Tag Archive for: multiple myeloma

Accessing Quality Myeloma Care | Advice for Overcoming Obstacles

Accessing Quality Myeloma Care | Advice for Overcoming Obstacles from Patient Empowerment Network on Vimeo.

How can you access the myeloma care that is best for YOU? Myeloma specialist Dr. Krina Patel shares advice for patients, including the importance of a second opinion and key questions to ask your doctor regarding your disease and treatment plan. 

Dr. Krina Patel is an Associate Professor in the Department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center in Houston, Texas. Dr. Patel is involved in research and cares for patients with multiple myeloma. Learn more about Dr. Krina Patel.

Related Resources:

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care?

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care?

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

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care?

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care? 

Transcript:

Katherine:

What hurdles to patients face when accessing quality overall myeloma care and what can be done to get over these obstacles?  

Dr. Krina Patel:

I talk about this a lot. 

So, again I think the biggest problem for me is that because myeloma care changes so fast, which is a good thing that we have all these options and we have so many new therapies, it’s really hard for people who don’t do just myeloma to keep up. I don’t think I would be able to. I don’t do breast cancer. I don’t do other cancers, so when I take my boards every 10 years, I have to learn a lot to take those.

So, it’s just a part of the system that this the problem. So, I think if you’re seeing a local oncologist that sees five myeloma patients a year, they’re gonna be stuck on what was the treatment when they did it last time for that last patient, which again might be very different now because things change so fast. 

And so, again, you want to get to a doctor quickly, and I understand that. When people hear “cancer,” they’re like “I gotta get treatment. I gotta go fast.” But part of it is, if you need treatment quickly to get to your doctor. But then, try to make a second-opinion appointment done, even virtually because we can do that now after COVID; we have so many more options for that.  

And get that second opinion just to say “Is this the right therapy for me? Going forward, what should I do?” So, patients, “Should I get a stem cell transplant?” if you’re newly diagnosed or not. “What kinda maintenance should I be doing? Do I have high-risk disease or not? What are the nuances of my myeloma versus everybody else that we need to be careful about? Should we dose reduce?” There’s a lot of those types of hurdles. Patients, if they have kidney failure form their myeloma, we should be decreasing the dose of some of the medications; those types of things that really we can help with to make sure those outcomes are in the best. 

And that first treatment really does matter so that we can reverse as much as possible, for patients who have kidney involvement versus bone involvement, to decrease the pain really quickly. Do we need to get our radiation doctors involved to get radiation to help make sure you don’t get a fracture from a potential bone lesion. So, I think, again, I understand the urgency of seeing somebody, of getting diagnosed, and starting therapy.   

But quickly get to a second opinion so that they can help. And then, again, some of these patient advocacy groups are amazing for myeloma. And I think there’s just so much information there that you don’t want to get overwhelmed, but at the same time you want to start going a little bit at a time at those things so that you can learn more about what you need to be asking and doing.  

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

Key Advice for Myeloma Patients | Questions to Ask About a Care Plan from Patient Empowerment Network on Vimeo.

How can newly diagnosed myeloma patients be proactive in their care? Dr. Krina Patel shares key advice for patients, including the importance of making notes before office visits and the role that a care partner can play in overall support. 

Dr. Krina Patel is an Associate Professor in the Department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center in Houston, Texas. Dr. Patel is involved in research and cares for patients with multiple myeloma. Learn more about Dr. Krina Patel.

Related Resources:

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care?

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care?

Accessing Quality Myeloma Care | Advice for Overcoming Obstacles

Accessing Quality Myeloma Care | Advice for Overcoming Obstacles

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care?

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care? 

Transcript:

Katherine:

For newly diagnosed patients, what key advice would you share with them? And are there specific questions they should be asking their doctor about their care plan? 

Dr. Krina Patel:

Yeah. So, I know it’s hardest for newly diagnosed patients. Most people have not even heard what multiple myeloma is. They’re learning how to spell it correctly and making sure it’s not melanoma. And this is a conversation I have with so many of my new patients that I think it’s really hard your visit, and maybe even your second visit, to ask all the right questions. So, really, coming home and every time you’re on a treatment or you’re talking about a treatment and you have a question, write it down because I know it’s really hard when we’re only there for 15, 30 minutes to talk to you.  

For us, we have MyChart, so my patients will send questions as they think of them through that. And I think that’s really important. Sometimes it’s hard to know what questions to ask when you have no idea what’s about to happen, and that’s okay. But I think as you’re going through therapy, really making sure that you ask about alternative therapies that might be available and why someone is picking one versus the other, making sure you know what supportive medications you really need.

And I will say that, with myeloma, a lot of our treatments are patient-friendly but they do cause side effects and infections, so, we have a lot of supportive medications we use; so, again, anti-shingles, potentially if you could get a blood clot, we have you on some type of blood thinner.  

We have people on against steroids because of all of our initial therapies have steroids. We wanna make sure you don’t get ulcers in your stomach, so we have patients on proton pump inhibitors. There’s a lot of things we do to again decrease that toxicity. So, that’s important.  

And then, I think the next part is when you’re on treatment, whatever symptoms you’re having keep a log of that. Some things are, okay, maybe it’s just a little bit here and there, that you’re feeling fatigued but then you’re better. But there are certain things that cause a lot of side effects that my patients sometimes don’t tell me about. So, the steroids can cause major insomnia for some of my patients where they don’t sleep for three days, and that’s not okay. We can decrease those.

So, there are ways to manipulate the treatments as we’re going through to make sure that not only are you having a great response but that you’re not having major side effects that are actually gonna hurt your health down the road. So, really important to discuss those things that you’re having as you’re going through.  

Katherine:

There’s also the importance of a care partner in your life –  

Dr. Krina Patel:

Yes. 

Katherine:

– right?  

Dr. Krina Patel:

I agree. So, I joke with my patients but it’s real; there’s actually a study that shows that men with three and a half women in their lives do much better in healthcare in general than those who don’t. So, I’m like “Go get more women in your life” – 

Katherine:

I love that.  

Dr. Krina Patel:

– or just caregivers in general.  

Men are great caregivers too, but really having someone there that can listen for you and write down those things because it is overwhelming. And when you’re on treatment there are a lot of times when you just can’t pay attention. You can’t focus. You can’t listen to everything. And so, the more people that are there, they’ll pick up other things.

So, a lot my patients will even have their loved ones on their phone with them, even if they can’t be there in person so that they can record. And a lot of my patients will record things and they’ll ask me; so, definitely as whoever you’re talking to if it’s okay to record. But most of us will say “Yes, it’s completely fine” so that you can listen to it again when you go home.   

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care?

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care? from Patient Empowerment Network on Vimeo.

How does bispecific antibody therapy impact the outlook for myeloma care and treatment? Dr. Krina Patel discusses how this treatment, and CAR T-cell therapy, are revolutionizing myeloma care.

Dr. Krina Patel is an Associate Professor in the Department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center in Houston, Texas. Dr. Patel is involved in research and cares for patients with multiple myeloma. Learn more about Dr. Krina Patel.

Related Resources:

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

Accessing Quality Myeloma Care | Advice for Overcoming Obstacles

Accessing Quality Myeloma Care | Advice for Overcoming Obstacles

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care?

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care? 

Transcript:

Katherine:

What role do you foresee bispecific antibody therapies playing the future of myeloma care?  

Dr. Krina Patel:

So, I think bispecifics are phenomenal. I’m a CAR T girl, but in terms of access, I will say, that more of our patients around the world are going to have access to bispecifics.  

And it’s off the shelf, so you don’t have to worry about taking cells out, making it, waiting and hoping to get those cells back. So, many more patients are going to be able to get it. And I think ideally if everybody could get a CAR T, my goal would be a CAR T first and then a bispecific until we can cure myeloma. Unfortunately, for the most part right now with these therapies, as single agents we haven’t seen that the majority of patients are cured.  

So, my goal is to make sure that I have all the treatment options possible to keep patients doing well for as long as possible. And so, again, ideally CAR T, then maybe a bispecific because of the way those resistance mechanisms happen. But if we don’t have the availability of CAR T for everybody or you’re not eligible, I do think bispecifics are a great therapy. And I have again patients who are frail, who are older that we’ve been able to give bispecifics to and they’ve had amazing responses. And I think right now they’re single agents. But I do think that as we get these trials with combinations approved, we’ll see a lot more increase in use of those.  

Again, the side effects are still something we’re learning about. So, bispecifics with BCMA, infections are a really big risk, even more than CAR T.  

So, it’s really, really important that, if anyone has fevers or they don’t feel well, they see their doctor right way and make sure it’s not a strange infection that we don’t usually see that needs to be treated versus even a regular pneumonia that can be pretty dangerous when your immune system’s down.  

So, that’s important. And then, the GPRC5D, as I said, it’s the taste and the weight loss and things like on skin that we really wanna make sure we do as much supportive care for that as possible.   

What’s Next in Myeloma Research and Treatment?

What’s Next in Myeloma Research and Treatment? from Patient Empowerment Network on Vimeo.

What are the next generation myeloma therapies? Dr. Krina Patel shares an update on new agents, such as CelMoDs, and discusses how combination treatment and sequencing of therapy will evolve in the future of myeloma care.

Dr. Krina Patel is an Associate Professor in the Department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center in Houston, Texas. Dr. Patel is involved in research and cares for patients with multiple myeloma. Learn more about Dr. Patel.

See More from Evolve Myeloma

Related Resources:

Next Generation Myeloma Treatment Options

Next Generation Myeloma Treatment Options 

Evolving Myeloma Treatment Options | Bispecific Antibody Therapy

Evolving Myeloma Treatment Options | Bispecific Antibody Therapy 

Evolving Myeloma Treatment Options | CAR T-Cell Therapy

Evolving Myeloma Treatment Options | CAR T-Cell Therapy 

Transcript:

Katherine:

Dr. Patel, for the last few years advances in myeloma treatment have been focused on the cellular therapies like CAR T. Can you share what’s next in myeloma research and treatment? 

Dr. Krina Patel:

Yes, I think it’s been really exciting. The last 10, 15 years, really, we’ve just catapulted in the whole world of immunotherapies; so, from monoclonal antibodies to even IMids and CelMoDs and things that we’ll talk about a little bit now, and cell therapy as well as just other ways of engaging T-cells with the bispecific therapies too. So, I think what’s really exciting, that we have not just new mechanism of action that’s coming down the road but new targets.  

So, again, coming back to really the big stuff like immunotherapy that I really like a lot and what I really am excited about, we have different ways of using the immune cells to help fight myeloma.  

And so, things like IMids, lenalidomide, and pomalidomide that are older drugs that we’ve had since 2006, but really there’s newer ones called CelMoDs that are coming out that are being evaluated in clinical trials. One is called iberdomide. Another is called mezigdomide.  

So, we’re really excited about this really in combination therapy, just like the prior iMids were used. And what it really does is it improves your immune system; it activates it to a point where things like monoclonal antibodies, such as daratumumab or isatuximab or elotuzumab, can work better in synergy.  

But even new trials with some of our CAR Ts that we already have available, the BCMA therapies, combining it with these to see if we can make those T-cells work better. 

So, once you get the CAR T-cell infusion, can we give some of these therapies now to improve how long it lasts, how well they work. And the same thing with the bispecifics.  

These are therapies that are using the T-cells that are already in your body. Can we combine it with some of these of other immune therapies that will help the T-cells already there get activated, and then the bispecific takes them to the myeloma to really get treated. And I think those combination studies that are coming down are really, really exciting. And then, I think the new antigens, as I mentioned, not just BCMA therapy but we have GPRC5D and we have something called FcRH5.  

And to my patients, I say it’s alphabet and number soup basically but they’re really targets for myeloma that we’re finding. It’s pretty amazing, considering that we didn’t have a target for the longest time, like lymphoma when they had their CD19 and we were jealous. And now we’re finding all these targets and now we’re figuring out how do we combine different mechanism of action for different targets so that we can hopefully kill every last myeloma cell.  

Care Partners | What Should You Know About the Bispecific Antibody Treatment for Myeloma?

Care Partners | What Should You Know About the Bispecific Antibody Treatment for Myeloma? from Patient Empowerment Network on Vimeo.

What are care partners essential when a loved one is undergoing bispecific antibody therapy for myeloma? Nurse practitioner Alexandra Distaso from Dana-Farber Cancer Institute explains the bispecific antibody process, reviews potential patient side effects, and shares resources that can help support care partners.

Alexandra Distaso, MSN, FNP-BC is on the Multiple Myeloma Nursing Team at Dana-Farber Cancer Institute.

Download Resource Guide

See More from the Care Partner Toolkit

Related Resources:

What Is the Role of Bispecific Antibody Therapy in the Future of Myeloma Care?

What Is the Role of Bispecific Antibody Therapy in the Future of Myeloma Care?

