Main

IMF Patient & Family Webinar 2023 - Frontline Therapy

This webinar covers both the basics of diagnosis and early treatment as well as excitement around new immune therapies. The importance of expert management of infections is an area of particular focus. Dr. Rafat Abonour led a segment on Frontline Therapy - Including a focus on autologous stem cell transplant, and - A discussion on triplets vs quadruplets in treatment Download Slides: https://www.myeloma.org/videos/imf-patient-family-webinar-2023-understanding-new-myeloma-landscape _______________ Improving Lives | Finding the Cure Founded in 1990, the International Myeloma Foundation (IMF) is the first and largest organization focusing specifically on multiple myeloma. The IMF’s reach extends to more than 525,000 members in 140 countries worldwide. The IMF is dedicated to improving the quality of lives of myeloma patients while working toward prevention and a cure through our four founding principles: Research, Education, Support, and Advocacy. Subscribe to our channel: https://www.youtube.com/c/IMFMyeloma Visit our website at: https://www.myeloma.org Find us online: Facebook: @myeloma | https://facebook.com/myeloma Twitter: @IMFMyeloma | https://twitter.com/IMFmyeloma Instagram: @imfmyeloma | https://www.instagram.com/imfmyeloma LinkedIn: https://www.linkedin.com/company/international-myeloma-foundation Support the IMF | Donate Now! https://secure.myeloma.org/page/40697/donate/1 Category Nonprofits & Activism License Standard YouTube License In most cases, captions are autogenerated by YouTube.

International Myeloma Foundation

9 months ago

Dr. Brian Durie: I would very much like to welcome Dr. Abonour, who  will talk about frontline therapy, the initial therapy for myeloma patients. Very,  very important first step for myeloma patients, so very pleased that Dr. Abonour can be with  us today to present this topic. Welcome Rafat. Dr. Rafat Abonour: Thank you, Brian. Good afternoon and good evening wherever you are. Thank you so  much, Brian. This is an exciting time in multiple myeloma. We've been doing this for a while. And  we hav
en't seen such amazing drugs and amazing results in a long time, so it's a good time I  guess. And hopefully I'll show you some of the progress that we've made in upfront therapy. Okay,  so we are going to try to move the slide flicking. Dr. Brian Durie: There you go. Dr. Rafat Abonour: Here we go. I guess when you diagnosed with multiple myeloma, what are the goals of  therapy? I mean, I think two things. Obviously you are diagnosed because you have symptoms related to  the myeloma. You have bo
ne pain, you have anemia, you have kidney failure. We're so excited about  the slide here. Let's see what's going on. Let's go back to the previous slide. My clicker does not  want to cooperate. Okay, previous slide please. Previous, previous, yeah, let's go here. All  right. Symptoms control. That's really important, so we need to get you under control quickly. And  probably part of the important thing is that if you have kidney failure, high calcium, we're  going to hydrate you, we're going to
give you steroid. Believe me, steroids are very important  in the first month or two of therapy. After that, we can ditch them. And obviously you want to  control the disease, you want to make sure that you get rid of as much myeloma as you  can and you want to minimize the toxicity. And the goal is to live as long as possible and we  need to achieve that. So I think what's evolving is that the depth of response matters, how much  myeloma you get rid of is very important. In the old days, the b
est we could do is partial  response, get rid of 50% of your myeloma. And when that happens, the patient didn't live that  long. The median survival 20 years ago used to be two to three years and then 10 years ago, five  to seven years, and now it's getting better. Why? I believe because of the depth of response.  Because look at it. If you leave PR here, that's partial response on the left. What you're  seeing is that it's taking not much that long to go back to where you started. But if you ge
t rid  of it, complete remission is going to take longer. But if you start getting rid of a lot of  myeloma, stringent complete remission, your myeloma protein is gone, your free light chain  is normal, no myeloma cells in the bone marrow, it's going to take much longer to relapse. But  then what happened we start looking at something you heard from Dr. Brian Durie, minimal residual  disease detections, we start improving on that. And when we improve on that and we get rid of the  majority of mi
nimal residual disease, you can see the curve. It takes much longer to relapse And  hopefully when you reach these amazing minimal residual disease undetectable, your relapse may  not happen for 30 years and that means hopefully you are cured, so getting to that level of  response I think is really very important. Okay. So for some reason it is... Dr. Brian Durie: It's working, it's working, yeah. Dr. Rafat Abonour: It's working? Dr. Brian Durie: Yeah, there's a slight delay I think. Dr. Rafat A
