

Lisa Hatfield:

That is true. Yeah. Well, thank you so much for that important overview, Dr. Maddocks. It’s that time where we answer questions that we’ve received from you in the audience. Remember, as patients, we should always feel empowered to ask our healthcare providers any and all questions we might have about our treatment and prognosis. Please remember, this program is not a substitute for medical care. Always consult with your own medical team.

So we have a patient who is asking, “What is the recommended frequency and length of imaging PET-CT as a diagnostic tool?” And I wonder if it might be helpful, since we have a broad range of…our audience kind of runs the gamut of newly diagnosed to people who are in remission, people in watch and wait, maybe you can explain the frequency and length of imaging for those who are watching and waiting, and also for those who are maybe in remission, and those who are currently in active treatment.

Dr. Kami Maddocks:

Yeah, so I think this is a great question. I think the important thing about PET scans is to recognize where and when they have a role in follicular lymphoma, because they’re not used as monitoring tools in follicular lymphoma long term in either watch and wait, typically, or in patients who’ve had treatment. So follicular lymphoma, when patients have a diagnosis, we like to get a PET scan, because it helps us stage the follicular lymphoma a little bit better than just generalized CT scans. When patients are being monitored and they have a change in symptoms, a change on maybe routine CT scans and their physical exam in their labs, then you may want a PET scan. We always want to get one before a patient requires treatment, and then after a patient has treatment to help determine the response by PET scan.

As far as monitoring in patients who are on watch and wait or monitoring in a patient who’s received treatment, routine PET imaging is actually not recommended. It’s not recommended by the guidelines. And honestly, they’re not usually approved by insurance for routine monitoring, because they’re not recommended. So what is recommended is seeing your physician, usually at a three to six-month window, depending on where you are in your journey. And then usually, for routine monitoring, CT scans are done. If somebody’s in watch and wait, oftentimes they might initially be done at three months, then six months, then even yearly, and same with after treatment.

Lisa Hatfield:

All right, thank you. So another person is asking, “How long does it take for the immune system to really start bouncing back after follicular lymphoma treatment? And what blood test results indicate a weakening immune system?”

Dr. Kami Maddocks

Yeah, so this is a great question. [chuckle] It also can be a complicated question with many different answers. So one, it can depend on the treatment that a patient receives. Two, it can actually depend on their different parts to the immune system. So different parts of the immune system can recover at different time periods from treatment. So acutely, our neutrophils are something that often gets…they’re bacteria infection fighting cells. Those are the cells that during chemotherapy, when that count gets low and patients are counseled on if you have a fever during your treatment, you need to be evaluated and be seen because if you have an infection and a fever during chemo or some of these treatments, your blood counts are low, you might need to be in the hospital on IV antibiotics.

So those neutrophil part of it are usually quicker to recover, so they drop with treatment and then recover pretty quickly with each cycle, including after an ended treatment cycle. Sometimes when patients have been treated with several different therapies, it can be harder for those cells to recover. They can stay lower for longer. Then there’s a component of the immune system, so we are ripping out the lymphocytes, because that’s what the cancers have.

And so things targeted. Chemotherapy in general kills the lymphocytes but there also are targeted therapies like rituximab bispecific antibodies CAR T-cells those are particularly wiping…targeted towards proteins on the lymphocytes and wiping them out. Those can be for a more prolonged time. In general, we usually think of about a six-month period so patients can be at increased risk for viral infections in that six-month period may not respond as well to vaccines in that period.

But for some patients it takes longer and some patients recover quicker. It also can depend on where patients are at in their journey because every therapy that they’ve had can take a little bit longer to recover. The last part I’ll add is just sometimes when the lymphocytes are wiped out for a long time people’s proteins, their immunoglobulins that help fight infection get low. And so sometimes we actually will end up giving patients replacement of IVIG to help if they’re having lots of infections.

Lisa Hatfield:

All right. Thank you. Another good question, and this comes up with many blood cancers or a lot of cancers. Should patients be mindful of beauty products such as shampoos, soaps, and sunblocks when in remission for follicular lymphoma?

Dr. Kami Maddocks:

That’s another great question. I am not aware of any data connecting those specific things.I think patients definitely should be wearing sunblock, because we know that a lot of patients with blood cancers can get secondary malignancies. So being careful of being…we also know, I should say, even patients who are getting treated can have a more sensitivity to the sun. So being careful with sun precautions, either avoiding the sun or wearing sunblock, making sure you’re covered when you go outside. I’ll even say I’ve seen a few patients who during treatment have gotten bad windburns. So your skin definitely can be more sensitive when you’re receiving therapies.

Lisa Hatfield:

All right, thank you. Let’s see here. This patient is asking if you are in remission for a long period of time after follicular lymphoma treatment, can you technically be cured in some cases, or are you considered to still have the cancer?

Dr. Kami Maddocks:

So that is a great question. There’s a term that’s used in follicular lymphoma called a functional cure. So we have patients that essentially get treated, and they live long enough that they die from something else without their follicular lymphoma ever relapsing. So while we say from what we know if somebody lives long enough that this disease is likely going to relapse at some point, there are patients that will be treated, and the disease will never come back.

Lisa Hatfield:

Okay. That’s helpful, thank you. Can patients facing follicular lymphoma be considered immunocompromised if they’re in remission?

Dr. Kami Maddocks:

I think this kind of goes back to when we talked about the immune system recovery that this can be a little bit of a complicated question, because it depends a little bit on the treatment that they got, how far out from the treatment they are and how many treatments they’ve had in the past. So, in general, if I have a patient that has received therapy, their counts have recovered, they in general look like…their lab work looks like their immune system, then in general I would say that they have an immune system that’s likely similar to somebody who didn’t have the follicular lymphoma, and they’re going to be able to fight infections and respond to vaccines.

I think what we do know is sometimes when patients get rituximab maintenance or obinutuzumab (Gazyva) maintenance or some of the chemotherapies there are some patients that can have a longer time that they’re immunosuppressed. So I think this is always something that’s good to ask your doctor for. In your specific situation with the treatment you received, when do you expect to have a regularly functioning immune system?

Lisa Hatfield:

For follicular lymphoma, what are the predictors of transformation and relapse, and what symptoms should patients be looking out for and tell their doctor about?

Dr. Kami Maddocks:

Yeah, so I think this is a great question. As far as just in everybody predicting when they’re going to progress, when they’re going to relapse, we don’t actually have great ways to do that right now. One of the things that has been shown to potentially predict things is for patients who do receive treatment if they have an early relapse, that suggests that their disease is going to behave more aggressively. As far as looking for relapse, things that people want to look for, not all patients will have symptoms but certainly if patients feel any lumps or bumps if they start…I like to tell my patients if you…patients usually know if something’s wrong.

So everybody’s going to have aches and pains, everybody’s going to have the normal infections, but if you’re not feeling well significant fatigue night sweats fevers are always something that we look for but that’s not something that everybody has. New pains, not feeling well, just kind of the inability to feel like you can keep up with what you’re doing daily, those are always things that you should at least call in to see if you should be evaluated. It’s important to know that follicular lymphoma patients are followed. As I said, you are followed forever. We do also watch your blood counts to make sure that we’re not seeing changes in blood counts, changes in lactate dehydrogenase which is a non-specific marker but something that we follow in lymphoma.

Lisa Hatfield:

Okay. Thank you. And one follow-up to that question also. So are there follicular lymphoma specialists? If a patient is maybe in an area that doesn’t have a large academic center or a large cancer institution, do you recommend they see somebody who specializes in follicular lymphoma or can they see even for a consult or do you think that their local hematologist oncologist is very familiar with that themselves? Do you have recommendations?

Dr. Kami Maddocks:

Yeah, so that’s a great question. Local, I think follicular lymphoma is common enough that a lot of our general oncologists who see everything see follicular lymphoma. I think it never hurts of course to ask about clinical trials. So if that’s something that might be available. If it’s available, it might be worth going to a specialist for. If there’s concerns, I think it’s always a good idea to get a second opinion to make sure that a patient is comfortable.

I think if a patient seems to have a more aggressive behaving follicular or if they’ve had a lot of different treatments, that’s also if you’re seeing a general oncologist at a time, that it’s good to see if there are clinical trials or if a specialist has anything new or different.

Lisa Hatfield:

Okay. Thanks. So we have a person asking, “Does lymphoma recurrence always happen in an aggressive manner?”

Dr. Kami Maddocks:

That’s a great question. The answer is no on that, and in fact lymphoma recurrence doesn’t always need to be treated just because it does recur. So when you look at follicular lymphoma, there are patients who are in a watch-and-wait period. When they’re diagnosed, they’ll eventually progress to requiring treatment or most…well, there are patients who might not. Once they require treatment, they get a time period without…most of them will get a time period without disease.

There are patients who will…that you’ll find lymph nodes growing on CT scans maybe that you’re doing monitoring for, but the patient will otherwise feel well. They won’t have, necessarily, very big lymph nodes. Their blood counts will be okay and you may say, okay, just like you had watch-and-wait to start with, we’re going to watch and wait right now with this relapse, because you don’t have any indications that are saying we need to treat this.

And again that doesn’t necessarily make our patient live longer. So you want to balance their quality of life and toxicities of treatment. There are patients who will…when they relapse, they will have indications for treatments, and then there are patients who will potentially have more aggressive relapses and be very symptomatic or have larger lymph nodes.

Lisa Hatfield:

Okay. All right. So one person says, “I’m currently in remission, what can I expect in my future? How long does remission last? And is treatment after remission the same as initial treatment?”

Dr. Kami Maddocks:

So that is very dependent on what a patient receives. So there are different kind…of a lot of our treatments we look at median times. When patients have relapse, that can be a little bit different for single agent antibody therapy versus antibody in combination with chemoimmunotherapy for how long that treatment remission lasts.