What Myeloma Patients Need to Know About Bispecific Antibodies

What Myeloma Patients Need to Know About Bispecific Antibodies

Evolving Myeloma Treatment Options | Bispecific Antibody Therapy

Evolving Myeloma Treatment Options | Bispecific Antibody Therapy

Transcript:

Katherine:

Hello and welcome. I’m your host, Katherine Banwell.

Being a care partner can be a demanding role. From understanding a loved one’s diagnosis to participating in treatment decisions, navigating care with a loved one can be challenging. That’s why the Patient Empowerment Network created the Care Partner Toolkit series to provide care partners with advice and information so that they can feel empowered and confident during the process. In today’s program, we’re going to learn about bispecific antibodies, how this myeloma approach works, who it’s right for, and the important role that care partners play throughout the process.

Before we meet our guest, though, let’s review a few important details. The reminder email you received about this program contains a link to program materials. If you haven’t already, click that link to access the guide to help follow along during the webinar. At the end of this program, you’ll receive a link to a program survey. Please take a moment to provide feedback about your experience today in order to help you plan future webinars. And finally, before we get into the discussion, please remember that this program is not a substitute for seeking medical advice. Please refer to your healthcare team about what might be best for you.

Well, let’s meet our guest, nurse practitioner Alexandra Distaso. Alexandra, welcome. Would you please introduce yourself?

Alexandra:        

Hi, Katherine. Thank you for having me. Yes, my name is Alexandra Distaso. I am a nurse practitioner at Dana-Farber Cancer Institute in Boston. I’ve been a nurse practitioner for just about nine years, and I’ve been at Dana-Farber for eight of those years.

Katherine:         

Well, thank you so much for joining us today. I’m looking forward to having a discussion with you. Alexandra, you work with myeloma patients and their care partners on a regular basis. What is your role on the healthcare team?

Alexandra:        

Yeah, so as a nurse practitioner, I’m seeing patients every day in the clinic that I’m here. And we’re seeing patients for both their kind of monthly check-in visits prior to treatment, sick visits, symptom management visits. So, either that’s once a month for your routine visit, or a couple of times a month if you’re going through a transition of treatment or having some sort of side effects we need to work on a little bit more closely.

Katherine:         

Well, as I mentioned today, we’re going to be focusing on bispecific antibody therapy. It’s a relatively new approach. What is it exactly?

Alexandra:        

Yeah. So, bispecific antibodies are a really exciting new therapy in myeloma that we’ve had for within the last year.

So, we have three bispecific antibodies that are currently approved right now. And the way that they work is, the medication binds to the tumor site on your plasma cell, where the myeloma cell is, and it binds to a T cell, which is your immune system cell. And it kind of works to redirect the T cell, your immune system, to kill off the cancer cells in your body.

Katherine:         

Okay. How has this treatment impacted the state of myeloma treatment and care?

Alexandra:        

This has been a great option for patients who are now triple-class refractory and further into their myeloma journey. The development of these new drugs represents really kind of a new era in myeloma. We’re having a lot of patients who are now exposed to more therapies with using three or four drug therapies in the first-line setting. So, having an extra line of therapy now further down the road has been a great option for a lot of patients.

Katherine:         

Well, who is this treatment approach approved for, and what are the eligibility requirements?

Alexandra:        

So, one thing that’s great about bispecific antibodies is that there is not a lot of restriction on who we can use these therapies for. So, these are great for patients who are a little bit more frail or maybe aren’t up for something like a CAR T, or whose disease is a little further along, and they don’t have time to wait for something like CAR T, which requires collecting of cells and manufacturing. What’s great about these medications is that they’re off the shelf. They’re ready to go kinda when you need them. There are restrictions in terms of how many lines of therapy that you need to have had before you can currently get bispecifics.

So, right now, you need to have four prior lines of therapy, and that needs to include an immunomodulatory agent. So, something like a lenalidomide (Revlimid) or a pomalidomide (Pomalyst), a proteasome inhibitor like bortezomib (Velcade), and a monoclonal antibody like daratumumab (Darzalex) before you’re eligible for these.

Katherine:         

Have there been any recent bispecific antibody research developments that patients should know about?

Alexandra:        

So, there are at least three bispecific antibodies that are hopefully coming into approval in the next several months to year, cevostamab being one of them. It’s a very exciting time for myeloma with all of these medications being approved. Teclistamab (Tecvayli), elranatamab (Elrexfio), and talquetamab (Talvey) in the last year. There’s still a lot of research on bispecific antibodies, especially trying to bring them all outpatient instead of just having inpatient treatment, and in addition, looking at them with other medications, such as teclistamab with daratumumab.

Katherine:         

Okay. Well, thank you for that, Alexandra. This gives us a good idea of what the therapy is, and how it works to treat myeloma. So, let’s dive into the process. How is this treatment administered, and what’s the frequency?

Alexandra:        

So, currently, all of the bispecifics are given as subcutaneous injections.

And all of them do require a current hospitalization visit, somewhere between four and 10 days, depending on which medication you’re getting and what schedule you’ll be on. So, everyone is required to be in the hospital. Again, we’re trying to move that outpatient to minimize patients’ times in the hospital if we have to. And you get a lower dose with that first exposure to each of the medications, and then we build up the dose for the doses in the hospital into what will eventually be your outpatient weekly, or biweekly dosing.

Katherine:         

Okay. Are there only certain medical centers that have this therapy? How widely available is it?

Alexandra:        

So, right now, the step-up dosing, the inpatient hospitalization part of bispecifics is primarily only at academic medical centers. So, it is a little bit more restricted in that initial therapy. But what we are seeing is that a lot of the community practices are able to enroll and give these medications in the community.

So, some patients will come see us for a consult and the initial step up, but then they’re able to go back to their primary team after the first cycle so that they’re not commuting back and forth to Boston all the time.

Katherine:         

That’s good to know. So, once the therapy has been given to a patient, what happens next?

Alexandra:        

When you’re admitted for these initial step-up dosings, we closely monitor you for reactions in the hospital. That’s why we kind of are doing this in in-patient settings to monitor very closely for CRS and neurotoxicity, which we’ll talk about a little bit later. While you’re in the hospital, they’re checking your labs every day, they’re monitoring your vital signs, they’re doing silly questions like, “Do you know your name and the year,” to kind of monitor how you’re functioning. Once you have passed kind of the step-up dosing, either you’ll come back to me and your primary team at Dana-Farber, or we’ll communicate with your local team to set up your schedule for moving forward.

Katherine:         

What are the short-term side effects associated with bispecific therapy?

Alexandra:        

Yeah. So, the short-term side effects that we’re watching for are these reactions in the hospital called CRS, cytokine release syndrome, and neurotoxicity.

So, the CRS is an inflammatory response where cytokines are released and usually cause a fever. We monitor and make sure that the fever isn’t being caused by some sort of infectious process or there’s no other cause for the fever. And if not, then there are medications we can give to help reverse these side effects while you’re in the hospital.

So, the way that we treat that is, again, we’ll make sure that there’s no sort of infection or other reason for a fever. And if the patient continues to have the fevers and they have low blood pressure and changes in their oxygen needs, which is kinda what happens if this inflammation progresses, is we’ll give things like dexamethasone, a steroid, or another medication called tocilizumab to help kind of reverse the effects of the cytokine release.

Katherine:         

Who else is on the healthcare team when someone receives these therapies?

Alexandra:        

Yep. So, you’ll always meet with your oncologist or an oncologist at the academic medical center where you’re going to be getting the medication to go over potential side effects, what the treatment entails, and consent. We have nurses here that are specific to bispecific antibodies, that help coordinate with your local team if you’re going back to your local practice. We have the infusion nursing team who are the ones who are actually giving the bispecific antibody therapies. They explain kind of what to watch for at the site where the injection goes. And then we have pharmacists who are also available to meet with you and go over any questions you may have about the treatment.

Katherine:         

What do we know about long-term side effects? Are there any?

Alexandra:        

So, long term, what we’re really seeing is risk for infections. So, all of these medications lower your blood cell counts, and we have to watch for these opportunistic infections, fungal, bacterial, viral.

Which is why it’s important that we have everyone on supportive medications to try and prevent that from happening. But long term, that is certainly something that we’re seeing. With the talquetamab, there can also be some skin and taste changes, and those are not necessarily right at the inpatient dosing, but we can see that. But those are things we’re also managing in the months after the initial therapy.

Katherine:         

Okay. Why is it so important that care partners let the healthcare team know of any changes that they see in their loved ones?

Alexandra:        

I say this to my patients and their families all the time. They know their family member best, and they may be one to notice that they’ve been more tired, or their energy just isn’t the same, or they do have a little cough that maybe the patient hasn’t even really noticed. And those are all things that we want your observation, we want you to speak up about, because the sooner we address some of these problems, the less complications the patients may have.

Katherine:         

What are the supportive medications for somebody who might be having side effects?

Alexandra:        

Yeah, so with the talquetamab, which we’re primarily seeing a lot of skin side effects and mouth discomfort, a lot of the time we have special mouthwashes to prevent discomfort and irritation. Things like biotin to just keep the mouth moisturized. Steroid creams and nail ointments to help with sometimes some peeling of the skin. And then for all bispecifics, we have everyone on viral prophylaxis. Something like acyclovir (Sitavig or Zovarax) or valacyclovir (Valtrex). PJP prophylaxis. So, something like sulfamethoxazole and trimethoprim (Bactrim) or dapsone (Aczone). And almost all of our patients are on an IVIG infusion once a month to help support their immune system and prevent against infections.

Katherine:         

Alexandra, you mentioned care partner looking for a cough, for instance, in a patient. What other things should care partners be looking for?

Alexandra:        

Any kind of change in the patient’s baseline is always helpful to know. So, if people are feeling much more tired, even if you’re not due for your therapy, sometimes calling to say that they just don’t seem themselves, we can check their blood counts. And again, sometimes they might need a blood transfusion, or their white count might be quite low, and they might need some Neupogen or filgrastim to help kind of support their blood counts. So, really kind of notifying us, even if it doesn’t seem like a big thing, it’s always better to call.

Katherine:         

Yeah. How long will a patient be on a therapy like this?

Alexandra:        

So, we still don’t know exactly the long-term duration of response. I think the most recent update we have was a median of 18 to 22 months was the last report. Which is a great response for what we have in myeloma.

Katherine:         

So, does the length of time a patient is on a therapy depend on the patient themselves, their comorbidities, et cetera?

Alexandra:        

Sometimes they’re comorbidities, but it is usually more just how their myeloma responds. So, every month when you’re coming in for therapy, even if your therapy is weekly or biweekly, every month, we’re monitoring your myeloma markers, and every month we’ll go over those markers to make sure we’re still seeing a good response. Usually, we’ll do a PET scan or a skeletal survey to also monitor everyone’s bones and any other lesions, they may have.

Katherine:         

What is considered an ideal response?

Alexandra:        

An ideal response. A lot of times we’re seeing everyone’s light chains go to even an undetectable level. So, even if we see some partial responses where the light chains were, let’s say they were 100 and they’re going down into the normal range, that’s still wonderful.

If it stayed like that for months, we wouldn’t make any changes. But best-case scenario, we see them go to a level that we can’t detect them in the blood work.

Katherine:         

As I mentioned, Alexandra, this program is aimed at helping care partners understand the process and how they can support their loved ones. What do you feel is the care partner’s role?

Alexandra:        

I feel like the best way for care partners to support patients is to kind of take the time to learn about the myeloma and the therapy and try to do the best they can to just be there for not just moral and emotional support, but the other little things that they may need. Coordinating rides, if it’s a family member, asking friends for help.

And then other things like insurance phone calls can be incredibly time-consuming and taxing. Or waiting at the pharmacy to pick up medications. Any, even little things like that, I think take a huge load off the patients who are doing this day in and day out.

Katherine:         

Yeah. Are there key questions that care partners should ask as they begin the process?

Alexandra:        

I think the best thing that patients and caregivers can do is if they have questions prior to the visit is to make a list. ‘Cause, I’m guilty of this myself, as soon as I show up at my doctor’s office, I completely forget what I wanted to say. So, making a list prior to the appointment, I think, is hugely beneficial. And then I don’t think that it’s ever a bad thing to ask for the doctor or nurse practitioner or pharmacist, whoever you’re meeting with, to see if they can repeat themselves. We’re putting a lot of information into a very short period of time.

And to try and keep track of, again, schedule, supportive care medication, when you’re going to need to be in the hospital, how long, what your follow up will be, taking notes during the visit or asking to hear it again is always helpful. Not even just for the caregiver, but sometimes for the patient who’s still trying to wrap their head around some of the change in therapy.

Katherine:         

Right. Yeah. That’s really good advice. As we’ve covered, it’s not always easy being a care partner. What would be your best advice for those who are caring for someone undergoing bispecific antibody treatment?

Alexandra:        

I would say it’s hard to put out how important care members are to the entire care team. Again, not just for the logistical, getting the patients to appointments, getting their medication, but really having someone the patient feels comfortable to be able to lean on. And again, they may say, “I’m so overwhelmed in these visits, and I really need some help.