bonour: Yeah, there is delay. I don't know why. Anyway, so basically what we're trying to do here is to how  we detect minimal residual disease. And there are two ways to do it, but why is it important to get  minimal residual disease? I think it's emerging as an important marker of lasting clinical benefit,  so when we look at clinical trials that have looked at minimal residual disease, patients  who achieve minimal residual disease tend to have longer remission and longer survival. And  we us
e two ways. One is a flow cytometry that the IMF really sort of brought to America from Europe.  The flowcytometry using eight colors and 10 colors flowcytometry has a decent sensitivity of one in a  hundred thousand. And in good hands it can be even one in a million. And it's really nice because  what the pathologists, when they're doing the flow, they're looking at actually abnormal plasma  cells and they can tell you if they have one cells in a million or 10 cells in a million. And that's  us
ually informative and it's easy because you don't have to have your initial bone marrow  to know that you have abnormal plasma cells. Abnormal plasma cells are there when you are  diagnosed, when you relapse, and when you still have disease. The other more sensitive assays  is the next-generation sequencing. You have to have the original myeloma samples When you are  diagnosed, we generate a sequence specific for your disease and then we follow you. And that's  after transplant, we can use these
sequences to look at these cells and find them. And the  sensitivity is usually one in a million, so one log more sensitive, so why is MRD useful?  Because it predicts the outcome. Here there are two data's showing if the patients achieve,  this is a large set of patients, if you achieve minimal residual disease negative, you are going  to enjoy a better time without relapse and you're going to enjoy a longer survival. That one on  the right is looking at overall survival. And on the one on the
left, that graph is looking at  progression-free survival, those times without relapse. And the more sensitive the assay, the  results are more actually important and reliable. Okay, so now the thing about it is that if you  get minimal residual disease, it may lead you to enjoy a very long progression-free survival,  living without the disease. And what you can see on top is that even patients with high risk  disease who achieve minimal residual disease are behaving like patients, what we call
standard risk  myeloma, so when we say you are high risk versus standard risk, risk of what? The risk is for a  short remission and short survival and you can overcome that by getting rid of the myeloma to  a level undetectable by these sensitive assay. Because if you don't have the myeloma cells  around you get rid of the high risk clone, that's a good thing because they're not going to  become more aggressive and cause relapse quickly, so that's very important things. Obviously when  you don'
t achieve that you're going to have a sort of shorter time to relapse, so we need to  work on that group of patient to try to improve their outcome. And the next slide, basically these  are data showing effect of sort of maintenance. And this is a study that was done in France  looking at patients who had received induction treatment, high-dose chemotherapy, and  autologous stem cell transplant. And if they achieve MRD negativity, they enjoy and  they had a longer time with good survival. And wh
at it means that achieving MRD negativity  can save the patient taking medication, side effect related to the medications,  and also cost, so this is great things. So most patients with just a year of maintenance  enjoy that. Again, the thing about it is that there is a great benefit of maintenance  but can be a downside for it. And that's why for example some of the work that is  being done, and I'll show you example of clinical trials that focus on achieving minimal  residual disease that is s
ustained and showed that actually that's maybe a different way of  managing patients. All right? And this is just, I'm trying to make the case that minimal residual  disease negativity is very important. And again, when you achieve a sustained minimal residual  disease, the survival is.. Look, it's flat. I mean there's nothing better than seeing a curve  that is flat, when you start seeing a drop-off when people are not doing well either relapsing  or not surviving, that's a not good thing, so M
RD negativity is so important for  improving overall survival. All right, so you saw a similar slide earlier. The evolution  of myeloma therapy is incredible and you can see when few years ago we used to say RVd is a good  thing, Revlimid/ Velcade, Kyprolis/Revlimid, things like that. Now we start using instead  of three drug regimen, we're using four drug regimens. We're seeing amazing result. A lot of  patients still getting consolidation with stem cell transplant and then they post-transplant