As far as we don’t typically reuse a treatment once we have used it before, although there is data in follicular lymphoma when patients receive single agent antibody. So rituximab alone, if they do well with that single agent immunotherapy for a long period, they may receive re-treatment with just that so long as they don’t have disease that requires more aggressive treatment.

Lisa Hatfield:

Okay. So is that more likely to happen then if a patient maybe wasn’t refractory to it, if they just stopped using it for some reason? Would that be more common for that to happen to go back on that same drug?

Dr. Kami Maddocks:

So with rituximab, we use it alone and in combination. So there are some patients that don’t necessarily have what we call a large tumor, and they don’t have a lot of lymph nodes or they don’t have large lymph nodes but they might be symptomatic from them or the location might be problematic. And so once these lymph nodes get a certain size, they usually don’t have as good of a response to single agent antibody therapy. But there are patients who have small lymph nodes that aren’t as big but again are causing a problem that can get completely…you give a short course of the rituximab, and it can last for a very long time and then you would consider again using a short course of that rituximab.

The chemotherapies we have, we don’t reuse chemotherapy, for the most part. Some of that, for a while, there was bendamustine if patients got five, six, 10-year remissions out of it. Sometimes they would re-get that chemotherapy. But I think we’ve just seen so many newer therapies approved in the last five six years. Like the bispecifics, the EZH2 inhibitors, lenalidomide, CAR T, we had different PI3K inhibitors available for a while. And so I think it was just that you had the ability to offer a patient something that they never had before, and that is more appealing.

Lisa Hatfield:

Okay. Thank you. So you spoke a little bit about IVIG infusions before and this person is saying that, “I’m having to do IVIG infusions, which started years after my treatment due to my IgG numbers being low. Are nausea and headache side effects common?”

Dr. Kami Maddocks:

Yeah, so some patients can have nausea, headache, myalgia, body aches, some get fevers and some infusion reactions. For some patients, they can have that from the start, some patients can develop it. That’s always a good thing to talk to your doctor about. There are different products for IVIG, and sometimes patients are able to switch products. I will say IVIG can be very insurance-dependent, so it’s also sometimes what…the formulation that insurance…an individual’s insurance covers. But yes, these are side effects, they’re worth, if they’re getting worse or lasting a long time, making sure that it’s discussed with the physician prescribing it.

Lisa Hatfield:

Okay. All right. One person is saying, “I’m in watch and wait currently. Is it possible that I’ll never need treatment or how long do you wait and what am I waiting for?”

Dr. Kami Maddocks:

That is a great question. There are patients in watch and wait who will never require treatment. Watching and waiting, we’re watching blood counts, watching the size of lymph nodes. So things that we’re watching for and you’re watching for are changes in lymph nodes size, so are they growing? Are they becoming more symptomatic? Is there a rapid change in them? Are we seeing a change in the blood counts? Are patients starting to have a drop in their blood counts which can happen if somebody’s spleen is getting bigger if they have lymphoma in their bone marrow and that’s progressing, watching for if the lymph nodes are causing a problem, you notice somebody have one in a location like the neck that’s starting to make swallowing difficult or changes in voice, that’s something you want to treat. And then there’s something called B symptoms that we watch for. So if the patient had night sweats…

 …night sweats are like drenching night sweats, soak the bed, have to change clothes potentially sheets, fevers, so daily fevers that occur, or significant or rapid weight loss for no reason.  All those are kinds of things that we want people to watch for. And we discussed a little bit too if patients start having extreme fatigue, not feeling well, not being able to eat, not having appetites if they have a new pain. And again everybody can have aches and pains. But if you’re having pain that’s not going away or some sort of symptom that’s not improving, those are all things we want to definitely have checked out.

Lisa Hatfield:

Okay. Thank you. I imagine with some of your patients in that mode, there’s what I call the mental gymnastics of thinking, okay, I have this cancer, but I can’t do anything about it, and these symptoms are really vague that come up. So do you allow your patients just to contact you if they’re saying, “I think I have these symptoms, I’m nervous about this.” Can they come in and have a visit with you or contact you at any time?

Dr. Kami Maddocks:

Oh, yes. So we have a 24-hour triage line. I recommend that if patients have a question or concern, it’s better to ask us because if we don’t know about it, we can’t help is the first thing. Usually, we talk to the patient and say, okay, how long has this been going on and see if it’s a red flag like you need to come in right now or is this something that maybe we might recommend getting a set of labs to look at certain labs to see if they’ve changed at all. We might say okay this seems like something we should actually see you for, but I want CT scans too so let’s order them so I can have that information when you see me.

So, yeah, I think people should always call with any signs, symptoms, concerns, and then it can be addressed. Now, there are some things that we might say, okay, we think based on everything that new cough is probably more likely a respiratory infection. It’s okay to see your PCP, but we also go through that as well. So yes, I think it’s always best to check in and not let something go.

Lisa Hatfield:

Okay. Thank you. I’m guessing that’s challenging for some of those people in that mode, just thinking, well, I’m just waiting here, so that’s got to be a little bit more challenging.

Dr. Kami Maddocks:

I think you’re absolutely right. And sometimes there’s a benefit to…certainly like rituximab therapy when there is a disease there, and it is a challenge to think that it’s not being treated.

Lisa Hatfield:

Okay. Thank you. This is I guess the last or second to the last question we have. Dr. Maddocks, can you speak to maintenance therapy and monitoring and follicular lymphoma, and what signs of infection should patients and care partners be aware of during treatment?

Dr. Kami Maddocks:

Yeah, so maintenance therapy, in follicular lymphoma is something, so maintenance antibody therapy after initial chemo…usually, chemoimmunotherapy is something that’s been studied that’s shown in some patients to provide a benefit as far as keeping disease away longer and a remission longer. But then it’s also been shown to be associated with a higher risk of infection, because you’re keeping those lymphocytes wiped out, particularly when it’s given as maintenance after certain chemotherapies in addition to the immunotherapy. So it can be a balance. I think maintenance isn’t something that every follicular lymphoma patient gets, but it’s something also that is used.

So that’s the first thing in discussion with your doctor, is this something that maintenance is recommended? Why or why not? Then watching during maintenance usually if people start to have more infections, which are oftentimes sinus respiratory infections, then we’re thinking about, okay, is this somebody that has low immunoglobulins? Do we need to check those? Are we worried about them? Needing to stop maintenance, potentially needing IVIG.

Lisa Hatfield:

Okay. Thank you. And then this is a Lisa question, “Do you know if there’s any data to suggest that the follicular lymphoma or any type of non-Hodgkin’s lymphoma has a familial or hereditary component?”

Dr. Kami Maddocks:

So there are very small number of…in non-Hodgkin’s lymphoma, there are some small familial components. Unlike, however, say breast cancer where there’s specific genes to test for, we don’t have that as a screening here in non-Hodgkin’s lymphoma.

Lisa Hatfield:

Dr. Maddocks, 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 very much, Dr. Maddocks.

Dr. Kami Maddocks:

Lisa, thank you so much for having me. It’s been a great conversation and hopefully it can help some people.

Lisa Hatfield:

I hope so too. I’m Lisa Hatfield. Thank you for joining this Patient Empowerment Network program.


Share Your Feedback:

Create your own user feedback survey

Follicular Lymphoma Monitoring and Maintenance: An Expert Weighs In

Follicular Lymphoma Monitoring and Maintenance: An Expert Weighs In from Patient Empowerment Network on Vimeo.

What do follicular lymphoma patients need to know about monitoring and maintenance therapy? Expert Dr. Sameh Gaballa shares research about maintenance treatment, maintenance therapy risks versus benefits, and virtual visits for consults and second opinions.

Dr. Sameh Gaballa is a hematologist/oncologist specializing in treating lymphoid malignancies from Moffitt Cancer Center. Learn more about Dr. Gaballa.

See More from START HERE Follicular Lymphoma

Related Resources:

Follicular Lymphoma Disease Transformation and Secondary Cancer Risk

Follicular Lymphoma Disease Transformation and Secondary Cancer Risk 

How Can Follicular Lymphoma Treatment Side Effects Be Reduced?

Understanding Follicular Lymphoma Disease Progression Symptoms and Monitoring


Transcript:

Lisa Hatfield:

Can you speak to maintenance therapy and monitoring in follicular lymphoma? And what signs of infection should patients and care partners be aware of during treatment?

Dr. Sameh Gaballa:  

Yeah, so there have been randomized studies in slow-growing lymphomas that show that if you do, after you get your standard treatment for follicular lymphoma, if you do what we call a maintenance treatment, usually with rituximab, which is an immune therapy, where you do it every two to three months for about two years, we have data showing that that decreases or delays the risk of relapse. However, it doesn’t change the overall survival, meaning that it just has patients in remission longer. When their disease comes back, they just get treated again at that point, and it doesn’t really affect survival.

So it’s one of those shared decision-making with the patients. I usually go over the risks and benefits of maintenance therapy. It’s optional. It’s not a must. During COVID, we pretty much stopped all maintenance treatments, because the risks were outweighing the benefits because maintenance treatment is…will suppress the immune system more, is associated with more infections. And these infections can be anything. I mean, it could be a pneumonia, could be recurrent urinary infections. It could be any type of infection. So there’s always this risk and benefit that we have to discuss with the patient.

Lisa Hatfield:

One thing that comes up, patients, I live in a state that we don’t have a lot of specialists for my type of cancer, for myeloma. If a patient wanted to consult with you, do you see patients via Zoom? Do you do consultations or maybe, I don’t want to call it a second opinion, but a consultation virtually for patients?

Dr. Sameh Gaballa:

So COVID has changed a lot of things. And that’s one of those things that have changed the way that we work, meaning offering virtual visits. And yes, sometimes we do, someone just wants to make sure they’re on the right track. They want us to review the records, review the diagnosis. We’ll get the biopsy slides reviewed here by our pathologist. And just to go over and make sure that they’re on the right track.