Could you ask these things?” Helping them keep track of all these medication changes and appointments and visits and any sort of even small things like grabbing them a water so that they’re staying hydrated. Those little things make such a difference to people. And I think doing those things, no matter how small they feel, really helps support the patients through these changes in therapy.

Katherine:         

Besides yourself, what other staff members can care partners turn to for support?

Alexandra:        

So, at our office, we have an amazing group of triage nurses who are available Monday to Friday all during office hours. We have after-hours. If your patient or family member spikes a fever and you’re worried they just don’t look good, there’s always a doctor on 24/7 that you can page to kinda ask for advice and see what to do from there. And again, we have infusion nurses who are giving these injections every day.

And they are wonderful resources on what you might feel later in the day, what that site might look like, side effects that might pop up. So, really, anyone wants to be there to answer questions to make it easier for the patient or the family.

Katherine:         

Are there social workers or psychologists on the team as well?

Alexandra:        

Yes. We have an amazing group of social workers at Dana-Farber. And one thing that I really like about the way our program is run is that we have a social worker who is dedicated specifically to myeloma. So, they’re very familiar with these medications and the hospitalization requirements, which can be extremely tough. And so having resources and just someone to talk to, both through social work and our psychosocial oncology department, is also a wonderful resource to have.

Katherine:         

What about online resources? Do you have any recommendations?

Alexandra:        

So, I think that the IMF, the International Myeloma Foundation, and the NCI, have amazing resources for patients.

Actually, the IMF has an entire caregiver support page with kind of caregiver self-help, and ways that you can care for the patient. Care for the caregiver, and care for the patient. I think the MMRF has wonderful resources, and they also have a lot of online forum videos about bispecific antibodies and the different treatments for myeloma that are available if you want to learn more. And then same thing with The Leukemia & Lymphoma Society. Excellent resources online.

Katherine:         

Thank you so much for all of that. What about self-care for the care partner? Why is that so important?

Alexandra:        

I feel like this is such a hard thing for people because it always gets put on the back burner. And I know a lot of the times it’s like when you’re on an airplane and they say, “Put your oxygen mask on yourself before you put it on anyone around you,” because you can’t help others if you’re running on empty. You really need to take care of yourself.

Make sure you’re not just functioning, that you’re eating and sleeping and hydrating and taking care of all your own personal needs, but also that you’re taking time for you to kind of reflect and have some time to decompress from everything you are trying to do to help your family member or loved one.

Katherine:         

Why should care partners speak up and ask questions about how they’re taking care of the patient, what they can do to help the patient and themselves?

Alexandra:        

Yeah, I think that these bispecific antibodies are new, and one great thing about them is that overall they are really well-tolerated in general. So, hopefully, it means the patient is feeling pretty good and having a really nice response to their therapy. But if they’re not feeling good, we want you to speak up at home. Again, sometimes patients are the last person that want to tell you there’s a problem ‘cause they’re worried about missing their therapy.

And I always tell patients it’s sometimes not safe, if you do have a cough, if you had had a fever, we want to be safe and maybe hold a dose of therapy to address maybe something else that’s going on and avoid further complications another week. So, if you’re noticing something, I always encourage people to speak up and let us know of any concerns they’re seeing at home.

Katherine:         

Alexandra, we received some questions from audience members prior to the program. Amelia wants to know, are bispecific antibodies covered by Medicare?

Alexandra:        

I believe they are. I would have to double-check, but I’m fairly certain some of our patients have Medicare and have had bispecific antibodies.

Katherine:     

Okay, so for the patient who is getting the bispecific antibody treatment, what are the lifestyle alterations that we as care partners need to make? Any changes to their diet?

Alexandra:        

No. I mean, a lot of patients definitely want to maximize anything they can do to make themselves feel better and help their myeloma respond. But what we’ve seen is that there’s not one particular diet or cutting out one particular food that’s going  to make a long-term or significant impact on any cancer therapy. The best thing that you can do in terms of diet or lifestyle is to try and just maintain a healthy lifestyle to balance all your other medical needs. You want to make sure your blood pressure is in good control.

You want to make sure if you have diabetes, that your blood glucose is in a good range. Because having those things be in good control is going to make your therapy and potential complications more manageable. 

Katherine:         

Okay. Can bispecific antibodies cause anemia?

Alexandra:        

Yes. So, bispecific antibodies, all of the three that are approved, can cause lower blood counts in all of your blood counts. Red cells, white cells, and platelets.

I will say we’ve usually seen that happen more at the beginning of therapy, and then as patients are on the therapy longer, their counts do tend to recover. So, whether that is from just the initial disease response, or it might be from the cytokine release syndrome, we see low blood counts with that, we don’t always 100 percent know. But it certainly can lower all of your blood counts.

Katherine:         

Okay. Gina asks, is there any home equipment we will need to help during treatment?

Alexandra:        

Nothing is required. So, you’re not required to have any sort of medical devices at home. Well, I’ll take it back. I would like everyone to have a thermometer so that if you do feel sick, you can at least check your temperature. Sometimes having a blood pressure cuff or an oxygen monitor at home, that can be helpful if you’re not feeling well, just to see if things are out of range. But there’s definitely not a requirement for those things at home.

Katherine:         

Okay. What can care partners do to help the patient have a more positive outlook during therapy, especially when they’re feeling down and depressed?

Alexandra:        

Sometimes I think the best thing that you can do is acknowledge that this is really hard. I mean, changing therapy, having myeloma, going through a hospital stay is really challenging. And sometimes patients just need to hear, “This is a really hard situation, and you’re doing a great job. Taking all your medication, going to the hospital for these treatments, coming to your follow-ups.” Even those small things, giving encouragement and acknowledging how hard that is, even if it seems like it’s not a big thing, can really give a patient a different perspective on how things are going.

Katherine:         

Are there support groups specifically for care partners?

Alexandra:        

We do have a care group, a support group here at Dana-Farber that is for care partners, run by our social work team. I think it is both now virtual and live, but I’m not positive if they’ve gone back to in-person support groups yet. But I do think the American Cancer Society has some good online groups as well.

Katherine:         

Okay, that’s good to know. Thank you. So, before we end the program, Alexandra, I’d like to get your thoughts about the topic. What message do you want to leave the audience with?

Alexandra:        

I would tell them that bispecific antibody therapies are a great option for patients in the current myeloma setting. I know that being in the hospital for a week is an incredibly big ask, especially after everything patients are going through with their treatments and then having their disease progress. But the inpatient stay for the long term, hopefully, outpatient benefit of a quick injection every other week with minimal toxicity is certainly worth it. And so I would try to keep an open mind about bispecifics and get excited about them.

Katherine:         

Thank you so much for joining us today, Alexandra. We really appreciate it.

Alexandra:        

Thank you so much for having me.

Katherine:         

And thank you to all of our partners. If you would like to watch this webinar again, there will be a replay available soon. You’ll receive an email when it’s ready. And don’t forget to take the survey immediately following the webinar.

It will help us as we plan programs in the future. To access tools to help you become a proactive care partner, visit powerfulpatients.org. I’m Katherine Banwell. Thanks for being with us.

PODCAST: Myeloma Patient Expert Q&A: Dr. Sikander Ailawadhi

 

 
In this START HERE myeloma program, Dr. Sikander Ailawadhi from Mayo Clinic spotlights priorities in the rapidly expanding myeloma treatment landscape. Watch as Dr. Ailawadhi addresses pressing questions submitted by patients and families, providing invaluable guidance and reassurance in navigating the complexities of myeloma care.

Download Guide  |  Descargar Guía

See More from START HERE Myeloma

Transcript:

Lisa Hatifield:

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

Joining me today is Dr. Ailawadhi, back by popular demand. Dr. Ailawadhi is a respected multiple myeloma expert from Mayo Clinic. Dr. Ailawadhi’s career focus includes the treatment of plasma cell disorders like myeloma and understanding the epidemiology and pathophysiology of this disorder. It’s always such a pleasure having you, Dr. Ailawadhi. I’m really excited you’re joining us again. So thank you for joining us.

Dr. Sikander Ailawadhi:

And thanks a lot for having me, Lisa. This is excellent. I look forward to this next iteration of the Patient Empowerment Network START HERE program.

Lisa Hatfield:

Thank you. So before we dive into today’s discussion, please take a moment to download the program resource guide using the QR code. This guide contains pertinent information to guide you both before and after the program. And this program will provide you with a comprehensive update on the latest myeloma news and its implications for you and your family. Following that, we’ll launch into some questions that we have received from you.

So let’s start here. Dr. Ailawadhi, at this juncture in myeloma history, we are witnessing unprecedented activity, a surge of new treatment options, and a wealth of insights. Today, we are privileged to have your expertise to help us decipher these developments and shed light on the advancements shaping the landscape of myeloma care. First, we’re going to get a high-level update from Dr. Ailawadhi on what the latest myeloma news means for you and your family. And then we’re going to talk about some questions that you’ve sent in. So let’s get started with the high-level update, Dr. Ailawadhi. Can you speak to the latest news and priorities in the rapidly expanding myeloma treatment landscape?

Dr. Sikander Ailawadhi:

Excellent. I think, Lisa, that’s an excellent and important question. Because as you rightly mentioned, there is such a large amount of data that is coming through for myeloma all the time. I mean, it’s almost, we kind of talk about the fact that every time you turn your shoulder or look over your shoulder, there is a new drug approved. So I can imagine this can be very overwhelming. So what I’ll say is that in my opinion, there are some categories of new data that are pertinent and important for our patients. 

The two or three out of them that come to my mind, one is what’s called CELMoDs, or there are a couple of agents there called iberdomide, mezigdomide. These are showing some interesting data. Important to keep in mind that they are somewhat related to the immunomodulatory drugs lenalidomide (Revlimid), pomalidomide (Pomalyst), but they’re showing benefit in patients who have had len and pom before and have progressed. So exciting stuff there.

We’re also seeing some interesting data about newer CAR Ts and bispecific antibodies. They are all coming up with some benefits in some cases. I think it’s important to keep in mind that the bispecifics are landing at the 60 to 70 percent response rate, and CAR Ts are typically landing at the 80 to 90 percent response rate, but there are more agents expected.

There are also some newer bispecifics in different classes, like one of them is called cevostamab, which is an FcRH5 inhibitor or targeting bispecific. So newer bispecific, not just more of the same category. And there has also been recent data about Bcl-2 inhibitors, which have been traditionally used for patients with translocation 11;14.

There have been some negative data, negative as in trials, which did not pan out with a drug called venetoclax (Venclexta), but there are two other drugs that had some recent data shown from different companies, which were exciting information. So in my mind, those are kind of the broad new drug categories. There is another, a couple of other quick things that I’ll mention.

One is we’re getting more and more information about real world experience with these new drugs. It’s good to see that CAR Ts are panning out very similar in the real world as they are in clinical trials. We’re also seeing that the side effect profile of a lot of these newer novel immunotherapy drugs is similar as seen in the clinical trials.

Racial ethnic disparities are something which are very close to my heart, and there is more and more information coming out in that. Unfortunately, highlighting the disparities more still rather than yet coming up with solutions. And I think the last thing that I feel which has been recent has been at the American Society of Hematology meeting in 2023, which was in December in San Diego.

One of the myeloma studies actually became a plenary session presentation, which is a pretty big deal for any disease area. So one thing is that it gets highlighted. Secondly, it was a combination of a regimen called isatuximab-irfc (Sarclisa) with carfilzomib (Kyprolis), lenalidomide, and dexamethasone (Decadron) in newly diagnosed patients.

It’s a randomized trial, Phase III, which was presented. I think the important part is we saw unprecedented deep responses and patients in much, much higher numbers than before becoming MRD-negative. So very deep responses. So these are kind of some very broad, but lots of highlights that I talked about.

Lisa Hatfield:  

All right. Thank you. So can you also talk about some of the newer tools for myeloma progression and relapse and what patients might want to know about that? And in particular, maybe talk a bit about MRD testing and the role of MRD testing for patients who relapse.

Dr. Sikander Ailawadhi:

Excellent question. Lisa, I think the first and foremost thing an important part for our patients to learn is what are their “tumor markers,” for the, or disease markers for myeloma. We can follow myeloma by either the M spike or monoclonal protein, by light chains, by monoclonal protein in the urine or blood. And it’s important to keep that in mind because every now and then we’ll see patients who say, Hey, my ratio changed. So I’m progressing. Well, that comes after the light chains change. So it’s important to understand the role of these things.

So M spike in the urine, M spike in the serum and light chains. One of them is typically the marker for a patient. Now the MRD status or minimal residual disease that is looking for one cell, one myeloma cell from amongst 100,000 cells in the bone marrow. So it is looking at a very deep level. The most important benefit of MRD testing right now is to understand that if a patient turns MRD negative, then they have a superior outcome. Their prognosis is better. Their progression free survival, or the time before their disease comes back is longer. 