  setting, you're getting maintenance. It used to be Revlimid alone. Now Revlimid/Daratumumab and  you can see the number of immunotherapies. So we've been around for a long time and we  used to say okay, if we look at Revlimid, it used to be in the relapse setting now is  upfront. Revlimid and Velcade used to be in the relapse setting. Now upfront DARA used to  be just in the relapse setting is now upfront. I really think some of these drugs in the  relapse setting, in the rescue setting, becau
se we understand how well they work and  because we understand their side effect profiles, we probably going to move a lot of these drugs  that Dr. Usmani is going to talk about, earlier on in the course of the disease. And I think that  will generate better responses, more patient getting minimal residual disease undetected, and  hopefully we can cure a large number of patient. What's really important is that in the old days we  used to think about, okay, let's sequence drugs, okay we're going
to use Revlimid Dex first and  then maybe use Velcade Dex. But the most important thing is that using combination therapy.  Combination therapy, that's what made the difference in the survival of patients. Just using  one or two drugs is not enough, three drugs is a great and using four drugs is maybe even better  like adding Daratumumab to RVd, Revlimid/Velcade, it's a great thing, so I think more to come  and it's going to change quickly here, okay. All right, so you heard about the trial that
Dr. Brian Durie  actually led and presented several times now, it's basically we used to say Revlimid Dex is a  good induction treatment for patient not going to transplant. Can we use Velcade/Revlimid Dex? Is  it three drugs better than two drugs? And yeah, I mean you know you saw earlier results that he  Dr. Brian and 101 showed you and basically what you can see is that time to relapse and overall  survival was in favor of using a three-drug regimen, so that makes the case, yes, we need a  t
hree drug regimen because we need to try to get rid of all the different clones that the patient  has because not all myeloma patients have the same sort of correct, the baby, the myeloma  cells coming from the original cells, they don't all behave the same way,  so some will benefit from drug A but not drug B and vice versa, so the  combination therapy makes a difference. The next thing was, okay, can we add something  called Daratumumab, the monoclonal antibody that bind to CD38 cells to Revli
mid Dex, would it be  better than Revlimid Dex? And so this is called the MAIA trial and was done mostly in Europe  and also in North America. But these patients were not transplant eligible, they thought they  are now going to be able to get a transplant, so can we improve their survival? And basically  what it showed here is that there is significant improvement in the duration of response and the  overall response, and a lot of patients who were able to stay on therapy without having to stop 
treatment. If you look at the overall response that mean how many patient responded well to the  combination of Dara/Revlimid Dex, you can see it's 93% versus 81%. But if you look at the quality of  the response was much better when Daratumumab was added that mean you're getting more patient into  complete remission, stringent complete remission. And so the initial data looks great and the follow  up data looks even good and better. For example, if you look at complete remission and stringent 
complete remission was 51% versus 30% in the Revlimid arm, so clearly combination therapy does  make a difference, does lead to a better response, deeper response. And again translates to better  time without having to worry about relapse of your disease. You can see that the difference  was at 60 months of follow up, yes five years of follow up was 52% versus 28%, so a lot of  patients in the Revlimid arm have relapsed and a lot of patients in the Daratumumab-Revlimid  arm, the combination of t
hree drugs led to a better duration of response and it does look  like it translated to improvement in overall survival. More patients are alive when they get  the three drug regimen versus two drug regimen. And they follow them for some times and the median  time to next treatment, that mean because you can judge the regimen is that how long it took  you to get to a different line of therapy? You know progress because you're not responding  was the DARA arm was only 42 months before you needed
something. I mean not reach for the  DARA arm and for the Revlimid was 42 months, so clearly you can stay without needing a new  therapy much longer when you use a three drug regimen versus two drug regimen. All right?  That's I think an important observation, and the subsequent therapy is just based  on what's available in these countries. All right, so I think the conclusion is that  I think Daratumumab added to Revlimid Dex does improve overall survival and does improve  time without disease