So yes, we routinely do those second opinions sometimes. Obviously in-person is usually better, because then we could do our own testing, and we could also examine the patients and see how they are. But yes, a virtual option is available. There are some restrictions sometimes. We cannot now see out-of-state patients as we used to, but during COVID that was not the case. But now we would have to see patients only in our state.


Share Your Feedback:

Create your own user feedback survey

Relapsed/Refractory Follicular Lymphoma Treatments and Bispecific Antibodies

Relapsed/Refractory Follicular Lymphoma Treatments and Bispecific Antibodies from Patient Empowerment Network on Vimeo.

What can relapsed/refractory follicular lymphoma patients expect for current and future treatment options? Expert Dr. Sameh Gaballa explains what treatments are currently available and ones that are being studied for the future.

Dr. Sameh Gaballa is a hematologist/oncologist specializing in treating lymphoid malignancies from Moffitt Cancer Center. Learn more about Dr. Gaballa.

See More from START HERE Follicular Lymphoma

Related Resources:

Follicular Lymphoma Monitoring and Maintenance: An Expert Weighs In

Follicular Lymphoma Monitoring and Maintenance: An Expert Weighs In

How Can Follicular Lymphoma Treatment Side Effects Be Reduced?

Understanding Follicular Lymphoma Disease Progression Symptoms and Monitoring


Transcript:

Lisa Hatfield:

Another patient is asking if you can speak to emerging treatment options for patients with relapsed/refractory follicular lymphoma?

Dr. Sameh Gaballa:

Yeah. So the field of follicular lymphoma is changing rapidly. I always tell patients that sometimes the best treatment is actually on a clinical trial because those are going to be the next generation of treatments that are going to get approved in the next few years. But right now we have the most effective therapy really is CAR T-cell therapy. CAR T-cell therapy by far is the most effective treatment we have at this time. It’s approved for patients who have had two or more lines of prior therapies. We also are investigating this. I actually have a trial here at Moffitt where we’re looking at CAR T-cell therapy as early as in the second line, in patients who have what we call the high-risk ones, the POD24. So a patient with POD24 follicular lymphoma relapsed in less than two years. We have a trial to investigate the role of CAR T-cell therapy in this setting. The other very promising group of treatments, again, is bispecific antibodies, again, currently approved in the third line, mosunetuzumab-axgb (Lunsumio).

But there are others coming up and have data on epcoritamab-bysp (Epkinly), as well as a lot of other bispecifics, as well as combinations. I mean, epcoritamab-bysp has also data presented with combination with lenalidomide. And right now, the follow-up duration is not very long, but so far, it looks extremely promising with very high response rates. So those also might be coming very soon. And, of course, once something works in the relapsed/refractory setting, we start looking at earlier lines of therapy. And actually, we’re now looking at trials in the first-line setting with some of these agents as well. Tazemetostat (Tazverik) is a pill. It’s also approved in the third-line setting, but we’re also investigating it. We have a trial here where we’re looking at combining it with standard rituximab (Rituxan), lenalidomide (Revlimid), so tazemetostat plus rituximab, lenalidomide as early as in the second line. So that also is interesting. And as I mentioned before, BTK inhibitors currently being looked at in trials might also have a role in follicular lymphoma very soon.

Lisa Hatfield:

And this patient is asking about the significance of bispecific antibody treatment. And you touched on that a little bit. It looks like she’s also asking if there are specific genetic or molecular markers that can predict a patient’s response. And if I try to translate that, maybe she might be asking about targeted therapy.

Dr. Sameh Gaballa:

Yeah, so bispecific antibodies and CAR T-cell therapy, they target something called CD, either CD19 or CD20, and that’s almost universally expressed on B cells. So most of your follicular lymphoma patients are going to be expressing CD19 or CD20. Tazemetostat is the pill that I talked about. It inhibits an enzyme called EZH2. Some patients have an EZH2 mutation where it seems to work very well. However, tazemetostat also works in patients who don’t have that mutation. So that’s why it’s not very important to check for the mutation.

It seems maybe it works better in patients who do have the mutation, but it does work as well in patients who do not have that mutation. So unlike other malignancies and other cancers, biomarkers are not yet driving a lot of our treatment decisions in follicular lymphoma as of right now.

Lisa Hatfield:

How exactly do bispecific antibodies engage the patient’s immune system to target and eliminate follicular lymphoma cells?

Dr. Sameh Gaballa:

So bispecific antibodies are a very interesting class of medicines. It’s an antibody that has two ends to it. So one end would target the patient’s own immune cells, meaning that they would attach the antibody to the patient’s own immune cell and then the other end of the antibody engages the cancer cell. So it’s basically hand-holding the patient’s own immune system to go and kill the cancer cell. And this is not just in lymphoma. It’s looked at in multiple myeloma as well, approved therapies there. And a lot of other cancers, we have bispecific antibodies being developed in clinical trials right now. 


Share Your Feedback:

Create your own user feedback survey

How Can Follicular Lymphoma Treatment Side Effects Be Reduced?

How Can Follicular Lymphoma Treatment Side Effects Be Reduced? from Patient Empowerment Network on Vimeo.

Follicular lymphoma patients may experience treatment side effects, but how can they be minimized? Expert Dr. Sameh Gaballa shares diet and lifestyle advice to help reduce the impact of treatment side effects.

Dr. Sameh Gaballa is a hematologist/oncologist specializing in treating lymphoid malignancies from Moffitt Cancer Center. Learn more about Dr. Gaballa.

See More from START HERE Follicular Lymphoma

Related Resources:

Follicular Lymphoma Monitoring and Maintenance: An Expert Weighs In

Follicular Lymphoma Monitoring and Maintenance: An Expert Weighs In

Relapsed/Refractory Follicular Lymphoma Treatments and Bispecific Antibodies

Relapsed/Refractory Follicular Lymphoma Treatments and Bispecific Antibodies

Understanding Follicular Lymphoma Disease Progression Symptoms and Monitoring


Transcript:

Lisa Hatfield:

We have another question about treatment profiles, “What can I do to reduce side effects during active treatment?”

Dr. Sameh Gaballa:

So it depends on what the treatment that you’re getting. If it’s immune therapy, like rituximab (Rituxan) alone, those typically don’t really have a lot of side effects. I mean, sometimes with the first one or two treatments, you might get an allergic reaction, an infusion allergic reaction, which is very common, but subsequently it shouldn’t really cause a lot of side effects.

dIf the patient is getting chemotherapy, well, it depends on which chemotherapy they’re getting. But in general, it’s always good to stay hydrated and to stay physically active. So if the patient goes in with a healthy body, well-hydrated, you eat fresh fruits and vegetables, walking 30 to 60 minutes a day, your body is going to handle the side effects much better than if you’re going in, you’re very weak, and your general health is not adequate.


Share Your Feedback:

Create your own user feedback survey

Newly Diagnosed With Follicular Lymphoma? Start Here

Newly Diagnosed with Follicular Lymphoma? Start Here from Patient Empowerment Network on Vimeo.

How are follicular lymphoma treatment options commonly explained to patients? Expert Dr. Sameh Gaballa shares how he walks patients through treatment options, POD24 and FLIPI tests that help guide treatment options, and follicular lymphoma staging.

Dr. Sameh Gaballa is a hematologist/oncologist specializing in treating lymphoid malignancies from Moffitt Cancer Center. Learn more about Dr. Gaballa.

See More from START HERE Follicular Lymphoma

Related Resources:

What Exactly is Follicular Lymphoma? An Expert Explains

What Exactly is Follicular Lymphoma? An Expert Explains

Follicular Lymphoma Patient Expert Q&A: Start Here

Follicular Lymphoma Patient Expert Q&A: Start Here

What Follicular Lymphoma Treatments Are Available?


Transcript:

Lisa Hatfield:

So, Dr. Gaballa, let’s start here. How do you explain follicular lymphoma treatment options and prognosis to your newly diagnosed patients? And what does shared decision-making look like in your office?

Dr. Sameh Gaballa:

Oh, absolutely. So follicular lymphoma, you really have to explain to the patient what, how are we coming to the recommendation that we’re currently giving. So if we think this is, this patient is a good candidate for a watch-and-wait approach, for example, we really have to walk them through why that really is the best option and not why should we jump on treatments and vice versa, if we think this patient needs to be treated, how do we really…the patient really has to understand all the other treatment options and why this needs to be treated. Because a lot of patients initially, sometimes when you present them with a watch-and-wait approach, if they don’t know all the background, they might not feel very comfortable because they might think, “Well, I have this cancer in me, and we’re not doing anything about it, and that doesn’t really sound too…something I should be doing.”

But then when you explain to them, “Well, you see, you don’t have a lot of disease, those studies have already been done in the past where patients who were treated or not treated, the survival was the same, so there, you might get side effects from the treatment, but not necessarily have benefits. And in the future, should this need to be treated, we have a lot of things to do.” So, really, so this is kind of the shared decision portion where you just have to walk the patients through why that will be the best situation. There is data with single-agent rituximab (Rituxan), even in patients who are asymptomatic, and we have the UK data, and that’s an option.

And that is also offered to some of the patients, even if they’re not symptomatic and they don’t have a lot of disease, if that’s what really the patient wants, if they’re not really comfortable with a watch and wait. And there’s again some data to help justify that. Again, there’s no advantage in overall survival, but sometimes the patients would kind of feel more in control. They feel like, “Okay, I did something about it.” So that’s the shared approach.