But when a patient is MRD-negative and is being followed or maintenance or whatever, if the bone marrow turns MRD-positive, then that might be the sign that the disease might be coming back. Right now, we do not keep switching drugs to get to MRD-negative. That is not the goal of treatment. The way to think about it is we want to get to MRD-negative, but that means it’s incumbent upon us to try and pick a regimen that is more likely to get to MRD-negative. That’s the way to think about it.

Let’s pick a regimen more likely to get us into MRD-negative and hope that we get to MRD-negative. We see every now and then that the patients keep switching regimens just to get to MRD-negative. That’s not the way to go because you’re just using up options too quickly, too fast. A common question that patients ask is, well, does that mean I need to get annual bone marrow biopsies and MRD testing? Probably not.

That’s too much testing. So what I suggest is that once somebody has turned MRD-negative, it’s important to keep an eye on every single thing. Now, change in any of the routine labs, imaging, new symptoms, etcetera. That’s when we switch to the bone marrow again and see if the patient has turned MRD-positive. There are clinical trials going on right now which are stopping drugs based on repeat MRD negativity or starting drugs on MRD positivity. But those are clinical trial questions.

Lisa Hatfield:

Okay. Thank you for that. So along those same lines, since you’re a Mayo physician, I’m curious about the mass spec testing. So if a patient say has been MRD-negative for some time, still wants to monitor at a deeper level, even though it’s not commercialized yet, do you see a role for mass spec testing on a regular basis in the future and being rolled out to community facilities also?

Dr. Sikander Ailawadhi:

Absolutely, Lisa. I did not specifically bring it up because mass spec is not, like you rightly said, is not yet commercially available. Now we’re doing mass spec quite frequently at Mayo Clinic. Basically mass spec is taking up a blood sample. Important to keep in mind, it’s not a bone marrow test, it’s a blood test, but it looks for those abnormal proteins based on the protein weight at a much, much lower level. Our SPEP or serum protein electrophoresis does not pick up very small quantities of the protein mass spec does. So in an essence, the mass spec, if somebody is negative on that, turning mass spec negative to mass spec positive may be an earlier sign of the disease coming back rather than the SPEP yet turning positive. But as you rightly said, it is not yet commercially available. I do see the benefit of it.

There is more and more data coming in favor of it, and there was data that was also at ASH. So I do see that in the future we’ll be able to most likely have it available more widely. At this point, it is just a blood test to attempt to check the disease level at a much deeper level and be able to notice if the disease is progressing sooner than our currently available tools.

Lisa Hatfield:

Great. Thank you. And as a patient, I like to have one more data point that they can get from my blood, not from my bone marrow to assess the disease. So thank you for explaining that. Regarding survivorship, patients are living longer with myeloma in general because of the novel therapies that have come out in the past few years. So how is myeloma survivorship evolving, and what’s different now than it was five or 10 years ago in terms of treatment planning?

Dr. Sikander Ailawadhi:

Yeah, I think it’s very important to keep that in mind. When I see a newly diagnosed patient, I’m not just telling them, “Hey, this is your induction therapy, and your transplant is the goal.’ We’re trying our best to decide that patient’s life journey with myeloma over the next 10, 15 and hopefully more years. So we’re trying to pick and choose the regimen that is most likely going to help the patient the most today and most likely will give a longer duration of the response. So when you say survivorship, that also very importantly brings up the point that patients are living with myeloma longer. We have to manage their health overall. So looking for any side effects from treatment, managing them very well so the patient is able to stay on the treatment and maintain good quality of life.

There are actually, are clinical trials looking at stopping treatment when there is a very deep, prolonged response. Again, going towards survivorship and giving the patient’s quality of life. There is looking for other cancers. In fact, I had a patient in the clinic and we were talking about just myeloma in general and I was telling them, “Okay, please remember you may not want to do a colonoscopy, but you already have one myeloma cancer diagnosis. The risk of subsequent cancers is always there in any cancer patient.” So that was a male person. So I said, “Okay, please do not miss your colonoscopy. Please do not miss your prostate screening and whatever is age-appropriate must be done.” So managing everything because myeloma is not a sprint, it’s a marathon.

We want to make sure that we pace ourselves well so we manage all the symptoms, all the signs. Bone health becomes much, much more important because the same bone structure is now going to carry us longer and many more years. And as you rightly said, planning, which treatment comes first, which comes next, when does CAR T come? It’s not that everybody must get CAR T today. That’s not the answer. So what to use when becomes extremely more important.

Lisa Hatfield:

Thank you for that. And thank you, Dr. Ailawadhi, for that important reminder. All of you watching, get your regular screenings, like he said, prostate cancer, mammograms, colonoscopies, get it done. So thank you for that.

One of the things that comes up with that regular, not regular screening, but monitoring after certain therapies for future malignancies, there’s been some discussions about post-CAR T, particularly with T-cell malignancies and monitoring for that. Can you just give a little description of that and any concerns that you have with that or any encouragement you have regarding that and whether that weighs into your treatment options that you give to patients when they are asking about CAR T therapy?

Dr. Sikander Ailawadhi:

Absolutely. Extremely important question, Lisa. This really had a lot of discussion going on. It’s been going on for the past few months now. Okay. So first let’s explain the landscape. The FDA reviewed CAR T-cell treatment because of the fact that there were about 19 T-cell malignancies noted in several thousand patients.

Out of those 19 cases of T-cell malignancies, there was one case of multiple myeloma to the best of my knowledge. Now, risk of subsequent cancers is something, unfortunately, every cancer patient lives with, but in myeloma, we have known about that, especially from our historical knowledge of second malignancies with lenalidomide-based maintenance therapy post-transplant. So subsequent malignancies have always been a risk. There is some risk that is being talked about with CAR T, but frankly speaking, the way I look at it, the risk is significantly lesser than the potential benefit.

Because remember when these CAR T therapies, the two agents got approved in myeloma, they were approved in a situation that there was no standard therapy. And we saw somewhere about 70, 75 percent response rate with one of them and about 98 percent response rate with the other one. So in a setting where there was nothing, you can see the degree of benefit. And the risk of second malignancies is relatively small. So we must discuss this.

A patient must be aware of it, but I think the benefit is way more than the risk. So we document, we discuss, we have specific documentation that we do and specific information that we share with patients, but I think still the benefit is significantly more than the risk.

Lisa Hatfield:

Great. Thank you so much for explaining that. And for any of you out there watching this, Dr. Ailawadhi is a myeloma specialist, and I highly encourage anybody who is looking at CAR T therapy or even for a first consult for myeloma, seek out even one consult from a myeloma specialist. It is so important in trying to understand these therapies and any fears you may have regarding those therapies and the risks of that. So really appreciate that, Dr. Ailawadhi. Thank you. So I think it’s time now to start answering questions from patients that we received from all of you in the audience.

Please remember, this is not a substitute for medical care. Always consult with your medical team. And we are going to jump right in, Dr. Ailawadhi. We have a lot of questions from patients here and I’ll just start with the first one. This patient is asking, my M spike keeps rising in spite of chemo. What can I do?

Dr. Sikander Ailawadhi:

Very important question, Lisa. Every patient must understand what their disease marker is. This patient is asking about the M spike, which is the monoclonal spike, whether it’s in the blood or in the urine. And if the M spike is continuing to increase and there is a significant increase, significant is defined by at least 25 percent from the nadir or from the bottom most point with the, at least a absolute increase of 0.5 gram per deciliter. So half a gram per deciliter. So we want a 25 percent increase, but we also want at least 0.5 gram per deciliter.

So if somebody had an M spike of one at their best point, then the increase to 1.5 is significant. If somebody had the M spike of 0.2, then it’s not the 25 percent increase, it’s the 0.5 that must happen. So they hit 0.7 and that’s a significant increase. So that’s how we think about M spike, 25 percent with an absolute of at least 0.5 gram per deciliter.

If that is indeed happening, this would be considered a biochemical progression. And at that point, it should be considered to switch around the treatment because we don’t want the disease to grow to the point that there are actually symptoms showing up or organ damage happening. We want to be able to capture the disease progression sooner and act upon it.

Lisa Hatfield:

Great, thank you. Do you have any recommendations for people who, as we might have some patients watching this, who are light chain only? Any guidelines on if those numbers are rising?

Dr. Sikander Ailawadhi:

That’s an excellent question too. So if somebody has light chains as their marker, we are looking at an increase in the involved serum free light chain. So if somebody has kappa as their marker, the kappa is going up, or if they have lambda as the marker, the lambda is going up. Typically, if both of them go up, that is not disease progression. That could be coming from kidney dysfunction. Somebody is dehydrated and they get labs checked. Both kappa and lambda might be elevated. Again, a 25 percent increase in the absolute. But at the same time, we are also looking at at least 10 milligram per deciliter change.

So if somebody had a light chain of two milligram per deciliter, if it goes to 12, that might be a significant change. But I can say that light chains are a little bit more volatile and they do get affected by our fluid status. So if I ever notice a patient with a light chain increase, I’m more likely to repeat the test very soon, maybe even at a couple of days, one week interval, just to make sure that there is a trend rather than just a fluctuating light chain.

Lisa Hatfield:

Okay. Thank you for that information.

Dr. Sikander Ailawadhi:

And I should maybe, very quickly add, we do not check light chains in the urine. Light chains should be checked in the blood. Urine light chains are very nonspecific and there’s no need to test them.

Lisa Hatfield:

Okay. That’s helpful also. So patients don’t have to walk around with their big orange jugs full of fluids. So thank you. All right. This might be a complicated question to answer. But in general terms, for those who relapse for the first time, what are the best treatment options?

Dr. Sikander Ailawadhi:

I think that’s a very important, and I can imagine a scary situation. So somebody who relapses in general, not just even the first time, the factors that are taken into account for deciding what treatment they should get, there are broadly three categories of factors. Patient factors deciding what’s the age, what’s the other comorbidities, are they diabetic, are they heart disease, kidney dysfunction, because those things go into the decision of what may or may not be given. So patient factors.

Also importantly, how close are you to your treatment center? Can you come in for infusion or injection drugs time? And again, can you prefer or do you prefer oral drugs only? Et cetera. Those things become important. Then that…so that’s patient factors and disease factors. How fast is the progression? Is it high-risk disease, standard risk disease? Is it biochemical progression like the previous person asked?

Or is it actually a clinical progression in which there’s kidney dysfunction or anemia or bone disease? Because the choices and the urgency of treatment may change. So patient factors, disease factors, and then drug factors are the third class or third category, which is what have you had before? How long have you been on it? Are you on maintenance or not? Is your disease considered refractory to a certain agent, meaning resistant to a certain agent? Typically, if you were on a treatment and your disease is progressing, that same drug may not be used again.

And there are some times that we will reuse a drug, but generally not. We can use the same class, but we may not typically use the same drug. So I think the choice of treatment depends on all of those factors put in. And then we come up with one or two or three regimens and we discuss them with patients. And, of course, being an academic, physician, I must say there is always, you must always seek out good clinical trials if they’re available to you. That is the way our field moves forward.

Lisa Hatfield:

Yes, thank you for that information. So we have another patient asking, “Do myeloma patients require multiple prior therapies prior to being eligible for CAR T?” And what’s the rationale for not implementing CAR T immediately, which probably has to do with FDA approval based on clinical trials at this point?

Dr. Sikander Ailawadhi:

Absolutely. You’re absolutely right, Lisa. So any drug, let alone CAR T, any drug can only be given in the situation that it is approved by the FDA. So basically in accordance with that drug’s FDA approval label. Currently, CAR T-cell therapy is approved in the U.S. after at least four prior lines of therapy. And the patient must have had treatment with at least one proteasome inhibitor,  for which we have three drugs, bortezomib (Velcade), ixazomib (Ninlaro), and carfilzomib. They must have been treated with at least one prior immunomodulatory drug; lenalidomide, thalidomide (Thalomid), pomalidomide. And they must have been previously treated by at least one monoclonal antibody, daratumumab (Darzalex) or Isatuximab (Sarclisa).

Once the patient has had all these criteria met, they’ve become a candidate for CAR T-cell therapy. Frankly, we cannot just use a drug anywhere because we cannot use a drug where it has not shown to be of benefit. And importantly, it has not shown to be of any risk. So CAR T-cell therapy in the first line setting is being studied in clinical trials, but is not FDA-approved. Currently approved only after four prior lines, but the FDA is reviewing data for both the CAR T’s to see if they may be available sooner. As of right now, that approval is still pending.

Lisa Hatfield:

Okay, thank you. We’re hopeful that that will happen soon.

Dr. Sikander Ailawadhi:

Hopeful.

Lisa Hatfield:

Yeah. So interesting question from a patient, “Does CAR-T therapy actually change one’s DNA?”