and it takes much longer to needing a new therapy, all right?  Now we are going to go to see the patient who will probably need a transplant  and we already established that RVd, the regimen that Dr. Brian Durie studied is a  really good treatment compared to Revlimid Dex. Can we make it even better? Can we add Daratumumab  to it, the monoclonal antibody to it? Can we make the results better? And this is a sort of  phase two randomized study called the GRIFFIN, Dr. Usmani and his colleagues or w
ere the  leader when he was in North Carolina on this. And basically what they did is that  they added Daratumumab to RVd and compare it to a group of patient who get just RVd  and they give him induction, four cycles, consolidation with two cycles with  transplant in the middle between the induction and consolidation and the patient  get maintenance Revilmid Dex versus Revlimid. And this study's been going on for a while so  we have some mature results, so what does this mature result show? So
first of all, let me show  you the DARA arm is on the left here. The kind of purplish color and the orange-ish color is on the  right is the just Revlimid/Velcade/Dexamethasone. And what you can see is that with each step  the number of patients getting stringent complete remissions increasing and after one  year it reached 63% and at two years 66%. In the RVd arm that stringent complete remission  is lower, so this regimen adding now Daratumumab to RVd for drug regimen is inducing more  respons
es that are deep quality responses. And this is the result of minimal  residual disease testing we talked about earlier that MRD negativity is very  important and what you can see here is that getting MRD negativity increased with each step  in DARA arm from 22% to 64% in the Revilmid arm went from 80% to 30%. So twice as many patients  after two years of therapy are achieving minimal residual disease. That's unheard of, I mean so I  think it makes sense that the four drug regimen are producing
deeper response to a level that  is undetectable in the bone marrow. All right. Dr. Brian Durie: Okay. Dr. Rafat Abonour: I have to figure out my clicker here and you can see the time to MRD and activity  is faster in DARA/Revlimid arm and you can see larger and more patients achieved that  while the people who just get Revlimid Dex, they get there but not as high the percentage and  not as fast. So basically the beta duration to MRD negatively was 8.5 months in DARA RVd and took 34  months for
the Revilmid/Valcade Dex to get there, so you get there, you get MRD negativity  faster, and more patient gets that, okay? Dr. Brian Durie: All right. Dr. Rafat Abonour: I may have done something to my, and obviously  the progression-free survival based on intention to treat was also in favor of the patient who  get DARA RVd, you can see almost a flat line. The majority of the patients have sustained a sort  of lasting remission, so that's what we want. We want to get more patients getting MRD n
egative  and more patients have a sustained control of their disease. We don't want a patient start  relapsing in a year or two, the longer the better, all right? The second thing I think that  is really exciting is this approach by Dr. Luciano Costa from Alabama a very, he recruited  other academic centers and came up with this trial called the MASTER trial and I start calling  him Master Luciano because I think it's really a beautiful way of looking at how we should be  treating multiple myelo
ma. And basically what he did is that, all right we know that four drug  regimen is good and we believe that Carfilzomib is a very good PI, so can we use Daratumumab,  Carfilzomib, Lanalidomide, and Dexamethasone, a combination in a way that will use it for  induction followed by transplant and then get guided by MRD negativity to decide what we  are going to do next, okay, so what did he do? What did he do? Basically what he did is four cycles of induction  with this four drug regimen DARA, Car
filzomib, Lanalidomide, and Dexamethasone followed by  autologous stem cell transplant. He did check MRD after each sort step but didn't make a  decision what to do until after transplant. And if you got MRD assure or MRD-SURE that means  you know get tested twice you're MRD negative, we are going to just watch you. But if you still  have MRD positive cells, minimal residual disease still detected in the bone marrow, he gives  another consolidation four cycles and then decided he's going to do f