In terms of your other question about prognosis, unfortunately that’s an area of an unmet need. I mean, we have some tools to help us differentiate follicular lymphoma patients from each other, which patient is high-risk, meaning those are the patients who might relapse quickly, or they might not respond well to treatments. Unfortunately, we don’t have great tools. We have something called a FLIPI score, which is, we use a number of parameters including clinical parameters like stage or age and some other parameters as well, and we have a scoring system. But it doesn’t 100 percent predict if this is going to be a high-risk follicular lymphoma or a low-risk.

Unfortunately, the best predictor of prognosis for follicular lymphoma, you would know about retrospectively, it’s something called POD24, progression of disease in 24 months. Meaning that if you have a patient who’s treated with chemotherapy and immune therapy, and then they go into remission, and then they relapse again in less than 24 months, progression of disease within 24 months, those are the, those represent about 20 percent of follicular   lymphoma patients, and those represent a high-risk group of patients. That’s the best tool that we have. But unfortunately, if you’re diagnosed today, you’re not going to know if you’re in this group or not until you actually need to be treated and not just treated with immune therapy.

It has to be with chemotherapy as well. And then if you relapse within two years, then we know that this is a high-risk entity. There is genetic testing, there is something called a FLIPI-m7 scoring system. But again, these tools are not great to tease out the low risk from the high-risk follicular lymphoma patients. But 80 percent of patients who are not going to be POD24, meaning that they get treated, they’re in remission for two years or more, and actually those patients have very similar survival to the general population. So, yeah, so a lot of times we don’t know right away, but we do have some tools to kind of give us an idea.

Lisa Hatfield:

Great. Thank you for that information. It’s kind of hard for cancer patients to only know what their prognosis is retrospectively, but that’s a great explanation. Thank you. Okay, another patient question, “How does the staging of follicular lymphoma impact treatment choices?”

Dr. Sameh Gaballa:

Yeah, so as you saw, I didn’t really stress too much about staging, because it’s a blood disease. So the vast majority of patients are going to be what we call stage III to IV disease. So, obviously when you see a patient if if they, they might think that, “Oh my God, I have a stage III to IV cancer,” because that’s really what they’re familiar with. But follicular lymphoma is a blood disease, so by default it’s going to be in a lot of lymph nodes, it might be in the bone marrow as well, but stage III to IV disease follicular lymphoma doesn’t, that does not mean that this is a terminal cancer. Patients could live completely in normal life, even with a stage III to IV follicular lymphoma. This is not like a breast cancer or colon cancer where stage is everything.

But why do we have a staging system? Obviously, there’s a need to have staging system for all cancers, but clinically, the only time it makes a difference is there’s a small group of patients who have a truly stage I or II disease, meaning just one group of lymph nodes on one side of the diaphragm that may fit within one radiation field. So if you have someone who’s just coming in with one or a few groups of lymph nodes all in one place, we call that a stage I or II follicular lymphoma, not common, because again, most patients are stage III to IV. The only difference there is you can potentially offer those patients radiation therapy if it’s truly localized, but then you would need to do a bone marrow biopsy and confirm that it’s not in the bone marrow.

And if it is localized within one radiation field, that can be offered and we can sometimes give after radiation therapy, either observe it or consider giving rituximab afterwards. But that’s the only time where we’re going to mention staging, again, uncommon because most, the vast majority of patients are going to be stage III to IV. So why would we do that? Why would we irradiate if it’s only one group of lymph nodes? Because there’s about, I mean, if you irradiated, those lymph nodes will go away, but there’s about maybe a, it’s different. The number is different between studies, but about maybe a third of patients, if you irradiate that group of lymph nodes or one lymph node, it actually might not come again in the future. So you might have very long remissions/possible cure if you…and this is the only situation where we would consider treating someone who does not have symptoms, because you could have very long remissions with radiation. 


Share Your Feedback:

Create your own user feedback survey

Can CLL Remission Occur With Rheumatoid Arthritis Drug Treatments?

Can CLL Remission Occur With Rheumatoid Arthritis Drug Treatments? from Patient Empowerment Network on Vimeo.

Is it possible for chronic lymphocytic leukemia (CLL) remission to occur from rheumatoid arthritis treatments? Expert Dr. Ryan Jacobs explains what he’s observed in his CLL patients who also have RA and take RA treatments.

Dr. Ryan Jacobs is a hematologist/oncologist specializing in chronic lymphocytic leukemia from Levine Cancer Institute. Learn more about Dr. Jacobs.

See More from START HERE CLL

Related Programs:

Why Does Bruising Occur in Chronic Lymphocytic Leukemia?

Which Oncologists Are Chronic Lymphocytic Leukemia Experts?

Which Oncologists Are Chronic Lymphocytic Leukemia Experts?

Can Bone Marrow Return to Normal After CLL Treatment?

Can Bone Marrow Return to Normal After CLL Treatment?


Transcript:

Lisa Hatfield:

Do any rheumatoid arthritis medications help prevent CLL from returning after treatment with FCR?

Dr. Jacobs:  

I do have a fair number of patients that have rheumatologic conditions and some with rheumatoid arthritis. There are some approvals there, and I in no way pretend to be an expert in rheumatoid conditions. But I do know that there happens to be some agents that are monoclonal antibodies directed against CD20 used to treat some rheumatoid conditions. So I do have some patients that are on drugs like rituximab (Rituxan) to suppress their rheumatoid condition and help prevent recurrences.

And then kind of two birds, one stone also are keeping their CLL in a clinically asymptomatic remission, I’m sure I would say, or stable disease. And it comes with the known risk for long-term antibodies, that there are some increased infections there that was particularly concerned during COVID, the worst parts of COVID. But yeah, so there are some potential treatments like that.


Share Your Feedback

Create your own user feedback survey

CLL and Vaccines | Vital Advice for Protecting Patients

CLL and Vaccines | Vital Advice for Protecting Patients from Patient Empowerment Network on Vimeo.

 What do chronic lymphocytic leukemia (CLL) patients need to know about vaccines? Expert Dr. Ryan Jacobs explains CLL treatments that reduce vaccine response and his vaccine recommendations.

Dr. Ryan Jacobs is a hematologist/oncologist specializing in Chronic Lymphocytic Leukemia from Levine Cancer Institute. Learn more about Dr. Jacobs.

Download Resource Guide   |  Descargar Guía en Español

See More from START HERE CLL

Related Programs:

How Can I Ensure My CLL Doesn't Progress to Richter's Transformation?

How Can I Ensure My CLL Doesn’t Progress to Richter’s Transformation?

CLL and BTK Inhibitor Treatment: What Are the Risk Factors?

CLL Genetic Markers: What Should I Ask About Prognostic Factors?

CLL Genetic Markers: What Should I Ask About Prognostic Factors?


Transcript:

Lisa Hatfield:

So we have another patient who has asked a series of questions. Her first question is, “Can you speak to immune vulnerability and the importance of regular vaccination for CLL patients?”

Dr. Ryan Jacobs:

Yes. So we know that having active CLL reduces a patient’s ability to respond to vaccination and increases redirection, we know being on treatment for CLL also produces varying risk depending on the treatment. The drugs that seem to do the most damage to the immune system, and specifically in terms of their ability to respond to vaccination or the antibody treatment like rituximab (Rituxan) and obinutuzumab (Gazyva), and their effects last for many months after that treatment is finished. Unlike the oral drugs which have a short half-life, the antibodies hang around for many months after being administered.

I in general am recommending, as does the CDC, to get boosted every six months for patients with any level of immune suppression and having CLL qualifies you as that. And then I recommend all of the general vaccines that come with age, like, for example, the Shingrix vaccine for shingles is now safe to give to CLL patients because it’s a conjugate vaccine, it’s not a live virus vaccine.

So we’re lucky now with just standard vaccines in the U.S., there are no live virus vaccines that the CLL patient has to worry about anymore, so I definitely encourage shingles, pneumonia vaccines, boosting for COVID. We’ll see if we get an RSV vaccine, that sounds like it’s on the horizon. Flu, of course. And the patient should just be aware based on what kind of treatment that they’re on, they may not have a good chance at responding to these vaccines, but I still try with my patients. The other important element to think about when you’re considering an infection risk and everything is just kind of what’s…obviously, the pandemic has been a very dynamic thing, and certain times there’s been a lot more risk than others. Thankfully, at the time of this recording, we’re doing on probably as good as we’ve done since the onset of COVID. So you have to make your decisions on the situations you put yourself into, based on your personal situation and what’s going on in the bigger picture, risk-wise. Flu season, COVID season, a lot of RSV going around or something like that.


Share Your Feedback

Create your own user feedback survey

Diagnosed With CLL? Start Here

Diagnosed with CLL? Start Here from Patient Empowerment Network on Vimeo.

What do newly diagnosed chronic lymphocytic leukemia (CLL) patients need to know? Expert Dr. Ryan Jacobs explains how CLL occurs and provides an overview of treatment types. 

Dr. Ryan Jacobs is a hematologist/oncologist specializing in Chronic Lymphocytic Leukemia from Levine Cancer Institute. Learn more about Dr. Jacobs.

Download Resource Guide   |  Descargar Guía en Español

See More from START HERE CLL

Related Programs:

CLL Patient Expert Q&A: Start Here

CLL Patient Expert Q&A: Start Here

IGHV-Mutated vs IGHV-Unmutated CLL | What’s the Difference

Is It Aging or My CLL?

Is It Aging or My CLL?


Transcript:

Lisa Hatfield:

There’s a lot going on in terms of novel therapies and new options. But before we jump into all of that, the tool box that’s expanding, can you introduce CLL and provide an explanation of what it is and what that means for a newly diagnosed patient?

Dr. Ryan Jacobs:

So chronic lymphocytic leukemia, or CLL, is the most common chronic lymphoma/leukemia. It is really both most of the time. It presents with what we call lymphocytosis, meaning the white blood cell count, and specifically the lymphocyte count is high or elevated. To call it CLL, to make that formal cancer diagnosis, we can generally take a patient’s blood sample and put it through a fancy machine that we call a flow cytometer, that looks for characteristic markers on the outside of the cells, and if there’s a bunch of those cells, we call that a monomorphic population.

