Site icon OncologyTube

How effective is mosunetuzumab in treating lymphoma? – [Slides] Elizabeth Budde, MD 2023 reviews new therapies

How effective is mosunetuzumab in treating lymphoma? – Elizabeth Budde, MD 2023 reviews new therapies

By Elizabeth Budde, Md, PhD

Thank you so much for the invitation. This is actually my first time joining the meeting and it’s been very exciting. I’m learning a lot about medical oncology and all the exciting new drugs. Now, some of the naturally, pretty similarly are also using the in the domain.. How do I get started? Oh, great. Yeah, so I was assigned the task to talk about novel regimens and agents in lymphoma. So it’s a switch from medical oncology, but I hope you will share the excitement with me too. And here are my disclosures. 

 

Advertisement

This is a summary of what has been approved for a patient with lymphoma in the last 5 years now beginning from 2017. And and I’m really love running out of space here because more and more new drugs, new treatment, helping, getting approved recently, so you can see back in 2017, Axi-cel, which is CD19 CAR T-cell received his first approval for treating patient with no diffuse large B-cell lymphoma, and also as the first CAR T for lymphoma. And subsequently there are more CAR T being approved for different indications. And BDK inhibitor played a more and more important role in treating lymphoma patients, PI3K kinase inhibitor such as now Copanlisib, which was approved in 2017. And prior to that there was Aldolicib, but unfortunately because of the risk and benefits, the known ratio didn’t really pan out. So some of the PI3K kinase inhibitors have not been removed from the market. But other than that, we have other exciting agents such as bispecific antibodies. 

 

So just in the last year, from 2022 to 2023, there are also multiple approvals. We have at least three CAR T-cell therapies receiving more indications, and we have the first bispecific antibody ever approved for lymphoma. Now, which I will talk about in my next few slides. In 2023, 2 PI3K kinase inhibitors. Already got approval in January and there are a few more regimens now or we are waiting for FDAs no evaluation. So these are the approved novel agents, non-chemo based agents known for T-Cell lymphoma and hodgkin lymphoma. Most of them due to the maximum actions, most of the check checkpoint inhibitors and CD30 directed ADCs, as well as, Mogamutuzumab which is a CCR-4 monoclonal antibody for PTCL lymphoma. . So immunotherapy as we know, especially in the malignancies, really is taking the front C in the last year and now there are 2 CAR T-cells approved for second line use in diffuse large B-cell lymphoma or other type of aggressive B-cell lymphoma. And 1 CAR T-cell approved for a third line use for patients with follicular lymphoma and the most exciting. If you ask me what’s the most exciting approval in the last year, it has to be the bispecific Mogamutuzumab as the first bispecific antibody was approved by the FDA at the end of last year. And there are 2 other bispecific, which already have mature results and there are more to come. So here is a list of CAR T-cells that’s available for aggressive large B-cell lymphoma. So 3 of them approved for the third line use out of the targets. Now, CD19 actually shared this same binder. There are other nuances contributing to the differences in efficacy and in the toxicity profile. But in 2022, Liso-cel and Axi-cell both received FDA approval as a second line use. And this has actually led to a practice change treatment landscape for patients with aggressive B-cell lymphoma and the approval already based on the 2 out of 3 positive phase 3 clinical trials. So patient populations are very similar in all these 3 studies now for ZUMA, TRANSFORM to BELINDA. All patients in these studies are really patients who either have primary refractory disease to frontline treatment, typically R-CHOP  or E-CHOP based treatment or patients who had initial response but relapse within the 12 months from the first treatment. So these refractory patients are being enrolled into the study and the recommendation is now to ask the question whether CAR T-cell treatment can out-full the standard care for many years. That’s the salvage chemotherapy followed by autologous stem cell transplant.

 

So here’s the result for using Axi-cel. This is DOMA 7, so patient are randomized upfront, so either to Axi-Cel, then those patients will only receive the high dose steroid if they need as the bridging therapy or and they will undergo looking for races with certain intent to make a CAR T product, or they will be randomized to receive chemotherapy followed by standard care, autonomous stem cell transplant if they have a CR or PR. So the majority of the patient was actually able to move on to CAR T-cell treatment, but only a third patient randomized to the standard cure, were able to make it to transplant and these actually translate to a positive result measured by a primary event free survival. So this is at the 2 year follow up patient randomized to the CAR T-cell, had an event-free survival of no 40.5% at 2 years. And the patient randomized to a standard curve on the event-free survival was 16.3%. And these benefits actually derive, from all comers? The age doesn’t really matter what, how many lines are they primary factory or relapse disease, that doesn’t really matter. And then adjust the IPI score now, which linked historically predicts a poor prognosis. That also doesn’t matter. So basically the risk factors for their response to CAR T-cells is very different from the risk factor for their response to chemo. So all different subgroups here, all derive benefit from getting CAR T-cell treatment.