our more cycle of induction.  And if they are MRD negative, we're going to call it a day. But if you still have disease,  you're going to get Lanalidomide maintenance, so what happened? Let's see what happened. First of all, if you look at all patients by  sensitive next-generation sequencing assay, he's getting a lot of patients who achieve MRD  negativity and when he looked at patients who use several, the top is the flow, the red and  the blue is the next-generation sequencing. And what he sh
owed is that you're getting  really large numbers of patients getting MRD negativity. And if you look at patients  who have no high risk cytogenetic or one high risk cytogenetic like 17p Deletion,  the response is still really good. The patients who do not achieve such good  results are the patient who have what we call double head myeloma that mean they are two  different kind of high risk features present. And so if you look at the patients who  in term of progression-free survival, the patien
ts who have one high risk cytogenetic or  none actually looks like majority of them stayed without progression with the now follow up of  a couple years or more, but the people who have high risk still not enjoying the same duration of  disease free survival. And the overall survival looks better for the patients who have standard  risk myeloma or one high risk cytogenetic, so the majority of the patients are benefiting  from such approach that is MRD directed go or no go based on where you are
after you get your  induction in transplant, so this is an opportunity for us to say does everybody need maintenance?  Does everybody need consolidation? I think it's generating a lot of questions that ought to be  tested in a bigger trial and I think there's a lot of interest in those kind of trials and  a lot of cooperative group are testing that. I think the conclusion from the MASTER trial is  that next-generation sequence response adaptive therapy is feasible in the majority of the  patient
s and in the multicenter and 72%, 72% of the patient enrolled on this  trial achieve what we call MRD-SURE. And patient with standard high-risk  cytogenetic have similar depth of response and low risk MRD, so I think this is a good  group. If you have one high-risk cytogenetic, you're going to do good and that's an  improvement because we use this thing that these patients are going to relapse in  a year or two, so I think quadruple therapy achieving MRD negative will able to explore this  conce
pt. Where are we still struggling I think, is the ultra high risk when you have for  example, gain a point Q and 17B deletion or a P deletion or some of the translocation 4;14  and things like I mean the 6;14 and like that. I think it's a really amazing trial, so we don't  have really time to talk about all the trials that have been going on, but I'm trying to give  you a flavor of some of the other things that are going on. For example, this is going on in  Germany where they added isatuximab i
s the other anti‐CD38 antibody to the RVd regimen in newly  diagnosed patients, so what did the German show? So basically randomized them a large number of  patients to induction, stem-cell transplant, and then maintenance either with Revlimid alone  or with Revlimid and the antibody Cetuximab. And the goal is to see how many patients will  achieve minimal residual disease. And you can see that by next-generation sequence  flow, which is detecting one cells in a hundred thousand cells. What you
can see is  that when you use the four drug regimen, a monoclonal antibody with RVd, you're getting more  patients enjoying a better overall response rate. And with each step the number of patient  achieving complete remission is increasing. And you see in when you add an antibody,  there are more patients achieving very good partial response, more patients achieve stringent  complete remission, so it is again another trial showing that four drug regimen improve overall  response rate and improv
e the depth of response. And this is just a sort of a breakdown of some of  the side effect that we're seeing. And obviously when you add more drugs, you're going to see  more side effect. Some of it is related to drop in a blood count, some of the related  to infection. For example, in the four drug regimen you're going to see more, for example  infection 43% versus 34%. But I think we are aware that this is happening and we can manage  those things safely. Okay, where are we next? I really thi
nk, I mean the upfront  therapy is evolving and I didn't say, I mentioned to you trials that was testing  three drugs in transplant ineligible patients. I show you trials that testing for drugs in  transplant eligible patients where the induction is for drug regimen and followed by transplant.  And basically what I'm showing you here is that right now I think commonly used in the US for  patients who are going through transplant, at least a four drug regimen, you add a  monoclonal antibody again