So a bunch of those cells that look the same, and they’re B cells, that’s the type of lymphocyte count they are, and they’re over this threshold of 5,000. Then that is the diagnosis right there, we don’t need an invasive procedure. You generally do not need a bone marrow biopsy or a lymph node biopsy. There is in about 15 percent of cases, the disease presents with just in large nymph nodes, and the white count is normal. We call that small lymphocytic lymphoma. It’s considered an overlap with CLL, and I’ve had many patients that have started off with SLL and then eventually manifest elevated lymphocyte count later in their disease course. So it is considered an overlap, the treatment is the same for both of those disease entities.

So that’s the diagnosis of CLL and how it generally shows up initially. In a nutshell, it’s a cancer of the aging population, average age is 70. I have a lot of patients that ask me, “Why did I get CLL?” And the answer is, “We don’t know.” It’s that way with most cancers, unfortunately, we don’t know why one person gets a cancer and the other person doesn’t.

But it obviously has something to do with the aging effect on the DNA of the B lymphocytes because of how much more common it is as patients get older. 

Lisa Hatfield:

We know that therapies are evolving faster, hopefully faster than patients are relapsing, which is a good thing. So when they do relapse, chances are there will be a new option for this patient. But a CLL cure still remains elusive.

So, Dr. Jacobs, if you can speak to…we’ll just jump right into some of the newer, the novel therapies and things that are being investigated with CLL treatment. If you can just speak to some of those newer therapies, the novel pathways and targets that are currently under investigation with CLL, we’d appreciate that.

Dr. Ryan Jacobs:

Sure. We’ve come a long way in how we are managing this common cancer that’s benefiting a lot of patients. And as mentioned, with this cancer being one that is more common in the older population, we do know that the population of the United States specifically is getting older, there’s going to be more 70-year-olds. So these breakthroughs are helping a growing number of CLL patients.

Before 2014, really outside of a clinical trial, the only way we could treat CLL Is with combinations of chemotherapy and an immune therapy, like a monoclonal antibody called rituximab (Rituxan), that was kind of our first, what we would call targeted treatment outside of chemotherapy that we had, and it was, like targeted treatments are, well tolerated. It was an antibody that targets B cells specifically.

So we were combining it with chemo, we would call that chemoimmunotherapy, and it helped a lot of CLL patients. But for many, those were poor prognostic markers in particular, and those with relapse disease, chemotherapy was not very helpful, and it was quite toxic in many circumstances.

So we’ve been fortunate that since 2014, we’ve had a lot of new treatment options, and they’re targeted therapies. It’s not like non-specific cytotoxic chemotherapy, these are treatments that have been developed with specific targets in mind that are unique to the B-cell neoplasm, the B-cell cancer, the CLL. And the first of these that really changed everything was a BTK inhibitor called ibrutinib (Imbruvica), that we got in 2014.

Initially, we can only use it in the relapse setting, but eventually, in 2016, we could start treating patients as a first line of therapy with ibrutinib.  And then in 2019, we had a newer version of BTK inhibitor, we call those second-generation BTK inhibitors. That drug was acalabrutinib. And it eventually was shown in a head-to-head study to be just as effective as ibrutinib in a relapsed patient population, but it had less side effects than ibrutinib. So in, specifically, atrial fibrillation, hypertension. Cardiac toxicities overall were one that they really focused on in that study.

So as a whole, when we were choosing BTK inhibitors, we were shifting away from ibrutinib, shifting to acalabrutinib, and then just as…earlier this year, we had a third BTK inhibitor, zanubrutinib (Brukinsa), that was approved. It’s also considered a second generation BTK inhibitor like ibrutinib and acalabrutinib (Calquence). It treats CLL in the same way, in how it inhibits BTK or Bruton’s tyrosine kinase, that’s over expressed in CLL cells.

But it also has a favorable toxicity profile when compared head to head with ibrutinib, and now we have two second-generation options between acalabrutinib and zanubrutinib. And it’s not really easy for us to know between those two, which is “better.” When we decide to treat with a BTK inhibitor, we’re usually choosing between those two at this point, and we’re trying to personalize the decision for the patient, and there are some different factors they can get involved in that complicated decision.

Luckily, we are not limited to BTK as a target. I mentioned earlier, we have monoclonal antibody, rituximab like the one I mentioned, but a newer version of rituximab, a more potent version, obinutuzumab. Is one that we have available along with a Bcl-2 inhibitor, venetoclax (Venclexta). That is now, as of 2018, improved in the frontline CLL setting, also approved in the relapsed setting, of course, since 2016.  And we use venetoclax with a monoclonal antibody like obinutuzumab (Gazyva), and together they are a very potent combination that cause pretty rapid cancer cell death, as opposed to the BTK inhibitors that more put the cancer to sleep and require daily dosing indefinitely for as long as the drugs work.

And remarkably, the data on BTK inhibitors tells us that should work for many, many years. They’ve been following some of the patients that got treatment in the first line setting, and eight years out, there’s still more than 50 percent of the patients are free of progression, so they can’t even quote an average response time yet. With eight years of follow-up on the early ibrutinib patients.  The difference with venetoclax combined with a monoclonal antibody like obinutuzumab, is because it causes a more rapid cell death, you can give it on a time-defined schedule. So we tend to give it for one year in the first-line setting and two years in the relapse setting, and the antibody portion is just given for the first six months. 

The BTK inhibitors and venetoclax are oral treatments, so that’s a big win for patients to avoid the infusion center for those treatments, but the IV antibody treatments will still require some trips to the infusion center if you’re doing that combination with venetoclax. For most patients, those two targets are what we’re choosing between, and we try to personalize the decision to the patient. And again, that’s a very complicated discussion on what is “best.” And we use things like the prognostic work-up, medical problems that the patients already have, medicines that patients are on, to help make the best treatment decision for our CLL patients. But those…for in terms of how well that treats the CLL, both of those are considered equivalent options for the large majority of CLL patients.

We’ve got some things on the horizon, but in general, those are two targets that we have at this point. There is for relapsed patients, PI3 kinase inhibitors that are still FDA-approved at this time, that aren’t quite as effective and more toxic, so we sometimes think about using one of those targeted therapies if a patient has already progressed on a BTK inhibitor in venetoclax class. In the future, we are looking towards combining BTK and Bcl-2 inhibitor. Like, for example, there’s been studies already done that I’ve put many patients on, with ibrutinib-venetoclax, and I believe there’s a question about that later.

There’s also an ongoing study that I have opened at my institution that’s looking at acalabrutinib and venetoclax. So taking these two pills together in a time-defined manner, so you don’t have to take the BTK inhibitor indefinitely. And then there are some therapies that have already been improved in other lymphomas, and we wonder if they’re going to have a role in CLL eventually. So we now have bispecific antibodies, so that’s taking a drug like rituximab or obinutuzumab and adding a T-cell engager to it, so it has two targets or it’s bispecific.

And we have that drug, mosunetuzumab (Lunsumio) available in follicular lymphoma and there’s several others in development, and we’ll see how their role comes into play in CLL as well. As well as CAR T-cell therapy, where we take a patient’s T cells and genetically engineer them to attack the cancer. That’s now an approved therapy for many different kinds of lymphomas and multiple myeloma as well.

So we wonder if that’s going to have a role in CLL. But I think for the foreseeable future, it’s going to be looking first at BTK and then Bcl-2 inhibition, or vice versa. And we don’t really know which is better to go first, we think they’re both…they can both be sequenced one after the other. And then maybe it will have some of these other breakthroughs coming in and helping for after patients need something beyond those therapies.

And there’s probably going to be a lot of patients that never need anything beyond those options, those initial couple of targets, because they do so well. I think in the most immediate future, the approval that is going to give us a new great option is going to be for an alternative site BTK inhibitor, or it’s also called a non-covalent BTK inhibitor.

And there’s this drug called pirtobrutinib (Jaypirca), it has been approved in mantle cell lymphoma and likely will get approved in CLL this year. And that drug specifically is a BTK inhibitor that still works even in patients that have, say, progressed on ibrutinib or acalabrutinib or zanubrutinib. That will be a new target available for CLL patients and probably pretty quickly become one of the go-to drugs that we use for relapsed CLL patients that have already been treated with a BTK…with a traditional BTK inhibitor. So growing number of options and it’s really great for our CLL patients. 


Share Your Feedback

Create your own user feedback survey

CLL Patient Expert Q&A: Start Here

CLL Patient Expert Q&A: Start Here from Patient Empowerment Network on Vimeo.

The START HERE program bridges the CLL expert and patient voice, whether you are newly diagnosed, in active treatment or in watch and wait. In this webinar, Empowerment lead Lisa Hatfield and expert Dr. Ryan Jacobs  provide an overview of the latest in CLL, managing CLL side effects and options for CLL progression.

Dr. Ryan Jacobs is a hematologist/oncologist specializing in Chronic Lymphocytic Leukemia from Levine Cancer Institute. Learn more about Dr. Jacobs.

Download Resource Guide   |  Descargar Guía en Español

See More from START HERE CLL

Related Programs:

Diagnosed with CLL? Start Here

Diagnosed with CLL? Start Here

Emerging CLL Research: Understanding the CAPTIVATE and MAJIC Studies

Can CLL Treatment Cause Gastrointestinal Side Effects?

Can CLL Treatment Cause Gastrointestinal Side Effects?