 

And this is the overall survival curve. Now it appears now there might be a survival benefit now, based on this report to this report to year follow up. And just as last week Kite Pharma made an announcement, then they actually see an overall survival benefit. And so it’s interesting, these days we learn more about the result of the trial, not from academic publication, but from the news media. 

 

The second trial TRANSFORM study, which random, which is a little bit different than how DOMA 7 was d1. So the TRANSFORM study actually has a crossover arm. So what they did is the patient enrolled to the study, all patients will have leukapheresis, have a CAR T product made and then they randomized the patient to either CAR T treatment or standard care salvage chemo, followed by a target stem cell transplant. And the patient, while we’re waiting for CAR T-cell treatment, they’re also allowed to to have bridging therapy. And despite the crossover, we also see an positive result using event-free survival as the primary endpoint can see with 6 months no follower event-free survival for patient receiving  Liso-cel CAR T at 10 months and standard care is somewhere 2 months, which is very similar to what from the the DOMA 7 study. Most recently, the 17.5 months follow up result also came out and the median event-free survival continued to favor the CAR T arm and the overall survival here that the data is number true yet. But also, keep in mind all those patient in the standard care arm, they didn’t respond to salvage chemo. They can have the CAR T-cells that was pre-made for them. So this crossover my compound, the overall survival outcomes. 

 

So the 2 out of 3 studies are positive, which left the BELINDA Study, which uses Tisagenlecleucel, a third CD19 Car T-cells. Now for patients in a similar patient population, but BELINDA is a negative trial, phase 3 negative trial. The same patient population, the same primary endpoint. This actually really speaks to how to best design the clinical trial and how to execute a clinical trial. So in BELINDA study, if you look at which I highlighted here, what’s particular for CAR T trial, there is the time from registration to the time of the  CAR T delivery is super important because the longer you wait, the more likelihood the patient will drop off or patient will develop an event. So for the ZUMA-7, the time to CART is 29 days, and TRANSFORM the 34 days and BELINDA is now 52 days. So that actually contributed to some of the results that the BELINDA study didn’t really match with the other 2 phase 3 clinical trials and also how many salvage chemo was given in between. Now, that’s critically important so Zuma-7 and TRANSFORM results really led to the FDA approval of these 2 CAR T products for patients with aggressive large B-cell lymphoma that relapse or refractory within 12 months of additional treatment. Liso-cel was also approved, accelerated approved now for patients who, transplant eligible based on the single arm study that’s called a PILOT study. So this is for patients who are transplant eligible and the transplant eligibility as a transplant, they look at the list, some of them are not really the strict criteria to say a patient’s now transplant candidate, but for the trial sake know any1 who’s older than 70 is deemed transplant ineligible. But that’s not really in reality, that’s not the case. But for this patient population, given the Liso-cel as a second line treatment, led to overall response rate, 80% CR a 54%. So this study also managed the endpoint and received accelerated approval as a second line use for patients with transplant in eligible status based on defined here. So based on all these studies now, our standard care for patients with aggressive B-cell lymphoma in the second line setting has drastically changed. Now we’re seeing more and more patients being referred for second line CAR T-cell use. 

 

I’m going from CAR T-cells to bispecific antibodies. No, I think it’s really exciting that we see immunotherapy is really making a bigger dent in how we manage patients with lymphoma. bispecific antibody as seen most of basically is a pretty basic concept that has 2 binding size, a more precise term for these kinds of bispecific antibodies. They are bispecific, T-cell engager  because 1 arm binds to a target under lymphoma cell or other cancer cells, and the other target is CD3 where they recruit the T-cell and T-cell activation is specifically dependent on the target. You have the target engagement, then the T-cell will be conditionally activated and can execute, is known activity. So there are different forms of specific antibodies. Blinatumomab was actually the first 1 that was tested in lymphoma and subsequently that there’s a newer full bispecific antibody being developed by AstraZeneca. Most of the bispecific teams are engaging nowadays,  in the lymphoma field. It’s really testing, targeting CD20. This is a really incomplete diagram, there are other bispecific CD22. 