st CD38 to VRD or KRD and we have the thing is feasible with  limited toxicity, you're getting deeper response and you're getting more patients achieving  minimal residual disease, negative disease. And I think MRD is becoming an important  measures that we probably start to include it in our assessment of how well our patient  is doing and use it maybe to escalate, deescalate the therapy and guide us where to go  with high risk disease that's still a challenge, but it seems like maybe not your
typical high  risk, the ultra high risk continue to be a group of patients that need more attentions, more  clinical trials to try to answer the question. And I didn't spend a lot of time about transplant.  Is it important? I think if you look at all of the study that's showing much deeper response, more  MRD negativity, better overall survival has really included transplant high-dose melphalan  still play a role until we have better trials in the era of better induction treatments and  better m
onitoring, it still has a room for it, so we still recommend it to our patients who are  eligible. The question is that will we replace it one day with something else? We don't know.  We don't have the data yet, so we still include it. I think that may be my last slide. Let  me see. Okay, so let me just go to one more sort of futuristic trial that is called  the MASTER-2 and it just addresses what I was saying earlier here in the last slide is  that can we now say okay, what he did is that he us
ed consolidation when he doesn't get MRD  negative after the induction and transplant. Well you're going to hear from Dr. Usmani about  the bispecific and the CAR T-cells and he's going to tell you how we are able now to manage a side  effect. How can we control cytokine release and neurotoxicity? They would becoming familiar with  these side effect. We can manage them and we can probably bring those therapy early on. Why should  we wait until the patient has a horrible myeloma that we cannot co
ntrol? If we have a high risk  patient's double head myeloma, we are not getting m MRD negativity, we can consider some of these  new therapy because you're going to hear about the mechanisms of action. They're totally different  than the drug we use upfront. If you're going to keep using the same image, the same monoclonal  antibody, you're just not changing anything, you're not changing the biology. But when you  use the T-cell specific by specific engagement, for example, what you're doing he
re is that you  are making the immune cells work in your favor. You're trying to get your myeloma cells to be  recognized by the T-cells. You're making the T-cells angry, get rid of the myeloma cells  and hopefully get rid of all the ugly cells that are left behind and the patient then will  achieve a sustained MRD negativity and perhaps cured, why not? All right, I think I am here  toward the end, I have to figure out why. Okay, so you better watch it myeloma, so we are going to  take care of y
ou. And so there are three myeloma patients here and one guy trying to pretend  that he's a cyclist, but we're going to take care of myeloma. I think this is a really exciting  time. Some of the drug you hear from Dr. Usmani, I really think they should be used early on  and we start thinking about myeloma as history, not as a continuum that we have to deal  with all the time. With that, I'm done. Dr. Brian Durie: All right, so thank you so much Rafat, and a number of questions have been coming i
n  and most of them you've touched on and made it clear that they are actually still questions.  One question is about high risk. Were high risk patients included in the GRIFFIN study? And then  obviously the MASTER trial really evaluated high risk with the finding that the ultra-high  risk really still need extra attention, so maybe you and Asad might want to  comment on the current status of high risk. I think earlier you mentioned Asad  that there were seven trials for high risk going on. Som
ebody wanted to know,  well, where are those seven trials? Dr. Saad Usmani: I'd mentioned that there have been seven trials that have been reported and there  are others that are actively being planned. And we will have three frontline high-risk studies open  in the United States by early part of next year. One is going to be a cellular therapy based  concept or clinical trial where we are going to use CAR-Ts in early alliance. There's another  one which will incorporate bispecific antibodies. A
nd then there's a third a consortium  platform study which will be exploring different questions, whether we need  to change something in induction, whether we need to change something in  consolidation, so I think those are coming, so there's a lot of hope for high risk patients,  but yeah, I mean some of the newer therapies that I'll talk about for relapse patients, we are going  to be moving them up for high risk patients soon. Dr. Brian Durie: Yeah, maybe you can just touch on that as you go