Transcript:

Lisa Hatfield:  

Hello and welcome. My name is Lisa Hatfield, your host for this Patient Empowerment Network program. In this important dialogue, we bridge the expert and patient voice 

to enable you and me to feel comfortable asking questions of our healthcare teams with more precision. The world is complicated, as is a cancer diagnosis, but understanding your CLL doesn’t have to be. The goal is to create actionable pathways for getting the most out of CLL treatment and survivorship. Joining me today is Dr. Ryan Jacobs, a CLL expert from Levine Cancer Institute. Thank you very much for joining us today, Dr. Jacobs, we really appreciate you being here and your time and expertise.

Dr. Ryan Jacobs:

Thanks for having me, Lisa.

Lisa Hatifield:

Before we get started, 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. Okay, let’s get started. So, Dr. Jacobs, I’d like to talk about what’s on the chronic lymphocytic leukemia radar, and rather than saying that entire phrase each time, I’m going to refer to it as CLL, because I’m pretty sure I’ll fumble that up. There’s a lot going on in terms of novel therapies and new options, but before we jump into all of that, the tool box that’s expanding, can you introduce CLL and provide an explanation of what it is and what that means for a newly diagnosed patient?

Dr. Ryan Jacobs: 

So chronic lymphocytic leukemia, or CLL, is the most common chronic lymphoma/leukemia. It is really both most of the time. It presents with what we call lymphocytosis, meaning the white blood cell count, and specifically the lymphocyte count is high or elevated. To call it CLL, to make that formal cancer diagnosis, we can generally take a patient’s blood sample and put it through a fancy machine that we call a flow cytometer, that looks for characteristic markers on the outside of the cells, and if there’s a bunch of those cells, we call that a monomorphic population.

So a bunch of those cells that look the same, and they’re B cells, that’s the type of lymphocyte count they are, and they’re over this threshold of 5,000. Then that is the diagnosis right there, we don’t need an invasive procedure. You generally do not need a bone marrow biopsy or a lymph node biopsy. There is in about 15 percent of cases, the disease presents with just in large nymph nodes and the white count is normal. We call that small lymphocytic lymphoma. It’s considered an overlap with CLL, and I’ve had many patients that have started off with SLL and then eventually manifest elevated lymphocyte count later in their disease course. So it is considered an overlap, the treatment is the same for both of those disease entities.So that’s the diagnosis of CLL and how it generally shows up initially. In a nutshell, it’s a cancer of the aging population, average age is 70. I have a lot of patients that ask me, “Why did I get CLL?” And the answer is, we don’t know. It’s that way with most cancers, unfortunately, we don’t know why one person gets a cancer and the other person doesn’t. But it obviously has something to do with the aging effect on the DNA of the B lymphocytes because of how much more common it is as patients get older.

Lisa Hatifield:

Thank you for that overview, Dr. Jacobs. We do have CLL patients who are watching this who are newly diagnosed, they may be in active treatment, they may be in remission, they may be managing their CLL just fine right now in their lives. So we’re along the whole spectrum of CLL, so thank you for that overview. We know that therapies are evolving faster, hopefully faster than patients are relapsing, which is a good thing. So when they do relapse, chances are there will be a new option for this patient. But a CLL cure still remains elusive. So, Dr. Jacobs, if you can speak to…we’ll just jump right into some of the newer, the novel therapies and things that are being investigated with CLL treatment. If you can just speak to some of those newer therapies, the novel pathways and targets that are currently under investigation with CLL, we’d appreciate that.

Dr. Ryan Jacobs:

Sure. We’ve come a long way in how we are managing this common cancer that’s benefiting a lot of patients. And as mentioned, with this cancer being one that is more common in the older population, we do know that the population of the United States specifically is getting older, there’s going to be more 70-year-olds. So these breakthroughs are helping a growing number of CLL patients.

Before 2014, really outside of a clinical trial, the only way we could treat CLL Is with combinations of chemotherapy and an immune therapy, like a monoclonal antibody called Rituximab, that was kind of our first, what we would call targeted treatment outside of chemotherapy that we had, and it was, like targeted treatments are, well tolerated.  It was an antibody that targets B cells specifically. So we were combining it with chemo, we would call that chemoimmunotherapy, and it helped a lot of CLL patients. But for many, those were poor prognostic markers in particular, and those with relapse disease, chemotherapy was not very helpful, and it was quite toxic in many circumstances. So we’ve been fortunate that since 2014, we’ve had a lot of new treatment options, and they’re targeted therapies. It’s not like non-specific cytotoxic chemotherapy, these are treatments that have been developed with specific targets in mind that are unique to the B-cell neoplasm, the B cell cancer, the CLL.

And the first of these that really changed everything was a BTK inhibitor called ibrutinib, that we got in 2014. Initially, we can only use it in the relapse setting, but eventually, in 2016, we could start treating patients as a first line of therapy with ibrutinib (Imbruvica). And then in 2019, we had a newer version of BTK inhibitor, we call those second-generation BTK inhibitors. That drug was acalabrutinib (Calquence). And it eventually was shown in a head-to-head study to be just as effective as ibrutinib in a relapsed patient population, but it had less side effects than ibrutinib. So in specifically, atrial fibrillation, hypertension. Cardiac toxicities overall were one that they really focused on in that study. So as a whole, when we were choosing BTK inhibitors, we were shifting away from ibrutinib, shifting to acalabrutinib, and then just as…earlier this year, we had a third BTK inhibitor, zanubrutinib (Brukinsa), that was approved. It’s also considered a second-generation BTK inhibitor like ibrutinib and acalabrutinib.

It treats CLL in the same way, in how it inhibits BTK or Bruton’s tyrosine kinase, that’s overexpressed in CLL cells. But it also has a favorable toxicity profile when compared head to head with ibrutinib, and now we have two second generation options between a acalabrutinib and zanubrutinib. And it’s not really easy for us to know between those two, which is “better.” When we decide to treat with a BTK inhibitor, we’re usually choosing between those two at this point, and we’re trying to personalize the decision for the patient, and there’s some different factors they can get involved in that complicated decision. Luckily, we are not limited to BTK as a target. I mentioned earlier, we have monoclonal antibody, rituximab (Rituxan) like the one I mentioned, but a newer version of Rituximab, a more potent version, obinutuzumab (Gazyva). Is one that we have available along with a Bcl-2 inhibitor, venetoclax. That is now, as of 2018, improved in the frontline CLL setting, also approved in the relapse setting, of course, since 2016.

And we use venetoclax with a monoclonal antibody like obinutuzumab, and together they are a very potent combination that cause pretty rapid cancer cell death, as opposed to the BTK inhibitors that more put the cancer to sleep and require daily dosing indefinitely for as long as the drugs work. And remarkably, the data on BTK inhibitors tells us that should work for many, many years. They’ve been following some of the patients that got treatment in the first line setting, and eight years out, there’s still more than 50 percent of the patients are free of progression, so they can’t even quote an average response time yet. With eight years of follow-up on the early ibrutinib patients. The difference with venetoclax combined with a monoclonal antibody like obinutuzumab, is because it causes a more rapid cell death, you can give it on a time-defined schedule. So we tend to give it for one year in the first-line setting and two years in the relapse setting, and the antibody portion is just given for the first six months.

The BTK inhibitors and venetoclax are oral treatments, so that’s a big win for patients to avoid the infusion center for those treatments, but the IV antibody treatments will still require some trips to the infusion center if you’re doing that combination with venetoclax. For most patients, those two targets are what we’re choosing between, and we try to personalize the decision to the patient. And again, that’s a very complicated discussion on what is “best”. And we use things like the prognostic work-up, medical problems that the patients already have, medicines that patients are on, to help make the best treatment decision for our CLL patients. But those…for in terms of how well that treats the CLL, both of those are considered equivalent options for the large majority of CLL patients. We’ve got some things on the horizon, but in general, those are two targets that we have at this point. There is for relapsed patients, PI3 kinase inhibitors that are still FDA approved at this time, that aren’t quite as effective and more toxic, so we sometimes think about using one of those targeted therapies if a patient has already progressed on a BTK inhibitor in venetoclax class.

In the future, we are looking towards combining BTK and Bcl-2 inhibitor. Like for example, there’s been studies already done that I’ve put many patients on, with ibrutinib-venetoclax, and I believe there’s a question about that later. There’s also an ongoing study that I have opened at my institution that’s looking at acalabrutinib and venetoclax. So taking these two pills together in a time-defined manner, so you don’t have to take the BTK inhibitor indefinitely. And then there’s some therapies that have already been improved in other lymphomas, and we wonder if they’re going to have a role in CLL eventually. So we now have bispecific antibodies, so that’s taking a drug like Rituximab or obinutuzumab and adding a T-cell engager to it so it has two targets or it’s bispecific. And we have that drug, mosunetuzumab (Lunsumio) available in follicular lymphoma, and there are several others in development, and we’ll see how their role comes into play in CLL as well. As well as CAR T-cell therapy, where we take a patient’s T cells and genetically engineer them to attack the cancer. That’s now an approved therapy for many different kinds of lymphomas and multiple myeloma as well. So we wonder if that’s going to have a role in CLL.

But I think for the foreseeable future, it’s going to be looking first at BTK and then Bcl-2 inhibition, or vice versa. And we don’t really know which is better to go first, we think they’re both…they can both be sequenced one after the other. And then maybe it will have some of these other breakthroughs coming in and helping for after patients need something beyond those therapies. And there’s probably going to be a lot of patients that never need anything beyond those options, those initial couple of targets, because they do so well. I think in the most immediate future, the approval that is going to give us a new great option is going to be for an alternative site BTK inhibitor, or it’s also called a non-covalent BTK inhibitor.

And there’s this drug called pirtobrutinib, it has been approved in mantle cell lymphoma and likely will get approved in CLL this year. And that drug specifically is a BTK inhibitor that still works even in patients that have, say, progressed on ibrutinib or acalabrutinib or zanubrutinib. That will be a new target available for CLL patients and probably pretty quickly become one of the go-to drugs that we use for relapsed CLL patients that have already been treated with a BTK…with a traditional BTK inhibitor. So growing number of options and it’s really great for our CLL patients.