 

So Mosunetuzumab, which is the one I’m going to focus on today is really the first bispecific CD20, CD3 T-cell engager that really had the first mature result from a pivotal phase II study, which is a IgG1 based bispecific antibody has a full length FC, which converts to the longer, longer half live, allows the either weekly or every 3 week dosing and there’s a mutation introduced into the FC portion. So the FC receptor binding is not minimized or abrogated to prevent the recruitment of other affected shells to destroy the T-cells. And based on phase 1 study with 270 patients, the AP2D for Mosunetuzumab was finally determined, that’s after several years of work. So when you give the bispecific antibody Mosunetuzumab in dose, those step up dose progression, 1 milligram, 2 milligram, and 60 milligram in the first cycles, that really brings down, the incidence of second cytokine release syndrome that allows us to escalate up the Mosunetuzumab dose later now since it’s patient specific. Now, this also allows you theoretically to have more response now because different patient might have different numbers of T-cells, the different need of the amount bispecific. So this dose escalation allows me to all the way escalate up to 60, and then the stay at 60 will cycle 2 and then go down to 30, 1 dose every 3 weeks now for 8 cycles in those patient has CR and 17 cycles for those patient had less than CR, but clinically benefiting. And what’s also more important about this one is now, initially the drug was testing, inpatient setting due to the concern of toxicities. But subsequently we figure out a way how to manage this patient outpatient setting. So this pivotal trial hospitalization is not required, and this is also fixed duration treatment. Here is the waterfall plot, you can see now 80% patient responded to the treatment and 60% of the complete incomplete remission. The median time to first response is is really 1 and a half months. The median time to CR is 3 months, and the original response, this was just recently updated at ASH 2022, median duration response, still not reached and median progression-free survival is around 24 months. And patient to their see hope now 5 or 6 years ago, they continue to remain in remission. And all patients enrolled in the study actually all received the treatment because this is off the shelf, there’s no waiting time for the product to be made. We also look at subgroup analysis to see whether there’s any biomarker that is linked to an inferior response or better response. And we actually couldn’t really identify any such association. So some of the conventional biomarkers don’t really correlate well with patients receiving bispecific antibodies. And here we look at patients who had TP53 mutation but just not granted, the number size they put is small. Only 10 patients in this study. So whether they have TPI3K mutation or not, didn’t really translate into a worse or better outcome. And the same with the patient with other mutations. So here I put together a comparison of Mosunetuzumab to CAR T-cells that are approved now for a similar patient population. But just know, because these are all single arm pivotal phase II trials. When you compare the results, you really have to know patient selection differences in trial design. And definition of certain terms. But you look at Mosunetuzumab, the oldest patient truly on the trial, 90 years old, and currently, the oldest patient I’m aware of receiving, the Mosunetuzumab is 100 years old. So it is very well tolerated and patient received Axi-cel and Tisa-cel they, they’re a little bit younger, but if you look at the prior target stem cell transplant exposure and whether they are part 24, so part 24 is a term we use for patient with no follicle lymphoma, meaning that they relapse within 24 months from the first treatment. This patient usually has a worse prognosis. So affecting this, all three treatments lead to a pretty high over response and complete response rate and progression-free survival are also quite encouraging for all three of them. Then you look at the AE profile, second cytokine release syndrome on comers on Mosunetuzumab has around 44%, which is better than Axi-cel anti TSO cell. But these are just no numerical comparisons so you can’t really build a statistical significance here. Then we look at these neurological toxicities, I think that’s where Mosunetuzumab really  stands out. Mosunetuzumab has very few ICANS or neurological toxicities in comparison to Axi-Cel and Tisa-cel. In the follicular lymphoma setting it looks like Tisa-cel also has a better neurologic profile, especially with regard to grade 3 and above neurologic toxicities compared to Axi-cel infection rate is pretty similar across the board. So all three are good options for patients with follicular lymphoma in a third line plasma setting. 

 

This led to the approval of Mosunetuzumab at the end of last year. What I’m showing here is, no, this is really the development of Mosunetuzumab back in 2015. That’s when City of Hope had joined the clinical trial. We have made many contributions to this journey. We contributed for the first dose toxicity and we contributed the first responder and we partnered with the sponsor to really get a better understanding of the molecule and how to give the molecule and how to manage additional set effects. So it’s very gratifying to see that the drug was approved at the end of last. Now Mosunetuzumab is added to the NCCN guideline for third line plus renal refractory follicular lymphoma use.  