Dr. Sikander Ailawadhi:

No. The CAR T therapy does not change one’s DNA. What happens is, there are T cells taken out of a patient. The DNA of those T cells is modified and then those T cells are given back to the patient. Those T cells do not go and integrate into your other healthy body cells or your stem cells, et cetera. Those T cells, it’s almost like giving a boost of immunity, which is targeted against your myeloma. So those T cells go in and they fill those myeloma cells. Now we hope that those T cells perpetuate and teach or create some memory T cells and that immunity lasts a little bit longer. But all of that genetic modification stays within the T cells. It does not integrate anywhere else.

Now, I know there was a previous question about T-cell lymphomas, that is related to this question in a way because the risk that is theoretical is, that that genetic modification in the T cells might make those T cells replicate uncontrollably leading to a T-cell cancer or T-cell lymphoma. But I’m saying that this is theoretical because while it is possible, it happens extremely rarely and even in the cases where the cancer happened, it has been seen that the cancer may not come from that portion of the DNA that was…that’s where the modification was done. So, low risk.

Lisa Hatfield:

Okay. Thank you very much for that.

Lisa Hatfield:

Okay. So what would be the next steps, Dr. Ailawadhi for a patient who’s had CAR T and reaches a relapse state or is relapsed?

Dr. Sikander Ailawadhi:

Yep. This is something, unfortunately is the truth of the matter in myeloma at least that we are, we don’t seek cures. We have had some long remissions. I have, for example, patients who are now reaching three, three-and-a-half years of remission on CAR T treatment who received it on clinical trials even before they got FDA-approved.

But, unfortunately, the disease does come back. So what happens is, we are seeing data that the novel, other novel immunotherapies like bispecific antibodies, even the ones who go after the same target as CAR T, BCMA targeting bispecifics, they do have some response rates, good response rates in post CAR T setting. So the bispecific antibodies by themselves may give us 60 to 65 percent response, but in the post CAR T setting, that response might go down to 40, 45 percent. So less responses, but still possible.

There are also bispecific antibody. There is one available, which is not against BCMA, it is against GPRC5D. That’s a bispecific called talquetamab-tgvs (Talvey). So a novel target. There is…there are of course a lot of clinical trials. There are some clinical trials that are even looking at CAR T post-CAR T. So different kind of a CAR T. Those clinical trials are going out. So what I would suggest is that if your disease progresses after CAR T-cell treatment, you should very strongly consider getting to a specialist myeloma center and get an opinion like you mentioned, Lisa.

That is so important because the choice of treatment is extremely important at that time. And we are trying our best to sequence all the options we have, in a way, actually one of my patients mentioned, one of these days, Hey, does that mean that I’m basically buying time till something new and exciting comes along? And I said in a way that is true. That we are trying to stretch all our treatments and get to the point that something new and promising just like CAR T comes, and hopefully we get longer benefits again.

Lisa Hatfield:

Thank you for that. So when you say there’s a possibility of CAR T and then a post-CAR T maybe a second CAR T. Would that be a different target then?

Dr. Sikander Ailawadhi:

So there could be a different target. I have, in fact, just yesterday I saw a patient who had received one CAR T in a clinical trial and then they were subsequently able to receive a CAR T standard of care, which had been FDA approved. So they used different CAR Ts, but one was in clinical trials and one was standard of care.

Lisa Hatfield:

Oh, great. Okay. Again, important to see a myeloma specialist to tease out all this information. Thank you. All right. This patient is asking, “I’m 81 and living with comorbidities. The myeloma was diagnosed after bone marrow test. How is treatment fitness determined?” And also a question about that is if you’re given an ECOG status of something you don’t like it, can that be improved after you’ve had treatment?

Dr. Sikander Ailawadhi:

Absolutely.

Lisa Hatfield:

Maybe be eligible for a trial or something.

Dr. Sikander Ailawadhi:

Correct. Correct. That is so important. When this patient mentioned that they’re 81-year-old and they’re living with comorbidities, I think, so when I’m talking to a patient who’s new to me, it’s very important for me to try to tease out what was their performance status or their fitness status prior to myeloma. Because my goal is to try to get them as close to that as possible. Now if this patient is saying that they were already quite frail before the diagnosis of myeloma and myeloma is added to the frailty, then it becomes a little tricky because we’re starting in a difficult spot. We do determine fitness by asking questions, simple questions like, what can a patient do at baseline? Can they do grocery store or grocery shopping by themselves? Can they walk around the block? Do they get short of breath? Et cetera.

And frankly, there are 81-year-olds who are playing golf every day and are fitter than me. So I’m just saying that age by itself is not the criteria. And, Lisa, like you rightly mentioned, if there are fitness issues coming from the disease itself, then that’s the time that we actually have to work with the treatment, get the treatment started, and then assess the fitness a couple of months later, a couple of cycles later. Because the treatment may have worked and may have improved the fitness quite a bit.

Lisa Hatfield:

Great thank you for that. So this person is asking, their husband is starting maintenance therapy, so I am assuming they just finished induction therapy, having leg pains mostly at night. Could this be a form of peripheral neuropathy or is maybe from bisphosphonates or from any of the medications that maybe were used during induction?

Dr. Sikander Ailawadhi:

So, excellent question. So, this is almost going back to that survivorship question that we discussed earlier, that it’s so important to manage the side effects and maintain quality of life. So, a lot of patients with myeloma will say that I have cramping or symptoms or some pins and needles at night more so. Part of it is because body’s at rest, relaxed, things, symptoms become more focused. Yes, it could be peripheral neuropathy, but at the same time certain drugs caused muscle cramping or what’s called myalgias, sometimes maintenance therapies can cause that.

It’s important for somebody to be able to determine is it coming from muscles or nerves? Is it coming because some electrolytes are abnormal. Like one of the common things is low magnesium or low potassium can cause neuropathy, for example, or cramping. I’ve had patients who will get some over-the-counter lotions or some forms et cetera, which are infused with some electrolytes and say that they feel some benefit. So topical things are good. So I think it’s important to figure out is it muscle or nerve and is it coming from drugs or disease? And that’s where your physician can help tease it out.

Lisa Hatfield:

Okay, thank you. So we have a patient who is talking about her genetic abnormalities, but has been through both auto and allo stem cell transplant in the last two years and has relapsed. And is asking, “Can CAR T-cell therapy help me?” And would she even be eligible for CAR T therapy given the allotransplant?

Dr. Sikander Ailawadhi:

That’s an important question. So first of all, sorry to hear that, that your disease is behaving that aggressively, that you’ve had both the transplants in the past two years and still having issues. So yes, CAR T can still be used after an allotransplant. There are some criteria. You should not be on any graft versus host suppressive medications, and you should not have any active graft versus host disease going on. So depending on those, yes patients can get CAR T post. And, in fact, I’ve had a couple of patients who’ve had CAR T after allotransplant.

Lisa Hatfield:

Great, thank you. I’m sure that’ll give this patient some hope. Are there any studies showing that treatment can be tapered? Tapered to by daily, once 90 percent reduction in myeloma has occurred with various therapies. So in general, you may know what medication this patient’s talking about, but is that possible to do that, to taper therapy?

Dr. Sikander Ailawadhi:

So, absolutely, first of all, in myeloma care, Lisa, you had mentioned initially that as somebody went to maintenance, they have had induction. So there are these terms used for categories of treatment, induction, consolidation, maintenance. But if the disease gets controlled adequately at a certain time point, the treatment can be modified to a maintenance. It depends on the regimen.

Some regimens, for example, we are able to get rid of the steroids after a certain time and then in certain regimens the drugs can be reduced in dose or frequency, et cetera. All of the drugs we use have maintenance regimens and maintenance doses. But I should put a word of caution there. I see very frequently that the moment the labs improve, this quote unquote “maintenance” is brought in.

That’s not the right way to do things. The right way is to go back to the clinical trial based on which this regimen was started. And according to that clinical trial, after however many cycles of treatment the maintenance was supposed to happen, it should happen. So if I’ll very quickly say if somebody stays, starts on a regimen and within four months their M spike comes down, and now it has plateaued. Because our drugs are so good that they work that fast. And somebody says, “Okay, four months of that is enough, let’s save it for the future. Let’s go to maintenance.” I would say, “Absolutely not.”

In fact, there is data suggested from a couple of regimens that if significant modifications were made prior to one year of the regimen, then the outcomes were inferior. And I’m not going in specific regimens and I’m not saying that that is applicable to everything, but what I’m saying is, yes, maintenance and tapering is possible. In fact, there are clinical trials looking at even stopping medication. But when and how that change is to be made is very very important. It’s critical. If your physician is not comfortable about that time point, reach out to a myeloma specialist. They should be able to guide when and how to reduce or taper or put on maintenance.

Lisa Hatfield:

Thank you. And that’s very important what you said about induction therapy. Go back to the clinical trial and look and see what the clinical trial said as far as how long that treatment should last because it is exciting as a patient when you start seeing those numbers dropping exponentially. They’re just plummeting, and you want to go off it, you don’t feel great. It’s hard to stay on a therapy for 6 to 12 months that you don’t really enjoy and nobody really does. So that’s important. And then maybe talk about maintenance therapy later. It would be nice to have limited duration maintenance, sometime in the future for induction therapy. Stick with what the clinical trial says. So, okay, this patient is asking another really important question, “I have myeloma and now my daughter does as well. She’s 37, is multiple myeloma hereditary?”

Dr. Sikander Ailawadhi:

I’m sorry to hear about this situation and I’m so sorry that your daughter who’s 37 got diagnosed. There is a small, very small number of very young patients and I’m saying using this term very young, which is just a generic thing that I’ve said because myeloma median age of diagnosis 68. I saw a patient who was diagnosed at 33 and they’re 40 now and they’ve already gone through every single thing that they can think of. And we were talking about clinical trials. So, typically myeloma is not hereditary. It is not something that is passed along through the generations. But what I would say is that there is, if this sort of a situation is happening that you have myeloma and now your daughter has it at a young age, it is important for you to consider getting genetic counseling.

So a genetic counsel for them to be able to look deeper into it. There is not a very standard specific test, so for me to say, Hey, you go and get this genetic test done and that’ll find out this mutation, whatever. But it’s important to get, go through some genetic counseling for them to be able to look a little bit deeper, some next-generation sequencing, what is called germline testing or somatic testing. They should be able to compare both the parent and the daughter’s disease as well as what’s called germline, which is their native DNA, which they were born with, to see if there is anything that jumps out of that. But that would be important to go through at a larger cancer center or if that service is available through your local physician also. That would be great.

Lisa Hatfield:

Great, thank you. Well, I think that’s it for our questions. That’s all that we have time for. But Dr. Ailawadhi, thank you so much for once again, being part of our Patient Empowerment Network START HERE Program. Because it really is these kinds of conversations that help patients, me included, feel more empowered to take questions back to our providers and our healthcare team. So thank you so much for joining us and thank you out there to everybody who’s watching this program, we appreciate you and we appreciate your time and expertise.

Dr. Sikander Ailawadhi:

Thanks and I look forward to the next time.

Lisa Hatfield:

Thank you. I’m Lisa Hatfield. Thank you for joining this Patient Empowerment Network Program and we look forward to seeing you again soon.

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care? Resource Guide

Download Resource Guide

PEN-199_Elevate_Guide_040924

Download Resource Guide

See More from Elevate Myeloma

Elevate | What Role Can YOU Play in Your Myeloma Treatment and Care?

How can you elevate your overall myeloma care and treatment? Myeloma expert Dr. Sikander Ailawadhi discusses advances in myeloma care, the importance of patient participation in myeloma treatment decisions, and shares key advice and resources for self-advocacy.
 
Dr. Sikander Ailawadhi is a hematologist and oncologist specializing in myeloma at Mayo Clinic in Jacksonville, Florida. Learn more about Dr. Ailawadhi.

Related Resources:

Self-Advocacy in Myeloma Care | Advice From an Expert

Self-Advocacy in Myeloma Care | Advice From an Expert 

Myeloma Combination Therapy _ What Patients Should Know

Myeloma Combination Therapy | What Patients Should Know 

What Should You Know About Emerging Myeloma Treatment Options? 

Transcript:

Katherine:

Hello and welcome. I’m your host, Katherine Banwell. It’s no secret that the quality of care that patients receive can vary depending on a number of factors and patients who are educated about their condition and involved in their care may have improved outcomes. That’s why the Patient Empowerment Network developed the Elevate Series, which aims to help patients and care partners be informed about their disease and more confident participating in conversations with their healthcare team.

In today’s program, we’re going to hear from an expert to learn more about myeloma and hear tips and advice for accessing better overall care. Before we get into the discussion, please remember that this program is not a substitute for seeking medical advice. Please refer to your healthcare team about what might be best for you. Well, let’s meet our guest today. Joining us is Dr. Sikandar Ailawadhi. Dr. Ailawadhi, welcome. Would you please introduce yourself?