Lisa Hatfield:

Thank you for that overview again, Dr. Jacobs. It does sound like there are a lot of new therapies coming out, especially for relapsed patients, super exciting for them. And this is actually a great time to jump right into questions. We have many questions from patients that different patients have submitted. But first, I want to remind everybody that this program is not a substitute for medical care. Please consult with your medical team for advice on your own condition or disease. And, Dr. Jacob, I was taking notes as you were talking, because you had spoken a little bit about a combination of the BTK inhibitor and Bcl-2 inhibitor with venetoclax. And I did a little research last night before I talked with you, and it sounds like that is something that the CAPTIVATE trial is investigating. 

So that’s exciting, and a patient asked about that, what that trial is. And it’s music to my ears as a cancer patient to hear something like “fixed duration,” it’s also investigating a fixed duration so patients and have maybe a bit of a medication vacation. So can you speak to that trial a little bit and explain what it is a little bit on how that might benefit patients with CLL?

Dr. Ryan Jacobs:

Yeah. So one of the best elements of treating with venetoclax is that it produces a deep level of remission in many patients. In fact, when given with the monoclonal antibody obinutuzumab, to CLL patients receiving that treatment as a first line of therapy for their CLL, about three-quarters of CLL patients will get to so deep of a remission that we call them minimal residual disease-negative. And that’s a blood test or a bone marrow test, but more easily done as a blood test, where we can look to a sensitivity of one in 10,000 white cells and determine if there’s any CLL in those 10,000 cells. We can actually go deeper than that, but we say, we call patients negative if they’re less than one in 10,000. And so 75 percent of patients will get to that depth of remission just with obinutuzumab for six months along with venetoclax for a year. So when researchers saw that, they recognized that we could probably stop treatment in those patients getting venetoclax because venetoclax yields these deep responses. And then the next kind of thought was, well, could we give a BTK inhibitor with venetoclax, but also over a defined treatment timeline and maybe get some of the remarkable benefits of treating with a BTK inhibitor but not get stuck being on therapy for years and years.

So the CAPTIVATE study was the first really to, in a large Phase II manner, look at that combination in a younger patient population, it was for patients 70 and younger. And it wasn’t in a high risk or anything, it was all comers. But they did have to be 70 and younger and getting treatment as a first-line therapy. So the combination was very effective. As of the last American Society of Hematology meeting in December, four years of data was reported and a large percentage of patients were still free of progression, over 80 percent still free of progression. And that’s three years off therapy at that point.

It was well-tolerated, not many patients had to come off due to toxicity. It was, in fact, less than 10 percent had really significant toxicities requiring discontinuation. So it was a well-tolerated effective treatment.

I do have one of those studies to open at my institution, the acalabrutinib-venetoclax combination, it’s called the MAJIC trial, and it is a large Phase III study that if it’s successful, I think would lead to the approval of giving those two drugs together. But then the extra credit question is, who should get the combination and who should get the drugs separately? And we don’t have an answer for that right now, and that’s a long topic of debate among CLL specialists.

Lisa Hatfield:

Great. Well, thank you. So for that trial you spoke of that you’re conducting right now, is that…is it only relapsed patients who are eligible for that? Or is that for front-line therapy?

Dr. Ryan Jacobs:

No, this is a first-line therapy that the MAJIC study is.

Lisa Hatifield:

Oh good. That’s promising for patients too.

Dr. Ryan Jacobs:

And it has a really good comparator arm, so that won’t be a problem that the standard arm on that study is venetoclax plus obinutuzumab, so it’s comparing against one of our best treatments, and so we really will get the answer of does it look better to use the BTK with the Bcl-2? Or is it not really that much better than just giving an venetoclax with obinutuzumab? And then the one obvious element that I didn’t mention that would be nice for most patients in addition to being efficacious and well-tolerated is if you could get an all-oral combination. Of course, venetoclax with obinutuzumab, you’re still getting quite a few infusions with the obinutuzumab over the first six months. So that’s a lot of time in the infusion center that you could avoid with just the combination of two oral targeted agents. So that would be a breakthrough for patients too, I think.

Lisa Hatfield:

Well, you commented also on something that’s really important for patients to know, and that is that if you go into a clinical trial, you won’t be given nothing for cancer clinical trials, you’re going to be given the standard of care or whatever it’s being compared to. So for patients who are considering that.

Dr. Ryan Jacobs:

That’s a Phase III. Yeah, for Phase III. If you go on an earlier phase trial, you know exactly what you’re getting. There’s usually not any randomization for earlier phase studies, you just get the intended treatment.

Lisa Hatfield:

Okay, great. Well, thank you so much for explaining that. So we have some pretty specific questions, and we have a patient who wrote in and asked, “What is the difference between IGHV-mutated and IGHV-unmutated CLL? And can you talk about treatment considerations for those?”

Dr. Ryan Jacobs:

Yeah. So that’s part of a bigger discussion around the prognostic work-up of CLL and not all CLL is the same, and we’ve done a really good job of figuring out tests to separate out the CLL patients that tend to behave more aggressively and respond to certain kind of therapies, versus those that are more of what we call indolent or slow growing and respond to other kinds of therapies. I do want to say, I haven’t mentioned it yet, we still don’t treat CLL if it’s not causing any problems. And about half of patients get diagnosed as sort of an accident, and they get a blood test for something else, and their white count is elevated, and that leads to a diagnosis, but they feel fine. We still leave those patients alone. Even with these good treatment options we have, we recognize that there are a select percentage of CLL patients that don’t ever need treatment, and so we don’t just want to start treatment in everybody.

But I do still like to check this prognostic work-up, even if I’m not going to start treatment, but I make sure and ask the patient if that’s what…iIn line with what they want. But certainly, if you’re going to start treatment, you’re required by guidelines to check a prognostic work-up, and I would really encourage the CLL patients tuning in to ask their oncologist, “What is my prognostic work-up?” if they’re going to start treatment.  Because of the oncologists, unfortunately, that have to deal with lots of other cancers, maybe don’t always know the right test to send. I’m very spoiled in that I get to just treat lymphoma and specifically focus a lot of my research in CLL and get to stay up with all this. I don’t know how a general oncologist keeps up with everything, honestly.

But the big three tests are going to be the FISH analysis, fluorescence in situ hybridization. And then IGHV mutational analysis, and then also a TP53 mutation analysis. And I don’t really have time to go through all of those, but IGHV is the question I get a lot. “What is that?” It’s one of these rare findings where it’s actually normal to have a mutation at the IGHV. IGHV stands for immune globulin heavy chain variable region, and it is usually mutated in B lymphocytes because it’s part of the process of a mature lymphocyte that is able to make a lot of different kinds of antibodies. And it undergoes somatic hypermutation, is what it’s called, as the B cell matures. Generally in oncology, the more mature a cancer is, the less aggressive it behaves and usually the easier it is to manage, and that is the case with CLL. So think of an unmutated IGHV CLL cancer as a more primitive or a more immature cancer clone, and as such, it is harder to treat.

In about half of patients will be found to be unmuted at the IGHV and historically, all we had was chemo and we knew these patients weren’t going to respond for near as long as the IGHV-mutated patients were to chemo. What’s nice is, with our targeted treatments, particularly the long-term data with the BTK inhibitors, it doesn’t look like it matters whether you’re mutated or you’re unmutated. So that’s one of the really great things with our new treatments for CLL, is it has, the people that have benefited the most are the ones that were doing the worst, so that’s great. It’s not just the patients that were already doing well, that are doing even better.

Lisa Hatfield:

So I just want to take a step back and kind of looking at this through the lens of a newly diagnosed CLL patient. You’d mention that sometimes you don’t treat every CLL patient. So is there something, if you find a patient who does not need treatment, is there something you tell the patients as far as regular monitoring? Will you monitor them to see if it progresses to the point where it requires treatment?

Dr. Ryan Jacobs:

Yeah. And we’re fortunate that this is a blood cancer that most of the time we can follow with a simple blood count and follow the white count, follow how the…follow the health of the bone marrow by looking at things like anemia, low red cell count, or a low platelet count that we call thrombocytopenia. So that’s the easiest thing to follow, but I’m also talking with my patients and examining my patients. I want to know if their length nodes are causing them a lot of pain, because we should treat that, there’s no reason they should live in pain.I want to know if they’re waking up drenched in sweat all the time, if their quality of life has been really affected by that. Or are a dramatic amount of fatigue that we can’t explain by some other cause. And I also, of course, examine the nodes myself and make sure that there’s no alarming findings there. So that’s really what’s involved with checking on a CLL patient that’s on active surveillance, that’s what we call it. And there’s a list of criteria that the oncologist should know in terms of deeming who needs treatment and who doesn’t. And so we’re kind of following the same rules, so to speak, in terms of who gets treated for CLL.

Lisa Hatfield:

Okay, thank you. So we have a patient who asked a series of questions here, and I think you already…you spoke pretty well to the role of the BTK inhibitors in treating CLL. I’m going to kind of clump these together.  So I guess three questions. What treatments do you think are the most beneficial for patients whose CLL has relapsed? What are the poor prognostic indicators for CLL? And along the same lines, what are the high-risk genetic markers for CLL?

Dr. Ryan Jacobs:

It’s a little more complicated discussion in the first line setting because both are options. At this point in time, we haven’t been…at least those that are, I would say, staying up to date on the CLL data, we have not been using chemotherapy for a long time. So most of the relapsed patients will have seen either one of the BTK inhibitors or venetoclax. And so what we do in the second-line setting is just use the other option that they haven’t seen. The data tells us, when you look at what treatments are being prescribed, most patients are going on BTK inhibitors, and they have been around longer than venetoclax in general. So for a lot of patients, that relapsed treatment is going to be venetoclax. Because that has the best data in terms of treating patients that have progressed on a BTK inhibitor like ibrutinib or acalabrutinib or zanubrutinib.