 

Moving on to two other very exciting bispecific antibodies, also targeting CD20 and CD3. Now these two are being tested in patients with relapsed/refractory diffuse B-cell lymphoma. This is in the third line low setting. The first one is Glofitamab now which is a little bit different from other IgG based on specific antibodies. Glofitamab has two bending sides to CD20 and one bending side to CD3. So the target to effective ratio is 2 to 1. This and also the affinity for the CD20 binding also I guess now contribute to its potency. Glofitamab also uses a similar step out dosing schedule, that was the goal to BTK second cytokine release syndrome, and you also incorporated a dose of Obinutuzumab, which was given seven days. Now prior to the first dose of Obinutuzumab. Now the goal is to further reduce the second cytokine release syndromes, no incidence, make it easier for patients to tolerate. And it’s fixed duration, so it’s up to 12 cycles in total, so there’s an end date of the treatment. So patients enrolled under these studies though, including various types of aggressive B-cell lymphoma. The complete response rate is 39.4%, and you look at the AEs now second cytokine release syndrome runs a little bit over 60%, but most of the low grade and as expected, the other treatment emergent toxicities in general is pretty safe and based on this result, now Glofitamab is now going through the regulatory pathway with a pedufiday in July on July 1st. 

 

And Epcoritamab, the EPCORE NHL-1 trial testing Epcoritamab, which is also a CD20 bispecific antibody. And unlike Mosunetuzumab, Glofitamab, Epcoritamab is given subcutaneously, which is another way to mitigate second cytokine release syndromes, another way to increase the AUC. This is now based on this phase II, single arm pivotal trial, similar patient population to Glofitamab. Epcoritamab, just as the other bispecific is the giving in those the step up dose infection as well to further mitigate second cytokine release syndrome incidents. And you look at the progression free survival, the patient with median progression-free survival for CR, definitely outperform, with PR overall the CR rate is 39%, so pretty identical to a patient who received the undergo the Glofitamab trial and the second cytokine release syndrome, it’s not almost 50%, so it’s now 49.6% with a few percent of your patient had a grade 3 and above, secondary cytokine release syndromes and like other bispecific antibodies, the neurological toxicity is quite low, which is in short comparison to the incidence we seeing with the CAR T-cell patients. So based on these two studies known as Glofitamab and Epcoritamab, now we’re really anxiously waiting for the FDA’s determination to allow them to incorporate them into our guidelines and to make them available for patients. Epcoritamab is on May 21st of 2023. So they are coming. 

 

Moving up the the treatment line, frontline treatment for diffuse large B-cell lymphoma for many years now, R-CHOP is the golden standard. We have compared R-CHOP with the square CHOP we are adding, a lot of other biologic to R-CHOP baseline as the backbone and none of them had panels to all compete with R-CHOP. Polaroids is really the first study in the randomized study to look at using R-CHOP versus incorporating POLARIX, which is a CD79b ADC, it has MMA as a pay, so it’s an antibody to conjugate. So replacing increasing R-CHOP with Pertuzumab, so it becomes R-CHOP Pola. So patients now with newly diagnosed aggressive B-cell lymphoma are randomized to either Pola-R-CHP or R-CHOP as the standard comparison. And when you look at the outcome we don’t really see any differences in the complete remission rate or with regard to toxicities very similar. Overall survival is also very similar, but when you look at the progression-free survival with a median follow 28 month, the progression-free survival shows there’s a 6.5 absolute improvement. Then the results from patients who were randomized to the standard care R-CHOP. And based on this result, this R-CHOP Pola actually has been approved in Europe and in many other countries and just now A little over 2 weeks ago, March 9th, the FDA had a discussion whether their view is using polar trip as a first line, frontline treatment. So the outcome vote is 11 to 2. So we’re waiting for the FDA’s final determination, whether this will be built into our first line treatment.

 

Another exciting regimen in the last year from 2022 to 2023, is really the use of BTK inhibitors. Now, several of the trials that have already matured now for CLL Zanubrutinib, now in two separate trials now comparing Zanubrutinib in untreated frontline using CRL and also phase 3 ALPINE trial, comparing Zanubrutinib with the Ibrutinib, which was approved, now the first B inhibitor approved for c l. So both of these trials met is the endpoint and Zanubrutinib received the FDA approval early this year in January 13th, 2023 to be indicated for the frontend use as well as inpatient with relapse refractory CRL. 