Dr. Ailawadhi:

Yes. Thanks a lot, Katherine. Thanks for this opportunity. To all our viewers, listeners, I’m Sikandar Ailawadhi. I’m one of the hematologists oncologists at Mayo Clinic in Jacksonville, Florida. So, at this Jacksonville, Florida site of Mayo, I lead our myeloma group, and we have a very comprehensive program with inpatient outpatient treatment and lots of clinical trials, cell therapy, et cetera. I look forward to the discussion.  

Katherine:

Well, thank you so much for joining us today. I know you’re a busy man. I’d like to start by discussing your role as a researcher. You’re on the front lines of advancements in the myeloma field. So, what led you here, and why is it so important to you?   

Dr. Ailawadhi:

Yeah. So, Katherine, thanks for asking that question because before jumping into the disease state and clinical trials and data, et cetera, I think it’s important for all of us to keep in mind what kind of brought us here or what kind of keeps driving us further. So, I think for me, the decision to come to the field of multiple myeloma was very strongly influenced by my mentors.

My mentor who shaped my career and also got me interested in this area. I think during my training, it was the time that newer drugs were just beginning to come about. So, the field of myeloma was just beginning to change. And since then, obviously, there have been lots of advancements, lots of research, clinical trials, new drugs, so that the outlook for not only the myeloma patients has improved quite a bit.   

But also, for physicians, researchers, us academicians who work in this field, the opportunities are much, much more. And because I trained at a large academic center, I – and again, with working with my mentor, I got so interested in the clinical research because frankly giving the available drugs is one thing but being at that cutting edge where you can bring newer drugs to life, newer drugs to our patients’ lives, that was what was most important for me and is the driving force for my work today.  

Katherine:

Thank you for that explanation, Dr. Ailawadhi. I appreciate it. So, when it comes to choosing therapy for myeloma, it’s important to work with your healthcare team to identify what might be best for you. How would you define shared decision-making and why is this so critical to properly managing life with myeloma?  

Dr. Ailawadhi:

Excellent question, Katherine. Shared decision-making or a process in which the physician, the health care team, and the patient, their caregivers, everybody comes together, shared, to make a decision that we feel is in the best interest for that patient at that time. That is the whole concept.  

Whenever we think about treatment decisions, in our mind, the three main components that have to be considered every single time. Not just newly diagnosed or relapsed or third line or whatever, every single time a treatment decision has been taken, we must consider patient-related factors. What is their preference? What are their goals? Do they have caregiver support? How far do they live? Do they want IV? Pills? Any side effects that are there?  

Comorbidities? Other issues? Financial conditions? Everything comes into play, patient-related factors. Then, there are disease-related factors. How fast is the disease growing? Is this new? Is this old disease, high-risk, low-risk, or standard risk? Or what has been given before, et cetera. So, patient and disease-related. And the number three is the treatment-related factors. What is being considered for the patient? What are the ins and outs, pros, and cons?   

All of this has to be laid out in front of the patient and preferably also their caregiver if the patient has someone who they can share their decision with.  

And when we put all of that in the mix, we come up with a decision which is hopefully in the patient’s best interest. They are more likely to go through with it. They are informed. They are involved in their care. And then, hopefully, if the patient starts on a treatment that they are interested in, knowledgeable about, and committed to, we’ll be able to keep the patient on that longer term and get the best benefit out of it.   

So, in my mind, the main reason for shared decision-making is to make sure my patient is committed to that treatment. They understand that treatment. And we make this kind of bond between us as clinicians and our teams and the patient and their home team, their family team, their caregiver team so that everybody is working together with a singular goal. Right treatment for the right patient at the right time because it must be patient-centric, not research or clinician, or drug-centric. 

Katherine:

Yeah. Okay. That’s good advice. What are myeloma treatment goals, and how are they determined?   

Dr. Ailawadhi:

So, I think the myeloma treatment goals can be very different depending on what vantage points you’re looking from. My treatment goal is to provide the best treatment for my patient that has least side effects, gets a deep control, and my patient’s able to live long with a good quality of life. Okay. But that’s my goal.  I need to figure out what my patient’s goals are, and sometimes our patient’s goals are very different. A patient’s goal might be that they want to really avoid side effects. Well, they want to live, lead their quality of life, and keep traveling. And this happens on a day-to-day basis.  

Just the other day, one of the patients said, “Well, I really want to keep driving around in my RV with my wife, because that is what we had wanted to do at this point of our life. What can you do to help me control my disease, but keep me driving my RV?” And we literally had to figure out where all they were traveling. We identified clinics close to them and connected with physicians so that they could continue their treatment wherever they were. So, the patient’s goals are very important, and in fact, I would say they are paramount. So, understanding what the patient wants. They may be wanting to control pain. They may be wanting to just live longer.  

They may be wanting to delay treatment so that they could watch their daughter’s soccer game. I’m just saying that the goals can be very different. It is important to lay them out. Every time you’re making a treatment decision, the goals should be laid out into short-, mid-, and long-term goals. I should bring my goals to the discussion. The patient should bring their goals to the discussion, and we come up with whatever is the best answer for them that suits them.  

Katherine:

So, you’re trying to maintain an open dialogue, an open line of communication, yeah.  

Dr. Ailawadhi:

Absolutely.  

Katherine:

What sort of tests should be done following a myeloma diagnosis?   

Dr. Ailawadhi:

Generally, when myeloma is suspected, we need to know what the basic blood counts are, something that is called a CBC, complete blood count. We’re looking for anemia, low white blood cells, low clotting cells, or platelets. We want to do serum chemistries or blood chemistries, looking for kidney function, liver function, electrolytes, calcium, et cetera.   

Then, we want to do some kind of an imaging of the body. Generally, routine X-rays are no longer done, and the most preferred is a PET-CT scan, a PET scan. We do PET-MRIs frequently. So, there are different tests available, but you want a good test to know what’s the state of bones and presence of any lesions or tumors. And then, the important question comes is doing a bone marrow biopsy.   

The reason for doing a bone marrow biopsy, and even if somebody has had a biopsy done from a compression fracture, et cetera, that diagnosed myeloma, a bone marrow biopsy still should be done. It gives us a lot of pieces of information.  

It tells us what is the percentage of plasma cells in the bone marrow. So, what is the disease burden we are starting with? Secondly, that bone marrow biopsy specimen can be sent for what is called a FISH testing, which is fluorescent in situ hybridization.  

It is basically looking for any mutations in the cancer cells. Based on those mutations, myeloma can be classified into standard or high-risk myeloma. And sometimes our treatment choices are differed based on whether somebody is standard or high-risk. So, blood work, basic counts – and I skipped over one of the things. Right after chemistries, I wanted to add also are myeloma markers.  

There are typically three lab tests of myeloma markers. One is called protein electrophoresis. It can be run in blood and urine. Ideally, it should be run in both. One is immunoglobulin levels, which gives us the level of IgG, IgA, IgM, et cetera. And the third one is serum-free light chains, which is kappa and lambda light chains. Neither one – none of these tests eliminates the needs for the other.

So, everybody, in the beginning, should have complete blood count, blood chemistries, SPEP or serum protein electrophoresis, urine electrophoresis, immunoglobulins, light chains, imaging, and then a bone marrow. This completes the workup. Then, based on that, the treatment can be determined.  

Katherine:

Well, you mentioned lab work. How often should tests and blood work be done?   

Dr. Ailawadhi:

Good question. Very, very important question because we see very frequently that the patients come in, they’re getting treatment somewhere, and every single time the patient steps foot in the door of that institution or wherever they’re going, they got a blood draw. That’s how they start their day. It’s needed more frequently in the beginning but needed less frequently later on.

Generally, the myeloma markers, those protein electrophoresis, immunoglobulins, light chains, they are frequently done just about every month. Generally, in myeloma, one month, three to four weeks is one cycle. So, at the beginning of every cycle, you want to know how good your response was. So, the myeloma markers once a month.  

The blood counts and chemistries in the first month, first one to two months, they can be done every other week or so just to make sure counts are fine, no need for transfusions, kidney/liver is okay, et cetera. But after the first couple of months, when the body is used to the drugs when the patient is settled with the treatment, frankly, once-a-month labs are good enough. We don’t really need labs on every single treatment visit. Because the other thing that happens is some of these drugs can lower the blood counts normally during treatment, but they have a rest period at the end of the cycle when the counts recover.

So, if somebody does labs in the middle of the cycle when the counts are expected to be down but not an issue, treatments are stopped, and growth factors are given. And this is done, but that is not really necessary. So, first couple of one to two cycles, maybe every other week to make sure counts are okay. Myeloma markers monthly, but after the first couple of months when things are settled, once a month should be sufficient.   

Katherine:

Okay. What questions should patients be asking about their test results?   

Dr. Ailawadhi:

Yeah. Very, very, very important. In fact, whenever I’m speaking in a patient caregiver symposium or anything, I spend a lot of time on these test results because frankly, a lot of times it sounds like jargon and the people talk about, “Oh, my ratio is going up,” or the doctor is saying, “Hey, your immunoglobulins are normal. You’re in remission.” But so, I think the patients need to understand and ask from their doctors, “What is my marker of the disease that you will be following?” And I’ll tell you that immunoglobulins, that IgG or IgA level, is nearly never the marker. It’s either M spike or light chains, generally one of those.  

So, the patients need to understand what is their marker. They also need to know what did their bone marrow show. What was the percentage and what was the FISH result or cytogenetic result? I think other than the tests, I will also add the patients need to ask their doctor a lot of these questions that you’re asking me. How frequently are the labs going to be done? Why is it important? Why was a certain treatment selected? What is the expected outcome? What are the chances that I can go into remission? How long does the intense treatment stay?  

When does it go to some kind of a maintenance? Et cetera, et cetera. Basically, you want to understand everything about the disease and its treatment. It is overwhelming. This is a lot of information. A lot of times the patients may say, “Well, I got a diagnosis. I got a treatment started. I just need to move on.”  That’s right. But once you spend all that time initially understanding your diagnosis and the treatment and the disease, it’ll make the rest of the journey much, much easier.  

Katherine:

That’s really helpful as we drill down a little further. What are the types of treatments available for people with myeloma?   

Dr. Ailawadhi:

So, myeloma has a lot of treatments available. We can classify these treatments into different classes of drugs, or we can classify the treatment as early lines or late lines of therapy. Or we can classify these treatments into cellular therapy or targeted therapy or chemotherapy. There are ways of classifying it.  What I would suggest is we should think about classes of drugs. We have something called proteasome inhibitors. That class has three drugs FDA-approved. We have something called immunomodulatory drugs. That class has three drugs also approved, but generally, we use two.   

Then, there are something called monoclonal antibodies. There are three drugs approved there as well.   

There are cellular therapies or CAR T-cell therapy. There are two of them approved. There is also a stem cell transplant that is used as a part of treatment sometimes but is different from CAR T. Then, there are other immunotherapy, something called T-cell engagers, in which also there are three drugs approved. In fact, as I’m saying to you, I’m trying to think…yeah, wow. Every class has three drugs. That’s so weird. And then, there are some other classes of drugs. There is something called exporting inhibitors. There is a drug there. All said and done, there are these different classes of drugs.  

There are some guiding principles for myeloma treatment. Generally, three to four drug combinations or regimens are better than two drugs. So, a patient should be in the initial therapy or later lines. Also, preferably be getting a three-drug combo. And I forgot to mention steroids, which are an important part of every regimen in myeloma, almost every regimen. So, three drugs or four drugs are better than two. That’s important to keep in mind. Longer durations of treatment are generally considered better. We should not tinker with the regimen’s recipe too soon. As long as the patient is tolerating for a longer duration before making any major changes like maintenance.    

Generally, maintenance in myeloma is not a response-assessed thing like, “Oh, you’ve responded in two months. We should go to maintenance.” Generally, in myeloma, maintenance transition is a time-dependent thing. Okay, you’ve had six or nine months or 12 months. We can go to maintenance, sort of a thing. So, even if somebody has responded, they may need the same treatment for a longer period of time to keep the disease quiet.  

And so, I think these are the different categories of drugs. We pick and choose from different categories to combine and make a regimen. The CAR T-cell therapy, the two CAR Ts that are approved, or the three T-cell engagers that are approved, they are all currently used as single agents. They are not combined with anything, not even with steroids. 

Katherine:

Yeah, I see. How do clinical trials fit into a treatment plan?   

Dr. Ailawadhi:

Okay, that’s an extremely important question, and you’re asking it from a person in my clinic about two-thirds of the patients who are on treatment at any given time are on clinical trials. So, I am very heavily, I shouldn’t say biased, but a proponent of clinical trials. In my opinion, clinical trials are a part and parcel of treatment for every single patient. In fact, when you look at the NCCN guidelines, which are National Comprehensive Cancer Network, which is large institutions across the country, and they make guidelines for all cancers, it is mentioned in every single setting that clinical trials should always be considered.  