In the near future, we’ll have pirtobrutinib (Jaypirca) and so maybe, maybe some will get that drug before venetoclax, and that’s probably okay. And so we’ll have that additional option. The complicated patients, and I’ve alluded to this, or what do we do after BTK and Bcl-2? What are we left with? I mentioned PI3 kinase, that’s not a great option. There’s still stem cell transplant out there for young patients that are running out of options. Clinical trial is really what I would like to emphasize there.  If you’re a patient that can get to a high volume referral cancer center with a CLL specialist, I would do that. If you have seen BTK inhibitor and venetoclax and are looking for other options.

Lisa Hatfield:  

Great, thank you. So the next question is actually a really good question, I think we can broaden it a little bit. But the question is, “How can I ask my doctor to make sure I am being tested for serum markers?” And more broadly, I think a lot of patients are a little bit nervous about asking questions of their doctor, because they don’t want to feel like they’re questioning their expertise or doubting them. So how in general can we ask our doctor questions if we hear something? Or how we approach our doctor with those types of questions?

Dr. Ryan Jacobs:

So I mentioned asking your doctor, “What’s my prognostic markers?” I think this is probably the easiest way to get that information. And your doctor should be checking those. The question comes up like, what are the “high-risk” markers? We talked about mutated versus unmutated. Thankfully, our novel treatments that doesn’t seem to matter. Same goes with…there’s on FISH there used to be, if you found three copies of chromosome 12, that’s called trisomy 12, that doesn’t seem to matter With our newer treatments. A deletion at chromosome 11, again, used to not do as well with chemo. Novel therapies…doesn’t seem to matter. The one that is still potentially affecting outcomes, even with our novel treatments, are chromosome 17 aberrations, which stately are rare in the initial diagnostic setting. That or a TP53. A deletion at 17p or TP53 mutation probably is only going to be around 10 percent of patients or so. And in the relapse setting though, that number goes up because of the more aggressive cancers emerge, we call that clonal evolution. So maybe in the 20-ish percent range. These patients, we tend to prioritize indefinite therapies first, because it seems like these patients do better if you keep treatment going, as opposed to interrupted therapies like venetoclax. And so we tend to treat those patients with a drug like acalabrutinib or zanubrutinib first and then think about the venetoclax later for those patients.

Lisa Hatfield:

Okay. Okay. And just to clarify, for patients too, I know that a lot of cancers, there are discussions about the 17 deletion, 17p, and then also the TP53 gene. So if I understand correctly, the TP53 gene is housed on chromosome number 17. So if that is missing, then that patient may be missing that gene, that is considered a tumor suppressor gene, which we want. Is that correct?

Dr. Ryan Jacobs:

Right. So it’s either missing, which is what we see on FISH with a deletion, or it can be mutated and that’s the next gen sequencing, and often it will be both in those patients.

We think with indefinite, there’s some really good data that was just released with zanubrutinib. When they looked at 17p-deleted patients, there’s some long-term follow-up with ibrutinib-treated 17p-deleted patients. With chemo these patients would only get about a year or so, but we’re getting maybe even close to normal outcomes with long-term BTK. But we do know if you just give them a year of venetoclax and obinutuzumab for six months and then stop, they do relapse quicker than the other patients. So they relapse after about four years. As opposed to with five years of follow-up with that first line venetoclax approach, there are 62 percent of patients are still free of progression.

Lisa Hatfield:

Oh wow, okay. Thanks for explaining that too. I know that that chromosome 17 and the TP53 gene, that’s talked about in a lot of different cancers and it often come up, “How are those connected?” So thanks for just describing that a bit. So this patient is asking, “For patients who may be eligible for BTK inhibitors, are there specific comorbidities that might contribute to adverse side effects?”

Dr. Ryan Jacobs:

Yeah, so we screen…all BTK inhibitors have some cardiac toxicity. They have been shown with the second-generation BTK inhibitors to have less cardiac toxicity than ibrutinib, specifically atrial fibrillation. So if you have atrial fibrillation, maybe that’s a reason why you might go on venetoclax first as opposed to a BTK inhibitor. But it’s not a contraindication to getting a BTK inhibitor if the atrial fibrillation is under good control.  Other cardiac risk factors would include difficult to control hypertension at baseline, or heart failure. These are all things that might make us think twice about using a BTK inhibitor as our first therapy, because venetoclax has no cardiac toxicities. The other thing to consider is BTK inhibitors all to a degree have, and I describe it to patients, like an aspirin-like effect on the platelets. They do interfere with the platelet binding, which so universally, patients will know to varying levels some easier bruising.

And if patients are on, because of say, they’ve had a heart attack in the past and they’re on aspirin at baseline, or what would even be more concerning if they were on a drug like Plavix because they’ve had a stent placed, that would be something that would really concern me and would definitely push me more towards venetoclax, that again, doesn’t have those anti-platelet interactions. Also, patients who are on blood thinners because of a history of blood clot or atrial fibrillation, there is the potential increased risk for bleeding and bruising there as well. None of these are absolute contraindications, they’re just all what goes into the blender, if you will, of putting lots of information in and coming up with the best treatment decision as personalized for the CLL patient. We’re blessed to have multiple options, but it does make it more of a challenge to find the “best” option.

Lisa Hatfield:

Yeah. Thank you for that. We have several questions from a couple of patients regarding side effects. So the question, “How long will my side effects of my CLL treatment last? And what can be done to reduce those?” And specifically, a patient is asking if there’s a connection with CLL and gastrointestinal issues?

Dr. Ryan Jacobs:

So all of the treatments, including venetoclax, the BTK inhibitors, will have diarrhea listed as a possible side effect. It’s usually low grade. But generally, I have found the gastrointestinal toxicities abate some over time. So if they are present earlier, if you’re able to stick with therapy, they do tend to get better. For the once daily meds, I encourage those patients to try to take the drug in the evening. The GI tract tends to be less active later in the day, and you can sleep off some of the potential gastrointestinal issues. So I’ve had success there. Sometimes we have to lower the dose to just find the best dose to help mitigate some of these. There’s the antidiarrheals that can help if you need them. Imodium. I had a patient I saw earlier this week that Imodium didn’t really work, but good old Pepto Bismol did the trick from time to time. So certainly though, if the gastrointestinal issues are significantly affecting quality of life, we need to come up with a new plan, whether that’s reducing the dose or changing to a different option. Specifically, what’s nice about the BTK inhibitors is they all have data that show if you’re having problems with one, you can switch to the other and likely not have the same problem occur. So that’s nice.

Lisa Hatfield:

Have you ever seen any uncharacteristic side effects several times in your practice? Anything really unique? I’m just curious about that.

Dr. Ryan Jacobs:

Yeah. There’s always the patients, they can have a more severe form of maybe, of a more common side effect, like the…we were talking about diarrhea, I’ve had a patient that actually had a difficult, with venetoclax, had difficulties with the stool incontinence. So that was kind of a severe form of that. It wasn’t so much diarrhea that was the problem. But we were able to ultimately mitigate that with a dose reduction. I would say the way, particularly if it’s an unusual side effect, the best thing to do is to take a break. If it’s a serious side effect that needs to be addressed and it’s affecting quality of life or causing problems, take a break from the treatment. If you take a week off these treatments, particularly venetoclax, taking breaks doesn’t matter. We like not to take long breaks with the BTK inhibitors. But if you take a week off, these drugs don’t have very long half-lives. So if the issue is not getting any better and you’ve been off of treatment for a week, it’s unlikely that that issue is coming from the treatment. So that’s a way I try to sort through some…particularly if they’re unusual side effects sometimes. And certainly, if we deem that the issue is connected to the treatment, I’ll usually try lowering the dose before just giving up.

Lisa Hatfield:

Okay. Thank you. A patient had asked, and I love this question because I often wonder myself when I get up in the morning, my bones are creaking and popping, “How do you know the difference between,” this patient’s talking about fatigue. How does a patient discern, “Well, this is fatigue from my cancer or my treatment,” versus just normal aging? Whether it’s fatigue or bruising or any side effect.

Dr. Ryan Jacobs:  

Yeah. Fatigue is a really…I had an attending physician when I was in my training that said, “Treating fatigue makes me fatigued.” But it’s hard. If it’s really the only problem the CLL patient is having, it can be. All those other problems I had mentioned earlier, the low red cells, the low platelets, the painful nodes, the night sweats, I with close to 100 percent certainty know I can fix those with treatment.Fatigue, I’m not as confident when that’s the only issue that a patient’s having. I try to differentiate between fatigue from other causes and old age, and specifically to CLL. 

They try to put it as a metric and say, if you’re having to spend half the day or more just lying around and you’re not able to do your normal activities of daily living, like that’s a severe level of fatigue and treatment should be considered.I’m looking for somewhat of a precipitous decline, not necessarily just kind of the gradual fatigue that you might more relate to aging. The problem with treating fatigue is you’ll look, if you look at the possible side effects of all of these medicines I talked about, fatigue will be a potential side effect.So you’re sometimes trading one problem and getting another, or maybe the fatigue does get better, but then the patient has some different side effect that’s even worse than the fatigue. So it’s hard to really help when fatigue’s the only issue. But certainly, I have helped some patients with fatigue. We don’t have a test that we can do to know for sure is the fatigue coming from the cancer, or is it coming from something else. 

Lisa Hatfield:

Great. Well, that wraps up our program for today. Thank you so much for joining us, Dr. Jacobs.  I am Lisa Hatfield from Patient Empowerment Network.


Share Your Feedback

Create your own user feedback survey