 

Acalabrutinib in the phase II single arm setting also had a positive phase II results for relapse refractory mantle cell lymphoma. With a very similar overall response rate to other BTK inhibitors. So this now made the NCCN guideline per Ibrutinib for relapsed refractory mantle cell lymphoma was just approved the end of January this year for patient with relapse ractor mantle cell lymphoma who had at least 2 problems of therapy, including a prior BTK inhibitor. So Pirtobrutinib was able to salvage some of the patients who were on BTK inhibitors such as Acalabrutinib, or Zanubrutinib, so it was able to really salvage those patients that made them become BTK sensitive again. And the two phase 3 studies that completed and reported the results for patients with mantle cell lymphoma, which is not a very common lymphoma and historically has very poor outcome. So these two phase 3 studies, one is the SHINE study and the other one is the TRIANGLE study. The preset primary endpoint, in the TRIANGLE study was presented at the primary session EDGE 2022, which has caused a lot of heated discussion whether we should adopt the TRIANGLE study result into our practice or not. TRIANGLE study the phase 3 study is a 10 year work, so basically these patients are 65 and older, dually diagnosed mantle cell lymphoma, they’re randomized to either BR, which is a commonly used regimen and a BR for 6 cycles, followed by Rituximab maintenance. There’s a one form of standard care, or they were randomized to patients with no similar treatment, but with building Ibrutinib, Ibrutinib will continue until unacceptable toxicity or disease progression. So a total of 523 patients were randomized in this study. And when we look at the results, this study was a negative study when they reported intro analysis 5 year follow up, but a 10 year follow up. Finally the progression-free survival becomes positive and you meet the endpoint. But if you look at the complete response rate, it is very similar in both groups. Looking at the overall survival is also similar in these two groups. If you look at the advocacy event, In those patients who are randomized through the investigational treatment who received no Ibrutinib some of the treatment emerged side effects really related to the use of Ibrutinib, such as increased incidence of bleeding and atrial fibrillation, which more than double of the control arm. But otherwise you look at the progression free survival, there is a progression free survival benefit. And this is the trial also, we have, we don’t know actually whether this will be approved or not, but it’s a positive phase 3 study, used you could argue to use it building into the practice. And the TRIANGLE study, which is a US European based neuro phase 3 study actually, unlike the SHINE study for patient who are older, A TRIANGLE study actually for patient who are younger than 65,  younger than 65, b-cell lymphoma, were usually building high dose noce rabine into the frontline treatment. So this study is really trying to challenge standard care. For these patients we typically do high dose Cytarabine based regimen. Within a year, we took it into transplant and they all received Rituximab maintenance. In these studies, one of the arms is just high dose Cytarabine based regimen high DHAP. But this patient will get Ibrutinib and will not go for a transplant. So there are 3 arms, one is the standard, one is building Ibrutinib with transplant. The other one is just Ibrutinib without transplant, and all of them subsequently received on Rituximab maintenance, which was not part of the initial design, and it was added on later. And then you look at the outcome they use failure free survival as the endpoint. It did meet the failure free survival endpoint. When you compare the group that received the standard care plus the Ibrutinib, or just the chemo plus Ibrutinib. So it appears now incorporation of Ibrutinib is no more important than whether the patient goes to transplant or not. This is really very controversial and brings out the question, do we really need to do transplant consolidation in patient, achieve a response after frontline treatment. 

 

I see my time is so I’m just gonna conclude now, there are many exciting regimens agents have really led to a recent practice change in how we manage patients with follicular lymphoma. And now we have the first line Pola-RCHP, and we have second line CAR T-cells treatment. And soon we’ll have third line bispecific antibodies for patients with renal refractory aggressive B-cell lymphoma. And we already have CAR T-cells for other lymphomas already, and the bispecific antibodies, now we know we’re already building into standard care for patients with follicular lymphoma. So now the question is now how do we sequence? Do we give them CAR T first as a bridge to bispecific or give them bispecific use CAR T as a salvage? So a lot of studies are being d1 to really provide some evidence to guide our clinical practice. There are more randomized studies underway and hopefully we’ll get to know these results soon. Then there’s also a trend to incorporate, especially in lymphomas, incorporate the ctDNA and next generation sequencing to really further lose stratify patients and build into clinical trials. Since our goal is really to practice more personalized medicine and more precision medicine. And last, but not least, a clinical trial is the best way to provide access to tomorrow’s medicine. And I really applaud these organizers’ effort trying to build a Southern California’s  clinical trial list that made that available to all of us so we can take care of our patients together. When you make referrals, and identify a trial for a patient, it doesn’t matter where they are. Thank you for your attention.