So, I personally feel that whenever the patient is coming up with a treatment decision, we talked about shared decision-making in the beginning, it’s important for them to ask at every single juncture, “Do you have any clinical trials available for me? 

And if you don’t have any clinical trials available, are there any clinical trials that I should consider, even if it means going to a different place and getting an opinion?” I know logistically it’s challenging, but we should at least know our options. So, in my opinion, clinical trials should be considered at every single juncture, because that is how patients get access to either a new drug, a new treatment, or a different way of using the current drugs, which might actually improve upon their current state. So, everybody all the time should consider clinical trials.   

Katherine:

That’s great information, Dr. Ailawadhi. Thank you for that. I’d like to add that if you’re interested in learning more about emerging treatments, such as CAR T-cell therapy, PEN has a number of resources available for you, and you can find these at powerfulpatients.org/myeloma, or by scanning the QR code on your screen.   

So, the symptoms of myeloma, as well as the side effects of certain medications, can vary greatly among those being treated. How do you approach symptom management with your patients?   

Dr. Ailawadhi:

It is extremely important that we focus on the symptoms, whether it’s coming from the disease or it’s coming from the treatment. Because frankly, if a person is responding to the treatment, you want them to stay on the treatment for a longer duration of time, so the disease can stay controlled. 

If we don’t handle the symptoms from the treatment or the side effects that are happening or if the disease is causing too many symptoms, it is more likely that either we’ll start cutting down the drug too much or stopping the treatment, et cetera, and then the disease just comes back. In some cases, that is necessary, but generally we would like to modulate the treatment or address the symptoms.  

So, one important piece that we should do, or at least we try to do over here, is that every single time that we talk to the patient for any of the visits – while there is enough time spent on, “Well, these are your labs, your diseases responding markers, SPEP, and M spike, and light chain,” and all that stuff – we spend a lot of time asking about symptoms.  

It is, I understand, challenging to cover everything, but to familiarize what drugs cause what kind of symptoms, and at least making sure that we ask those from the patient. For example, IMiDs like lenalidomide (Revlimid) can cause some diarrhea, can cause fatigue, can cause sleepiness. Well, I must ask about diarrhea from all my lenalidomide patients.  

Bortezomib (Velcade) can cause neuropathy. It can give rise to shingles. I must ask my patients for every bortezomib-treated patient. “Hey, do you have any neuropathy numbness, or tingling?  

Are you taking your medication to prevent shingles, et cetera?” I’m just saying we may not be able to do a comprehensive review of every single symptom from every single patient, but whatever the target side effects are important to know every single time. We educate the patients about these side effects so that they are aware of them, and they can report these side effects. And then, if the side effects are happening, any symptoms are happening, then is it to the point that we need to stop the treatment?  

Frequently, we do take drug holidays for a few weeks just to make sure, okay, we know is it coming from the drug or the disease? And every now and then, we realize, well, the drug was not even causing the symptom, because we stopped it, and the symptom stayed. Or so then, why stop the drug? There’s no point stopping it if I can’t control the symptom. So, understanding whether it’s coming from disease or drug or something else, addressing them, making the changes appropriately to lower the dose, space them out, et cetera. All of that is done. And of course, like I said, importantly, educating the patient is so very important. I’ll add one quick thing. We focus on the drug-related effects.  

As you rightly mentioned, Katherine, the disease itself can cause a lot of symptoms. So, generally, when I see a new myeloma patient, in the first couple of visits, we’ve done all the testing, we’ve discussed the treatment, and we’ve addressed some of the basic symptoms like pain, for example. That is big in myeloma.

But then, when the patient has started treatment, generally within the first two months, the focus that our clinic has is we need to control any side effects, and we need to address any symptoms that are being left over from the disease. And that’s when we start referring patients to interventional radiology for any bone procedures or palliative care for pain control or neurology for neuropathy, whatever so that we are controlling all the symptoms.  

And that’s when we hopefully get the patient as close to their baseline as possible.   

Katherine:

I would like to talk more about self-advocacy, Dr. Ailawadhi, managing the worry associated with a diagnosis, concerns about relapse, side effects. It can lead to emotional symptoms like anxiety and fear for many. So, why is it important for patients to share any worries they’re having with their healthcare team?   

Dr. Ailawadhi:

Yes. Extremely important. See, nobody’s thinking, “Okay, I’m going to have cancer today.” Nobody’s prepared for it ever. Cancer is always a diagnosis that comes out of the blue, blindsides us, and then suddenly we have to change the rest of our life because of it.  Not only our life, our caregiver’s life, family’s life, everything changes.  

So, it is okay to admit that it is difficult. It is okay to admit that we need help. And, Katherine, I like your kind of the use of the word, self-advocacy, although I want to qualify it.  

A lot of times we say patients got to be their own advocates. But if a patient doesn’t know what to ask, they’re going to be lost. My thought is it is okay to – the first and foremost that a patient or their caregiver can do is please report your symptoms or how you’re feeling. And those symptoms could be physical, those could be psychological. 

Please report what are you feeling, what are the symptoms. On a drug, what are the side effects, et cetera, so that your healthcare team can try to address them. Don’t ever assume, “I am on chemotherapy. I should have diarrhea.” No. Don’t think, “I’m on chemotherapy. Other patients outside in the waiting room look sicker than I. I feel embarrassed to ask a question.”  

We hear this so many times. A lot of patients will say, “I feel embarrassed to ask that I’m going through this symptom, because I see sicker people outside.” Yeah, but know when I’m with you as a patient, you are it. I’m not thinking about anybody else. And I don’t want anybody else’s decision to obscure or cloud our relationship at that visit. Please report your symptoms. Please ask for help.  To me, that is good enough self-advocacy. Self-advocacy is not saying, “I should get this treatment, not that treatment.” But self-advocacy could mean, are there clinical trial options?  I know I live far away from a large center. Could I get a tele-visit with a large center? Could I get a second opinion from someone? Those are all very, very reasonable questions, and by asking those questions, a patient is advocating for themselves.  

Katherine:

As you alluded, there’s a whole healthcare team working with each patient, and there’ll be people on that team who can help support a patient’s emotional needs. So, one thing that’s on the mind of many viewers is the financial aspect of care. And you mentioned that earlier everyone’s situation is different, of course, but where can patients turn if they need resources for financial support

Dr. Ailawadhi:

Very important question. I can tell you every day when I come into my office, my nurse has a stack of documents ready for my signature. Every single day. Today, there was only one, but there could be different numbers. And these are generally from foundations from diagnosis confirmations, et cetera. Things that we are filling on and signing on behalf of our patients so that they are able to receive resources, whether it’s from a pharmaceutical manufacturer, a foundation, or society that has funding available, et cetera. I should start by saying, Katherine – and I feel embarrassed to admit this, but I should start by saying, I may not have all the answers for my patient during that visit.   

But I think the very important piece where we can start is asking the patient, “Is this causing any financial strain on you?” As I mentioned earlier, we don’t think about, “Oh, I’m going to have cancer today. Let me prepare for that.” Or “I’m going to have cancer five years down the road. Let me prepare for that.” We’re not always ready for this. It’s okay. It’s important for me to ask if there is a problem, and it’s important for the patient to admit there’s a problem or say, “Well, I’m having difficulty with copayments.” And whatever may be difficult for one may be okay for the other. So, I shouldn’t assume. So, that discussion must happen.

Generally, in our setup, what happens is if the patient brings up a concern, if I identify a concern, or if we think something may be going on, but we’re not very sure about it, we tend to bring in our social workers. The social workers are typically the ones who are able to do that discussion with the patient, talk about what are the resources available. What are the foundations that we can apply to? We have patient navigators who can do the similar things. So, the patient navigator, social worker, there are different individuals who will be able to provide much more granular information. I also strongly suggest patients to join support groups.

There are lots of resources, which I may not be aware of during our visit with a patient, but I can connect to the social worker, their patient navigators, and online support. 

Katherine:

Thank you for that. It’s great advice. As we close the program, what would you like to leave the audience with? Why are you hopeful?   

Dr. Ailawadhi:

First of all, I should admit, yes, I’m very, very hopeful for myeloma. I started my work with myeloma or my time working in this field in somewhere around the year 2000 or around that year. In the early 2000s, the average survival of a myeloma patient despite treatment was about two to three years.

Today, while national data is suggesting somewhere in the vicinity of 10ish years, give or take, all of us who are myeloma-focused physicians and have specialized centers that we work in, we have many patients who are living in excess of 10 years and pushing the envelope. In fact, my longest survivor is now maybe 34, 35 years with myeloma, and she’s not even been on treatment for a few years.  

This is what gives me hope. That it’s not only that patients are living longer, more and more patients are living with less disease burden, better quality of life, and in a lot of cases, not even on a treatment. Our myeloma world is now going from everybody should be treated forever and ever to there are many a clinical trial that are testing the hypothesis of, “Can we stop treatment? Who needs to be treated? Could we be getting closer to that elusive cure that we all are looking for in myeloma?”

So, to me, the hope is newer drugs that are better tolerated, providing better quality of life for patients. And in a lot of cases, the patients are not even on treatment. That is where we think we are making a change and making a difference. And you had started by asking me, “What is the driving force?” That to me is the driving force of why we get excited to come to work every morning, because we know that we can help someone else, and we can learn something new.

Katherine:

That’s very promising, Dr. Ailawadhi. Thanks so much for taking the time to join us today.  

Dr. Ailawadhi:

Absolutely. Thanks a lot for this opportunity.   

Katherine:

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

Care Partners | What Should You Know About the Bispecific Antibody Treatment for Myeloma? Resource Guide

Download Resource Guide

PEN-198_BACP_DownloadableGuide.F

Download Resource Guide

See More from the Care Partner Toolkit

Is Myeloma Research Examining Sequencing of CAR T and Bispecifics?

Is Myeloma Research Examining Sequencing of CAR T and Bispecifics? from Patient Empowerment Network on Vimeo.

Is the sequencing of CAR T-cell therapy and bispecific antibodies being examined in myeloma studies? Expert Dr. Ola Landgren from University of Miami Sylvester Comprehensive Cancer Center discusses what’s known about sequencing of CAR T and bispecifics and what needs further study in clinical trials.

Download GuideDescargar Guía

See More from START HERE Myeloma

Related Programs:

What Are the Latest Artificial Intelligence Advancements for Myeloma?

What Myeloma Patient Monitoring Occurs After Induction Therapy?

What Are the Benefits of Myeloma Consults and Second Opinions?


Transcript:

Lisa Hatfield:

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

Dr. Ola Landgren:

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

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

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

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

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

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

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


Share Your Feedback:

Create your own user feedback survey

What Myeloma Patient Monitoring Occurs After Induction Therapy?

What Myeloma Patient Monitoring Occurs After Induction Therapy? from Patient Empowerment Network on Vimeo.

What is involved in myeloma patient monitoring following induction therapy? Expert Dr. Ola Landgren from University of Miami Sylvester Comprehensive Cancer Center discusses some common treatment and monitoring scenarios that he carries out with his patients.

Download GuideDescargar Guía

See More from START HERE Myeloma

Related Programs:

What Are the Latest Artificial Intelligence Advancements for Myeloma?

Is Myeloma Research Examining Sequencing of CAR T and Bispecifics?

What Are the Benefits of Myeloma Consults and Second Opinions?


Transcript:

Lisa Hatfield:

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

Dr. Ola Landgren:

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

But in my mind, I think that week off is a very important week off for the patient. So I would rather do testing the third day of the treatment at the treatment unit. So if it’s week one, week two, week three, I would draw the myeloma labs that same day. And that would give the patient six more days off from injection, infusion that third week and the whole fourth week off. So I would give the patient 13 days off. Again, these are small things. These are things I’ve thought about a lot. I’ve practiced medicine for many years and I recognize that having time off like that, many patients travel, they go on vacation, they do different things. So I don’t want to just randomly put a blood test in the fourth week just because I want to check after week one, two, three, and then have the assessment.

I sort of underestimate the benefit of the therapy and then I start the next cycle, say back to back cycle two and cycle three and so forth. I would typically do blood tests once a month following these principles. I do baseline and I would do the last day of injection or infusion. For a newly diagnosed patient, you ask me, I would for baseline always do bone marrow biopsy and an aspirate. I would always do a PET-CT for every patient as my default.

Sometimes we end up doing MRI. So that could be other things that are happening, but that is what we do for the majority of our patients. After we have completed four to six cycles of treatment for patients that are candidates for consideration of transplant with chemotherapy with melphalan (Alkeran), we would usually do a biopsy after four to six cycles and we would use that to determine what’s the optimal mobilization protocol for stem cells. When we do that, we would run a MRD test.

We would run our in-house flow cytometry test that we developed when I used to work at Sloan Kettering and we have developed that here in Miami as well. We work closely with Sloan Kettering, and we have set up this assay in collaboration in the new 2.0 version. We will also send the aspirate for the clonoSEQ at Adaptive Biotech, which is the DNA-based sequencing for MRD. We would send the patient for collection of stem cells. When the patient is back, we will continue treating. So if you say we do it after four cycles, we would collect, if we do it after five or six, then we collect.