through a variety  of those new therapies in a moment. Yeah. Rafat, any other comments? I mean the two  comments really are the role for autologous stem cell transplant and basically you've said  that we're still doing that for the time being because it prolongs remission. There was a  question actually which was indicating that maybe the high-dose melphalan increases  genomic damage, which is something that one of our colleagues, Dr. Richardson frequently  talks about. I don't know if there ar
e real pros and cons to doing the stem cell transplant,  but you are still recommending it, right? Dr. Rafat Abonour: Yeah, I mean I was still recommended it. I mean obviously some of the data on the genomic  instability is probably related to having cells to become genomically unstable. I mean, getting rid  of it to a minimal residual disease undetected, you're not going to worry about genomic  instability, so if you look at all the trials that really had a high level of MRD negativity,  includ
ed induction with four drug regimen followed by transplant and maybe some consolidation or  maintenance, and that's when you're not going to see that, so I really think it's not totally  wrong to say that there's genomic instability. What I really think is wrong is to  think about myeloma is a disease, that we are going to be able to deal with  continuous therapy that also will generate genomic instability. Having myeloma cells around,  they're not going to be behaving. I mean, I always say that
when you live in a bad neighborhood,  you're going to become bad, so you know want to get rid of these cells, so that's why  I sort of like the idea that if it's feasible with a limited duration of treatment to get MRD  negatively, why not? What do you think, Asad? Dr. Saad Usmani: Yeah, no, I agree with you. I think there are two important things and when we listen  to other experts, it's very important. People say things in passing, so I want to be very specific  about the data. What we comme
nt on is this observation that actually came from the Memorial  Sloan Kettering data set, actually. It was a paper that was published by us in collaboration with  the Arkansas group, some European colleagues and with Ola and Francesca Mora in Miami now. And the  observation is that high-dose melphalan increases the gene mutations in myeloma cells, but there  is no data to suggest that that's a bad thing. Why? Because the PFS is always better  on the transplant arms. And the issue is that transpl
ant has set such a high bar of  progressions free survival. It's going to be very, it's a high bar and we'll have to beat it with  frontline studies, which we do not have now, so until then we have to follow the data because  if anything, one can actually argue the other case that if you have more genomic instability,  you actually create more new antigens that the immune system can take those cancer cells  out better, so I think that there are a few different ways in which we can look at that 
data, but I support what Dr. Abonour was saying. Dr. Brian Durie: All right. Great. Great. I'm going to cut you on, there's a couple of questions. Why  is MRD status not standardized? Well, I think that the definition of MRD negative is standardized in  that we have 10 to the minus five as the cutoff. You're not supposed to be MRD negative unless  you're at least 10 to the minus five. But what I would say is the reporting of it is all across the  board, 10 to the minus four, 10 to the minus five
, 10th of the minus six. And so 10 to the minus six  is obviously better in terms of outcome versus 10 to the minus five. But really we should not be  reporting MRD negative unless one has achieved at least 10 to the minus five, either by NGF  or by NGS. Then a question about the timing. I think that the best timing for doing MRD testing  is still a little bit up in the air. However, one question and some of the data you saw is  that with ongoing therapy response deepens and so about 10 to 15% o
f patients will achieve  their best level of MRD negative after or during the maintenance period. And so actually overall,  one of the best times to do an MRD test is after about 12 months of treatment where the maximum  number of patients will have had an opportunity to achieve MRD negative, if they're going to do  that. In a relapse setting, it is probably a good idea to test the MRD a little bit sooner  more in the six to nine month timeframe, so that maybe covers some of the things. What is
PFS? Yeah, some of these basic questions.  What is PFS? Progression-free survival. This is basically the time that a patient stays in  remission without the myeloma progressing, progression-free survival. How do we know  for high risk? That is based on either the fish testing, which shows those problems  with chromosome 17, chromosome number 1, 414, things like that. Or if relapse has  occurred in that one year timeframe or if some other things have happened like  plasma cell leukemia with myelo
ma in the blood or myeloma in soft tissue areas,  what we call extramedullary myeloma.

Comments