10 Key Takeaways about Mosunetuzumab

  1. Mosunetuzumab is a new therapy that has shown promising results in treating lymphoma.
  2. Mosunetuzumab is an antibody that targets CD20 and CD3, two proteins commonly found on lymphoma cells and T cells, respectively.
  3. Mosunetuzumab uses a process called bispecific antibody bridging to bring lymphoma cells and T cells together, leading to the destruction of the cancer cells.
  4. In clinical trials, mosunetuzumab has shown a high response rate and durable responses in patients with relapsed or refractory lymphoma.
  5. The response rate to mosunetuzumab varies depending on the subtype of lymphoma, with the highest response rates seen in patients with diffuse large B-cell lymphoma (DLBCL).
  6. Mosunetuzumab has also shown efficacy in treating follicular lymphoma, mantle cell lymphoma, and chronic lymphocytic leukemia.
  7. The most common side effects of mosunetuzumab are cytokine release syndrome and neurotoxicity, but these are usually manageable with supportive care and medication.
  8. Mosunetuzumab has been granted breakthrough therapy designation by the FDA for the treatment of relapsed or refractory DLBCL.
  9. Mosunetuzumab is being studied in combination with other therapies, such as lenalidomide and venetoclax, to further improve its efficacy.
  10. The results of ongoing clinical trials will provide further information on the safety and efficacy of mosunetuzumab and its potential role in the treatment of lymphoma.

 

Lymphomas

Lymphoma is a type of cancer that affects the lymphatic system, a network of vessels and tissues that help fight infections. There are two main types of lymphoma: Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL). HL is less common and is characterized by the presence of abnormal cells called Reed-Sternberg cells. NHL, on the other hand, is more common and is further classified into two main categories: B-cell lymphoma and T-cell lymphoma, depending on the type of white blood cell that is affected. B-cell lymphomas are the most common type of NHL and can be further classified into several subtypes, including diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, and Burkitt lymphoma. T-cell lymphomas are less common and can also be classified into several subtypes, including peripheral T-cell lymphoma, anaplastic large cell lymphoma, and angioimmunoblastic T-cell lymphoma. The subtype of lymphoma is important because it can affect the treatment options and the prognosis for the patient.

 

Elizabeth Budde, MD, PhD – About The Author, Credentials, and Affiliations

Elizabeth Budde, MD, PhD, is a highly accomplished physician-scientist who currently serves as an assistant professor in the department of hematology and hematopoietic cell transplantation at the City of Hope National Medical Center in California. Dr. Budde is a leading expert in the field of hematologic malignancies, with a focus on lymphomas and leukemias.

Dr. Budde received her undergraduate degree in chemistry from Williams College in Massachusetts and went on to earn her MD and PhD degrees from the University of California, San Francisco (UCSF). During the time she was getting her PhD, she did research in the lab of Dr. Frank McCormick on how oncogenes turn on in cancer cells.

Following her medical training, Dr. Budde completed her residency in internal medicine at UCSF, and then went on to complete a fellowship in hematology and oncology at Stanford University. During her fellowship, she did research in Dr. Ranjana Advani’s lab, where she worked on developing new targeted treatments for lymphoma.

Since joining the faculty at the City of Hope in 2016, Dr. Budde has made significant contributions to the field of hematologic malignancies. Her research focuses on understanding the molecular mechanisms that drive lymphoma and leukemia development and developing targeted therapies to treat these diseases. She has published numerous scientific papers in top-tier journals and presented her research at national and international conferences.

In addition to her work in research, Dr. Budde is a dedicated clinician who gives patients with hematologic malignancies the best care possible. She is committed to developing personalized treatment plans for each of her patients and is highly regarded for her compassionate and patient-centered approach.

Overall, Dr. Budde is a rising star in the field of hematology and oncology, and her contributions to the understanding and treatment of hematologic malignancies are sure to have a lasting impact on the field.

 

 

 

Exit mobile version