After that, we would typically resume therapy and for the majority of our patients, we actually give around eight cycles of therapy, and we have seen that you can deepen the response. You don’t increase the toxicity, but you deepen the response for the vast, vast majority of our patients. When we have used our best therapies, we have done it that way…

We have even published on this, over 70 percent of our patients are MRD-negative, and many of those patients, when they come to cycle eight, they ask, do I have to do the transplant? And that is a controversial topic. But I think there are two large randomized trials that have shown the same thing, that there is no survival benefit with transplant. But you can also say that there is, in those two trials, a progression free survival benefit, meaning that the disease would stay way longer with transplant. But many patients say, if I reach MRD-negative, both those two trials show that if you’re MRD-negative without transplant, or you’re MRD-negative with the transplant, PFS was actually the same.

And given that there is no survival, overall survival benefit, why would I subject myself to go to that? Why don’t I keep the cells in the freezer and go right to maintenance? And we will have a conversation with every patient, they would meet our transplant team, they would meet our myeloma expert team.

And the individual patient will make decisions. I think over time, more and more patients have chosen to keep the cells in the freezer. For patients that are MRD-positive, we would counsel towards transplant, but there are patients that don’t want to do that, and we are not forcing any patients to do that. We would give patient maintenance, and on some of our trials, we use the standard of care, which is lenalidomide (Revlimid) maintenance. And we are also developing new approaches where we have done daratumumab (Darzalex) added once a month with lenalidomide. We have gone one year, and we have started to do two years of that. And after that, we would stop daratumumab and just do lenalidomide maintenance.

Lastly, to answer your question fully here, we would do a PET-CT in the bone marrow after the eight cycles as a repeat, and we would offer a patient to check on maintenance on an annual basis, and this is in accord with the NCCN guidelines. So a lot of details here, but you asked me how we do testing.


Share Your Feedback:

Create your own user feedback survey

Targeting of Myeloma Disease Progression and Bispecific Antibody Advantages

Targeting of Myeloma Disease Progression and Bispecific Antibody Advantages from Patient Empowerment Network on Vimeo.

How can myeloma progression and bispecific antibodies be used for myeloma care? Expert Dr. Ola Landgren from University of Miami Sylvester Comprehensive Cancer Center discusses how disease progression and genomic features can be targeted and the role that he perceives for bispecific antibodies in myeloma care. 

Download GuideDescargar Guía

See More from START HERE Myeloma

Related Programs:

What Myeloma Patient Monitoring Occurs After Induction Therapy?

Is Myeloma Research Examining Sequencing of CAR T and Bispecifics?

What Are the Benefits of Myeloma Consults and Second Opinions?


Transcript:

Lisa Hatfield:

Dr. Landgren, what are the key biological processes driving disease progression and evolution of multiple myeloma, and how can we target these processes to prevent disease relapse and improve long-term outcomes?

Dr. Ola Landgren:

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

Plasma cells make immunoglobulins. For reasons that are not entirely clear. The translocations in myeloma that include IgH, they are partnering up with oncogenes. There is a list of oncogenes, there’s MATH, there’s three MATHs, A, B, C. There’s FGFR3, MMSET, and there’s also Cyclin D1 that are on the list. So these are the different types of structural variants that you can see with FISH probes. What people have understood less about are something called mutational signatures. And myeloma is made up by eight distinct mutational signatures that you can see in every single patient. And what that means is that you can, if you conduct whole genome sequencing and you look at all the base pairs, you can see there are certain number of combinations. C can be swapped for A and C can be swapped for G or C can be swapped for T, T can be A and T can be C and T can also be G.

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

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

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

It doesn’t matter if it’s immunotherapy, chemotherapy or small molecule therapy, there are some fundamental programs the cells can turn on. We need to understand that better and we are spending a lot of time trying to drill into this. Lastly, I also want to say there was a fourth class of genomic events called complex events that you can see in myeloma, something called chromothripsis. That’s a very severe genomic lesion, is a ripple effect through the genome. There are a lot of havoc going on. And the first time we saw that, we thought this has to be something wrong with this sample. But when we look through more and more samples, we see that about a quarter of the patients actually have this chromothripsis.

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

Lisa Hatfield:

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

Dr. Ola Landgren:

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

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

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

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

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

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


Share Your Feedback:

Create your own user feedback survey

What Are the Latest Artificial Intelligence Advancements for Myeloma?

What Are the Latest Artificial Intelligence Advancements for Myeloma? from Patient Empowerment Network on Vimeo.

What artificial intelligence advancements have emerged for myeloma? Expert Dr. Ola Landgren from University of Miami Sylvester Comprehensive Cancer Center discusses the IRMMa prediction model for myeloma care, factors that go into the IRMMaa model, and potential AI advancements for myeloma.

Download GuideDescargar Guía

See More from START HERE Myeloma

Related Programs:

Targeting of Myeloma Disease Progression and Bispecific Antibody Advantages

Is Myeloma Research Examining Sequencing of CAR T and Bispecifics?

What Are the Benefits of Myeloma Consults and Second Opinions?


Transcript:

Lisa Hatfield:

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

Dr. Ola Landgren:

Yeah. So you mentioned the study we just published. We published a model that we call IRMMa and that stands for individual risk prediction for patients with multiple myeloma. So what we were thinking was at the current time, all the existing models are pretty much providing the average patient’s predicted outcome. So think about it is like it’s a probability measure.

So you say, if I take this about therapy, what’s the predicted average outcome for patients that take this therapy, say, five years later? So on average, say 70 percent of patients are free from progression. That sounds pretty good. The problem is that you don’t know if you are in the group, 70 percent group that didn’t progress or if you’re in the 30 percent that did progress.

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

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

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

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

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

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

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

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

Lisa Hatfield:

Yeah, that’s wonderful. So if you have a newly diagnosed patient coming in to see you, do you use this model and explain to them what came back to you? Or is it right now just collecting the data for this dataset?

Dr. Ola Landgren:

So at the current time, we have to be cautious. We cannot promise things that we cannot deliver. So we have clearly said this is a research tool. It was just published less than a month ago in the Journal of Clinical Oncology. It is publicly available. The paper is available. Anyone can go there and download the paper and anyone can also in this paper find there’s a website. You can actually see the database as well. And there is a lot of corresponding material online on how to interpret. So for now, it is a research tool. But I think it’s possible in the future that we would start considering using it. And if other people find it useful, maybe they will do that, too. But for now, it is a research tool.

Lisa Hatfield:

As a patient, I would be very intrigued with that and what might come back. Just like you said, it’s a tool to maybe help identify new treatments or whatever. I did ask ChatGPT if there’s a cure for myeloma, if there will be one in the next 10 years. I didn’t really love the answer. It was a little bit vague. But yeah, I like looking into AI a little bit and ChatGPT and all that. So thank you so much for that overview.

Dr. Ola Landgren:

This is exactly like ChatGPT. It works the same way, but it’s only centered around multiple myeloma. For now, the way we have done it is that we have to start somewhere. As I told you, it’s a four-year work effort with a lot of people. We have like 10, 15 people working day and night on this project. So we started on the newly diagnosed patients. But we intend to scale it up. We intend to build in a lot of new features. And, of course, we want to add more datasets to it. And last thing I want to say to you, I find it very, very fascinating how you can, as a human being, as a researcher, you can ask the computer, and it will give you answers back that you didn’t think about yourself.

So you talk about ChatGPT. So we are using our model. We can have the model looking at biopsies. We can ask the computer, what is this biopsy? What’s going on? And the computer will say this and this and this genetic feature is going on. And then we ask the question, how did you conclude that? And then the computer will say, look here. So it would then label areas in the biopsy and say, I looked here. It doesn’t yet tell us what it found, but it tells us where it found it or where it looked to come to its conclusion.

So when it finds the right conclusion, we are looking in these areas to see what’s going on in these areas and how does it look different from other areas or in other samples. So having a dialogue with the computer can give us new insight. It’s almost like taking a young kid and you go out for a walk and you look and you see a lot of buildings and the kid looks down and looks and finds a little flower on the ground. And you say, oh, my gosh, I missed that one. The kid would not miss it. The computer is the same way.

Lisa Hatfield:

Yeah, that’s a great analogy, too. And I think we could have a 10-hour conversation about that, particularly with myeloma, because it’s so complicated, complex. So I hope we can in the future again talk about that. 


Share Your Feedback:

Create your own user feedback survey

What Are the Benefits of Myeloma Consults and Second Opinions?

What Are the Benefits of Myeloma Consults and Second Opinions? from Patient Empowerment Network on Vimeo.

What can myeloma consults and second opinions bring to patients? Expert Dr. Ola Landgren from University of Miami Sylvester Comprehensive Cancer Center discusses the benefits of consults, second opinions, and myeloma specialist centers.

Download GuideDescargar Guía

See More from START HERE Myeloma

Related Programs:

Targeting of Myeloma Disease Progression and Bispecific Antibody Advantages

Is Myeloma Research Examining Sequencing of CAR T and Bispecifics?

What Are the Latest Artificial Intelligence Advancements for Myeloma?


Transcript:

Lisa Hatfield:

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

Dr. Ola Landgren:

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

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

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


Share Your Feedback:

Create your own user feedback survey

Self-Advocacy in Myeloma Care | Advice From an Expert

Self-Advocacy in Myeloma Care | Advice From an Expert from Patient Empowerment Network on Vimeo.

What questions should myeloma patients ask their doctor about a treatment plan? Myeloma expert Dr. Omar Nadeem shares key advice and emphasizes the importance of patients engaging with their healthcare team in treatment decisions.

Dr. Omar Nadeem is the Clinical Director of the Myeloma Immune Effector Cell Therapy Program and Associate Director of the Multiple Myeloma Clinical Research Program at the Dana-Farber Cancer Institute. Learn more about Dr. Nadeem.

Download Resource Guide

See More from Evolve Myeloma

Related Resources:

Clinical Trials for Myeloma Treatment | Essential Information for Patients

Clinical Trials for Myeloma Treatment | Essential Information for Patients

What Should Myeloma Patients Ask About Developing Research

What Should Myeloma Patients Ask About Developing Research?

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Transcript:

Katherine:

What questions should patients be asking their provider about a proposed treatment plan?  

Dr. Nadeem:

Yeah. I think because myeloma therapy’s so nuanced and much of this is still in clinical trials or under investigation about what to do with some of these results, I would say, as a whole, it’s important to know which tests the physician looks at to determine how you’re doing, and kind of what their assessment of that result is. So, for example, if somebody’s had a 50 percent reduction in the amount of abnormal protein in the blood, is that sufficient, or should we be aiming for a number that’s much higher than that? 

Some of that depends on kind of where they are in their treatment course, but that’s a very sort of reasonable question to ask your physician is that where do you see my response now, let’s say six months into therapy, and is this adequate, and what is now, after we have all this information, what is my roadmap going forward to try to keep this disease in check? 

Katherine:

Yeah. Well, that’s great advice, Dr. Nadeem. Thank you. I’d like to turn to self-advocacy, Dr. Nadeem. Why is it so important that patients engage in their care treatment decisions?  

Dr. Nadeem:

Yeah. As I mentioned, myeloma therapy is so individualized now and we can sit here, look at the trial data, get very into the weeds and technical about this therapy with this approach as X or Y higher response rate.  

Or MRD-negative rate, but in reality, we’re dealing with people and we’re dealing with people that have lives.  

They have all their priorities, and until you share that with us, it’s very difficult for us to know exactly what’s important to you. So, what I may consider to be kind of the “best therapy” for you may not make sense for you because of all the priorities that you may have, and I think it’s so important to advocate for yourself and not be afraid to bring that up to your physician because I think many patients kind of hold that stuff in for a long time because they don’t want it to impact their care. But I would argue the other way around.  

Tell us. Tell us exactly what you prioritize. Tell us if you can’t be out of commission for work for X amount of time because of a stem cell transplant. We now have options. We now have options for patients because of all these amazing new therapies for myeloma and we can come up with a very individualized treatment plan for you based on your priorities.  

Katherine:

If a patient is feeling like they’re not getting the best care or they’re uncomfortable with the care they’re receiving, what steps should they take to change that?  

Dr. Nadeem:

Yeah, I think that’s very difficult because this is a complex system. Medical systems are getting even more and more complex. They’re busy. Everybody’s busy: busy offices, labs, radiology. We’re all feeling that. It doesn’t matter where you are. So, I think it’s important to raise those concerns, number one, to your practice that you’re being seen at because they would like to see that feedback, right? So, kind of see what is something that they can perhaps improve upon. I think it’s always important, like we just said, to advocate for yourself and raise some of these issues and not be afraid of that.  

We’re all in this together, right, so I think ultimately, we’re all trying to take the best care of you and we would need to know which part of that may or may not be working so well.